1
|
Rana R, Mukherjee R, Mehan S, Khan Z, Das Gupta G, Narula AS. Molecular mechanisms of neuroprotection: The interplay of Klotho, SIRT-1, Nrf2, and HO-1 in neurological health. Behav Brain Res 2025; 485:115545. [PMID: 40120944 DOI: 10.1016/j.bbr.2025.115545] [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: 01/12/2025] [Revised: 03/06/2025] [Accepted: 03/16/2025] [Indexed: 03/25/2025]
Abstract
Neurological disorders significantly impair neuronal function and lead to cognitive and motor deficits. This review manuscript explores the therapeutic potential of key proteins-Klotho, SIRT-1, Nrf2, and HO-1-in combating these disorders. Neurological conditions encompass neurotraumatic, neurodegenerative, and neuropsychiatric diseases, all characterized by neuronal loss and dysfunction. The complex functions of Klotho, an anti-aging protein, and SIRT-1, a histone deacetylase, highlight their roles in neuronal survival and neuroprotection through the enhancement of antioxidant defences and the modulation of stress responses. Nrf2 functions as the principal regulator of the antioxidant response, whereas HO-1 facilitates the control of oxidative stress and the resolution of inflammation. Evidence suggests that the interplay between these proteins facilitates neuroprotection by decreasing oxidative damage and promoting cognitive function. The study emphasises the significance of signalling pathways, particularly the Nrf2/HO-1 axis, which are essential in mitigating oxidative stress and inflammation linked to neurodegenerative disorders. Future therapeutic strategies must consider personalized approaches, innovative drug delivery systems, and early intervention to optimize outcomes. This review provides a comprehensive framework for understanding how targeting these pathways can mitigate the burden of neurological disorders, advancing the development of effective interventions for enhancing brain health.
Collapse
Affiliation(s)
- Ravi Rana
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India; Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India
| | - Ritam Mukherjee
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India; Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India; Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India.
| | - Zuber Khan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India; Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India
| | - Ghanshyam Das Gupta
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, India; Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India
| | - Acharan S Narula
- Narula Research, LLC, 107 Boulder Bluff, Chapel Hill, NC 27516, USA
| |
Collapse
|
2
|
Previti S, Calcaterra E, Di Chio C, Natale B, Waqas M, Bogacz M, Schirmeister T, Cavelier F, Calabrò ML, Cosconati S, Ettari R, Zappalà M. Identification of constrained peptidomimetics carrying a Michael acceptor warhead as antitrypanosomal agents. Eur J Med Chem 2025; 288:117389. [PMID: 39970727 DOI: 10.1016/j.ejmech.2025.117389] [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: 12/06/2024] [Revised: 02/07/2025] [Accepted: 02/10/2025] [Indexed: 02/21/2025]
Abstract
In this structure-activity relationship (SAR) study, we report the development of rhodesain-targeting peptidomimetics with antitrypanosomal activity. The new compounds (SPR65-SPR80) feature the 1,2,3,4-tetrahydroisoquinoline-3-carboxylic acid (Tic) moiety as conformationally constrained Phe analog. Various substituents were inserted at the P1 and P3 positions, and the methyl vinyl ketone moiety was introduced as warhead. The incorporation of Tic resulted in reduced affinity against rhodesain compared to the parent compounds containing Phe (2a-m), suggesting that its rigidity negatively affects target binding. Nevertheless, promising EC50 values ranging from 0.42 to 1.35 μM were observed in cell-based assays, probably due to better pharmacokinetic properties and/or interactions with additional protozoal targets. CC50 values > 100 μM were observed. Therefore, while Tic is less tolerated by rhodesain, its incorporation in peptidomimetic Michael acceptors led to antitrypanosomal effects that were comparable or slightly better than those of the parent compounds and no cytotoxicity up to 100 μM. These findings could be taken into consideration in future SAR studies aimed at the development of antitrypanosomal agents.
Collapse
Affiliation(s)
- Santo Previti
- Department of Chemical, Biological, Pharmaceutical, and Environmental Sciences, University of Messina, Viale Stagno d'Alcontres 31, 98166, Messina, Italy.
| | - Elsa Calcaterra
- Department of Chemical, Biological, Pharmaceutical, and Environmental Sciences, University of Messina, Viale Stagno d'Alcontres 31, 98166, Messina, Italy
| | - Carla Di Chio
- Department of Chemical, Biological, Pharmaceutical, and Environmental Sciences, University of Messina, Viale Stagno d'Alcontres 31, 98166, Messina, Italy
| | - Benito Natale
- DiSTABiF, University of Campania Luigi Vanvitelli, Via Vivaldi 43, 81100, Caserta, Italy
| | - Muhammad Waqas
- DiSTABiF, University of Campania Luigi Vanvitelli, Via Vivaldi 43, 81100, Caserta, Italy
| | - Marta Bogacz
- Institute of Organic Chemistry & Macromolecular Chemistry, Friedrich-Schiller-University of Jena, 07743, Jena, Germany
| | - Tanja Schirmeister
- Institute of Pharmaceutical and Biomedical Sciences, University of Mainz, 55128, Mainz, Germany
| | - Florine Cavelier
- Institut des Biomolécules Max Mousseron IBMM, UMR 5247, CNRS, Université Montpellier, ENSCM, Pôle Chimie Balard, 1919 Route de Mende, 34293, Montpellier, France
| | - Maria Luisa Calabrò
- Department of Chemical, Biological, Pharmaceutical, and Environmental Sciences, University of Messina, Viale Stagno d'Alcontres 31, 98166, Messina, Italy
| | - Sandro Cosconati
- DiSTABiF, University of Campania Luigi Vanvitelli, Via Vivaldi 43, 81100, Caserta, Italy
| | - Roberta Ettari
- Department of Chemical, Biological, Pharmaceutical, and Environmental Sciences, University of Messina, Viale Stagno d'Alcontres 31, 98166, Messina, Italy
| | - Maria Zappalà
- Department of Chemical, Biological, Pharmaceutical, and Environmental Sciences, University of Messina, Viale Stagno d'Alcontres 31, 98166, Messina, Italy
| |
Collapse
|
3
|
Chu CT, Uruno A, Katsuoka F, Yamamoto M. Role of NRF2 in Pathogenesis of Alzheimer's Disease. Antioxidants (Basel) 2024; 13:1529. [PMID: 39765857 PMCID: PMC11727090 DOI: 10.3390/antiox13121529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 01/15/2025] Open
Abstract
Alzheimer's disease (AD) is a polygenic, multifactorial neurodegenerative disorder and remains the most prevalent form of dementia, globally. Despite decades of research efforts, there is still no effective cure for this debilitating condition. AD research has increasingly focused on transcription factor NRF2 (nuclear factor erythroid 2-related factor 2) as a potential therapeutic target. NRF2 plays a crucial role in protecting cells and tissues from environmental stressors, such as electrophiles and reactive oxygen species. Recently, an increasing number of studies have demonstrated that NRF2 is a key regulator in AD pathology. NRF2 is highly expressed in microglia, resident macrophages in the central nervous system, and contributes to neuroinflammation, phagocytosis and neurodegeneration in AD. NRF2 has been reported to modulate microglia-induced inflammation and facilitate the transition from homeostatic microglia to a disease-associated microglia subset. Genetic and pharmacological activation of NRF2 has been demonstrated to improve cognitive function. Here, we review the current understanding of the involvement of NRF2 in AD and the critical role that NRF2 plays in microglia in the context of AD. Our aim is to highlight the potential of targeting NRF2 in the microglia as a promising therapeutic strategy for mitigating the progression of AD.
Collapse
Affiliation(s)
- Ching-Tung Chu
- Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank Organization, Tohoku University, Sendai 980-8573, Japan; (C.-T.C.); (A.U.)
| | - Akira Uruno
- Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank Organization, Tohoku University, Sendai 980-8573, Japan; (C.-T.C.); (A.U.)
| | - Fumiki Katsuoka
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai 980-8573, Japan;
- Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University, Sendai 980-8573, Japan
| | - Masayuki Yamamoto
- Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank Organization, Tohoku University, Sendai 980-8573, Japan; (C.-T.C.); (A.U.)
| |
Collapse
|
4
|
Izumi Y, Koyama Y. Nrf2-Independent Anti-Inflammatory Effects of Dimethyl Fumarate: Challenges and Prospects in Developing Electrophilic Nrf2 Activators for Neurodegenerative Diseases. Antioxidants (Basel) 2024; 13:1527. [PMID: 39765855 PMCID: PMC11727036 DOI: 10.3390/antiox13121527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 11/26/2024] [Accepted: 12/03/2024] [Indexed: 01/15/2025] Open
Abstract
The NF-E2-related factor 2 (Nrf2)-antioxidant response element (ARE) pathway is a potential therapeutic target for central nervous system diseases. This review emphasizes the role of oxidative stress and neuroinflammation in neurodegenerative diseases, highlighting the therapeutic potential of Nrf2 activators such as dimethyl fumarate (DMF). DMF, initially administered for treating psoriasis, has demonstrated efficacy in multiple sclerosis and is metabolized to monomethyl fumarate, which may exert significant therapeutic effects. DMF activates the Nrf2-ARE pathway, and recent studies have indicated that its anti-inflammatory effects occur through Nrf2-independent mechanisms. Electrophilic Nrf2 activators, such as DMF, covalently bind to cysteine residues in proteins and modulate their function. We discuss the implications of cysteine residue modifications by DMF, which may cause both therapeutic benefits and potential off-target effects. Furthermore, we propose a chemical proteomics-based drug discovery approach to achieve desired therapeutic effects by selectively covalently modifying cysteines in target proteins. These findings advocate for a broader understanding of the Nrf2-independent mechanisms of electrophilic Nrf2 activators, thereby improving drug discovery strategies that target neurodegenerative diseases while minimizing toxicity.
Collapse
Affiliation(s)
- Yasuhiko Izumi
- Laboratory of Pharmacology, Kobe Pharmaceutical University, 4-19-1 Motoyamakita-machi, Higashinada-ku, Kobe 658-8558, Japan;
| | | |
Collapse
|
5
|
Cade S, Prestidge C, Zhou X, Bobrovskaya L. The effects of a bioavailable curcumin formulation on Alzheimer's disease pathologies: A potential risk for neuroinflammation. IBRAIN 2024; 10:500-518. [PMID: 39691427 PMCID: PMC11649387 DOI: 10.1002/ibra.12187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 11/18/2024] [Accepted: 11/21/2024] [Indexed: 12/19/2024]
Abstract
Alzheimer's disease (AD) is a common cause of dementia characterized by the presence of two proteinaceous deposits in the brain. These pathologies may be a consequence of complex interactions between neurons and glia before the onset of cognitive impairments. Curcumin, a bioactive compound found in turmeric, is a promising candidate for AD because it alleviates neuropathologies in mouse models of the disease. Although its clinical efficacy has been hindered by low oral bioavailability, the development of new formulations may overcome this limitation. The purpose of this study was to determine the effects of a bioavailable curcumin formulation in a mouse model of AD. The formulation was administered to mice in drinking water after encapsulation into micelles using a previously validated method. A neuropathological assessment was performed to determine if it slows or alters the course of the disease. Cognitive performance was not included because it had already been assessed by a previous study. The bioavailable curcumin formulation was unable to alter the size or number of amyloid plaques in a transgenic mouse model. In addition, mechanisms that regulate amyloid beta production were unchanged, suggesting that the disease had not been altered. The number of reactive astrocytes in the hippocampus and dentate gyrus was not altered by curcumin. However, protein levels of glial fibrillary acidic protein were increased overall in the brain, suggesting that it may have aggravated neuroinflammation. Therefore, a higher dosage, despite its enhanced oral bioavailability, may have a potential risk for neuroinflammation.
Collapse
Affiliation(s)
- Shaun Cade
- Health and Biomedical Innovation, Clinical and Health SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| | - Clive Prestidge
- Center for Pharmaceutical Innovation, Clinical and Health SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| | - Xin‐Fu Zhou
- Health and Biomedical Innovation, Clinical and Health SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| | - Larisa Bobrovskaya
- Health and Biomedical Innovation, Clinical and Health SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| |
Collapse
|
6
|
Sánchez-Sanz A, Coronado-Albi MJ, Muñoz-Viana R, García-Merino A, Sánchez-López AJ. Neuroprotective and Anti-Inflammatory Effects of Dimethyl Fumarate, Monomethyl Fumarate, and Cannabidiol in Neurons and Microglia. Int J Mol Sci 2024; 25:13082. [PMID: 39684792 DOI: 10.3390/ijms252313082] [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: 10/31/2024] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024] Open
Abstract
Dimethyl fumarate (DMF) is an immunomodulatory treatment for multiple sclerosis (MS) that can cross the blood-brain barrier, presenting neuroprotective potential. Its mechanism of action is not fully understood, and there is a need to characterize whether DMF or its bioactive metabolite monomethyl fumarate (MMF) exerts neuroprotective properties. Moreover, the combination of adjuvant agents such as cannabidiol (CBD) could be relevant to enhance neuroprotection. The aim of this study was to compare the neuroprotective and immunomodulatory effects of DMF, MMF, and CBD in neurons and microglia in vitro. We found that DMF and CBD, but not MMF, activated the Nrf2 antioxidant pathway in neurons. Similarly, only DMF and CBD, but not MMF, prevented the LPS-induced activation of the inflammatory pathway NF-kB in microglia. Additionally, the three drugs inhibited the production of nitric oxide in microglia and protected neurons against apoptosis. Transcriptomically, DMF modulated a greater number of inflammatory and Nrf2-related genes compared to MMF and CBD in both neurons and microglia. Our results show that DMF and MMF, despite being structurally related, present differences in their mechanisms of action that could be relevant for the achievement of neuroprotection in MS patients. Additionally, CBD shows potential as a neuroprotective agent.
Collapse
Affiliation(s)
- Alicia Sánchez-Sanz
- Neuroimmunology Unit, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana, 28222 Madrid, Spain
| | - María José Coronado-Albi
- Confocal Microscopy Core Facility, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana, 28222 Madrid, Spain
| | - Rafael Muñoz-Viana
- Bioinformatics Unit, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana, 28222 Madrid, Spain
| | - Antonio García-Merino
- Neuroimmunology Unit, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana, 28222 Madrid, Spain
- Department of Neurology, Hospital Universitario Puerta de Hierro Majadahonda, 28222 Madrid, Spain
- Department of Medicine, Universidad Autónoma de Madrid, 28049 Madrid, Spain
- Red Española de Esclerosis Múltiple (REEM), 08028 Barcelona, Spain
| | - Antonio J Sánchez-López
- Neuroimmunology Unit, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana, 28222 Madrid, Spain
- Red Española de Esclerosis Múltiple (REEM), 08028 Barcelona, Spain
- Biobank, Instituto de Investigación Sanitaria Puerta de Hierro-Segovia de Arana, 28222 Madrid, Spain
| |
Collapse
|
7
|
Koike S, Tsurudome S, Okano S, Kishida A, Ogasawara Y. Dimethyl Fumarate Reduces Methylglyoxal-derived Carbonyl Stress Through Nrf2/GSH Activation in SH-SY5Y Cells. Neurochem Res 2024; 50:28. [PMID: 39576418 DOI: 10.1007/s11064-024-04255-0] [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: 10/04/2024] [Accepted: 10/15/2024] [Indexed: 11/24/2024]
Abstract
Carbonyl stress refers to the excessive accumulation of advanced glycation end products (AGEs) in mammalian tissues. This phenomenon plays a significant role in the pathogenesis of various diseases, including diabetes, chronic renal failure, arteriosclerosis, and central nervous system (CNS) disorders. We have previously demonstrated that an increase in glutathione concentration, dependent on the nuclear factor erythroid 2-related factor 2 (Nrf2) system, provides a potent cytoprotective effect against Methylglyoxal (MGO)-induced carbonyl stress. Meanwhile, dimethyl fumarate (DMF), known for its Nrf2-activating effects, was recently approved as a treatment for multiple sclerosis (MS), a neurodegenerative disease. DMF is a first line therapy for relapsing-remitting MS and may also be effective for other neurodegenerative conditions. However, the detailed mechanisms by which DMF mitigates neurodegenerative pathologies remain unclear. This study investigates the impact of DMF on anticarbonyl activity and its underlying mechanism focusing on the accumulation of carbonyl protein in the cell. MGO, a glucose metabolite, was used to induce carbonylation in the neuronal cell line. MGO is a typical carbonyl compound that readily reacts with arginine and lysine residues to form AGE-modified proteins. Methylglyoxal-derived hydroimidazolone 1 (MG-H1) often forms uncharged, hydrophobic residues on the protein surface, which can affect protein distribution and lead to misfolding. Our findings indicate that DMF increases levels of glutathione (GSH), glutamate cysteine ligase modifier subunit (GCLM), and nuclear Nrf2 in SH-SY5Y cells. Importantly, DMF pretreatment significantly reduced the accumulation of MG-H1-modified proteins. Furthermore, this effect of DMF was diminished when Nrf2 expression was suppressed and when GCL, a rate-limiting enzyme in GSH synthesis, was inhibited. Thus, the increase in GSH levels, leading to the activation of the Nrf2 pathway, a key factor in DMF's ability to suppress the accumulation of MG-H1-modified proteins. This study is the first to demonstrate that DMF possesses strong anticarbonyl stress activity in neuronal cells. Therefore, future research may extend the application of DMF to other CNS diseases associated with carbonyl stress, such as Alzheimer's and Parkinson's disease.
Collapse
Affiliation(s)
- Shin Koike
- Department of Analytical Biochemistry, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo, 204-8588, Japan
| | - Satori Tsurudome
- Department of Analytical Biochemistry, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo, 204-8588, Japan
| | - Saki Okano
- Department of Analytical Biochemistry, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo, 204-8588, Japan
| | - Atsushi Kishida
- Graduate School of Pharmaceutical Sciences, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo, 204-8588, Japan
| | - Yuki Ogasawara
- Department of Analytical Biochemistry, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo, 204-8588, Japan.
| |
Collapse
|
8
|
Filippone A, Mannino D, Cucinotta L, Calapai F, Crupi L, Paterniti I, Esposito E. Rebalance of mitophagy by inhibiting LRRK2 improves colon alterations in an MPTP in vivo model. iScience 2024; 27:110980. [PMID: 39635134 PMCID: PMC11615202 DOI: 10.1016/j.isci.2024.110980] [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: 01/31/2024] [Revised: 05/10/2024] [Accepted: 09/13/2024] [Indexed: 12/07/2024] Open
Abstract
Mutations in the leucine-rich repeat kinase 2 (LRRK2) gene are common genetic causes of Parkinson's disease (PD). Studies demonstrated that variants in LRRK2 genetically link intestinal disorders to PD. We aimed to evaluate whether the selective inhibitor of LRRK2, PF-06447475 (PF-475), attenuates the PD induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) in central nervous system (CNS) and in the gastrointestinal system. CD1 mice received four intraperitoneal injections of MPTP (20 mg/kg, total dose of 80 mg/kg) at 2 h intervals (day 1). After 24 h PF-475 was administered intraperitoneally at the doses of 2.5, 5, and 10 mg/kg for seven days. LRRK2 inhibition reduced brain α-synuclein and modulated mitophagy pathway and reduced pro-inflammatory markers and α-synuclein aggregates in colonic tissues through the modulation of mitophagy proteins. LRRK2 inhibition suppressed MPTP-induced enteric dopaminergic neuronal injury and protected tight junction in the colon. Results suggested that PF-475 may attenuate gastrointestinal dysfunction associated to PD.
Collapse
Affiliation(s)
- Alessia Filippone
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31-98166 Messina, Italy
| | - Deborah Mannino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31-98166 Messina, Italy
| | - Laura Cucinotta
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31-98166 Messina, Italy
| | - Fabrizio Calapai
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy
| | - Lelio Crupi
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31-98166 Messina, Italy
| | - Irene Paterniti
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31-98166 Messina, Italy
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31-98166 Messina, Italy
| |
Collapse
|
9
|
Sheng L, Bhalla R. Biomarkers and Target-Specific Small-Molecule Drugs in Alzheimer's Diagnostic and Therapeutic Research: From Amyloidosis to Tauopathy. Neurochem Res 2024; 49:2273-2302. [PMID: 38844706 PMCID: PMC11310295 DOI: 10.1007/s11064-024-04178-w] [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: 01/16/2024] [Revised: 03/31/2024] [Accepted: 05/22/2024] [Indexed: 08/09/2024]
Abstract
Alzheimer's disease (AD) is the most common type of human dementia and is responsible for over 60% of diagnosed dementia cases worldwide. Abnormal deposition of β-amyloid and the accumulation of neurofibrillary tangles have been recognised as the two pathological hallmarks targeted by AD diagnostic imaging as well as therapeutics. With the progression of pathological studies, the two hallmarks and their related pathways have remained the focus of researchers who seek for AD diagnostic and therapeutic strategies in the past decades. In this work, we reviewed the development of the AD biomarkers and their corresponding target-specific small molecule drugs for both diagnostic and therapeutic applications, underlining their success, failure, and future possibilities.
Collapse
Affiliation(s)
- Li Sheng
- Centre for Advanced Imaging, The University of Queensland, St Lucia, Brisbane, QLD, 4072, Australia.
| | - Rajiv Bhalla
- Centre for Advanced Imaging, The University of Queensland, St Lucia, Brisbane, QLD, 4072, Australia
| |
Collapse
|
10
|
Mayer C, Riera-Ponsati L, Kauppinen S, Klitgaard H, Erler JT, Hansen SN. Targeting the NRF2 pathway for disease modification in neurodegenerative diseases: mechanisms and therapeutic implications. Front Pharmacol 2024; 15:1437939. [PMID: 39119604 PMCID: PMC11306042 DOI: 10.3389/fphar.2024.1437939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 07/03/2024] [Indexed: 08/10/2024] Open
Abstract
Neurodegenerative diseases constitute a global health issue and a major economic burden. They significantly impair both cognitive and motor functions, and their prevalence is expected to rise due to ageing societies and continuous population growth. Conventional therapies provide symptomatic relief, nevertheless, disease-modifying treatments that reduce or halt neuron death and malfunction are still largely unavailable. Amongst the common hallmarks of neurodegenerative diseases are protein aggregation, oxidative stress, neuroinflammation and mitochondrial dysfunction. Transcription factor nuclear factor-erythroid 2-related factor 2 (NRF2) constitutes a central regulator of cellular defense mechanisms, including the regulation of antioxidant, anti-inflammatory and mitochondrial pathways, making it a highly attractive therapeutic target for disease modification in neurodegenerative disorders. Here, we describe the role of NRF2 in the common hallmarks of neurodegeneration, review the current pharmacological interventions and their challenges in activating the NRF2 pathway, and present alternative therapeutic approaches for disease modification.
Collapse
Affiliation(s)
| | - Lluís Riera-Ponsati
- NEUmiRNA Therapeutics, Copenhagen, Denmark
- Center for RNA Medicine, Aalborg University, Copenhagen, Denmark
| | - Sakari Kauppinen
- NEUmiRNA Therapeutics, Copenhagen, Denmark
- Center for RNA Medicine, Aalborg University, Copenhagen, Denmark
| | | | | | | |
Collapse
|
11
|
Rezaee N, Hone E, Sohrabi HR, Johnson S, Zhong L, Chatur P, Gunzburg S, Martins RN, Fernando WMADB. Sorghum Grain Polyphenolic Extracts Demonstrate Neuroprotective Effects Related to Alzheimer's Disease in Cellular Assays. Foods 2024; 13:1716. [PMID: 38890943 PMCID: PMC11171927 DOI: 10.3390/foods13111716] [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: 04/07/2024] [Revised: 05/23/2024] [Accepted: 05/23/2024] [Indexed: 06/20/2024] Open
Abstract
Sorghum grain contains high levels and a diverse profile of polyphenols (PPs), which are antioxidants known to reduce oxidative stress when consumed in the diet. Oxidative stress leading to amyloid-β (Aβ) aggregation, neurotoxicity, and mitochondrial dysfunction is implicated in the pathogenesis of Alzheimer's disease (AD). Thus, PPs have gained attention as possible therapeutic agents for combating AD. This study aimed to (a) quantify the phenolic compounds (PP) and antioxidant capacities in extracts from six different varieties of sorghum grain and (b) investigate whether these PP extracts exhibit any protective effects on human neuroblastoma (BE(2)-M17) cells against Aβ- and tau-induced toxicity, Aβ aggregation, mitochondrial dysfunction, and reactive oxygen species (ROS) induced by Aβ and tert-butyl hydroperoxide (TBHP). PP and antioxidant capacity were quantified using chemical assays. Aβ- and tau-induced toxicity was determined using the 3-(4,5-dimenthylthiazol-2-yl)-2,5-dimethyltetrazolium bromide (MTS) assay. The thioflavin T (Th-T) assay assessed anti-Aβ aggregation. The dichlorodihydrofluorescein diacetate (DCFDA) assay determined the levels of general ROS and the MitoSOX assay determined the levels of mitochondrial superoxide. Sorghum varieties Shawaya short black-1 and IS1311C possessed the highest levels of total phenolics, total flavonoids, and antioxidant capacity, and sorghum varieties differed significantly in their profile of individual PPs. All extracts significantly increased cell viability compared to the control (minus extract). Variety QL33 (at 2000 µg sorghum flour equivalents/mL) showed the strongest protective effect with a 28% reduction in Aβ-toxicity cell death. The extracts of all sorghum varieties significantly reduced Aβ aggregation. All extracts except that from variety B923296 demonstrated a significant (p ≤ 0.05) downregulation of Aβ-induced and TBHP-induced ROS and mitochondrial superoxide relative to the control (minus extract) in a dose- and variety-dependent manner. We have demonstrated for the first time that sorghum polyphenolic extracts show promising neuroprotective effects against AD, which indicates the potential of sorghum foods to exert a similar beneficial property in the human diet. However, further analysis in other cellular models and in vivo is needed to confirm these effects.
Collapse
Affiliation(s)
- Nasim Rezaee
- Centre of Excellence for Alzheimer’s Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA 6027, Australia (E.H.)
| | - Eugene Hone
- Centre of Excellence for Alzheimer’s Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA 6027, Australia (E.H.)
| | - Hamid R. Sohrabi
- Centre of Excellence for Alzheimer’s Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA 6027, Australia (E.H.)
- Department of Biomedical Sciences, Macquarie University, Sydney, NSW 2109, Australia
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, WA 6150, Australia
| | - Stuart Johnson
- School of Molecular and Life Sciences, Faculty of Science and Engineering, Curtin University, Perth, WA 6845, Australia
| | - Leizhou Zhong
- School of Molecular and Life Sciences, Faculty of Science and Engineering, Curtin University, Perth, WA 6845, Australia
| | - Prakhar Chatur
- School of Molecular and Life Sciences, Faculty of Science and Engineering, Curtin University, Perth, WA 6845, Australia
| | | | - Ralph N. Martins
- Centre of Excellence for Alzheimer’s Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA 6027, Australia (E.H.)
- Department of Biomedical Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - W. M. A. D. Binosha Fernando
- Centre of Excellence for Alzheimer’s Disease Research & Care, School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA 6027, Australia (E.H.)
| |
Collapse
|
12
|
Araújo AM, Marques SI, Guedes de Pinho P, Carmo H, Carvalho F, Silva JP. Identification of key neuronal mechanisms triggered by dimethyl fumarate in SH-SY5Y human neuroblastoma cells through a metabolomic approach. Arch Toxicol 2024; 98:1151-1161. [PMID: 38368281 PMCID: PMC10944387 DOI: 10.1007/s00204-024-03683-9] [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: 10/20/2023] [Accepted: 01/17/2024] [Indexed: 02/19/2024]
Abstract
Dimethyl fumarate (DMF) is an old drug used for psoriasis treatment that has recently been repurposed to treat relapse-remitting multiple sclerosis, mostly due to its neuro- and immunomodulatory actions. However, mining of a pharmacovigilance database recently ranked DMF as the second pharmaceutical most associated with cognitive adverse events. To our best knowledge, the signaling mechanisms underlying its therapeutic and neurotoxic outcomes remain mostly undisclosed. This work thus represents the first-hand assessment of DMF-induced metabolic changes in undifferentiated SH-SY5Y human neuroblastoma cells, through an untargeted metabolomic approach using gas chromatography-mass spectrometry (GC-MS). The endometabolome was analyzed following 24 h and 96 h of exposure to two pharmacologically relevant DMF concentrations (0.1 and 10 μM). None of these conditions significantly reduced metabolic activity (MTT reduction assay). Our data showed that 24 h-exposure to DMF at both concentrations tested mainly affected metabolic pathways involved in mitochondrial activity (e.g., citric acid cycle, de novo triacylglycerol biosynthesis), and the synthesis of catecholamines and serotonin by changing the levels of their respective precursors, namely phenylalanine (0.68-fold decrease for 10 μM DMF vs vehicle), and tryptophan (1.36-fold increase for 0.1 μM DMF vs vehicle). Interestingly, taurine, whose levels can be modulated via Nrf2 signaling (DMF's primary target), emerged as a key mediator of DMF's neuronal action, displaying a 3.86-fold increase and 0.27-fold decrease for 10 μM DMF at 24 h and 96 h, respectively. A 96 h-exposure to DMF seemed to mainly trigger pathways associated with glucose production (e.g., gluconeogenesis, glucose-alanine cycle, malate-aspartate shuttle), possibly related to the metabolism of DMF into monomethyl fumarate and its further conversion into glucose via activation of the citric acid cycle. Overall, our data contribute to improving the understanding of the events associated with neuronal exposure to DMF.
Collapse
Affiliation(s)
- Ana Margarida Araújo
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
- UCIBIO, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
- LAQV-REQUIMTE, Laboratory of Bromatology and Hydrology, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
| | - Sandra I Marques
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
- UCIBIO, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
| | - Paula Guedes de Pinho
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
- UCIBIO, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
| | - Helena Carmo
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
- UCIBIO, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
| | - Félix Carvalho
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal.
- UCIBIO, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal.
| | - João Pedro Silva
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal.
- UCIBIO, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal.
| |
Collapse
|
13
|
Chen WT, Dodson M. The untapped potential of targeting NRF2 in neurodegenerative disease. FRONTIERS IN AGING 2023; 4:1270838. [PMID: 37840813 PMCID: PMC10569223 DOI: 10.3389/fragi.2023.1270838] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 09/18/2023] [Indexed: 10/17/2023]
Abstract
Since its initial discovery almost three decades ago, the transcription factor nuclear factor erythroid 2-related factor 2 (NRF2) has been shown to regulate a host of downstream transcriptional responses and play a critical role in preventing or promoting disease progression depending on the context. Critically, while the importance of proper nuclear factor erythroid 2-related factor 2 function has been demonstrated across a variety of pathological settings, the ability to progress NRF2-targeted therapeutics to clinic has remained frustratingly elusive. This is particularly true in the case of age-related pathologies, where nuclear factor erythroid 2-related factor 2 is a well-established mitigator of many of the observed pathogenic effects, yet options to target this pathway remain limited. Along these lines, loss of nuclear factor erythroid 2-related factor 2 function has clearly been shown to enhance neuropathological outcomes, with enhancing nuclear factor erythroid 2-related factor 2 pathway activation to prevent neurodegenerative/neurological disease progression continuing to be an active area of interest. One critical obstacle in generating successful therapeutics for brain-related pathologies is the ability of the compound to cross the blood brain barrier (BBB), which has also hampered the implementation of several promising nuclear factor erythroid 2-related factor 2 inducers. Another limitation is that many nuclear factor erythroid 2-related factor 2 activators have undesirable off-target effects due to their electrophilic nature. Despite these constraints, the field has continued to evolve, and several viable means of targeting nuclear factor erythroid 2-related factor 2 in a neuropathological context have emerged. In this perspective, we will briefly discuss the key findings and promising therapeutic options that have been discovered to date, as well as highlight emerging areas of NRF2-neurodegeneration research that provide hope for successfully targeting this pathway in the future.
Collapse
Affiliation(s)
| | - Matthew Dodson
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
14
|
Rahman SO, Khan T, Iqubal A, Agarwal S, Akhtar M, Parvez S, Shah ZA, Najmi AK. Association between insulin and Nrf2 signalling pathway in Alzheimer's disease: A molecular landscape. Life Sci 2023:121899. [PMID: 37394097 DOI: 10.1016/j.lfs.2023.121899] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/17/2023] [Accepted: 06/27/2023] [Indexed: 07/04/2023]
Abstract
Insulin, a well-known hormone, has been implicated as a regulator of blood glucose levels for almost a century now. Over the past few decades, the non-glycemic actions of insulin i.e. neuronal growth and proliferation have been extensively studied. In 2005, Dr. Suzanne de La Monte and her team reported that insulin might be involved in the pathogenesis of Alzheimer's Disease (AD) and thus coined a term "Type-3 diabetes" This hypothesis was supported by several subsequent studies. The nuclear factor erythroid 2- related factor 2 (Nrf2) triggers a cascade of events under the regulation of distinct mechanisms including protein stability, phosphorylation and nuclear cytoplasmic shuttling, finally leading to the protection against oxidative damage. The Nrf2 pathway has been investigated extensively in relevance to neurodegenerative disorders, particularly AD. Many studies have indicated a strong correlation between insulin and Nrf2 signalling pathways both in the periphery and the brainbut merely few of them have focused on elucidating their inter-connective role in AD. The present review emphasizes key molecular pathways that correlate the role of insulin with Nrf2 during AD. The review has also identified key unexplored areas that could be investigated in future to further establish the insulin and Nrf2 influence in AD.
Collapse
Affiliation(s)
- Syed Obaidur Rahman
- Pharmaceutical Medicine, Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Tahira Khan
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Ashif Iqubal
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Shivani Agarwal
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Mohd Akhtar
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Suhel Parvez
- Neurobehavioral Pharmacology Laboratory, Department of Medical Elementology and Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Zahoor Ahmad Shah
- Department of Medicinal and Biological Chemistry, University of Toledo, 3000 Arlington Avenue, Toledo, OH 43614, USA
| | - Abul Kalam Najmi
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
15
|
ChemR23 signaling ameliorates cognitive impairments in diabetic mice via dampening oxidative stress and NLRP3 inflammasome activation. Redox Biol 2022; 58:102554. [PMID: 36446229 PMCID: PMC9703827 DOI: 10.1016/j.redox.2022.102554] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 11/22/2022] [Accepted: 11/23/2022] [Indexed: 11/27/2022] Open
Abstract
Diabetes mellitus is associated with cognitive impairment characterized by memory loss and cognitive inflexibility. Recent studies have revealed that ChemR23 is implicated in both diabetes mellitus and Alzheimer's disease. However, the impact of ChemR23 on diabetes-associated cognitive impairment remains elusive. In this study, we explored the longitudinal changes of ChemR23 expression and cognitive function in STZ-induced type 1 diabetic mice and leptin receptor knockout type 2 diabetic mice at different ages. We also treated diabetic mice with ChemR23 agonists RvE1 or chemerin-9 to explore whether ChemR23 activation could alleviate diabetes-associated cognitive impairment. The underlying mechanism was further investigated in diabetic mice with genetic deletion of ChemR23. The results showed that ChemR23 expression was decreased along with aging and the progression of diabetes, suggesting that abnormal ChemR23 signaling may be involved in diabetes-associated cognitive impairment. Administration of RvE1 or chemerin-9 ameliorated oxidative stress and inhibited NLRP3 inflammasome activation through Nrf2/TXNIP pathway, and ultimately alleviated cognitive impairment in diabetic mice. Depletion of ChemR23 in diabetic mice abolished the beneficial effects of RvE1 and chemerin-9, and exacerbated cognitive impairment via increasing oxidative stress and activating NLRP3 inflammasome. Collectively, our data highlight the crucial role of ChemR23 signaling in diabetes-associated cognitive impairment via regulating oxidative stress and NLRP3 inflammasome, and targeting ChemR23 may serve as a promising novel strategy for the treatment of diabetes-associated cognitive impairment.
Collapse
|
16
|
Zhang Y, Wang M, Chang W. Iron dyshomeostasis and ferroptosis in Alzheimer’s disease: Molecular mechanisms of cell death and novel therapeutic drugs and targets for AD. Front Pharmacol 2022; 13:983623. [PMID: 36188557 PMCID: PMC9523169 DOI: 10.3389/fphar.2022.983623] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/25/2022] [Indexed: 11/13/2022] Open
Abstract
Alzheimer’s disease (AD) is a degenerative disease of the central nervous system that is the most common type of senile dementia. Ferroptosis is a new type of iron-dependent programmed cell death identified in recent years that is different from other cell death forms. Ferroptosis is induced by excessive accumulation of lipid peroxides and reactive oxygen species (ROS) in cells. In recent years, it has been found that ferroptosis plays an important role in the pathological process of AD. Iron dyshomeostasis contribute to senile plaques (SP) deposition and neurofibrillary tangles (NFTs). Iron metabolism imbalance in brain and the dysfunction of endogenous antioxidant systems including system Xc- and glutathione peroxidase (GPX) are closely related to the etiopathogenesis of AD. Dysfunction of nuclear receptor coactivator 4 (NCOA4)-mediated ferritinophagy induced ferroptosis can accelerates the pathological process of AD. In addition, NRF2, through regulating the expression of a considerable number of genes related to ferroptosis, including genes related to iron and glutathione metabolism, plays an important role in the development of AD. Here, we review the potential interaction between AD and ferroptosis and the major pathways regulating ferroptosis in AD. We also review the active natural and synthetic compounds such as iron chelators, lipid peroxidation inhibitors and antioxidants available to treat AD by alleviating iron dyshomeostasis and preventing ferroptosis in mice and cell models to provide valuable information for the future treatment and prevention of AD.
Collapse
|
17
|
Heurtaux T, Bouvier DS, Benani A, Helgueta Romero S, Frauenknecht KBM, Mittelbronn M, Sinkkonen L. Normal and Pathological NRF2 Signalling in the Central Nervous System. Antioxidants (Basel) 2022; 11:1426. [PMID: 35892629 PMCID: PMC9394413 DOI: 10.3390/antiox11081426] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/18/2022] [Accepted: 07/19/2022] [Indexed: 11/17/2022] Open
Abstract
The nuclear factor erythroid 2-related factor 2 (NRF2) was originally described as a master regulator of antioxidant cellular response, but in the time since, numerous important biological functions linked to cell survival, cellular detoxification, metabolism, autophagy, proteostasis, inflammation, immunity, and differentiation have been attributed to this pleiotropic transcription factor that regulates hundreds of genes. After 40 years of in-depth research and key discoveries, NRF2 is now at the center of a vast regulatory network, revealing NRF2 signalling as increasingly complex. It is widely recognized that reactive oxygen species (ROS) play a key role in human physiological and pathological processes such as ageing, obesity, diabetes, cancer, and neurodegenerative diseases. The high oxygen consumption associated with high levels of free iron and oxidizable unsaturated lipids make the brain particularly vulnerable to oxidative stress. A good stability of NRF2 activity is thus crucial to maintain the redox balance and therefore brain homeostasis. In this review, we have gathered recent data about the contribution of the NRF2 pathway in the healthy brain as well as during metabolic diseases, cancer, ageing, and ageing-related neurodegenerative diseases. We also discuss promising therapeutic strategies and the need for better understanding of cell-type-specific functions of NRF2 in these different fields.
Collapse
Affiliation(s)
- Tony Heurtaux
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, 4367 Belvaux, Luxembourg; (S.H.R.); (M.M.); (L.S.)
- Luxembourg Center of Neuropathology (LCNP), 3555 Dudelange, Luxembourg; (D.S.B.); (K.B.M.F.)
| | - David S. Bouvier
- Luxembourg Center of Neuropathology (LCNP), 3555 Dudelange, Luxembourg; (D.S.B.); (K.B.M.F.)
- National Center of Pathology (NCP), Laboratoire National de Santé (LNS), 3555 Dudelange, Luxembourg
- Luxembourg Centre of Systems Biomedicine (LCSB), University of Luxembourg, 4367 Belvaux, Luxembourg
| | - Alexandre Benani
- Centre des Sciences du Goût et de l’Alimentation, AgroSup Dijon, CNRS, INRAE, Université Bourgogne Franche-Comté, 21000 Dijon, France;
| | - Sergio Helgueta Romero
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, 4367 Belvaux, Luxembourg; (S.H.R.); (M.M.); (L.S.)
- Luxembourg Center of Neuropathology (LCNP), 3555 Dudelange, Luxembourg; (D.S.B.); (K.B.M.F.)
| | - Katrin B. M. Frauenknecht
- Luxembourg Center of Neuropathology (LCNP), 3555 Dudelange, Luxembourg; (D.S.B.); (K.B.M.F.)
- National Center of Pathology (NCP), Laboratoire National de Santé (LNS), 3555 Dudelange, Luxembourg
| | - Michel Mittelbronn
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, 4367 Belvaux, Luxembourg; (S.H.R.); (M.M.); (L.S.)
- Luxembourg Center of Neuropathology (LCNP), 3555 Dudelange, Luxembourg; (D.S.B.); (K.B.M.F.)
- National Center of Pathology (NCP), Laboratoire National de Santé (LNS), 3555 Dudelange, Luxembourg
- Luxembourg Centre of Systems Biomedicine (LCSB), University of Luxembourg, 4367 Belvaux, Luxembourg
- Luxembourg Institute of Health (LIH), 1526 Luxembourg, Luxembourg
| | - Lasse Sinkkonen
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, 4367 Belvaux, Luxembourg; (S.H.R.); (M.M.); (L.S.)
| |
Collapse
|
18
|
Gao W, Guo L, Yang Y, Wang Y, Xia S, Gong H, Zhang BK, Yan M. Dissecting the Crosstalk Between Nrf2 and NF-κB Response Pathways in Drug-Induced Toxicity. Front Cell Dev Biol 2022; 9:809952. [PMID: 35186957 PMCID: PMC8847224 DOI: 10.3389/fcell.2021.809952] [Citation(s) in RCA: 110] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 12/29/2021] [Indexed: 12/12/2022] Open
Abstract
Nrf2 and NF-κB are important regulators of the response to oxidative stress and inflammation in the body. Previous pharmacological and genetic studies have confirmed crosstalk between the two. The deficiency of Nrf2 elevates the expression of NF-κB, leading to increased production of inflammatory factors, while NF-κB can affect the expression of downstream target genes by regulating the transcription and activity of Nrf2. At the same time, many therapeutic drug-induced organ toxicities, including hepatotoxicity, nephrotoxicity, cardiotoxicity, pulmonary toxicity, dermal toxicity, and neurotoxicity, have received increasing attention from researchers in clinical practice. Drug-induced organ injury can destroy body function, reduce the patients’ quality of life, and even threaten the lives of patients. Therefore, it is urgent to find protective drugs to ameliorate drug-induced injury. There is substantial evidence that protective medications can alleviate drug-induced organ toxicity by modulating both Nrf2 and NF-κB signaling pathways. Thus, it has become increasingly important to explore the crosstalk mechanism between Nrf2 and NF-κB in drug-induced toxicity. In this review, we summarize the potential molecular mechanisms of Nrf2 and NF-κB pathways and the important effects on adverse effects including toxic reactions and look forward to finding protective drugs that can target the crosstalk between the two.
Collapse
Affiliation(s)
- Wen Gao
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Lin Guo
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yan Yang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yu Wang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Shuang Xia
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Hui Gong
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Bi-Kui Zhang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Miao Yan
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Miao Yan,
| |
Collapse
|
19
|
A Distinct Hibiscus sabdariffa Extract Prevents Iron Neurotoxicity, a Driver of Multiple Sclerosis Pathology. Cells 2022; 11:cells11030440. [PMID: 35159249 PMCID: PMC8834068 DOI: 10.3390/cells11030440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/23/2022] [Accepted: 01/25/2022] [Indexed: 02/04/2023] Open
Abstract
Iron deposition in the brain begins early in multiple sclerosis (MS) and continues unabated. Ferrous iron is toxic to neurons, yet the therapies used in MS do not counter iron neurotoxicity. Extracts of Hibiscus sabdariffa (HS) are used in many cultures for medicinal purposes. We collected a distinct HS extract and found that it abolished the killing of neurons by iron in culture; medications used in MS were ineffective when similarly tested. Neuroprotection by HS was not due to iron chelation or anthocyanin content. In free radical scavenging assays, HS was equipotent to alpha lipoic acid, an anti-oxidant being tested in MS. However, alpha lipoic acid was only modestly protective against iron-mediated killing. Moreover, a subfraction of HS without radical scavenging activity negated iron toxicity, whereas a commercial hibiscus preparation with anti-oxidant activity could not. The idea that HS might have altered properties within neurons to confer neuroprotection is supported by its amelioration of toxicity caused by other toxins: beta-amyloid, rotenone and staurosporine. Finally, in a mouse model of MS, HS reduced disability scores and ameliorated the loss of axons in the spinal cord. HS holds therapeutic potential to counter iron neurotoxicity, an unmet need that drives the progression of disability in MS.
Collapse
|
20
|
He L, Sun Y. The potential role of Keap1-Nrf2 pathway in the pathogenesis of Alzheimer's disease, type 2 diabetes, and type 2 diabetes-related Alzheimer's disease. Metab Brain Dis 2021; 36:1469-1479. [PMID: 34129198 DOI: 10.1007/s11011-021-00762-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 06/06/2021] [Indexed: 12/30/2022]
Abstract
Kelch-like ECH associated-protein 1 (Keap1)-nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway is thought to be the key regulatory process defensing oxidative stress in multiple organs. Type 2 diabetes mellitus (T2DM) and Alzheimer's disease (AD) are both serious global health problems with high prevalence. A growing number of literatures have suggested a possible link between Keap1-Nrf2 signaling pathway and the pathological changes of T2DM, AD as well as T2DM-related AD. The current review mainly discusses how the damaged Keap1-Nrf2 signaling pathway leads to dysregulated redox molecular signaling, which may contribute to the pathogenesis of AD and T2DM-related cognitive dysfunction, as well as some compounds targeting this pathway. The further exploration of the mechanisms of this pathway could provide novel therapeutic strategies to improve cognitive function, through restoration of expression or translocation of Nrf2 and scavenging excessive free radicals.
Collapse
Affiliation(s)
- Ling He
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, China
| | - Yi Sun
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, China.
| |
Collapse
|
21
|
Scuderi SA, Lanza M, Casili G, Esposito F, Colarossi C, Giuffrida D, Irene P, Cuzzocrea S, Esposito E, Campolo M. TBK1 Inhibitor Exerts Antiproliferative Effect on Glioblastoma Multiforme Cells. Oncol Res 2021; 28:779-790. [PMID: 33741083 PMCID: PMC8420908 DOI: 10.3727/096504021x16161478258040] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Glioma are common malignant brain tumors, among which glioblastoma multiforme (GBM) has the worst prognosis. Different studies of GBM revealed that targeting nuclear factor B (NF-B) induced an attenuation tumor proliferation and prolonged cell survival. TBK1 {TANK [TRAF (TNF (tumor-necrosis-factor) receptor-associated factor)-associated NF-B activator]-binding kinase 1} is a serine/threonine protein kinase, and it is a member of the IB kinase (IKK) family involved in NF-B pathway activation. The aim of this study was to investigate the potential effect of BX795, an inhibitor of TBK1, in an in vitro and ex vivo model of GBM. GBM cell lines (U87 and U138) and primary GBM cells were treated with different concentrations of BX795 at different time points (24, 48, and 72h) to evaluate cell viability, autophagy, inflammation, and apoptosis. Our results demonstrated that BX795 10 M was able to reduce cell viability, showing antiproliferative effect in U87, U138, and primary GBM cells. Moreover, treatment with BX795 10 M increased the proapoptotic proteins Bax, p53, caspase 3, and caspase 9, whereas the antiapoptotic Bcl-2 expression was reduced. Additionally, our results showed a marked decrease in autophagy following BX795 treatment, reducing Atg 7, Atg 5/12, and AKT expression. The anti-inflammatory effect of BX795 was demonstrated by a significantly reduction in NIK, IKK, and TNF- expression, accompanied by a downregulation of angiogenesis. Furthermore, in primary GBM cell, BX795 10 M was able to reduce TBK1 pathway activation and SOX3 expression. In conclusion, these findings showed that TBK1 is involved in GBM proliferation, demonstrating that the inhibitor BX795, thanks to its abilities, could improve therapeutic strategies for GBM treatment.
Collapse
Affiliation(s)
- Sarah A. Scuderi
- *Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Marika Lanza
- *Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Giovanna Casili
- *Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | | | | | | | - Paterniti Irene
- *Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Salvatore Cuzzocrea
- *Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Emanuela Esposito
- *Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Michela Campolo
- *Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| |
Collapse
|
22
|
Laselva O, Allegretta C, Di Gioia S, Avolio C, Conese M. Anti-Inflammatory and Anti-Oxidant Effect of Dimethyl Fumarate in Cystic Fibrosis Bronchial Epithelial Cells. Cells 2021; 10:cells10082132. [PMID: 34440900 PMCID: PMC8391758 DOI: 10.3390/cells10082132] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 08/08/2021] [Accepted: 08/16/2021] [Indexed: 11/16/2022] Open
Abstract
Cystic Fibrosis (CF) is caused by mutations on the CF transmembrane conductance regulator (CFTR) gene and is associated with chronic infection and inflammation. Recently, it has been demonstrated that LPS-induced CFTR dysfunction in airway epithelial cells is due to an early oxidative stress. Dimethyl fumarate (DMF) is an approved anti-inflammatory and anti-oxidant drug for auto-immune and inflammatory diseases, but its role in the CF has never been investigated. In this study, we examined the effect of DMF on CF-related cytokines expression, ROS measurements and CFTR channel function. We found that DMF reduced the inflammatory response to LPS stimulation in both CF and non-CF bronchial epithelial cells, both as co-treatment and therapy, and restored LPS-mediated decrease of Trikafta™-mediated CFTR function in CF cells bearing the most common mutation, c.1521_1523delCTT (F508del). DMF also inhibited the inflammatory response induced by IL-1β/H2O2 and IL-1β/TNFα, mimicking the inflammatory status of CF patients. Finally, we also demonstrated that DMF exhibited an anti-oxidant effect on CF cells after different inflammatory stimulations. Since DMF is an approved drug, it could be further investigated as a novel anti-inflammatory molecule to ameliorate lung inflammation in CF and improve the CFTR modulators efficacy.
Collapse
|
23
|
KYP-2047, an Inhibitor of Prolyl-Oligopeptidase, Reduces GlioBlastoma Proliferation through Angiogenesis and Apoptosis Modulation. Cancers (Basel) 2021; 13:cancers13143444. [PMID: 34298658 PMCID: PMC8306782 DOI: 10.3390/cancers13143444] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/06/2021] [Accepted: 07/07/2021] [Indexed: 12/02/2022] Open
Abstract
Simple Summary Glioblastoma (GB) is the most aggressive brain tumor characterized by necrosis, excessive proliferation, and invasiveness. Despite relevant progress in conventional treatments, the survival rate for patients with GB remains low. The present study investigated the potential effect of KYP-2047, an inhibitor of the prolyl-oligopeptidase (POP or PREP), in an in vivo U87-xenograft model and in an in vitro study on human GB cells. This study demonstrated the abilities of KYP-2047 to counteract and reduce GB progression through angiogenesis and apoptosis modulation. Abstract Glioblastoma (GB) is the most aggressive tumor of the central nervous system (CNS), characterized by excessive proliferation, necrosis and invasiveness. The survival rate for patients with GB still remains low. Angiogenesis and apoptosis play a key role in the development of GB. Thus, the modulation of angiogenesis and apoptosis processes represent a possible strategy to counteract GB progression. This study aimed to investigate the potential effect of KYP-2047, an inhibitor of the prolyl-oligopeptidase (POP), known to modulate angiogenesis, in an in vivo U87-xenograft model and in an in vitro study on human GB cells. Our results showed that KYP-2047 at doses of 2.5 mg/kg and 5 mg/kg was able to reduce tumor burden in the xenograft-model. Moreover, KYP-2047 significantly reduced vascular endothelial-growth-factor (VEGF), angiopoietins (Ang) and endothelial-nitric-oxide synthase (eNOS) expression. In vitro study revealed that KYP-2047 at different concentrations reduced GB cells’ viability. Additionally, KYP-2047 at the concentrations of 50 µM and 100 µM was able to increase the pro-apoptotic protein Bax, p53 and caspase-3 expression whereas Bcl-2 expression was reduced. Thus, KYP-2047 could represent a potential therapeutic treatment to counteract or reduce GB progression, thanks its abilities to modulate angiogenesis and apoptosis pathways.
Collapse
|
24
|
Villavicencio Tejo F, Quintanilla RA. Contribution of the Nrf2 Pathway on Oxidative Damage and Mitochondrial Failure in Parkinson and Alzheimer's Disease. Antioxidants (Basel) 2021; 10:1069. [PMID: 34356302 PMCID: PMC8301100 DOI: 10.3390/antiox10071069] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 06/13/2021] [Accepted: 06/16/2021] [Indexed: 12/17/2022] Open
Abstract
The increase in human life expectancy has become a challenge to reduce the deleterious consequences of aging. Nowadays, an increasing number of the population suffer from age-associated neurodegenerative diseases including Parkinson's disease (PD) and Alzheimer's disease (AD). These disorders present different signs of neurodegeneration such as mitochondrial dysfunction, inflammation, and oxidative stress. Accumulative evidence suggests that the transcriptional factor nuclear factor (erythroid-derived 2)-like 2 (Nrf2) plays a vital defensive role orchestrating the antioxidant response in the brain. Nrf2 activation promotes the expression of several antioxidant enzymes that exert cytoprotective effects against oxidative damage and mitochondrial impairment. In this context, several studies have proposed a role of Nrf2 in the pathogenesis of PD and AD. Thus, we consider it important to summarize the ongoing literature related to the effects of the Nrf2 pathway in the context of these diseases. Therefore, in this review, we discuss the mechanisms involved in Nrf2 activity and its connection with mitochondria, energy supply, and antioxidant response in the brain. Furthermore, we will lead our discussion to identify the participation of the Nrf2 pathway in mitochondrial impairment and neurodegeneration present in PD and AD. Finally, we will discuss the therapeutic effects that the Nrf2 pathway activation could have on the cognitive impairment, neurodegeneration, and mitochondrial failure present in PD and AD.
Collapse
Affiliation(s)
| | - Rodrigo A Quintanilla
- Laboratory of Neurodegenerative Diseases, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago 8910060, Chile;
| |
Collapse
|
25
|
Paterniti I, Scuderi SA, Casili G, Lanza M, Mare M, Giuffrida R, Colarossi C, Portelli M, Cuzzocrea S, Esposito E. Poly (ADP-Ribose) Polymerase Inhibitor, ABT888, Improved Cisplatin Effect in Human Oral Cell Carcinoma. Biomedicines 2021; 9:biomedicines9070771. [PMID: 34356835 PMCID: PMC8301366 DOI: 10.3390/biomedicines9070771] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 06/16/2021] [Accepted: 06/28/2021] [Indexed: 11/16/2022] Open
Abstract
Cisplatin is one of the chemotherapeutic drugs used for the management of oral carcinoma, in which combined therapies are estimated to exert superior therapeutic efficacy compared with monotherapy. It is known that poly(ADP-ribosyl)ation is implicated in a multiplicity of cellular activities, such as DNA repair and cell death. Based on these, PARP inhibitors are used for the treatment of cancers; however, the capacity of PARP inhibitors associated to anti-cancer drugs have not been completely assessed in oral carcinoma. Here, we evaluated the effects of PARPi veliparib (ABT888) in combination with cisplatin on the survival of three human oral cancer cell lines HSC-2, Ca9-22 and CAL27 and we observed the effects of ABT888 alone or in combination with cisplatin on apoptosis and DNA damage repair mechanism. The results obtained showed that ABT888 induces a cytotoxicity effect on cell viability increasing the apoptotic pathway as well as DNA strand break; moreover, our results displayed the effects with cisplatin in a dose-dependent manner. Therefore, our results indicate PARP inhibitors as adjuvants for therapeutic strategy of oral cancer.
Collapse
Affiliation(s)
- Irene Paterniti
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31-98166 Messina, ME, Italy; (I.P.); (S.A.S.); (G.C.); (M.L.); (S.C.)
| | - Sarah Adriana Scuderi
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31-98166 Messina, ME, Italy; (I.P.); (S.A.S.); (G.C.); (M.L.); (S.C.)
| | - Giovanna Casili
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31-98166 Messina, ME, Italy; (I.P.); (S.A.S.); (G.C.); (M.L.); (S.C.)
| | - Marika Lanza
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31-98166 Messina, ME, Italy; (I.P.); (S.A.S.); (G.C.); (M.L.); (S.C.)
| | - Marzia Mare
- Istituto Oncologico del Mediterraneo, via Penninazzo 7, 95029 Viagrande, CT, Italy; (M.M.); (C.C.)
- IOM Ricerca Srl, via Penninazzo 11, 95029 Viagrande, CT, Italy;
| | | | - Cristina Colarossi
- Istituto Oncologico del Mediterraneo, via Penninazzo 7, 95029 Viagrande, CT, Italy; (M.M.); (C.C.)
| | - Marco Portelli
- Department of Biomedical and Dental Science, Morphological and Functional Images, University of Messina, via Consolare Valeria, 98125 Messina, ME, Italy;
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31-98166 Messina, ME, Italy; (I.P.); (S.A.S.); (G.C.); (M.L.); (S.C.)
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 31-98166 Messina, ME, Italy; (I.P.); (S.A.S.); (G.C.); (M.L.); (S.C.)
- Correspondence: ; Tel.: +39-090-676-5208
| |
Collapse
|
26
|
|
27
|
Timpani CA, Rybalka E. Calming the (Cytokine) Storm: Dimethyl Fumarate as a Therapeutic Candidate for COVID-19. Pharmaceuticals (Basel) 2020; 14:15. [PMID: 33375288 PMCID: PMC7824470 DOI: 10.3390/ph14010015] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 12/23/2020] [Accepted: 12/23/2020] [Indexed: 12/15/2022] Open
Abstract
COVID-19 has rapidly spread worldwide and incidences of hospitalisation from respiratory distress are significant. While a vaccine is in the pipeline, there is urgency for therapeutic options to address the immune dysregulation, hyperinflammation and oxidative stress that can lead to death. Given the shared pathogenesis of severe cases of COVID-19 with aspects of multiple sclerosis and psoriasis, we propose dimethyl fumarate as a viable treatment option. Currently approved for multiple sclerosis and psoriasis, dimethyl fumarate is an immunomodulatory, anti-inflammatory and anti-oxidative drug that could be rapidly implemented into the clinic to calm the cytokine storm which drives severe COVID-19.
Collapse
Affiliation(s)
- Cara A. Timpani
- Institute for Health and Sport, Victoria University, Melbourne, VIC 8001, Australia;
- Australian Institute for Musculoskeletal Science, St Albans, VIC 3021, Australia
| | - Emma Rybalka
- Institute for Health and Sport, Victoria University, Melbourne, VIC 8001, Australia;
- Australian Institute for Musculoskeletal Science, St Albans, VIC 3021, Australia
| |
Collapse
|
28
|
Campolo M, Lanza M, Casili G, Paterniti I, Filippone A, Caffo M, Cardali SM, Puliafito I, Colarossi C, Raciti G, Cuzzocrea S, Esposito E. TAK1 Inhibitor Enhances the Therapeutic Treatment for Glioblastoma. Cancers (Basel) 2020; 13:cancers13010041. [PMID: 33375627 PMCID: PMC7794959 DOI: 10.3390/cancers13010041] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 12/18/2020] [Accepted: 12/22/2020] [Indexed: 11/16/2022] Open
Abstract
Glioblastoma (GBM) is a brain tumor characterized by poor therapeutic response and overall survival. Despite relevant progress in conventional treatments represented by the clinical use of temozolomide (TMZ), a combination of approaches might be a possible future direction for treating GBM. Transforming growth factor-beta-activated kinase-1 (TAK1) is an essential component in genotoxic stresses-induced NF-κB-activation and mitogen-activated protein kinase (MAPK)-pathways; however, the role of TAK1 in GBM-chemoresistance remains unknown. This study aimed to verify, in GBM human cell lines, in an in vivo U87-xenograft model and in TMZ-treated-patients, the effect of TAK1 inhibition on the sensitivity of GBM cells to chemotherapy. In vitro model, using GBM cell lines, showed that 5Z-7-oxozeaenol augmented the cytotoxic effects of TMZ, blocking TMZ-induced NF-κB-activation, reducing DNA-damage and enhancing TMZ-induced apoptosis in GMB cell lines. We showed a reduction in tumor burden as well as tumor volume in the xenograft model following the treatment with 5Z-7-oxozaenol associated with TMZ. Our results showed a significant up-regulation in TAK1, p-p38, p-JNK and NF-κB in glioblastoma TMZ-treated-patients and denoted the role of 5Z-7-oxozeaenol in increasing the sensitivity of GBM cells to chemotherapy, proving to be an effective coadjuvant to current GBM chemotherapeutic regimens, suggesting a new option for therapeutic treatment of GBM.
Collapse
Affiliation(s)
- Michela Campolo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98122 Messina, Italy; (M.C.); (M.L.); (G.C.); (I.P.); (A.F.); (S.C.)
| | - Marika Lanza
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98122 Messina, Italy; (M.C.); (M.L.); (G.C.); (I.P.); (A.F.); (S.C.)
| | - Giovanna Casili
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98122 Messina, Italy; (M.C.); (M.L.); (G.C.); (I.P.); (A.F.); (S.C.)
| | - Irene Paterniti
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98122 Messina, Italy; (M.C.); (M.L.); (G.C.); (I.P.); (A.F.); (S.C.)
| | - Alessia Filippone
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98122 Messina, Italy; (M.C.); (M.L.); (G.C.); (I.P.); (A.F.); (S.C.)
| | - Maria Caffo
- Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, Unit of Neurosurgery, University of Messina, 98122 Messina, Italy; (M.C.); (S.M.C.)
| | - Salvatore M. Cardali
- Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, Unit of Neurosurgery, University of Messina, 98122 Messina, Italy; (M.C.); (S.M.C.)
| | - Ivana Puliafito
- Istituto Oncologico del Mediterraneo, Via Penninazzo 7, 95029 Viagrande, Italy; (I.P.); (C.C.)
| | - Cristina Colarossi
- Istituto Oncologico del Mediterraneo, Via Penninazzo 7, 95029 Viagrande, Italy; (I.P.); (C.C.)
| | - Gabriele Raciti
- IOM Ricerca S.r.l., Via Penninazzo 11, 95029 Viagrande, Italy;
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98122 Messina, Italy; (M.C.); (M.L.); (G.C.); (I.P.); (A.F.); (S.C.)
- Department of Pharmacological and Physiological Sciences, Saint Louis University School of Medicine, Saint Louis, MO 63104, USA
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98122 Messina, Italy; (M.C.); (M.L.); (G.C.); (I.P.); (A.F.); (S.C.)
- Correspondence: ; Tel.: +39-090-6765-208
| |
Collapse
|
29
|
Casili G, Lanza M, Campolo M, Siracusa R, Paterniti I, Ardizzone A, Scuderi SA, Cuzzocrea S, Esposito E. Synergic Therapeutic Potential of PEA-Um Treatment and NAAA Enzyme Silencing In the Management of Neuroinflammation. Int J Mol Sci 2020; 21:ijms21207486. [PMID: 33050589 PMCID: PMC7589809 DOI: 10.3390/ijms21207486] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/09/2020] [Accepted: 10/09/2020] [Indexed: 12/11/2022] Open
Abstract
Inflammation is a key element in the pathobiology of neurodegenerative diseases and sees the involvement of different neuronal and non-neuronal cells as players able to respond to inflammatory signals of immune origin. Palmitoylethanolamide (PEA) is an endogenous potent anti-inflammatory agent, in which activity is regulated by N-acylethanolamine acid amidase (NAAA), that hydrolyzes saturated or monounsaturated fatty acid ethanolamides, such as PEA. In this research, an in vitro study was performed on different neuronal (SH-SY5Y) and non-neuronal cell lines (C6, BV-2, and Mo3.13) subjected to NAAA enzyme silencing and treated with PEA ultra-micronized (PEA-um) (1, 3, and 10 μM) to increase the amount of endogenous PEA available for counteract neuroinflammation provoked by stimulation with lipopolysaccharide (LPS) (1 μg/mL) and interferon gamma (INF-γ )(100 U/mL). Cell viability was performed by MTT (3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide) staining, suggesting a protective effect of PEA-um (3 and 10 μM) on all cell lines studied. Western Blot analysis for inflammatory markers (Inducible nitric oxide synthase (iNOS) and cyclooxygenase 2 (COX-2)) was carried out in control and NAAA-silenced cells, highlighting how the concomitant treatment of the neuronal and non-neuronal cells with PEA-um after NAAA genic downregulation is satisfactory to counteract neuroinflammation. These in vitro findings support the protective role of endogenous PEA availability in the neuronal field, bringing interesting information for a translational point of view.
Collapse
Affiliation(s)
- Giovanna Casili
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98165 Messina, Italy; (G.C.); (M.L.); (M.C.); (R.S.); (I.P.); (A.A.); (S.A.S.); (E.E.)
| | - Marika Lanza
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98165 Messina, Italy; (G.C.); (M.L.); (M.C.); (R.S.); (I.P.); (A.A.); (S.A.S.); (E.E.)
| | - Michela Campolo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98165 Messina, Italy; (G.C.); (M.L.); (M.C.); (R.S.); (I.P.); (A.A.); (S.A.S.); (E.E.)
| | - Rosalba Siracusa
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98165 Messina, Italy; (G.C.); (M.L.); (M.C.); (R.S.); (I.P.); (A.A.); (S.A.S.); (E.E.)
| | - Irene Paterniti
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98165 Messina, Italy; (G.C.); (M.L.); (M.C.); (R.S.); (I.P.); (A.A.); (S.A.S.); (E.E.)
| | - Alessio Ardizzone
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98165 Messina, Italy; (G.C.); (M.L.); (M.C.); (R.S.); (I.P.); (A.A.); (S.A.S.); (E.E.)
| | - Sarah Adriana Scuderi
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98165 Messina, Italy; (G.C.); (M.L.); (M.C.); (R.S.); (I.P.); (A.A.); (S.A.S.); (E.E.)
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98165 Messina, Italy; (G.C.); (M.L.); (M.C.); (R.S.); (I.P.); (A.A.); (S.A.S.); (E.E.)
- Department of Pharmacological and Physiological Science, Saint Louis University, Saint Louis, MO 63103, USA
- Correspondence: ; Tel.: +39-090-6765208
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98165 Messina, Italy; (G.C.); (M.L.); (M.C.); (R.S.); (I.P.); (A.A.); (S.A.S.); (E.E.)
| |
Collapse
|
30
|
De Nuccio C, Bernardo A, Troiano C, Brignone MS, Falchi M, Greco A, Rosini M, Basagni F, Lanni C, Serafini MM, Minghetti L, Visentin S. NRF2 and PPAR-γ Pathways in Oligodendrocyte Progenitors: Focus on ROS Protection, Mitochondrial Biogenesis and Promotion of Cell Differentiation. Int J Mol Sci 2020; 21:E7216. [PMID: 33003644 PMCID: PMC7583077 DOI: 10.3390/ijms21197216] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/18/2020] [Accepted: 09/26/2020] [Indexed: 12/15/2022] Open
Abstract
An adequate protection from oxidative and inflammatory reactions, together with the promotion of oligodendrocyte progenitor (OP) differentiation, is needed to recover from myelin damage in demyelinating diseases. Mitochondria are targets of inflammatory and oxidative insults and are essential in oligodendrocyte differentiation. It is known that nuclear factor-erythroid 2-related factor/antioxidant responsive element (NRF2/ARE) and peroxisome proliferator-activated receptor gamma/PPAR-γ response element (PPAR-γ/PPRE) pathways control inflammation and overcome mitochondrial impairment. In this study, we analyzed the effects of activators of these pathways on mitochondrial features, protection from inflammatory/mitochondrial insults and cell differentiation in OP cultures, to depict the specificities and similarities of their actions. We used dimethyl-fumarate (DMF) and pioglitazone (pio) as agents activating NRF2 and PPAR-γ, respectively, and two synthetic hybrids acting differently on the NRF2/ARE pathway. Only DMF and compound 1 caused early effects on the mitochondria. Both DMF and pio induced mitochondrial biogenesis but different antioxidant repertoires. Moreover, pio induced OP differentiation more efficiently than DMF. Finally, DMF, pio and compound 1 protected from tumor necrosis factor-alpha (TNF-α) insult, with pio showing faster kinetics of action and compound 1 a higher activity than DMF. In conclusion, NRF2 and PPAR-γ by inducing partially overlapping pathways accomplish complementary functions aimed at the preservation of mitochondrial function, the defense against oxidative stress and the promotion of OP differentiation.
Collapse
Affiliation(s)
- Chiara De Nuccio
- Research Coordination and Support Service, Istituto Superiore di Sanità, 00161 Rome, Italy; (C.D.N.); (L.M.)
| | - Antonietta Bernardo
- National Center for Research and Preclinical and Clinical Evaluation of Drugs, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.B.); (A.G.)
| | - Carmen Troiano
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | | | - Mario Falchi
- National Research Center on HIV/AIDS, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Anita Greco
- National Center for Research and Preclinical and Clinical Evaluation of Drugs, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.B.); (A.G.)
| | - Michela Rosini
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy; (M.R.); (F.B.)
| | - Filippo Basagni
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy; (M.R.); (F.B.)
| | - Cristina Lanni
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (C.L.); (M.M.S.)
| | | | - Luisa Minghetti
- Research Coordination and Support Service, Istituto Superiore di Sanità, 00161 Rome, Italy; (C.D.N.); (L.M.)
| | - Sergio Visentin
- National Center for Research and Preclinical and Clinical Evaluation of Drugs, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.B.); (A.G.)
| |
Collapse
|
31
|
Ikram M, Park TJ, Ali T, Kim MO. Antioxidant and Neuroprotective Effects of Caffeine against Alzheimer's and Parkinson's Disease: Insight into the Role of Nrf-2 and A2AR Signaling. Antioxidants (Basel) 2020; 9:antiox9090902. [PMID: 32971922 PMCID: PMC7554764 DOI: 10.3390/antiox9090902] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 09/20/2020] [Accepted: 09/21/2020] [Indexed: 12/15/2022] Open
Abstract
This paper reviews the results of studies conducted on the role of caffeine in the management of different neurological disorders, such as Parkinson's disease (PD) and Alzheimer's disease (AD). To highlight the potential role of caffeine in managing different neurodegenerative diseases, we identified studies by searching PubMed, Web of Science, and Google Scholar by scrutinizing the lists of pertinent publications. According to the collected overall findings, caffeine may reduce the elevated oxidative stress; inhibit the activation of adenosine A2A, thereby regulating the accumulation of Aβ; reduce the hyperphosphorylation of tau; and reduce the accumulation of misfolded proteins, such as α-synuclein, in Alzheimer's and Parkinson's diseases. The studies have suggested that caffeine has promising protective effects against different neurodegenerative diseases and that these effects may be used to tackle the neurological diseases and/or their consequences. Here, we review the ongoing research on the role of caffeine in the management of different neurodegenerative disorders, focusing on AD and PD. The current findings suggest that caffeine produces potent antioxidant, inflammatory, and anti-apoptotic effects against different models of neurodegenerative disease, including AD, PD, and other neurodegenerative disorders. Caffeine has shown strong antagonistic effects against the adenosine A2A receptor, which is a microglial receptor, and strong agonistic effects against nuclear-related factor-2 (Nrf-2), thereby regulating the cellular homeostasis at the brain by reducing oxidative stress, neuroinflammation, regulating the accumulation of α-synuclein in PD and tau hyperphosphorylation, amyloidogenesis, and synaptic deficits in AD, which are the cardinal features of these neurodegenerative diseases.
Collapse
Affiliation(s)
- Muhammad Ikram
- Division of Life Science and Applied Life Science (BK21 plus), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea; (M.I.); (T.A.)
| | - Tae Ju Park
- Paul O’Gorman Leukaemia Research Centre, Institute of Cancer Sciences, University of Glasgow, Glasgow 0747 657 5394, UK;
| | - Tahir Ali
- Division of Life Science and Applied Life Science (BK21 plus), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea; (M.I.); (T.A.)
| | - Myeong Ok Kim
- Division of Life Science and Applied Life Science (BK21 plus), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea; (M.I.); (T.A.)
- Correspondence: ; Tel.: +82-55-772-1345; Fax: +82-55-772-2656
| |
Collapse
|
32
|
Bordoni L, Gabbianelli R. Mitochondrial DNA and Neurodegeneration: Any Role for Dietary Antioxidants? Antioxidants (Basel) 2020; 9:E764. [PMID: 32824558 PMCID: PMC7466149 DOI: 10.3390/antiox9080764] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/07/2020] [Accepted: 08/14/2020] [Indexed: 02/07/2023] Open
Abstract
The maintenance of the mitochondrial function is essential in preventing and counteracting neurodegeneration. In particular, mitochondria of neuronal cells play a pivotal role in sustaining the high energetic metabolism of these cells and are especially prone to oxidative damage. Since overproduction of reactive oxygen species (ROS) is involved in the pathogenesis of neurodegeneration, dietary antioxidants have been suggested to counteract the detrimental effects of ROS and to preserve the mitochondrial function, thus slowing the progression and limiting the extent of neuronal cell loss in neurodegenerative disorders. In addition to their role in the redox-system homeostasis, mitochondria are unique organelles in that they contain their own genome (mtDNA), which acts at the interface between environmental exposures and the molecular triggers of neurodegeneration. Indeed, it has been demonstrated that mtDNA (including both genetics and, from recent evidence, epigenetics) might play relevant roles in modulating the risk for neurodegenerative disorders. This mini-review describes the link between the mitochondrial genome and cellular oxidative status, with a particular focus on neurodegeneration; moreover, it provides an overview on potential beneficial effects of antioxidants in preserving mitochondrial functions through the protection of mtDNA.
Collapse
Affiliation(s)
- Laura Bordoni
- Unit of Molecular Biology, School of Pharmacy, University of Camerino, 62032 Camerino, Italy;
| | | |
Collapse
|
33
|
Rosito M, Testi C, Parisi G, Cortese B, Baiocco P, Di Angelantonio S. Exploring the Use of Dimethyl Fumarate as Microglia Modulator for Neurodegenerative Diseases Treatment. Antioxidants (Basel) 2020; 9:antiox9080700. [PMID: 32756501 PMCID: PMC7465338 DOI: 10.3390/antiox9080700] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/01/2020] [Accepted: 08/02/2020] [Indexed: 12/12/2022] Open
Abstract
The maintenance of redox homeostasis in the brain is critical for the prevention of the development of neurodegenerative diseases. Drugs acting on brain redox balance can be promising for the treatment of neurodegeneration. For more than four decades, dimethyl fumarate (DMF) and other derivatives of fumaric acid ester compounds have been shown to mitigate a number of pathological mechanisms associated with psoriasis and relapsing forms of multiple sclerosis (MS). Recently, DMF has been shown to exert a neuroprotective effect on the central nervous system (CNS), possibly through the modulation of microglia detrimental actions, observed also in multiple brain injuries. In addition to the hypothesis that DMF is linked to the activation of NRF2 and NF-kB transcription factors, the neuroprotective action of DMF may be mediated by the activation of the glutathione (GSH) antioxidant pathway and the regulation of brain iron homeostasis. This review will focus on the role of DMF as an antioxidant modulator in microglia processes and on its mechanisms of action in the modulation of different pathways to attenuate neurodegenerative disease progression.
Collapse
Affiliation(s)
- Maria Rosito
- Center for Life Nanoscience, Istituto Italiano di Tecnologia, 00161 Rome, Italy; (M.R.); (C.T.); (G.P.)
| | - Claudia Testi
- Center for Life Nanoscience, Istituto Italiano di Tecnologia, 00161 Rome, Italy; (M.R.); (C.T.); (G.P.)
| | - Giacomo Parisi
- Center for Life Nanoscience, Istituto Italiano di Tecnologia, 00161 Rome, Italy; (M.R.); (C.T.); (G.P.)
| | - Barbara Cortese
- Nanotechnology Institute, CNR-Nanotechnology Institute, Sapienza University, 00185 Rome, Italy;
| | - Paola Baiocco
- Department of Biochemical Sciences “A. Rossi Fanelli” Sapienza University, 00185 Rome, Italy
- Correspondence: (P.B.); (S.D.A.)
| | - Silvia Di Angelantonio
- Center for Life Nanoscience, Istituto Italiano di Tecnologia, 00161 Rome, Italy; (M.R.); (C.T.); (G.P.)
- Department of Physiology and Pharmacology, Sapienza University, 00185 Rome, Italy
- Correspondence: (P.B.); (S.D.A.)
| |
Collapse
|
34
|
POSCAbilities: The Application of the Prion Organotypic Slice Culture Assay to Neurodegenerative Disease Research. Biomolecules 2020; 10:biom10071079. [PMID: 32698402 PMCID: PMC7407827 DOI: 10.3390/biom10071079] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/06/2020] [Accepted: 07/16/2020] [Indexed: 12/14/2022] Open
Abstract
Prion diseases are fatal, transmissible neurodegenerative disorders whose pathogenesis is driven by the misfolding, self-templating and cell-to-cell spread of the prion protein. Other neurodegenerative diseases such as Alzheimer’s disease, Parkinson’s disease, amyotrophic lateral sclerosis and Huntington’s disease, share some of these prion-like features, with different aggregation-prone proteins. Consequently, researchers have begun to apply prion-specific techniques, like the prion organotypic slice culture assay (POSCA), to these disorders. In this review we explore the ways in which the prion phenomenon has been used in organotypic cultures to study neurodegenerative diseases from the perspective of protein aggregation and spreading, strain propagation, the role of glia in pathogenesis, and efficacy of drug treatments. We also present an overview of the advantages and disadvantages of this culture system compared to in vivo and in vitro models and provide suggestions for new directions.
Collapse
|
35
|
Scuderi SA, Ardizzone A, Paterniti I, Esposito E, Campolo M. Antioxidant and Anti-inflammatory Effect of Nrf2 Inducer Dimethyl Fumarate in Neurodegenerative Diseases. Antioxidants (Basel) 2020; 9:antiox9070630. [PMID: 32708926 PMCID: PMC7402174 DOI: 10.3390/antiox9070630] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/13/2020] [Accepted: 07/14/2020] [Indexed: 12/13/2022] Open
Abstract
Neurodegenerative diseases (NDs) represents debilitating conditions characterized by degeneration of neuronal cells in specific brain areas, causing disability and death in patients. In the pathophysiology of NDs, oxidative stress, apoptosis and neuroinflammation have a key role, as demonstrated by in vivo and in vitro models. Therefore, the use of molecules with antioxidant and anti-inflammatory activities represents a possible strategy for the treatment of NDs. Many studies demonstrated the beneficial effects of fumaric acid esters (FAEs) to counteract neuroinflammation and oxidative stress. Among these molecules, dimethyl fumarate (DMF) showed a valid therapeutic approach to slow down neurodegeneration and relieve symptoms in patients with NDs. DMF is a methyl ester of fumaric acid and acts as modulator of the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway as well as nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) translocation. Therefore, this review aims to examine the potential beneficial effects of DMF to counteract oxidative stress and inflammation in patients with NDs.
Collapse
|
36
|
Cores Á, Piquero M, Villacampa M, León R, Menéndez JC. NRF2 Regulation Processes as a Source of Potential Drug Targets against Neurodegenerative Diseases. Biomolecules 2020; 10:E904. [PMID: 32545924 PMCID: PMC7356958 DOI: 10.3390/biom10060904] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/03/2020] [Accepted: 06/09/2020] [Indexed: 12/12/2022] Open
Abstract
NRF2 acts by controlling gene expression, being the master regulator of the Phase II antioxidant response, and also being key to the control of neuroinflammation. NRF2 activity is regulated at several levels, including protein degradation by the proteasome, transcription, and post-transcription. The purpose of this review is to offer a concise and critical overview of the main mechanisms of NRF2 regulation and their actual or potential use as targets for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Ángel Cores
- Unidad de Química Orgánica y Farmacéutica, Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense, 28040 Madrid, Spain; (Á.C.); (M.P.); (M.V.)
| | - Marta Piquero
- Unidad de Química Orgánica y Farmacéutica, Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense, 28040 Madrid, Spain; (Á.C.); (M.P.); (M.V.)
| | - Mercedes Villacampa
- Unidad de Química Orgánica y Farmacéutica, Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense, 28040 Madrid, Spain; (Á.C.); (M.P.); (M.V.)
| | - Rafael León
- Instituto Teófilo Hernando y Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid, Spain;
- Instituto de Investigación Sanitaria, Servicio de Farmacología Clínica, Hospital Universitario de la Princesa, 28006 Madrid, Spain
| | - J. Carlos Menéndez
- Unidad de Química Orgánica y Farmacéutica, Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense, 28040 Madrid, Spain; (Á.C.); (M.P.); (M.V.)
| |
Collapse
|
37
|
Campolo M, Casili G, Paterniti I, Filippone A, Lanza M, Ardizzone A, Scuderi SA, Cuzzocrea S, Esposito E. Effect of a Product Containing Xyloglucan and Pea Protein on a Murine Model of Atopic Dermatitis. Int J Mol Sci 2020; 21:ijms21103596. [PMID: 32438777 PMCID: PMC7279434 DOI: 10.3390/ijms21103596] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/23/2020] [Accepted: 05/17/2020] [Indexed: 12/12/2022] Open
Abstract
Atopic dermatitis (AD) is a chronic inflammatory disease of the skin, characterized by dryness and more or less severe itching. The etiology of AD is complex and has not been fully clarified, involving genetic susceptibility, immunological abnormalities, epidermal barrier dysfunction, and environmental factors. Xyloglucan (XG) and pea protein (PP) are two compounds of natural origin characterized by the ability to create a physical barrier that protects mucosae membranes, reducing inflammation. The aim of the present study was to evaluate the potential beneficial effects of XG + PP in both a mouse model of AD and Staphylococcus aureus (S. aureus) infection- associated AD. Mice were topically treated with 200 μL of 0.5% oxazolone on the dorsal skin three times a week for AD induction. Mice received XG and PP by topical administration 1 h before oxazolone treatment. In S. aureus infection-associated AD, to induce a superficial superinfection of the skin, mice were also treated with 5 μL of 108 of a culture of S. aureus for 2 weeks; mice superinfected received XG and PP by topical administration 1 h before oxazolone + S. aureus. Four weeks later, the skin was removed for histological and biochemical analysis. Our results demonstrated the protective barrier effects of XG and PP characterized by a reduction in histological tissue changes, mastocyte degranulation, and tight junction permeability in the skin following oxazolone treatment. Moreover, XG + PP was able to preserve filaggrin expression, a hallmark of AD. Our data also support the effectiveness of XG + PP to reduce the damage by superinfection post AD induced by S. aureus. In conclusion, a future product containing XG and PP could be considered as a potentially interesting approach for the treatment of AD.
Collapse
|
38
|
Chen N, Wang J, He Y, Xu Y, Zhang Y, Gong Q, Yu C, Gao J. Trilobatin Protects Against Aβ 25-35-Induced Hippocampal HT22 Cells Apoptosis Through Mediating ROS/p38/Caspase 3-Dependent Pathway. Front Pharmacol 2020; 11:584. [PMID: 32508629 PMCID: PMC7248209 DOI: 10.3389/fphar.2020.00584] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 04/16/2020] [Indexed: 12/31/2022] Open
Abstract
Emerging evidence reveals that an aberrant accumulation of β-amyloid (Aβ) is the main reason of Alzheimer’s disease (AD) pathogenesis. Thus, inhibition of Aβ-induced neurotoxicity may be promising therapeutic tactics to mitigate AD onset and advance. The development of agent candidates by cultured neurons against Aβ-induced cytotoxicity is widely accepted to be an efficient strategy to explore the drug for AD patients. Previously, we have revealed that trilobatin (TLB), a small molecule monomer, derives from Lithocarpus polystachyus Rehd, possessed antioxidative activities on hydrogen peroxide-induced oxidative injury in PC12 cells. The present study was designed to investigate the effects and the underlying mechanism of TLB on Aβ-induced injury in hippocampal HT22 cells. The results demonstrated that TLB attenuated Aβ25–35-induced HT22 cell death, as evidenced by MTT assay and LDH release. Furthermore, TLB dramatically mitigated cell death after Aβ25–35 insulted via decreasing the intracellular and mitochondrial ROS overproduction and restoring antioxidant enzyme activities, as well as suppressing apoptosis. Of note, Aβ25–35 triggered increase in ratio of Bax/Bcl-2, activation of caspase-3, phosphorylation of tau, JNK, p38 MAPK, and decrease in Sirt3 expression, whereas TLB reversed these changes. Intriguingly, TLB could directly bind to p38, as evidenced by molecular docking and p38 inhibitor. Taken together, the results reveal that TLB effectively protects against Aβ25–35-induced neuronal cell death via activating ROS/p38/caspase 3-dependent pathway. Our findings afford evidence for the potential development of TLB to hinder neuronal death during AD.
Collapse
Affiliation(s)
- Nana Chen
- Department of Clinical Pharmacotherapeutics, School of Pharmacy, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, China
| | - Jiao Wang
- Department of Neurology, the Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yuqi He
- Department of Clinical Pharmacotherapeutics, School of Pharmacy, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Yingshu Xu
- Department of Clinical Pharmacotherapeutics, School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Yuchuan Zhang
- Department of Clinical Pharmacotherapeutics, School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Qihai Gong
- Department of Clinical Pharmacotherapeutics, School of Pharmacy, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Changyin Yu
- Department of Neurology, the Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jianmei Gao
- Department of Clinical Pharmacotherapeutics, School of Pharmacy, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| |
Collapse
|
39
|
Hennig P, Fenini G, Di Filippo M, Beer HD. Electrophiles Against (Skin) Diseases: More Than Nrf2. Biomolecules 2020; 10:E271. [PMID: 32053878 PMCID: PMC7072181 DOI: 10.3390/biom10020271] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 02/04/2020] [Accepted: 02/06/2020] [Indexed: 12/11/2022] Open
Abstract
The skin represents an indispensable barrier between the organism and the environment and is the first line of defense against exogenous insults. The transcription factor NRF2 is a central regulator of cytoprotection and stress resistance. NRF2 is activated in response to oxidative stress by reactive oxygen species (ROS) and electrophiles. These electrophiles oxidize specific cysteine residues of the NRF2 inhibitor KEAP1, leading to KEAP1 inactivation and, subsequently, NRF2 activation. As oxidative stress is associated with inflammation, the NRF2 pathway plays important roles in the pathogenesis of common inflammatory diseases and cancer in many tissues and organs, including the skin. The electrophile and NRF2 activator dimethyl fumarate (DMF) is an established and efficient drug for patients suffering from the common inflammatory skin disease psoriasis and the neuro-inflammatory disease multiple sclerosis (MS). In this review, we discuss possible molecular mechanisms underlying the therapeutic activity of DMF and other NRF2 activators. Recent evidence suggests that electrophiles not only activate NRF2, but also target other inflammation-associated pathways including the transcription factor NF-κB and the multi-protein complexes termed inflammasomes. Inflammasomes are central regulators of inflammation and are involved in many inflammatory conditions. Most importantly, the NRF2 and inflammasome pathways are connected at different levels, mainly antagonistically.
Collapse
Affiliation(s)
- Paulina Hennig
- Department of Dermatology, University Hospital of Zurich, Gloriastrasse 31, CH-8091 Zurich, Switzerland; (P.H.); (G.F.); (M.D.F.)
- Faculty of Medicine, University of Zurich, 8006 Zurich, Switzerland
| | - Gabriele Fenini
- Department of Dermatology, University Hospital of Zurich, Gloriastrasse 31, CH-8091 Zurich, Switzerland; (P.H.); (G.F.); (M.D.F.)
- Faculty of Medicine, University of Zurich, 8006 Zurich, Switzerland
| | - Michela Di Filippo
- Department of Dermatology, University Hospital of Zurich, Gloriastrasse 31, CH-8091 Zurich, Switzerland; (P.H.); (G.F.); (M.D.F.)
- Faculty of Medicine, University of Zurich, 8006 Zurich, Switzerland
| | - Hans-Dietmar Beer
- Department of Dermatology, University Hospital of Zurich, Gloriastrasse 31, CH-8091 Zurich, Switzerland; (P.H.); (G.F.); (M.D.F.)
- Faculty of Medicine, University of Zurich, 8006 Zurich, Switzerland
| |
Collapse
|
40
|
Serafini MM, Catanzaro M, Fagiani F, Simoni E, Caporaso R, Dacrema M, Romanoni I, Govoni S, Racchi M, Daglia M, Rosini M, Lanni C. Modulation of Keap1/Nrf2/ARE Signaling Pathway by Curcuma- and Garlic-Derived Hybrids. Front Pharmacol 2020; 10:1597. [PMID: 32047434 PMCID: PMC6997134 DOI: 10.3389/fphar.2019.01597] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 12/10/2019] [Indexed: 12/15/2022] Open
Abstract
Nrf2 is a basic leucine zipper transcription factor that binds to the promoter region of the antioxidant response element (ARE), inducing the coordinated up-regulation of antioxidant and detoxification genes. We recently synthesized a set of new molecules by combining the functional moieties of curcumin and diallyl sulfide, both known to induce the expression of antioxidant phase II enzymes by activating Nrf2 pathway. The aim of the study is to investigate the ability of such compounds to activate Keap1/Nrf2/ARE cytoprotective pathway, in comparison with two reference Nrf2-activators: curcumin and dimethyl fumarate, a drug approved for the treatment of relapsing-remitting multiple sclerosis. Furthermore, since Nrf2 pathway is known to be regulated also by epigenetic modifications, including key modifications in microRNA (miRNA) expression, the effects of the hybrids on the expression levels of selected miRNAs, associated with Nrf2 signaling pathway have also been investigated. The results show that compounds exert antioxidant effect by activating Nrf2 signaling pathway and inducing the ARE-regulated expression of its downstream target genes, such as HO-1 and NQO1, with two hybrids to a higher extent than curcumin. In addition, some molecules induce changes in the expression levels of miR-125b-5p, even if to a lesser extent than curcumin. However, no changes have been observed in the expression levels of mRNA coding for glutathione synthetase, suggesting that the modulation of this mRNA is not strictly under the control of miR-125b-5p, which could be influenced by other miRNAs.
Collapse
Affiliation(s)
- Melania Maria Serafini
- Department of Drug Sciences, University of Pavia, Pavia, Italy.,Scuola Universitaria Superiore IUSS, Pavia, Italy
| | | | - Francesca Fagiani
- Department of Drug Sciences, University of Pavia, Pavia, Italy.,Scuola Universitaria Superiore IUSS, Pavia, Italy
| | - Elena Simoni
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Roberta Caporaso
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Marco Dacrema
- Department of Pharmacy, University of Napoli Federico II, Naples, Italy
| | - Irene Romanoni
- Department of Drug Sciences, University of Pavia, Pavia, Italy
| | - Stefano Govoni
- Department of Drug Sciences, University of Pavia, Pavia, Italy
| | - Marco Racchi
- Department of Drug Sciences, University of Pavia, Pavia, Italy
| | - Maria Daglia
- Department of Pharmacy, University of Napoli Federico II, Naples, Italy
| | - Michela Rosini
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Cristina Lanni
- Department of Drug Sciences, University of Pavia, Pavia, Italy
| |
Collapse
|
41
|
Giannetto A, Esposito E, Lanza M, Oliva S, Riolo K, Di Pietro S, Abbate JM, Briguglio G, Cassata G, Cicero L, Macrì F. Protein Hydrolysates from Anchovy ( Engraulis encrasicolus) Waste: In Vitro and In Vivo Biological Activities. Mar Drugs 2020; 18:md18020086. [PMID: 32012959 PMCID: PMC7074155 DOI: 10.3390/md18020086] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 01/20/2020] [Accepted: 01/26/2020] [Indexed: 12/12/2022] Open
Abstract
Fish waste utilization to obtain protein hydrolysates has been demonstrated to be a useful strategy to face both environmental and economic impacts while obtaining high-value products with remarkable biological and nutritional properties. In the present study, protein hydrolysates obtained from anchovy Engraulis encrasicolus (APH) by-products were assessed for their potential biological activities in both in vitro and in vivo models. The treatment with APH exerted a significant protection against LPS-induced inflammation in RAW 264.7 cells, decreasing the protein expression of pro-inflammatory mediators (i.e., COX-2) and inhibiting the nuclear translocation of NF-κB through IκB-α. Moreover, APH modulated the expression of iNOS, MnSOD and HO-1, thus decreasing the severity of oxidative stress. The supplementation of APH in the diet of ApoE knockout mice down-regulated the proinflammatory cytokines (i.e., TNF-α, IL-1α, IL-1β, IL-6) in both aorta and heart tissues, and modulated the expression of oxidative stress-related genes (Cu/ZnSod, MnSod, Cat, Gpx and Ho), indicating that APH can exert a beneficial role, having anti-inflammatory and antioxidant activities. The nutritional properties of APH, together with their biological activities herein reported, highlight the possibility of obtaining bioactive molecules from fish waste and encourage their use as potential nutraceuticals in food and pharmaceutical industries in the next future.
Collapse
Affiliation(s)
- Alessia Giannetto
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences. University of Messina, Viale Ferdinando Stagno d’Alcontres, 31, 98166 Messina, Italy
- Correspondence: ; Tel.: +39-0906765810
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences. University of Messina, Viale Ferdinando Stagno d’Alcontres, 31, 98166 Messina, Italy
| | - Marika Lanza
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences. University of Messina, Viale Ferdinando Stagno d’Alcontres, 31, 98166 Messina, Italy
| | - Sabrina Oliva
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences. University of Messina, Viale Ferdinando Stagno d’Alcontres, 31, 98166 Messina, Italy
| | - Kristian Riolo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences. University of Messina, Viale Ferdinando Stagno d’Alcontres, 31, 98166 Messina, Italy
| | - Simona Di Pietro
- Department of Veterinary Sciences, University of Messina, Polo Universitario SS. Annunziata, 98168 Messina, Italy
| | | | | | - Giovanni Cassata
- Zooprophylactic Institute of Sicily “A. Mirri”, 90129 Palermo, Italy
| | - Luca Cicero
- Zooprophylactic Institute of Sicily “A. Mirri”, 90129 Palermo, Italy
| | - Francesco Macrì
- Department of Veterinary Sciences, University of Messina, Polo Universitario SS. Annunziata, 98168 Messina, Italy
| |
Collapse
|
42
|
Brandes MS, Gray NE. NRF2 as a Therapeutic Target in Neurodegenerative Diseases. ASN Neuro 2020; 12:1759091419899782. [PMID: 31964153 PMCID: PMC6977098 DOI: 10.1177/1759091419899782] [Citation(s) in RCA: 167] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 11/26/2019] [Accepted: 12/03/2019] [Indexed: 12/13/2022] Open
Abstract
Increased reactive oxygen species production and oxidative stress have been implicated in the pathogenesis of numerous neurodegenerative conditions including among others Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, Friedrich’s ataxia, multiple sclerosis, and stroke. The endogenous antioxidant response pathway protects cells from oxidative stress by increasing the expression of cytoprotective enzymes and is regulated by the transcription factor nuclear factor erythroid 2-related factor 2 (NRF2). In addition to regulating the expression of antioxidant genes, NRF2 has also been shown to exert anti-inflammatory effects and modulate both mitochondrial function and biogenesis. This is because mitochondrial dysfunction and neuroinflammation are features of many neurodegenerative diseases as well NRF2 has emerged as a promising therapeutic target. Here, we review evidence for a beneficial role of NRF2 in neurodegenerative conditions and the potential of specific NRF2 activators as therapeutic agents.
Collapse
Affiliation(s)
- Mikah S. Brandes
- Department of Neurology, Oregon Health and Science University, Portland, OR, USA
| | - Nora E. Gray
- Department of Neurology, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
43
|
Kerr F, Bjedov I, Sofola-Adesakin O. Molecular Mechanisms of Lithium Action: Switching the Light on Multiple Targets for Dementia Using Animal Models. Front Mol Neurosci 2018; 11:297. [PMID: 30210290 PMCID: PMC6121012 DOI: 10.3389/fnmol.2018.00297] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 08/03/2018] [Indexed: 12/12/2022] Open
Abstract
Lithium has long been used for the treatment of psychiatric disorders, due to its robust beneficial effect as a mood stabilizing drug. Lithium’s effectiveness for improving neurological function is therefore well-described, stimulating the investigation of its potential use in several neurodegenerative conditions including Alzheimer’s (AD), Parkinson’s (PD) and Huntington’s (HD) diseases. A narrow therapeutic window for these effects, however, has led to concerted efforts to understand the molecular mechanisms of lithium action in the brain, in order to develop more selective treatments that harness its neuroprotective potential whilst limiting contraindications. Animal models have proven pivotal in these studies, with lithium displaying advantageous effects on behavior across species, including worms (C. elegans), zebrafish (Danio rerio), fruit flies (Drosophila melanogaster) and rodents. Due to their susceptibility to genetic manipulation, functional genomic analyses in these model organisms have provided evidence for the main molecular determinants of lithium action, including inhibition of inositol monophosphatase (IMPA) and glycogen synthase kinase-3 (GSK-3). Accumulating pre-clinical evidence has indeed provided a basis for research into the therapeutic use of lithium for the treatment of dementia, an area of medical priority due to its increasing global impact and lack of disease-modifying drugs. Although lithium has been extensively described to prevent AD-associated amyloid and tau pathologies, this review article will focus on generic mechanisms by which lithium preserves neuronal function and improves memory in animal models of dementia. Of these, evidence from worms, flies and mice points to GSK-3 as the most robust mediator of lithium’s neuro-protective effect, but it’s interaction with downstream pathways, including Wnt/β-catenin, CREB/brain-derived neurotrophic factor (BDNF), nuclear factor (erythroid-derived 2)-like 2 (Nrf2) and toll-like receptor 4 (TLR4)/nuclear factor-κB (NFκB), have identified multiple targets for development of drugs which harness lithium’s neurogenic, cytoprotective, synaptic maintenance, anti-oxidant, anti-inflammatory and protein homeostasis properties, in addition to more potent and selective GSK-3 inhibitors. Lithium, therefore, has advantages as a multi-functional therapy to combat the complex molecular pathology of dementia. Animal studies will be vital, however, for comparative analyses to determine which of these defense mechanisms are most required to slow-down cognitive decline in dementia, and whether combination therapies can synergize systems to exploit lithium’s neuro-protective power while avoiding deleterious toxicity.
Collapse
Affiliation(s)
- Fiona Kerr
- Department of Life Sciences, School of Health & Life Sciences, Glasgow Caledonian University, Glasgow, United Kingdom
| | - Ivana Bjedov
- UCL Cancer Institute, University College London, London, United Kingdom
| | - Oyinkan Sofola-Adesakin
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, United Kingdom
| |
Collapse
|
44
|
Protective Effects of Sulforaphane on Cognitive Impairments and AD-like Lesions in Diabetic Mice are Associated with the Upregulation of Nrf2 Transcription Activity. Neuroscience 2018; 381:35-45. [DOI: 10.1016/j.neuroscience.2018.04.017] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Revised: 04/06/2018] [Accepted: 04/11/2018] [Indexed: 11/19/2022]
|