1
|
Basak B, Holzbaur ELF. Mitophagy in Neurons: Mechanisms Regulating Mitochondrial Turnover and Neuronal Homeostasis. J Mol Biol 2025:169161. [PMID: 40268233 DOI: 10.1016/j.jmb.2025.169161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 04/14/2025] [Accepted: 04/15/2025] [Indexed: 04/25/2025]
Abstract
Mitochondrial quality control is instrumental in regulating neuronal health and survival. The receptor-mediated clearance of damaged mitochondria by autophagy, known as mitophagy, plays a key role in controlling mitochondrial homeostasis. Mutations in genes that regulate mitophagy are causative for familial forms of neurological disorders including Parkinson's disease (PD) and Amyotrophic lateral sclerosis (ALS). PINK1/Parkin-dependent mitophagy is the best studied mitophagy pathway, while more recent work has brought to light additional mitochondrial quality control mechanisms that operate either in parallel to or independent of PINK1/Parkin mitophagy. Here, we discuss our current understanding of mitophagy mechanisms operating in neurons to govern mitochondrial homeostasis. We also summarize progress in our understanding of the links between mitophagic dysfunction and neurodegeneration, and highlight the potential for therapeutic interventions to maintain mitochondrial health and neuronal function.
Collapse
Affiliation(s)
- Bishal Basak
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Erika L F Holzbaur
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA.
| |
Collapse
|
2
|
Salgado KDCB, Nascimento RGF, Albuquerque ALS, Oliveira LAM, Pinto Coelho Nogueira KDO. Melatonin protects mouse hippocampal neurons from neurotoxicity induced by amyloid β-peptide 25-35. Brain Res 2025; 1859:149637. [PMID: 40222698 DOI: 10.1016/j.brainres.2025.149637] [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: 02/05/2025] [Revised: 04/07/2025] [Accepted: 04/08/2025] [Indexed: 04/15/2025]
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disorder and the leading cause of dementia in the elderly, as classified by the WHO. Its neuropathological hallmarks include the accumulation of amyloid-β (Aβ) plaques and intracellular tau tangles, which contribute to oxidative stress, mitochondrial dysfunction, lipid peroxidation, and neuronal death. Emerging evidence suggests that melatonin, a potent antioxidant produced by the pineal gland, plays a neuroprotective role in AD, yet its precise mechanisms remain underexplored. In this study, we utilized a physiologically relevant primary culture of hippocampal neurons to investigate melatonin's protective effects against toxicity induced by Aβ25-35. Our findings demonstrate that melatonin significantly enhances cellular metabolism and viability while reducing reactive oxygen species (ROS) levels and lipid peroxidation, thereby mitigating Aβ-induced neurotoxicity. These results provide mechanistic insights into melatonin's antioxidative and neuroprotective properties, reinforcing its potential as a therapeutic agent against oxidative stress in AD. This study underscores the promise of melatonin-based interventions in the development of novel antioxidant-targeted therapies for AD.
Collapse
Affiliation(s)
- Karen Del Carmen B Salgado
- Department of Biological Sciences, Laboratory of Neurobiology and Biomaterials (LNBio), Federal University of Ouro Preto, Ouro Preto, Brazil
| | - Rosiene G F Nascimento
- Department of Biological Sciences, Laboratory of Neurobiology and Biomaterials (LNBio), Federal University of Ouro Preto, Ouro Preto, Brazil
| | - Ana Luiza S Albuquerque
- Department of Biological Sciences, Laboratory of Neurobiology and Biomaterials (LNBio), Federal University of Ouro Preto, Ouro Preto, Brazil
| | - Laser A M Oliveira
- Department of Biological Sciences, Laboratory of Neurobiology and Biomaterials (LNBio), Federal University of Ouro Preto, Ouro Preto, Brazil
| | | |
Collapse
|
3
|
Sayehmiri F, Motamedi F, Batool Z, Naderi N, Shaerzadeh F, Zoghi A, Rezaei O, Khodagholi F, Pourbadie HG. Mitochondrial plasticity and synaptic plasticity crosstalk; in health and Alzheimer's disease. CNS Neurosci Ther 2024; 30:e14897. [PMID: 39097920 PMCID: PMC11298206 DOI: 10.1111/cns.14897] [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: 01/05/2024] [Revised: 06/19/2024] [Accepted: 07/18/2024] [Indexed: 08/06/2024] Open
Abstract
Synaptic plasticity is believed to underlie the cellular and molecular basis of memory formation. Mitochondria are one of the main organelles involved in metabolism and energy maintenance as plastic organelles that change morphologically and functionally in response to cellular needs and regulate synaptic function and plasticity through multiple mechanisms, including ATP generation, calcium homeostasis, and biogenesis. An increased neuronal activity enhances synaptic efficiency, during which mitochondria's spatial distribution and morphology change significantly. These organelles build up in the pre-and postsynaptic zones to produce ATP, which is necessary for several synaptic processes like neurotransmitter release and recycling. Mitochondria also regulate calcium homeostasis by buffering intracellular calcium, which ensures proper synaptic activity. Furthermore, mitochondria in the presynaptic terminal have distinct morphological properties compared to dendritic or postsynaptic mitochondria. This specialization enables precise control of synaptic activity and plasticity. Mitochondrial dysfunction has been linked to synaptic failure in many neurodegenerative disorders, like Alzheimer's disease (AD). In AD, malfunctioning mitochondria cause delays in synaptic vesicle release and recycling, ionic gradient imbalances, and mostly synaptic failure. This review emphasizes mitochondrial plasticity's contribution to synaptic function. It also explores the profound effect of mitochondrial malfunction on neurodegenerative disorders, focusing on AD, and provides an overview of how they sustain cellular health under normal conditions and how their malfunction contributes to neurodegenerative diseases, highlighting their potential as a therapeutic target for such conditions.
Collapse
Affiliation(s)
- Fatemeh Sayehmiri
- Neuroscience Research Center, Faculty of MedicineShahid Beheshti University of Medical SciencesTehranIran
| | - Fereshteh Motamedi
- Neuroscience Research Center, Faculty of MedicineShahid Beheshti University of Medical SciencesTehranIran
- Faculty of MedicineShahid Beheshti University of Medical SciencesTehranIran
| | - Zehra Batool
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological SciencesUniversity of KarachiKarachiPakistan
| | - Nima Naderi
- Department of Pharmacology and Toxicology, Faculty of PharmacyShahid Beheshti University of Medical SciencesTehranIran
| | | | - Anahita Zoghi
- Department of Neurology, Loghman Hakim HospitalShahid Beheshti University of Medical SciencesTehranIran
| | - Omidvar Rezaei
- Skull Base Research CenterLoghman Hakim Hospital, Shahid Beheshti University of Medical SciencesTehranIran
| | - Fariba Khodagholi
- Neuroscience Research Center, Faculty of MedicineShahid Beheshti University of Medical SciencesTehranIran
| | | |
Collapse
|
4
|
Ferdous KA, Jansen J, Amjad E, Pray E, Bloch R, Benoit A, Callahan M, Park HA. Mitochondrial protective potential of fucoxanthin in brain disorders. J Nutr Sci 2024; 13:e21. [PMID: 39776519 PMCID: PMC11704942 DOI: 10.1017/jns.2024.16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 04/15/2024] [Accepted: 05/09/2024] [Indexed: 01/11/2025] Open
Abstract
Mitochondrial dysfunction is a common feature of brain disorders. Mitochondria play a central role in oxidative phosphorylation; thus changes in energy metabolism in the brain have been reported in conditions such as Alzheimer's disease, Parkinson's disease, and stroke. In addition, mitochondria regulate cellular responses associated with neuronal damage such as the production of reactive oxygen species (ROS), opening of the mitochondrial permeability transition pore (mPTP), and apoptosis. Therefore, interventions that aim to protect mitochondria may be effective against brain disorders. Fucoxanthin is a marine carotenoid that has recently gained recognition for its neuroprotective properties. However, the cellular mechanisms of fucoxanthin in brain disorders, particularly its role in mitochondrial function, have not been thoroughly discussed. This review summarises the current literature on the effects of fucoxanthin on oxidative stress, neuroinflammation, and apoptosis using in vitro and in vivo models of brain disorders. We further present the potential mechanisms by which fucoxanthin protects mitochondria, with the objective of developing dietary interventions for a spectrum of brain disorders. Although the studies reviewed are predominantly preclinical studies, they provide important insights into understanding the cellular and molecular functions of fucoxanthin in the brain. Future studies investigating the mechanisms of action and the molecular targets of fucoxanthin are warranted to develop translational approaches to brain disorders.
Collapse
Affiliation(s)
- Khondoker Adeba Ferdous
- Department of Human Nutrition and Hospitality Management, College of Human Environmental Sciences, The University of Alabama, Tuscaloosa, AL, USA
| | - Joseph Jansen
- Department of Human Nutrition and Hospitality Management, College of Human Environmental Sciences, The University of Alabama, Tuscaloosa, AL, USA
| | - Emma Amjad
- Department of Human Nutrition and Hospitality Management, College of Human Environmental Sciences, The University of Alabama, Tuscaloosa, AL, USA
| | - Eliana Pray
- Department of Human Nutrition and Hospitality Management, College of Human Environmental Sciences, The University of Alabama, Tuscaloosa, AL, USA
| | - Rebecca Bloch
- Department of Human Nutrition and Hospitality Management, College of Human Environmental Sciences, The University of Alabama, Tuscaloosa, AL, USA
| | - Alex Benoit
- Department of Human Nutrition and Hospitality Management, College of Human Environmental Sciences, The University of Alabama, Tuscaloosa, AL, USA
| | - Meredith Callahan
- Department of Human Nutrition and Hospitality Management, College of Human Environmental Sciences, The University of Alabama, Tuscaloosa, AL, USA
| | - Han-A Park
- Department of Human Nutrition and Hospitality Management, College of Human Environmental Sciences, The University of Alabama, Tuscaloosa, AL, USA
| |
Collapse
|
5
|
Liu X, Guan Z, Liang S, Feng S, Zhou Y. Associations of cataract, cataract surgery with dementia risk: A systematic review and meta-analysis of 448,140 participants. Eur J Clin Invest 2024; 54:e14113. [PMID: 37874275 DOI: 10.1111/eci.14113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 10/06/2023] [Accepted: 10/10/2023] [Indexed: 10/25/2023]
Abstract
BACKGROUND Cataract is the leading cause of blindness around the world. Previous investigations have assessed the relationship between cataract, cataract surgery and dementia risk, but their results remain controversial. Herein, we conducted a meta-analysis to evaluate the associations between cataract, cataract surgery and the risk of dementia. METHODS We systemically screened the literature from three electronic databases PubMed, EMBASE and CENTRAL until April 2023. The data were collected by two independent researchers. The hazard ratios (HRs) or odds ratios (ORs) from eligible studies with 95% confidence intervals (CIs) were adjusted into the risk ratios (RRs), which were pooled using the random-effects model. RESULTS A total of nine studies with 448,140 participants reported the associations between cataract or cataract surgery and the risk of dementia were included in this meta-analysis. The outcomes of our pooled analysis indicated that cataract was associated with an increased risk of all-cause dementia (RR = 1.24, 95% CI, 1.14-1.35, p < .00001), Alzheimer's disease (RR = 1.22, 95% CI, 1.10-1.35, p = .0002) and vascular dementia (RR = 1.29, 95% CI, 1.01-1.66, p = .04). Cataract surgery is associated with a reduction of the dementia risk (RR = 0.74, 95% CI, 0.67-0.81, p < .00001). CONCLUSIONS Current evidence from the existing studies supports that cataract is associated with an increased risk of dementia, and cataract surgery may be instrumental in reducing the risk of dementia in patients with cataract.
Collapse
Affiliation(s)
- Xin Liu
- Department of Critical Care Medicine, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Zeyu Guan
- Faculty of Medicine, Macau University of Science and Technology, Macau SAR, China
| | - Shucheng Liang
- Faculty of Medicine, Macau University of Science and Technology, Macau SAR, China
| | - Shenghui Feng
- The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yue Zhou
- Department of Ophthalmology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
6
|
Wang L, Sang B, Zheng Z. The risk of dementia or cognitive impairment in patients with cataracts: a systematic review and meta-analysis. Aging Ment Health 2024; 28:11-22. [PMID: 37416949 DOI: 10.1080/13607863.2023.2226616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 06/04/2023] [Indexed: 07/08/2023]
Abstract
OBJECTIVES The aim of this study was to investigate whether cataract disease is associated with the risk of developing dementia or cognitive impairment. METHODS A systematic search of the literature in PubMed, the Extracts Database (Embase), the Cochrane Library and the Web of Science databases was performed from the inception data of each database until 1 September 2022. Sensitivity analyses were performed to assess the robustness and reliability of the overall findings. All extracted data were statistically analyzed using Stata software v.16.0. Publication bias was assessed using funnel plots and the Egger test. RESULTS There were 11 publications included in this study, which consisted of 489,211participants, spanning 10 countries from 2012 to 2022. Aggregation suggested that cataracts were associated with cognitive impairment (odds ratio [OR] = 1.32; 95% CI: 1.21-1.43; I 2 = 45.4.%; p = 0.000). The presence of cataracts is significantly associated with an increased risk of developing all-cause dementia (relative risk [RR] = 1.17; 95% CI: 1.08-1.26; I2 = 0.0%; p = 0.000). In subgroup analyses, having cataracts may increase the risk of Alzheimer's disease (hazard ratio [HR] = 1.28; 95% CI: 1.13-1.45; I2 = 0.0%; p = 0.000) and vascular dementia (HR = 1.35; 95% CI = 1.06-1.73; I2 = 0.0%, p = 0.015). The data from the Egger's test showed no significant evidence of publication bias. CONCLUSIONS Cataracts are associated with the risk of cognitive impairment and dementia, including Alzheimer's disease, and vascular dementia.
Collapse
Affiliation(s)
- Luping Wang
- Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Bowen Sang
- Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Zuyan Zheng
- Department of Acupuncture, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
7
|
Chavoshinezhad S, Beirami E, Izadpanah E, Feligioni M, Hassanzadeh K. Molecular mechanism and potential therapeutic targets of necroptosis and ferroptosis in Alzheimer's disease. Biomed Pharmacother 2023; 168:115656. [PMID: 37844354 DOI: 10.1016/j.biopha.2023.115656] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 10/01/2023] [Accepted: 10/04/2023] [Indexed: 10/18/2023] Open
Abstract
Alzheimer's disease (AD), a neurodegenerative condition, is defined by neurofibrillary tangles, amyloid plaques, and gradual cognitive decline. Regardless of the advances in understanding AD's pathogenesis and progression, its causes are still contested, and there are currently no efficient therapies for the illness. The post-mortem analyses revealed widespread neuronal loss in multiple brain regions in AD, evidenced by a decrease in neuronal density and correlated with the disease's progression and cognitive deterioration. AD's neurodegeneration is complicated, and different types of neuronal cell death, alone or in combination, play crucial roles in this process. Recently, the involvement of non-apoptotic programmed cell death in the neurodegenerative mechanisms of AD has received a lot of attention. Aberrant activation of necroptosis and ferroptosis, two newly discovered forms of regulated non-apoptotic cell death, is thought to contribute to neuronal cell death in AD. In this review, we first address the main features of necroptosis and ferroptosis, cellular signaling cascades, and the mechanisms involved in AD pathology. Then, we discuss the latest therapies targeting necroptosis and ferroptosis in AD animal/cell models and human research to provide vital information for AD treatment.
Collapse
Affiliation(s)
- Sara Chavoshinezhad
- Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| | - Elmira Beirami
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Esmael Izadpanah
- Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Marco Feligioni
- Laboratory of Neuronal Cell Signaling, EBRI Rita Levi-Montalcini Foundation, 00161 Rome, Italy; Department of Neurorehabilitation Sciences, Casa di Cura del Policlinico, 20144 Milan, Italy.
| | - Kambiz Hassanzadeh
- Robert Wood Johnson Medical School Institute for Neurological Therapeutics, and Department of Neurology, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA.
| |
Collapse
|
8
|
Atlante A, Valenti D. Mitochondrial Complex I and β-Amyloid Peptide Interplay in Alzheimer's Disease: A Critical Review of New and Old Little Regarded Findings. Int J Mol Sci 2023; 24:15951. [PMID: 37958934 PMCID: PMC10650435 DOI: 10.3390/ijms242115951] [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: 10/05/2023] [Revised: 10/30/2023] [Accepted: 10/31/2023] [Indexed: 11/15/2023] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder and the main cause of dementia which is characterized by a progressive cognitive decline that severely interferes with daily activities of personal life. At a pathological level, it is characterized by the accumulation of abnormal protein structures in the brain-β-amyloid (Aβ) plaques and Tau tangles-which interfere with communication between neurons and lead to their dysfunction and death. In recent years, research on AD has highlighted the critical involvement of mitochondria-the primary energy suppliers for our cells-in the onset and progression of the disease, since mitochondrial bioenergetic deficits precede the beginning of the disease and mitochondria are very sensitive to Aβ toxicity. On the other hand, if it is true that the accumulation of Aβ in the mitochondria leads to mitochondrial malfunctions, it is otherwise proven that mitochondrial dysfunction, through the generation of reactive oxygen species, causes an increase in Aβ production, by initiating a vicious cycle: there is therefore a bidirectional relationship between Aβ aggregation and mitochondrial dysfunction. Here, we focus on the latest news-but also on neglected evidence from the past-concerning the interplay between dysfunctional mitochondrial complex I, oxidative stress, and Aβ, in order to understand how their interplay is implicated in the pathogenesis of the disease.
Collapse
Affiliation(s)
- Anna Atlante
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council (CNR), Via G. Amendola 122/O, 70126 Bari, Italy;
| | | |
Collapse
|
9
|
Wu YH, Hsieh HL. Effects of Redox Homeostasis and Mitochondrial Damage on Alzheimer's Disease. Antioxidants (Basel) 2023; 12:1816. [PMID: 37891895 PMCID: PMC10604635 DOI: 10.3390/antiox12101816] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 09/21/2023] [Accepted: 09/28/2023] [Indexed: 10/29/2023] Open
Abstract
Bioenergetic mitochondrial dysfunction is a common feature of several diseases, including Alzheimer's disease (AD), where redox imbalance also plays an important role in terms of disease development. AD is an age-related disease and begins many years before the appearance of neurodegenerative symptoms. Intracellular tau aggregation, extracellular β-amyloid (Aβ) deposition in the brain, and even the APOE4 genotype contribute to the process of AD by impairing redox homeostasis and mitochondrial dysfunction. This review summarizes the evidence for the redox imbalance and mitochondrial dysfunction in AD and demonstrates the current therapeutic strategies related to mitochondrial maintenance.
Collapse
Affiliation(s)
- Yi-Hsuan Wu
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 333, Taiwan or
| | - Hsi-Lung Hsieh
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 333, Taiwan or
- Department of Nursing, Division of Basic Medical Sciences, Graduate Institute of Health Industry Technology, Chang Gung University of Science and Technology, Taoyuan 333, Taiwan
- Department of Neurology, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| |
Collapse
|
10
|
Wang Y, Wu S, Li Q, Sun H, Wang H. Pharmacological Inhibition of Ferroptosis as a Therapeutic Target for Neurodegenerative Diseases and Strokes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300325. [PMID: 37341302 PMCID: PMC10460905 DOI: 10.1002/advs.202300325] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 05/23/2023] [Indexed: 06/22/2023]
Abstract
Emerging evidence suggests that ferroptosis, a unique regulated cell death modality that is morphologically and mechanistically different from other forms of cell death, plays a vital role in the pathophysiological process of neurodegenerative diseases, and strokes. Accumulating evidence supports ferroptosis as a critical factor of neurodegenerative diseases and strokes, and pharmacological inhibition of ferroptosis as a therapeutic target for these diseases. In this review article, the core mechanisms of ferroptosis are overviewed and the roles of ferroptosis in neurodegenerative diseases and strokes are described. Finally, the emerging findings in treating neurodegenerative diseases and strokes through pharmacological inhibition of ferroptosis are described. This review demonstrates that pharmacological inhibition of ferroptosis by bioactive small-molecule compounds (ferroptosis inhibitors) could be effective for treatments of these diseases, and highlights a potential promising therapeutic avenue that could be used to prevent neurodegenerative diseases and strokes. This review article will shed light on developing novel therapeutic regimens by pharmacological inhibition of ferroptosis to slow down the progression of these diseases in the future.
Collapse
Affiliation(s)
- Yumin Wang
- Department of Respiratory and Critical Care MedicineAerospace Center HospitalPeking University Aerospace School of Clinical MedicineBeijing100049P. R. China
| | - Shuang Wu
- Department of NeurologyZhongnan Hospital of Wuhan UniversityWuhan430000P. R. China
| | - Qiang Li
- Department of NeurologyThe Affiliated Hospital of Chifeng UniversityChifeng024005P. R. China
| | - Huiyan Sun
- Chifeng University Health Science CenterChifeng024000P. R. China
| | - Hongquan Wang
- Tianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerTianjin's Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin300060P. R. China
| |
Collapse
|
11
|
Canet G, Zussy C, Hernandez C, Maurice T, Desrumaux C, Givalois L. The pathomimetic oAβ25–35 model of Alzheimer's disease: Potential for screening of new therapeutic agents. Pharmacol Ther 2023; 245:108398. [PMID: 37001735 DOI: 10.1016/j.pharmthera.2023.108398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 03/27/2023] [Indexed: 03/31/2023]
Abstract
Alzheimer's disease (AD) is the most common form of dementia in the elderly, currently affecting more than 40 million people worldwide. The two main histopathological hallmarks of AD were identified in the 1980s: senile plaques (composed of aggregated amyloid-β (Aβ) peptides) and neurofibrillary tangles (composed of hyperphosphorylated tau protein). In the human brain, both Aβ and tau show aggregation into soluble and insoluble oligomers. Soluble oligomers of Aβ include their most predominant forms - Aβ1-40 and Aβ1-42 - as well as shorter peptides such as Aβ25-35 or Aβ25-35/40. Most animal models of AD have been developed using transgenesis, based on identified human mutations. However, these familial forms of AD represent less than 1% of AD cases. In this context, the idea emerged in the 1990s to directly inject the Aβ25-35 fragment into the rodent brain to develop an acute model of AD that could mimic the disease's sporadic forms (99% of all cases). This review aims to: (1) summarize the biological activity of Aβ25-35, focusing on its impact on the main structural and functional alterations observed in AD (cognitive deficits, APP misprocessing, tau system dysfunction, neuroinflammation, oxidative stress, cholinergic and glutamatergic alterations, HPA axis dysregulation, synaptic deficits and cell death); and (2) confirm the interest of this pathomimetic model in AD research, as it has helped identify and characterize many molecules (marketed, in clinical development, and in preclinical testing), and to the development of alternative approaches for AD prevention and therapy. Today, the Aβ25-35 model appears as a first-intent choice model to rapidly screen the symptomatic or neuroprotective potencies of new compounds, chemical series, or innovative therapeutic strategies.
Collapse
|
12
|
Ebanks B, Chakrabarti L. Mitochondrial ATP Synthase is a Target of Oxidative Stress in Neurodegenerative Diseases. Front Mol Biosci 2022; 9:854321. [PMID: 35237666 PMCID: PMC8882969 DOI: 10.3389/fmolb.2022.854321] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 01/26/2022] [Indexed: 12/11/2022] Open
Abstract
The mitochondrial ATP synthase is responsible for the production of cellular ATP, and it does so by harnessing the membrane potential of the mitochondria that is produced by the sequential oxidation of select cellular metabolites. Since the structural features of ATP synthase were first resolved nearly three decades ago, significant progress has been made in understanding its role in health and disease. Mitochondrial dysfunction is common to neurodegeneration, with elevated oxidative stress a hallmark of this dysfunction. The patterns of this oxidative stress, including molecular targets and the form of oxidative modification, can vary widely. In this mini review we discuss the oxidative modifications of ATP synthase that have been observed in Alzheimer’s disease, Parkinson’s disease, and Huntington’s disease. Oxidative modifications of ATP synthase in Alzheimer’s disease are well-documented, and there is a growing body of knowledge on the subject in Parkinson’s disease. The consideration of ATP synthase as a pharmacological target in a variety of diseases underlines the importance of understanding these modifications, both as a potential target, and also as inhibitors of any pharmacological intervention.
Collapse
Affiliation(s)
- Brad Ebanks
- School of Veterinary Medicine and Science, University of Nottingham, Nottingham, United Kingdom
- *Correspondence: Brad Ebanks,
| | - Lisa Chakrabarti
- School of Veterinary Medicine and Science, University of Nottingham, Nottingham, United Kingdom
- MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, Nottingham, United Kingdom
| |
Collapse
|
13
|
Wilkins HM, Troutwine BR, Menta BW, Manley SJ, Strope TA, Lysaker CR, Swerdlow RH. Mitochondrial Membrane Potential Influences Amyloid-β Protein Precursor Localization and Amyloid-β Secretion. J Alzheimers Dis 2022; 85:381-394. [PMID: 34806611 PMCID: PMC9212216 DOI: 10.3233/jad-215280] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Amyloid-β (Aβ), which derives from the amyloid-β protein precursor (AβPP), forms plaques and serves as a fluid biomarker in Alzheimer's disease (AD). How Aβ forms from AβPP is known, but questions relating to AβPP and Aβ biology remain unanswered. AD patients show mitochondrial dysfunction, and an Aβ/AβPP mitochondria relationship exists. OBJECTIVE We considered how mitochondrial biology may impact AβPP and Aβ biology. METHODS SH-SY5Y cells were transfected with AβPP constructs. After treatment with FCCP (uncoupler), Oligomycin (ATP synthase inhibitor), or starvation Aβ levels were measured. β-secretase (BACE1) expression was measured. Mitochondrial localized full-length AβPP was also measured. All parameters listed were measured in ρ0 cells on an SH-SY5Y background. iPSC derived neurons were also used to verify key results. RESULTS We showed that mitochondrial depolarization routes AβPP to, while hyperpolarization routes AβPP away from, the organelle. Mitochondrial AβPP and cell Aβ secretion inversely correlate, as cells with more mitochondrial AβPP secrete less Aβ, and cells with less mitochondrial AβPP secrete more Aβ. An inverse relationship between secreted/extracellular Aβ and intracellular Aβ was observed. CONCLUSION Our findings indicate mitochondrial function alters AβPP localization and suggest enhanced mitochondrial activity promotes Aβ secretion while depressed mitochondrial activity minimizes Aβ secretion. Our data complement other studies that indicate a mitochondrial, AβPP, and Aβ nexus, and could help explain why cerebrospinal fluid Aβ is lower in those with AD. Our data further suggest Aβ secretion could serve as a biomarker of cell or tissue mitochondrial function.
Collapse
Affiliation(s)
- Heather M. Wilkins
- Department of Neurology University of Kansas Medical Center, Kansas City, KS, USA
- University of Kansas Alzheimer’s Disease Center, Kansas City, KS, USA
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS USA
| | - Benjamin R. Troutwine
- Department of Neurology University of Kansas Medical Center, Kansas City, KS, USA
- University of Kansas Alzheimer’s Disease Center, Kansas City, KS, USA
| | - Blaise W. Menta
- University of Kansas Alzheimer’s Disease Center, Kansas City, KS, USA
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS USA
| | - Sharon J. Manley
- Department of Neurology University of Kansas Medical Center, Kansas City, KS, USA
- University of Kansas Alzheimer’s Disease Center, Kansas City, KS, USA
| | - Taylor A. Strope
- University of Kansas Alzheimer’s Disease Center, Kansas City, KS, USA
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS USA
| | - Colton R. Lysaker
- University of Kansas Alzheimer’s Disease Center, Kansas City, KS, USA
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS USA
| | - Russell H. Swerdlow
- Department of Neurology University of Kansas Medical Center, Kansas City, KS, USA
- University of Kansas Alzheimer’s Disease Center, Kansas City, KS, USA
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS USA
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
14
|
Chen K, Jiang X, Wu M, Cao X, Bao W, Zhu LQ. Ferroptosis, a Potential Therapeutic Target in Alzheimer's Disease. Front Cell Dev Biol 2021; 9:704298. [PMID: 34422824 PMCID: PMC8374166 DOI: 10.3389/fcell.2021.704298] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 07/09/2021] [Indexed: 12/11/2022] Open
Abstract
Cell death is a common phenomenon in the progression of Alzheimer’s disease (AD). However, the mechanism of triggering the death of neuronal cells remains unclear. Ferroptosis is an iron-dependent lipid peroxidation-driven cell death and emerging evidences have demonstrated the involvement of ferroptosis in the pathological process of AD. Moreover, several hallmarks of AD pathogenesis were consistent with the characteristics of ferroptosis, such as excess iron accumulation, elevated lipid peroxides, and reactive oxygen species (ROS), reduced glutathione (GSH), and glutathione peroxidase 4 (GPX4) levels. Besides, some ferroptosis inhibitors can relieve AD-related pathological symptoms in AD mice and exhibit potential clinical benefits in AD patients. Therefore, ferroptosis is gradually being considered as a distinct cell death mechanism in the pathogenesis of AD. However, direct evidence is still lacking. In this review, we summarize the features of ferroptosis in AD, its underlying mechanisms in AD pathology, and review the application of ferroptosis inhibitors in both AD clinical trials and mice/cell models, to provide valuable information for future treatment and prevention of this devastating disease.
Collapse
Affiliation(s)
- Kai Chen
- Key Lab of Neurological Disorder of Education Ministry, Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Department of Neurosurgery, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaobing Jiang
- Department of Neurosurgery, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Moxin Wu
- Department of Jiujiang Clinical Research Center for Precision Medicine, Affiliated Hospital of Jiujiang University, Jiujiang, China
| | - Xianming Cao
- Department of Jiujiang Clinical Research Center for Precision Medicine, Affiliated Hospital of Jiujiang University, Jiujiang, China
| | - Wendai Bao
- Key Lab of Neurological Disorder of Education Ministry, Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, China
| | - Ling-Qiang Zhu
- Key Lab of Neurological Disorder of Education Ministry, Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
15
|
Perluigi M, Di Domenico F, Barone E, Butterfield DA. mTOR in Alzheimer disease and its earlier stages: Links to oxidative damage in the progression of this dementing disorder. Free Radic Biol Med 2021; 169:382-396. [PMID: 33933601 PMCID: PMC8145782 DOI: 10.1016/j.freeradbiomed.2021.04.025] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 04/15/2021] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia in the elderly population and has worldwide impact. The etiology of the disease is complex and results from the confluence of multiple mechanisms ultimately leading to neuronal loss and cognitive decline. Among risk factors, aging is the most relevant and accounts for several pathogenic events that contribute to disease-specific toxic mechanisms. Accumulating evidence linked the alterations of the mammalian target of rapamycin (mTOR), a serine/threonine protein kinase playing a key role in the regulation of protein synthesis and degradation, to age-dependent cognitive decline and pathogenesis of AD. To date, growing studies demonstrated that aberrant mTOR signaling in the brain affects several pathways involved in energy metabolism, cell growth, mitochondrial function and proteostasis. Recent advances associated alterations of the mTOR pathway with the increased oxidative stress. Disruption of all these events strongly contribute to age-related cognitive decline including AD. The current review discusses the main regulatory roles of mTOR signaling network in the brain, focusing on its role in autophagy, oxidative stress and energy metabolism. Collectively, experimental data suggest that targeting mTOR in the CNS can be a valuable strategy to prevent/slow the progression of AD.
Collapse
Affiliation(s)
- M Perluigi
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185, Roma, Italy
| | - F Di Domenico
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185, Roma, Italy
| | - E Barone
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185, Roma, Italy
| | - D A Butterfield
- Department of Chemistry, Sapienza University of Rome, Piazzale A. Moro 5, 00185, Roma, Italy; Department of Chemistry and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, 40506-0055, USA.
| |
Collapse
|
16
|
Clarke GA, Hartse BX, Niaraki Asli AE, Taghavimehr M, Hashemi N, Abbasi Shirsavar M, Montazami R, Alimoradi N, Nasirian V, Ouedraogo LJ, Hashemi NN. Advancement of Sensor Integrated Organ-on-Chip Devices. SENSORS (BASEL, SWITZERLAND) 2021; 21:1367. [PMID: 33671996 PMCID: PMC7922590 DOI: 10.3390/s21041367] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 02/10/2021] [Accepted: 02/11/2021] [Indexed: 02/06/2023]
Abstract
Organ-on-chip devices have provided the pharmaceutical and tissue engineering worlds much hope since they arrived and began to grow in sophistication. However, limitations for their applicability were soon realized as they lacked real-time monitoring and sensing capabilities. The users of these devices relied solely on endpoint analysis for the results of their tests, which created a chasm in the understanding of life between the lab the natural world. However, this gap is being bridged with sensors that are integrated into organ-on-chip devices. This review goes in-depth on different sensing methods, giving examples for various research on mechanical, electrical resistance, and bead-based sensors, and the prospects of each. Furthermore, the review covers works conducted that use specific sensors for oxygen, and various metabolites to characterize cellular behavior and response in real-time. Together, the outline of these works gives a thorough analysis of the design methodology and sophistication of the current sensor integrated organ-on-chips.
Collapse
Affiliation(s)
- Gabriel A. Clarke
- Department of Mechanical Engineering, Iowa State University, Ames, IA 50011, USA; (G.A.C.); (B.X.H.); (A.E.N.A.); (M.T.); (M.A.S.); (R.M.); (N.A.); (V.N.); (L.J.O.)
| | - Brenna X. Hartse
- Department of Mechanical Engineering, Iowa State University, Ames, IA 50011, USA; (G.A.C.); (B.X.H.); (A.E.N.A.); (M.T.); (M.A.S.); (R.M.); (N.A.); (V.N.); (L.J.O.)
| | - Amir Ehsan Niaraki Asli
- Department of Mechanical Engineering, Iowa State University, Ames, IA 50011, USA; (G.A.C.); (B.X.H.); (A.E.N.A.); (M.T.); (M.A.S.); (R.M.); (N.A.); (V.N.); (L.J.O.)
| | - Mehrnoosh Taghavimehr
- Department of Mechanical Engineering, Iowa State University, Ames, IA 50011, USA; (G.A.C.); (B.X.H.); (A.E.N.A.); (M.T.); (M.A.S.); (R.M.); (N.A.); (V.N.); (L.J.O.)
| | - Niloofar Hashemi
- Department of Materials Science and Engineering, Sharif University of Technology, Tehran 11365, Iran;
| | - Mehran Abbasi Shirsavar
- Department of Mechanical Engineering, Iowa State University, Ames, IA 50011, USA; (G.A.C.); (B.X.H.); (A.E.N.A.); (M.T.); (M.A.S.); (R.M.); (N.A.); (V.N.); (L.J.O.)
| | - Reza Montazami
- Department of Mechanical Engineering, Iowa State University, Ames, IA 50011, USA; (G.A.C.); (B.X.H.); (A.E.N.A.); (M.T.); (M.A.S.); (R.M.); (N.A.); (V.N.); (L.J.O.)
| | - Nima Alimoradi
- Department of Mechanical Engineering, Iowa State University, Ames, IA 50011, USA; (G.A.C.); (B.X.H.); (A.E.N.A.); (M.T.); (M.A.S.); (R.M.); (N.A.); (V.N.); (L.J.O.)
| | - Vahid Nasirian
- Department of Mechanical Engineering, Iowa State University, Ames, IA 50011, USA; (G.A.C.); (B.X.H.); (A.E.N.A.); (M.T.); (M.A.S.); (R.M.); (N.A.); (V.N.); (L.J.O.)
| | - Lionel J. Ouedraogo
- Department of Mechanical Engineering, Iowa State University, Ames, IA 50011, USA; (G.A.C.); (B.X.H.); (A.E.N.A.); (M.T.); (M.A.S.); (R.M.); (N.A.); (V.N.); (L.J.O.)
| | - Nicole N. Hashemi
- Department of Mechanical Engineering, Iowa State University, Ames, IA 50011, USA; (G.A.C.); (B.X.H.); (A.E.N.A.); (M.T.); (M.A.S.); (R.M.); (N.A.); (V.N.); (L.J.O.)
- Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA
| |
Collapse
|
17
|
N'Go PK, Ahami OTA, El Hessni A, Azzaoui FZ, Aboussaleh Y, Tako AN. Neuroprotective effects of the Chrysophyllum perpulchrum extract against an Alzheimer-like rat model of β amyloid 1-40 intrahippocampal injection. Transl Neurosci 2021; 12:545-560. [PMID: 34992853 PMCID: PMC8678622 DOI: 10.1515/tnsci-2020-0183] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 08/10/2021] [Accepted: 08/23/2021] [Indexed: 12/29/2022] Open
Abstract
Objective Alzheimer’s disease (AD) is a threatening disease for African populations in the upcoming years because of the increase in their expectancy of life. Here, we investigated whether natural products from Chrysophyllum perpulchrum as catechin and two dimeric procyanidins (catechin + hexose) could prevent progression of oxidative stress and cognitive changes using an AD-like rat model induced by Aβ1-40 injection into the hippocampal CA1 subfield. Methodology Adult male Wistar rats were either microinjected with 1% ammonia as a vehicle (10 µL) or aggregated Aβ1-40 at 10 µg bilateral hippocampus. On the 14th day of post-surgery, some Aβ rats were treated with melatonin (10 mg/kg i.p.) or with the Chrysophyllum perpulchrum extract (300 mg/kg p.o.), and some sham-operated rats received the extract alone. Cognitive abilities were tested with Y-maze, object recognition test and Morris Water Maze. Oxidative stress markers as well as the level of activated microglial cells were assayed in the brain. Results Aβ rats exhibited significant deficits of recognition memory and spatial learning. This was associated with an increase of microglia Iba 1 immunoreactivity as well as nitric oxide (NO), malondialdehyde and superoxide dismutase levels but not to the thiol content in the hippocampus, prefrontal cortex and septum of AD-like rats. The Chrysophyllum perpulchrum extract treatment mitigated Aβ-induced cognitive impairments and reversed microglia overactivation and subsequent generation of oxidative stress markers. Interestingly, the neuroprotective actions of the Chrysophyllum perpulchrum extract seem to be comparable to the control drug melatonin used albeit with some more beneficial effects. Conclusion These findings are preliminary and should be strengthened by more pharmacological studies of bioactive compounds of Chrysophyllum perpulchrum before being proposed as a promising drug against AD.
Collapse
Affiliation(s)
- Pacôme Kouadio N'Go
- Peleforo GON COULIBALY University, Training and Research Unit of Biological Sciences, Department of Animal Biology, PO Box 1328, Korhogo, Ivory Coast.,Clinical and Cognitive Neurosciences Group, Biology and Health Lab, Ibn Tofail University, PO Box 133, Kenitra, Morocco
| | - Omar Touhami Ahmed Ahami
- Clinical and Cognitive Neurosciences Group, Biology and Health Lab, Ibn Tofail University, PO Box 133, Kenitra, Morocco
| | - Aboubaker El Hessni
- Genetic, Neuroendocrinology and Biotechnology Team, Biology and Health Lab, Department of Biology, Ibn Tofail University, PO Box 133, Kenitra, Morocco
| | - Fatima-Zahra Azzaoui
- Clinical and Cognitive Neurosciences Group, Biology and Health Lab, Ibn Tofail University, PO Box 133, Kenitra, Morocco
| | - Youssef Aboussaleh
- Clinical and Cognitive Neurosciences Group, Biology and Health Lab, Ibn Tofail University, PO Box 133, Kenitra, Morocco
| | - Antoine Némé Tako
- Neurosciences Team, Biology and Health Lab, Department of Biosciences, Felix Houphouet Boigny University, 01 BPV 34 Abidjan 01, Abidjan, Ivory Coast
| |
Collapse
|
18
|
Parodi-Rullán R, Sone JY, Fossati S. Endothelial Mitochondrial Dysfunction in Cerebral Amyloid Angiopathy and Alzheimer's Disease. J Alzheimers Dis 2020; 72:1019-1039. [PMID: 31306129 DOI: 10.3233/jad-190357] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia. Cerebrovascular dysfunction is one of the earliest events in the pathogenesis of AD, as well as in vascular and mixed dementias. Cerebral amyloid angiopathy (CAA), the deposition of amyloid around cerebral vessels, is observed in up to 90% of AD patients and in approximately 50% of elderly individuals over 80 years of age. CAA is a strong contributor to vascular dysfunction in AD. CAA-laden brain vessels are characterized by dysfunctional hemodynamics and leaky blood-brain barrier (BBB), contributing to clearance failure and further accumulation of amyloid-β (Aβ) in the cerebrovasculature and brain parenchyma. Mitochondrial dysfunction is increasingly recognized as an important early initiator of the pathogenesis of AD and CAA. The objective of this review is to discuss the effects of Aβ on cerebral microvascular cell function, focusing on its impact on endothelial mitochondria. After introducing CAA and its etiology and genetic risk factors, we describe the pathological relationship between cerebrovascular amyloidosis and brain microvascular endothelial cell dysfunction, critically analyzing its roles in disease progression, hypoperfusion, and BBB integrity. Then, we focus on discussing the effect of Aβ challenge on endothelial mitochondrial dysfunction pathways, and their contribution to the progression of neurovascular dysfunction in AD and dementia. Finally, we report potential pharmacological and non-pharmacological mitochondria-targeted therapeutic strategies which may help prevent or delay cerebrovascular failure.
Collapse
Affiliation(s)
- Rebecca Parodi-Rullán
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Je Yeong Sone
- Department of Psychiatry, Center for Brain Health, NYU School of Medicine, New York, NY, USA
| | - Silvia Fossati
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| |
Collapse
|
19
|
Sarparast M, Dattmore D, Alan J, Lee KSS. Cytochrome P450 Metabolism of Polyunsaturated Fatty Acids and Neurodegeneration. Nutrients 2020; 12:E3523. [PMID: 33207662 PMCID: PMC7696575 DOI: 10.3390/nu12113523] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/08/2020] [Accepted: 11/10/2020] [Indexed: 12/11/2022] Open
Abstract
Due to the aging population in the world, neurodegenerative diseases have become a serious public health issue that greatly impacts patients' quality of life and adds a huge economic burden. Even after decades of research, there is no effective curative treatment for neurodegenerative diseases. Polyunsaturated fatty acids (PUFAs) have become an emerging dietary medical intervention for health maintenance and treatment of diseases, including neurodegenerative diseases. Recent research demonstrated that the oxidized metabolites, particularly the cytochrome P450 (CYP) metabolites, of PUFAs are beneficial to several neurodegenerative diseases, including Alzheimer's disease and Parkinson's disease; however, their mechanism(s) remains unclear. The endogenous levels of CYP metabolites are greatly affected by our diet, endogenous synthesis, and the downstream metabolism. While the activity of omega-3 (ω-3) CYP PUFA metabolites and omega-6 (ω-6) CYP PUFA metabolites largely overlap, the ω-3 CYP PUFA metabolites are more active in general. In this review, we will briefly summarize recent findings regarding the biosynthesis and metabolism of CYP PUFA metabolites. We will also discuss the potential mechanism(s) of CYP PUFA metabolites in neurodegeneration, which will ultimately improve our understanding of how PUFAs affect neurodegeneration and may identify potential drug targets for neurodegenerative diseases.
Collapse
Affiliation(s)
- Morteza Sarparast
- Department of Chemistry, Michigan State University, East Lansing, MI 48824, USA;
| | - Devon Dattmore
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA;
| | - Jamie Alan
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA;
| | - Kin Sing Stephen Lee
- Department of Chemistry, Michigan State University, East Lansing, MI 48824, USA;
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA;
| |
Collapse
|
20
|
The Ubiquitin System in Alzheimer's Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1233:195-221. [PMID: 32274758 DOI: 10.1007/978-3-030-38266-7_8] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia, most prevalent in the elderly population and has a significant impact on individuals and their family as well as the health care system and the economy. While the number of patients affected by various forms of dementia including AD is on the increase, there is currently no cure. Although genome-wide association studies have identified genetic markers for familial AD, the molecular mechanisms underlying the initiation and development of both familial and sporadic AD remain poorly understood. Most neurodegenerative diseases and in particular those associated with dementia have been defined as proteinopathies due to the presence of intra- and/or extracellular protein aggregates in the brain of affected individuals. Although loss of proteostasis in AD has been known for decades, it is only in recent years that we have come to appreciate the role of ubiquitin-dependent mechanisms in brain homeostasis and in brain diseases. Ubiquitin is a highly versatile post-translational modification which regulates many aspects of protein fate and function, including protein degradation by the Ubiquitin-Proteasome System (UPS), autophagy-mediated removal of damaged organelles and proteins, lysosomal turnover of membrane proteins and of extracellular molecules brought inside the cell through endocytosis. Amyloid-β (Aβ) fragments as well as hyperphosphorylation of Tau are hallmarks of AD, and these are found in extracellular plaques and intracellular fibrils in the brain of individuals with AD, respectively. Yet, whether it is the oligomeric or the soluble species of Aβ and Tau that mediate toxicity is still unclear. These proteins impact on mitochondrial energy metabolism, inflammation, as well as a number of housekeeping processes including protein degradation through the UPS and autophagy. In this chapter, we will discuss the role of ubiquitin in neuronal homeostasis as well as in AD; summarise crosstalks between the enzymes that regulate protein ubiquitination and the toxic proteins Tau and Aβ; highlight emerging molecular mechanisms in AD as well as future strategies which aim to exploit the ubiquitin system as a source for next-generation therapeutics.
Collapse
|
21
|
Lanzillotta C, Di Domenico F, Perluigi M, Butterfield DA. Targeting Mitochondria in Alzheimer Disease: Rationale and Perspectives. CNS Drugs 2019; 33:957-969. [PMID: 31410665 PMCID: PMC6825561 DOI: 10.1007/s40263-019-00658-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
A decline in mitochondrial function plays a key role in the aging process and increases the incidence of age-related disorders, including Alzheimer disease (AD). Mitochondria-the power station of the organism-can affect several different cellular activities, including abnormal cellular energy generation, response to toxic insults, regulation of metabolism, and execution of cell death. In AD subjects, mitochondria are characterized by impaired function such as lowered oxidative phosphorylation, decreased adenosine triphosphate production, significant increased reactive oxygen species generation, and compromised antioxidant defense. The current review discusses the most relevant mitochondrial defects that are considered to play a significant role in AD and that may offer promising therapeutic targets for the treatment/prevention of AD. In addition, we discuss mechanisms of action and translational potential of some promising mitochondrial and bioenergetic therapeutics for AD including compounds able to potentiate energy production, antioxidants to scavenge reactive oxygen species and reduce oxidative damage, glucose metabolism, and candidates that target mitophagy. While mitochondrial therapeutic strategies have shown promise at the preclinical stage, there has been little progress in clinical trials. Thus, there is an urgent need to better understand the mechanisms regulating mitochondrial homeostasis in order to identify powerful drug candidates that target 'in and out' the mitochondria to preserve cognitive functions.
Collapse
Affiliation(s)
- Chiara Lanzillotta
- Department of Biochemical Sciences, Sapienza University of Rome, 00185, Rome, Italy
| | - Fabio Di Domenico
- Department of Biochemical Sciences, Sapienza University of Rome, 00185, Rome, Italy
| | - Marzia Perluigi
- Department of Biochemical Sciences, Sapienza University of Rome, 00185, Rome, Italy
| | - D Allan Butterfield
- Department of Chemistry, University of Kentucky, Lexington, KY, 40506-0055, USA.
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, 40506-0055, USA.
| |
Collapse
|
22
|
Patricio-Martínez A, Sánchez-Zavaleta R, Angulo-Cruz I, Gutierrez-Praxedis L, Ramírez E, Martínez-García I, Limón ID. The Acute Activation of the CB1 Receptor in the Hippocampus Decreases Neurotoxicity and Prevents Spatial Memory Impairment in Rats Lesioned with β-Amyloid 25-35. Neuroscience 2019; 416:239-254. [PMID: 31400487 DOI: 10.1016/j.neuroscience.2019.08.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 07/31/2019] [Accepted: 08/01/2019] [Indexed: 12/23/2022]
Abstract
Given their anti-inflammatory properties, cannabinoids have been shown to be neuroprotective agents and to reduce excitotoxicity, through the activation of the Cannabinoid receptor type 1 (CB1r). These properties have led to CB1r being proposed as pharmacological targets for the treatment of various neurodegenerative diseases. Amyloid-β 25-35 (Aβ25-35) induces the expression of inducible nitric oxide synthase (iNOS) and increases nitric oxide (NO●) levels. It has been observed that increased NO● concentrations trigger biochemical pathways that contribute to neuronal death and cognitive damage. This study aimed to evaluate the neuroprotective effect of an acute activation of CB1r on spatial memory and its impact on iNOS protein expression, NO● levels, gliosis and the neurodegenerative process induced by the injection of Aβ(25-35) into the CA1 subfield of the hippocampus. ACEA [1 μM/1 μL] and Aβ(25-35) [100 μM/1 μL] and their respective vehicle groups were injected into the CA1 subfield of the hippocampus. The animals were tested for spatial learning and memory in the eight-arm radial maze, with the results revealing that the administration of ACEA plus Aβ(25-35) improves learning and memory processes, in contrast with the Aβ(25-35) group. Moreover, ACEA plus Aβ(25-35) prevented both the increase in iNOS protein and NO● levels and the reactive gliosis induced by Aβ(25-35). Importantly, neurodegeneration was significantly reduced by the administration of ACEA plus Aβ(25-35) in the CA1 subfield of the hippocampus. The data obtained in the present research suggest that the acute early activation of CB1r is crucial for neuroprotection.
Collapse
Affiliation(s)
- Aleidy Patricio-Martínez
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas-Benemérita Universidad Autónoma de Puebla, Puebla, Mexico; Facultad de Ciencias Biológicas-Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Rodolfo Sánchez-Zavaleta
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas-Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Isael Angulo-Cruz
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas-Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Liliana Gutierrez-Praxedis
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas-Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Eleazar Ramírez
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas-Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Isabel Martínez-García
- Laboratorio de Neuroquímica, Facultad de Ciencias Químicas-Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Ilhuicamina Daniel Limón
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas-Benemérita Universidad Autónoma de Puebla, Puebla, Mexico.
| |
Collapse
|
23
|
Evans CS, Holzbaur ELF. Quality Control in Neurons: Mitophagy and Other Selective Autophagy Mechanisms. J Mol Biol 2019; 432:240-260. [PMID: 31295455 DOI: 10.1016/j.jmb.2019.06.031] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 06/28/2019] [Accepted: 06/29/2019] [Indexed: 12/19/2022]
Abstract
The cargo-specific removal of organelles via selective autophagy is important to maintain neuronal homeostasis. Genetic studies indicate that deficits in these pathways are implicated in neurodegenerative diseases, including Parkinson's and amyotrophic lateral sclerosis. Here, we review our current understanding of the pathways that regulate mitochondrial quality control, and compare these mechanisms to those regulating turnover of the endoplasmic reticulum and the clearance of protein aggregates. Research suggests that there are multiple mechanisms regulating the degradation of specific cargos, such as dysfunctional organelles and protein aggregates. These mechanisms are critical for neuronal health, as neurons are uniquely vulnerable to impairment in organelle quality control pathways due to their morphology, size, polarity, and postmitotic nature. We highlight the consequences of dysregulation of selective autophagy in neurons and discuss current challenges in correlating noncongruent findings from in vitro and in vivo systems.
Collapse
Affiliation(s)
- Chantell S Evans
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104-6085, USA.
| | - Erika L F Holzbaur
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104-6085, USA.
| |
Collapse
|
24
|
Houben E, Hellings N, Broux B. Oncostatin M, an Underestimated Player in the Central Nervous System. Front Immunol 2019; 10:1165. [PMID: 31191538 PMCID: PMC6549448 DOI: 10.3389/fimmu.2019.01165] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 05/08/2019] [Indexed: 12/12/2022] Open
Abstract
For a long time, the central nervous system (CNS) was believed to be an immune privileged organ. In the last decades, it became apparent that the immune system interacts with the CNS not only in pathological, but also in homeostatic situations. It is now clear that immune cells infiltrate the healthy CNS as part of immune surveillance and that immune cells communicate through cytokines with CNS resident cells. In pathological conditions, an enhanced infiltration of immune cells takes place to fight the pathogen. A well-known family of cytokines is the interleukin (IL)-6 cytokine family. All members are important in cell communication and cell signaling in the immune system. One of these members is oncostatin M (OSM), for which the receptor is expressed on several cells of the CNS. However, the biological function of OSM in the CNS is not studied in detail. Here, we briefly describe the general aspects related to OSM biology, including signaling and receptor binding. Thereafter, the current understanding of OSM during CNS homeostasis and pathology is summarized.
Collapse
Affiliation(s)
- Evelien Houben
- Department of Immunology, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Niels Hellings
- Department of Immunology, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Bieke Broux
- Department of Immunology, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| |
Collapse
|
25
|
Deng L, Pushpitha K, Joseph C, Gupta V, Rajput R, Chitranshi N, Dheer Y, Amirkhani A, Kamath K, Pascovici D, Wu JX, Salekdeh GH, Haynes PA, Graham SL, Gupta VK, Mirzaei M. Amyloid β Induces Early Changes in the Ribosomal Machinery, Cytoskeletal Organization and Oxidative Phosphorylation in Retinal Photoreceptor Cells. Front Mol Neurosci 2019; 12:24. [PMID: 30853886 PMCID: PMC6395395 DOI: 10.3389/fnmol.2019.00024] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Accepted: 01/21/2019] [Indexed: 01/20/2023] Open
Abstract
Amyloid β (Aβ) accumulation and its aggregation is characteristic molecular feature of the development of Alzheimer's disease (AD). More recently, Aβ has been suggested to be associated with retinal pathology associated with AD, glaucoma and drusen deposits in age related macular degeneration (AMD). In this study, we investigated the proteins and biochemical networks that are affected by Aβ in the 661 W photoreceptor cells in culture. Time and dose dependent effects of Aβ on the photoreceptor cells were determined utilizing tandem mass tag (TMT) labeling-based quantitative mass-spectrometric approach. Bioinformatic analysis of the data revealed concentration and time dependent effects of the Aβ peptide stimulation on various key biochemical pathways that might be involved in mediating the toxicity effects of the peptide. We identified increased Tau phosphorylation, GSK3β dysregulation and reduced cell viability in cells treated with Aβ in a dose and time dependent manner. This study has delineated for the first-time molecular networks in photoreceptor cells that are impacted early upon Aβ treatment and contrasted the findings with a longer-term treatment effect. Proteins associated with ribosomal machinery homeostasis, mitochondrial function and cytoskeletal organization were affected in the initial stages of Aβ exposure, which may provide key insights into AD effects on the photoreceptors and specific molecular changes induced by Aβ peptide.
Collapse
Affiliation(s)
- Liting Deng
- Department of Molecular Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW, Australia
| | - Kanishka Pushpitha
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Chitra Joseph
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Veer Gupta
- School of Medicine, Deakin University, Waurn Ponds, VIC, Australia
| | - Rashi Rajput
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Nitin Chitranshi
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Yogita Dheer
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Ardeshir Amirkhani
- Department of Molecular Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW, Australia
- Australian Proteome Analysis Facility (APAF), Macquarie University, Sydney, NSW, Australia
| | - Karthik Kamath
- Department of Molecular Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW, Australia
- Australian Proteome Analysis Facility (APAF), Macquarie University, Sydney, NSW, Australia
| | - Dana Pascovici
- Department of Molecular Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW, Australia
- Australian Proteome Analysis Facility (APAF), Macquarie University, Sydney, NSW, Australia
| | - Jemma X. Wu
- Department of Molecular Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW, Australia
- Australian Proteome Analysis Facility (APAF), Macquarie University, Sydney, NSW, Australia
| | - Ghasem Hosseini Salekdeh
- Department of Molecular Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW, Australia
- Cell Science Research Center, Department of Molecular Systems Biology, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
| | - Paul A. Haynes
- Department of Molecular Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW, Australia
| | - Stuart L. Graham
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Vivek K. Gupta
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Mehdi Mirzaei
- Department of Molecular Sciences, Faculty of Science and Engineering, Macquarie University, Sydney, NSW, Australia
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
- Australian Proteome Analysis Facility (APAF), Macquarie University, Sydney, NSW, Australia
| |
Collapse
|
26
|
Zádori D, Veres G, Szalárdy L, Klivényi P, Vécsei L. Alzheimer's Disease: Recent Concepts on the Relation of Mitochondrial Disturbances, Excitotoxicity, Neuroinflammation, and Kynurenines. J Alzheimers Dis 2019; 62:523-547. [PMID: 29480191 DOI: 10.3233/jad-170929] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The pathomechanism of Alzheimer's disease (AD) certainly involves mitochondrial disturbances, glutamate excitotoxicity, and neuroinflammation. The three main aspects of mitochondrial dysfunction in AD, i.e., the defects in dynamics, altered bioenergetics, and the deficient transport, act synergistically. In addition, glutamatergic neurotransmission is affected in several ways. The balance between synaptic and extrasynaptic glutamatergic transmission is shifted toward the extrasynaptic site contributing to glutamate excitotoxicity, a phenomenon augmented by increased glutamate release and decreased glutamate uptake. Neuroinflammation in AD is predominantly linked to central players of the innate immune system, with central nervous system (CNS)-resident microglia, astroglia, and perivascular macrophages having been implicated at the cellular level. Several abnormalities have been described regarding the activation of certain steps of the kynurenine (KYN) pathway of tryptophan metabolism in AD. First of all, the activation of indolamine 2,3-dioxygenase, the first and rate-limiting step of the pathway, is well-demonstrated. 3-Hydroxy-L-KYN and its metabolite, 3-hydroxy-anthranilic acid have pro-oxidant, antioxidant, and potent immunomodulatory features, giving relevance to their alterations in AD. Another metabolite, quinolinic acid, has been demonstrated to be neurotoxic, promoting glutamate excitotoxicity, reactive oxygen species production, lipid peroxidation, and microglial neuroinflammation, and its abundant presence in AD pathologies has been demonstrated. Finally, the neuroprotective metabolite, kynurenic acid, has been associated with antagonistic effects at glutamate receptors, free radical scavenging, and immunomodulation, giving rise to potential therapeutic implications. This review presents the multiple connections of KYN pathway-related alterations to three main domains of AD pathomechanism, such as mitochondrial dysfunction, excitotoxicity, and neuroinflammation, implicating possible therapeutic options.
Collapse
Affiliation(s)
- Dénes Zádori
- Department of Neurology, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Szeged, Hungary
| | - Gábor Veres
- Department of Neurology, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Szeged, Hungary
| | - Levente Szalárdy
- Department of Neurology, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Szeged, Hungary
| | - Péter Klivényi
- Department of Neurology, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Szeged, Hungary
| | - László Vécsei
- Department of Neurology, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged, Szeged, Hungary.,MTA-SZTE Neuroscience Research Group, Szeged, Hungary
| |
Collapse
|
27
|
Nishikawa N, Sakae Y, Gouda T, Tsujimura Y, Okamoto Y. Structural Analysis of a Trimer of β 2-Microgloblin Fragment by Molecular Dynamics Simulations. Biophys J 2019; 116:781-790. [PMID: 30771855 DOI: 10.1016/j.bpj.2018.11.3143] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 06/08/2018] [Accepted: 11/06/2018] [Indexed: 01/22/2023] Open
Abstract
A peptide β2-m21-31, which is a fragment from residue 21 to residue 31 of β2-microgloblin, is experimentally known to self-assemble and form amyloid fibrils. In order to understand the mechanism of amyloid fibril formations, we applied the replica-exchange molecular dynamics method to the system consisting of three fragments of β2-m21-31. From the analyses on the temperature dependence, we found that there is a clear phase transition temperature in which the peptides aggregate with each other. Moreover, we found by the free energy analyses that there are two major stable states: One of them is like amyloid fibrils and the other is amorphous aggregates.
Collapse
Affiliation(s)
- Naohiro Nishikawa
- Department of Physics, Graduate School of Science, Nagoya University, Nagoya, Aichi, Japan; Department of Theoretical and Computational Molecular Science, Institute for Molecular Science, Okazaki, Aichi, Japan
| | - Yoshitake Sakae
- Department of Physics, Graduate School of Science, Nagoya University, Nagoya, Aichi, Japan
| | - Takuya Gouda
- Department of Physics, Graduate School of Science, Nagoya University, Nagoya, Aichi, Japan
| | - Yuichiro Tsujimura
- Department of Physics, Graduate School of Science, Nagoya University, Nagoya, Aichi, Japan
| | - Yuko Okamoto
- Department of Physics, Graduate School of Science, Nagoya University, Nagoya, Aichi, Japan; Structural Biology Research Center, Graduate School of Science, Nagoya University, Nagoya, Aichi, Japan; Center for Computational Science, Graduate School of Engineering, Nagoya University, Nagoya, Aichi, Japan; Information Technology Center, Nagoya University, Nagoya, Aichi, Japan; JST-CREST, Nagoya, Aichi, Japan.
| |
Collapse
|
28
|
When safeguarding goes wrong: Impact of oxidative stress on protein homeostasis in health and neurodegenerative disorders. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2018; 114:221-264. [PMID: 30635082 DOI: 10.1016/bs.apcsb.2018.11.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cellular redox status is an established player in many different cellular functions. The buildup of oxidants within the cell is tightly regulated to maintain a balance between the positive and negative outcomes of cellular oxidants. Proteins are highly sensitive to oxidation, since modification can cause widespread unfolding and the formation of toxic aggregates. In response, cells have developed highly regulated systems that contribute to the maintenance of both the global redox status and protein homeostasis at large. Changes to these systems have been found to correlate with aging and age-related disorders, such as neurodegenerative pathologies. This raises intriguing questions as to the source of the imbalance in the redox and protein homeostasis systems, their interconnectivity, and their role in disease progression. Here we focus on the crosstalk between the redox and protein homeostasis systems in neurodegenerative diseases, specifically in Alzheimer's, Parkinson's, and ALS. We elaborate on some of the main players of the stress response systems, including the master regulators of oxidative stress and the heat shock response, Nrf2 and Hsf1, which are essential features of protein folding, and mediators of protein turnover. We illustrate the elegant mechanisms used by these components to provide an immediate response, including protein plasticity controlled by redox-sensing cysteines and the recruitment of naive proteins to the redox homeostasis array that act as chaperons in an ATP-independent manner.
Collapse
|
29
|
Morsy A, Trippier PC. Amyloid-Binding Alcohol Dehydrogenase (ABAD) Inhibitors for the Treatment of Alzheimer’s Disease. J Med Chem 2018; 62:4252-4264. [DOI: 10.1021/acs.jmedchem.8b01530] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Ahmed Morsy
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas 79106, United States
| | - Paul C. Trippier
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas 79106, United States
- Center for Chemical Biology, Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, Texas 79409, United States
| |
Collapse
|
30
|
Shi R, Guberman M, Kirshenbaum LA. Mitochondrial quality control: The role of mitophagy in aging. Trends Cardiovasc Med 2018; 28:246-260. [DOI: 10.1016/j.tcm.2017.11.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 11/27/2017] [Accepted: 11/28/2017] [Indexed: 12/25/2022]
|
31
|
Reddy PH, Manczak M, Yin X. Mitochondria-Division Inhibitor 1 Protects Against Amyloid-β induced Mitochondrial Fragmentation and Synaptic Damage in Alzheimer's Disease. J Alzheimers Dis 2018; 58:147-162. [PMID: 28409745 DOI: 10.3233/jad-170051] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The purpose our study was to determine the protective effects of mitochondria division inhibitor 1 (Mdivi1) in Alzheimer's disease (AD). Mdivi1 is hypothesized to reduce excessive fragmentation of mitochondria and mitochondrial dysfunction in AD neurons. Very little is known about whether Mdivi1 can confer protective effects in AD. In the present study, we sought to determine the protective effects of Mdivi1 against amyloid-β (Aβ)- and mitochondrial fission protein, dynamin-related protein 1 (Drp1)-induced excessive fragmentation of mitochondria in AD progression. We also studied preventive (Mdivi1+Aβ42) and intervention (Aβ42+Mdivi1) effects against Aβ42 in N2a cells. Using real-time RT-PCR and immunoblotting analysis, we measured mRNA and protein levels of mitochondrial dynamics, mitochondrial biogenesis, and synaptic genes. We also assessed mitochondrial function by measuring H2O2, lipid peroxidation, cytochrome oxidase activity, and mitochondrial ATP. MTT assays were used to assess the cell viability. Aβ42 was found to impair mitochondrial dynamics, lower mitochondrial biogenesis, lower synaptic activity, and lower mitochondrial function. On the contrary, Mdivi1 enhanced mitochondrial fusion activity, lowered fission machinery, and increased biogenesis and synaptic proteins. Mitochondrial function and cell viability were elevated in Mdivi1-treated cells. Interestingly, Mdivi1 pre- and post-treated cells treated with Aβ showed reduced mitochondrial dysfunction, and maintained cell viability, mitochondrial dynamics, mitochondrial biogenesis, and synaptic activity. The protective effects of Mdivi1 were stronger in N2a+Aβ42 pre-treated with Mdivi1, than in N2a+Aβ42 cells than Mdivi1 post-treated cells, indicating that Mdivi1 works better in prevention than treatment in AD like neurons.
Collapse
Affiliation(s)
- P Hemachandra Reddy
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, USA.,Garrison Institute on Aging, South West Campus, Texas Tech University Health Sciences Center, Lubbock, TX, USA.,Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, USA.,Deparment of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, USA.,Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX, USA.,Department of Speech, Language and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX, USA.,Department of Public Health, Graduate School of Biomedical Sciences, Lubbock, TX, USA
| | - Maria Manczak
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - XiangLing Yin
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| |
Collapse
|
32
|
Navarro-Mabarak C, Camacho-Carranza R, Espinosa-Aguirre JJ. Cytochrome P450 in the central nervous system as a therapeutic target in neurodegenerative diseases. Drug Metab Rev 2018; 50:95-108. [DOI: 10.1080/03602532.2018.1439502] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Cynthia Navarro-Mabarak
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Rafael Camacho-Carranza
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Jesús Javier Espinosa-Aguirre
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, México
| |
Collapse
|
33
|
Oxidant/Antioxidant Imbalance in Alzheimer's Disease: Therapeutic and Diagnostic Prospects. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:6435861. [PMID: 29636850 PMCID: PMC5831771 DOI: 10.1155/2018/6435861] [Citation(s) in RCA: 157] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 12/18/2017] [Indexed: 12/16/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia and a great socioeconomic burden in the aging society. Compelling evidence demonstrates that molecular change characteristics for AD, such as oxidative stress and amyloid β (Aβ) oligomerization, precede by decades the onset of clinical dementia and that the disease represents a biological and clinical continuum of stages, from asymptomatic to severely impaired. Nevertheless, the sequence of the early molecular alterations and the interplay between them are incompletely understood. This review presents current knowledge about the oxidative stress-induced impairments and compromised oxidative stress defense mechanisms in AD brain and the cross-talk between various pathophysiological insults, with the focus on excessive reactive oxygen species (ROS) generation and Aβ overproduction at the early stages of the disease. Prospects for AD therapies targeting oxidant/antioxidant imbalance are being discussed, as well as for the development of novel oxidative stress-related, blood-based biomarkers for early, noninvasive AD diagnostics.
Collapse
|
34
|
Adiele RC, Adiele CA. Mitochondrial Regulatory Pathways in the Pathogenesis of Alzheimer's Disease. J Alzheimers Dis 2018; 53:1257-70. [PMID: 27392851 DOI: 10.3233/jad-150967] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD) is an age-associated neurodegenerative brain disorder with progressive cognitive decline that leads to terminal dementia and death. For decades, amyloid-beta (Aβ) and neurofibrillary tangle (NFT) aggregation hypotheses have dominated studies on the pathogenesis and identification of potential therapeutic targets in AD. Little attention has been paid to the mitochondrial molecular/biochemical pathways leading to AD. Mitochondria play a critical role in cell viability and death including neurons and neuroglia, not only because they regulate energy and oxygen metabolism but also because they regulate cell death pathways. Mitochondrial impairment and oxidative stress are implicated in the pathogenesis of AD. Interestingly, current therapeutics provide symptomatic benefits to AD patients resulting in the use of preventive trials on presymptomatic subjects. This review article elucidates the pathophysiology of AD and emphasizes the need to explore the mitochondrial pathways to provide solutions to unanswered questions in the prevention and treatment of AD.
Collapse
Affiliation(s)
- Reginald C Adiele
- Department of Physiology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Chiedukam A Adiele
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, University of Nigeria, Nsukka, Nigeria
| |
Collapse
|
35
|
Zheng Y, Pan Q, Mo L, Zhang W, Duan Y, Chen C, Chen H, Guo Y, Shi X, Yang J. Monascuspigment rubropunctatin derivative FZU-H reduces Aβ(1-42)-induced neurotoxicity in Neuro-2A cells. RSC Adv 2018; 8:17389-17398. [PMID: 35539257 PMCID: PMC9080402 DOI: 10.1039/c8ra02365d] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Accepted: 05/08/2018] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's disease (AD) is an extremely complex disease, characterized by several pathological features including oxidative stress and amyloid-β (Aβ) aggregation. Blockage of Aβ-induced injury has emerged as a potential therapeutic approach for AD. Our previous efforts resulted in the discovery of Monascus pigment rubropunctatin derivative FZU-H with potential neuroprotective effects. This novel lead compound significantly diminishes toxicity induced by Aβ(1-42) in Neuro-2A cells. Our further mechanism investigation revealed that FZU-H inhibited Aβ(1-42)-induced caspase-3 protein activation and the loss of mitochondrial membrane potential. In addition, treatment of FZU-H was proven to attenuate Aβ(1-42)-induced cell redox imbalance and Tau hyperphosphorylation which caused by okadaic acid in Neuro-2A cells. These results indicated that FZU-H shows promising neuroprotective effects for AD. Monascus pigment rubropunctatin derivative FZU-H shows promising neuroprotective effects for AD.![]()
Collapse
Affiliation(s)
- Yunquan Zheng
- College of Chemistry
- Fuzhou University
- Fuzhou 350116
- China
- Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology
| | - Qisheng Pan
- College of Chemistry
- Fuzhou University
- Fuzhou 350116
- China
| | - Liuda Mo
- College of Chemistry
- Fuzhou University
- Fuzhou 350116
- China
| | - Wenyi Zhang
- College of Chemistry
- Fuzhou University
- Fuzhou 350116
- China
| | - Yunjian Duan
- College of Chemistry
- Fuzhou University
- Fuzhou 350116
- China
| | - Chengqun Chen
- Department of Chemical Engineering
- Fuzhou University
- Zhicheng College
- Fuzhou 350002
- China
| | - Haijun Chen
- College of Chemistry
- Fuzhou University
- Fuzhou 350116
- China
| | - Yanghao Guo
- Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology
- Fuzhou University
- Fuzhou 350116
- China
| | - Xianai Shi
- Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology
- Fuzhou University
- Fuzhou 350116
- China
| | - Jianmin Yang
- Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology
- Fuzhou University
- Fuzhou 350116
- China
| |
Collapse
|
36
|
Jodeiri Farshbaf M, Kiani-Esfahani A. Succinate dehydrogenase: Prospect for neurodegenerative diseases. Mitochondrion 2017; 42:77-83. [PMID: 29225013 DOI: 10.1016/j.mito.2017.12.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 11/25/2017] [Accepted: 12/06/2017] [Indexed: 12/13/2022]
Abstract
Onset of Alzheimer's, Parkinson's and Huntington's diseases as neurodegenerative disorders is increased by age. Alleviation of clinical symptoms and protection of neurons against degeneration are the main aspects of researches to establish new therapeutic strategies. Many studies have shown that mitochondria play crucial roles in high energy demand tissues like brain. Impairments in mitochondrial activity and physiology can makes neurons vulnerable to stress and degeneration. Succinate dehydrogenase (SDH) connects tricarboxylic cycle to the electron transport chain. Therefore, dysfunction of the SDH could impair mitochondrial activity, ATP generation and energy hemostasis in the cell. Exceed lipid synthesis, induction of the excitotoxicity in neurodegenerative disorders could be controlled by SDH through direct and indirect mechanism. In addition, mutation in SDH correlates with the onset of neurodegenerative disorders. Therefore, SDH could behave as a key regulator in neuroprotection. This review will present recent findings which are about SDH activity and related pathways which could play important roles in neuronal survival. Additionally, we will discuss about all possibilities which candidate SDH as a neuroprotective agent.
Collapse
Affiliation(s)
| | - Abbas Kiani-Esfahani
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan 816513-1378, Iran
| |
Collapse
|
37
|
Fivenson EM, Lautrup S, Sun N, Scheibye-Knudsen M, Stevnsner T, Nilsen H, Bohr VA, Fang EF. Mitophagy in neurodegeneration and aging. Neurochem Int 2017; 109:202-209. [PMID: 28235551 PMCID: PMC5565781 DOI: 10.1016/j.neuint.2017.02.007] [Citation(s) in RCA: 270] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 02/16/2017] [Indexed: 12/17/2022]
Abstract
Mitochondrial dysfunction contributes to normal aging and a wide spectrum of age-related diseases, including neurodegenerative disorders such as Parkinson's disease and Alzheimer's disease. It is important to maintain a healthy mitochondrial population which is tightly regulated by proteolysis and mitophagy. Mitophagy is a specialized form of autophagy that regulates the turnover of damaged and dysfunctional mitochondria, organelles that function in producing energy for the cell in the form of ATP and regulating energy homeostasis. Mechanistic studies on mitophagy across species highlight a sophisticated and integrated cellular network that regulates the degradation of mitochondria. Strategies directed at maintaining a healthy mitophagy level in aged individuals might have beneficial effects. In this review, we provide an updated mechanistic overview of mitophagy pathways and discuss the role of reduced mitophagy in neurodegeneration. We also highlight potential translational applications of mitophagy-inducing compounds, such as NAD+ precursors and urolithins.
Collapse
Affiliation(s)
- Elayne M Fivenson
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Sofie Lautrup
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA; Danish Aging Research Center, Department of Molecular Biology and Genetics, University of Aarhus, 8000 Aarhus C, Denmark
| | - Nuo Sun
- Center for Molecular Medicine, National Heart Lung and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Morten Scheibye-Knudsen
- Center for Healthy Aging, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Tinna Stevnsner
- Danish Aging Research Center, Department of Molecular Biology and Genetics, University of Aarhus, 8000 Aarhus C, Denmark
| | - Hilde Nilsen
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, 1478 Lørenskog, Norway
| | - Vilhelm A Bohr
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA; Center for Healthy Aging, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark.
| | - Evandro F Fang
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| |
Collapse
|
38
|
The biological foundation of the genetic association of TOMM40 with late-onset Alzheimer's disease. Biochim Biophys Acta Mol Basis Dis 2017; 1863:2973-2986. [PMID: 28768149 DOI: 10.1016/j.bbadis.2017.07.031] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Revised: 07/13/2017] [Accepted: 07/28/2017] [Indexed: 02/06/2023]
Abstract
A variable-length poly-T variant in intron 6 of the TOMM40 gene, rs10524523, is associated with risk and age-of-onset of sporadic (late-onset) Alzheimer's disease. In Caucasians, the three predominant alleles at this locus are Short (S), Long (L) or Very long (VL). On an APOE ε3/3 background, the S/VL and VL/VL genotypes are more protective than S/S. The '523 poly-T has regulatory properties, in that the VL poly-T results in higher expression than the S poly-T in luciferase expression systems. The aim of the current work was to identify effects on cellular bioenergetics of increased TOM40 protein expression. MitoTracker Green fluorescence and autophagic vesicle staining was the same in control and over-expressing cells, but TOM40 over-expression was associated with increased expression of TOM20, a preprotein receptor of the TOM complex, the mitochondrial chaperone HSPA9, and PDHE1a, and increased activities of the oxidative phosphorylation complexes I and IV and of the TCA member α-ketoglutaric acid dehydrogenase. Consistent with the complex I findings, respiration was more sensitive to inhibition by rotenone in control cells than in the TOM40 over-expressing cells. In the absence of inhibitors, total cellular ATP, the mitochondrial membrane potential, and respiration were elevated in the over-expressing cells. Spare respiratory capacity was greater in the TOM40 over-expressing cells than in the controls. TOM40 over-expression blocked Ab-elicited decreases in the mitochondrial membrane potential, cellular ATP levels, and cellular viability in the control cells. These data suggest elevated expression of TOM40 may be protective of mitochondrial function.
Collapse
|
39
|
Interaction of misfolded proteins and mitochondria in neurodegenerative disorders. Biochem Soc Trans 2017; 45:1025-1033. [PMID: 28733489 DOI: 10.1042/bst20170024] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 06/20/2017] [Accepted: 06/23/2017] [Indexed: 12/17/2022]
Abstract
The number of the people affected by neurodegenerative disorders is growing dramatically due to the ageing of population. The major neurodegenerative diseases share some common pathological features including the involvement of mitochondria in the mechanism of pathology and misfolding and the accumulation of abnormally aggregated proteins. Neurotoxicity of aggregated β-amyloid, tau, α-synuclein and huntingtin is linked to the effects of these proteins on mitochondria. All these misfolded aggregates affect mitochondrial energy metabolism by inhibiting diverse mitochondrial complexes and limit ATP availability in neurones. β-Amyloid, tau, α-synuclein and huntingtin are shown to be involved in increased production of reactive oxygen species, which can be generated in mitochondria or can target this organelle. Most of these aggregated proteins are capable of deregulating mitochondrial calcium handling that, in combination with oxidative stress, lead to opening of the mitochondrial permeability transition pore. Despite some of the common features, aggregated β-amyloid, tau, α-synuclein and huntingtin have diverse targets in mitochondria that can partially explain neurotoxic effect of these proteins in different brain regions.
Collapse
|
40
|
Wilkins HM, Swerdlow RH. Amyloid precursor protein processing and bioenergetics. Brain Res Bull 2016; 133:71-79. [PMID: 27545490 DOI: 10.1016/j.brainresbull.2016.08.009] [Citation(s) in RCA: 147] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Revised: 08/15/2016] [Accepted: 08/17/2016] [Indexed: 02/08/2023]
Abstract
The processing of amyloid precursor protein (APP) to amyloid beta (Aβ) is of great interest to the Alzheimer's disease (AD) field. Decades of research define how APP is altered to form Aβ, and how Aβ generates oligomers, protofibrils, and fibrils. Numerous signaling pathways and changes in cell physiology are known to influence APP processing. Existing data additionally indicate a relationship exists between mitochondria, bioenergetics, and APP processing. Here, we review data that address whether mitochondrial function and bioenergetics modify APP processing and Aβ production.
Collapse
Affiliation(s)
- Heather M Wilkins
- Department of Neurology University of Kansas Medical Center, Kansas City, KS, USA; University of Kansas Alzheimer's Disease Center, Kansas City, KS, USA
| | - Russell H Swerdlow
- Department of Neurology University of Kansas Medical Center, Kansas City, KS, USA; University of Kansas Alzheimer's Disease Center, Kansas City, KS, USA; Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA; Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS USA.
| |
Collapse
|
41
|
Ghobeh M, Ahmadian S, Meratan AA, Ebrahim-Habibi A, Ghasemi A, Shafizadeh M, Nemat-Gorgani M. Interaction of Aβ(25-35) fibrillation products with mitochondria: Effect of small-molecule natural products. Biopolymers 2016; 102:473-86. [PMID: 25297917 DOI: 10.1002/bip.22572] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Revised: 09/18/2014] [Accepted: 10/06/2014] [Indexed: 12/25/2022]
Abstract
The 25-35 fragment of the amyloid β (Aβ) peptide is a naturally occurring proteolytic by-product that retains the pathophysiology of its larger parent molecule, whose deposition has been shown to involve mitochondrial dysfunction. Hence, disruption of Aβ(25-35) aggregates could afford an effective remedial strategy for Alzheimer's disease (AD). In the present study, the effect of a number of selected small-molecule natural products (polyphenols: resveratrol, quercetin, biochanin A, and indoles: indole-3-acetic acid, indole-3-carbinol (I3C)) on Aβ(25-35) fibrillogenesis was explored under physiological conditions, and interaction of the resulting structures with rat brain mitochondria was investigated. Several techniques, including fluorescence, circular dichroism, and transmission electron microscopy were utilized to characterize the aggregation products, and possible mitochondrial membrane permeabilization was determined following release of marker enzymes. Results demonstrate the capacity of Aβ(25-35) fibrils to damage mitochondria and suggest how small molecules may afford protection. While I3C appeared more effective in inhibiting the fibrillation process, all natural products behaved similarly in destabilizing preformed aggregates. It is concluded that elucidation of such protection may provide important insights into the development of preventive and therapeutic agents for AD.
Collapse
Affiliation(s)
- Maryam Ghobeh
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | | | | | | | | | | | | |
Collapse
|
42
|
Diabetes and Alzheimer’s disease crosstalk. Neurosci Biobehav Rev 2016; 64:272-87. [PMID: 26969101 DOI: 10.1016/j.neubiorev.2016.03.005] [Citation(s) in RCA: 166] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 01/26/2016] [Accepted: 03/04/2016] [Indexed: 12/12/2022]
|
43
|
Pahrudin Arrozi A, Wan Ngah WZ, Mohd Yusof YA, Ahmad Damanhuri MH, Makpol S. Antioxidant modulation in restoring mitochondrial function in neurodegeneration. Int J Neurosci 2016; 127:218-235. [PMID: 27074540 DOI: 10.1080/00207454.2016.1178261] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Alzheimer's disease (AD) and Parkinson's disease (PD) are the leading causes of disability associated with neurodegeneration worldwide. These diseases are influenced by multiple genetic and environmental factors and share similar mechanisms as both are characterized by accumulation and aggregation of misfolded proteins - amyloid-beta (Aβ) in AD and α-synuclein in PD. Over the past decade, increasing evidence has shown that mitochondrial dysfunction and the generation of reactive oxygen species (ROS) are involved in the pathology of these diseases, and the contributions of these defects to the cellular and molecular changes that eventually cause neuronal death have been explored. Using mitochondrial protective agents, such as antioxidants, to combat ROS provides a new strategy for neurodegenerative treatment. In this review, we highlight the potential of multiple types of antioxidants, including vitamins, phytochemicals, fatty acids and minerals, as well as synthetic antioxidants specifically targeting the mitochondria, which can restore mitochondrial function, in the treatment of neurodegenerative disorders at both the pre-clinical and clinical stages by focusing on AD and PD.
Collapse
Affiliation(s)
- Aslina Pahrudin Arrozi
- a Department of Biochemistry , Universiti Kebangsaan Malaysia Medical Center , Kuala Lumpur , Malaysia
| | - Wan Zurinah Wan Ngah
- a Department of Biochemistry , Universiti Kebangsaan Malaysia Medical Center , Kuala Lumpur , Malaysia
| | - Yasmin Anum Mohd Yusof
- a Department of Biochemistry , Universiti Kebangsaan Malaysia Medical Center , Kuala Lumpur , Malaysia
| | | | - Suzana Makpol
- a Department of Biochemistry , Universiti Kebangsaan Malaysia Medical Center , Kuala Lumpur , Malaysia
| |
Collapse
|
44
|
The Dual Function of Reactive Oxygen/Nitrogen Species in Bioenergetics and Cell Death: The Role of ATP Synthase. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:3869610. [PMID: 27034734 PMCID: PMC4806282 DOI: 10.1155/2016/3869610] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 02/15/2016] [Indexed: 01/11/2023]
Abstract
Reactive oxygen species (ROS) and reactive nitrogen species (RNS) targeting mitochondria are major causative factors in disease pathogenesis. The mitochondrial permeability transition pore (PTP) is a mega-channel modulated by calcium and ROS/RNS modifications and it has been described to play a crucial role in many pathophysiological events since prolonged channel opening causes cell death. The recent identification that dimers of ATP synthase form the PTP and the fact that posttranslational modifications caused by ROS/RNS also affect cellular bioenergetics through the modulation of ATP synthase catalysis reveal a dual function of these modifications in the cells. Here, we describe mitochondria as a major site of production and as a target of ROS/RNS and discuss the pathophysiological conditions in which oxidative and nitrosative modifications modulate the catalytic and pore-forming activities of ATP synthase.
Collapse
|
45
|
Almeida S, Alves MG, Sousa M, Oliveira PF, Silva BM. Are Polyphenols Strong Dietary Agents Against Neurotoxicity and Neurodegeneration? Neurotox Res 2016; 30:345-66. [PMID: 26745969 DOI: 10.1007/s12640-015-9590-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 12/10/2015] [Accepted: 12/12/2015] [Indexed: 12/21/2022]
Abstract
Life expectancy of most human populations has greatly increased as a result of factors including better hygiene, medical practice, and nutrition. Unfortunately, as humans age, they become more prone to suffer from neurodegenerative diseases and neurotoxicity. Polyphenols can be cheaply and easily obtained as part of a healthy diet. They present a wide range of biological activities, many of which have relevance for human health. Compelling evidence has shown that dietary phytochemicals, particularly polyphenols, have properties that may suppress neuroinflammation and prevent toxic and degenerative effects in the brain. The mechanisms by which polyphenols exert their action are not fully understood, but it is clear that they have a direct effect through their antioxidant activities. They have also been shown to modulate intracellular signaling cascades, including the PI3K-Akt, MAPK, Nrf2, and MEK pathways. Polyphenols also interact with a range of neurotransmitters, illustrating that these compounds can promote their health benefits in the brain through a direct, indirect, or complex action. We discuss whether polyphenols obtained from diet or food supplements are an effective strategy to prevent or treat neurodegeneration. We also discuss the safety, mechanisms of action, and the current and future relevance of polyphenols in clinical treatment of neurodegenerative diseases. As populations age, it is important to discuss the dietary strategies to avoid or counteract the effects of incurable neurodegenerative disorders, which already represent an enormous financial and emotional burden for health care systems, patients, and their families.
Collapse
Affiliation(s)
- Susana Almeida
- Department of Microscopy, Laboratory of Cell Biology and Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Marco G Alves
- CICS-UBI, Health Sciences Research Centre, Faculty of Health Sciences, University of Beira Interior, Av. Infante D. Henrique, 6200-506, Covilhã, Portugal
| | - Mário Sousa
- Department of Microscopy, Laboratory of Cell Biology and Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal.,Centre for Reproductive Genetics Prof. Alberto Barros, Porto, Portugal
| | - Pedro F Oliveira
- Department of Microscopy, Laboratory of Cell Biology and Unit for Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal.,I3S - Institute of Health Research and Innovation, University of Porto, Porto, Portugal
| | - Branca M Silva
- CICS-UBI, Health Sciences Research Centre, Faculty of Health Sciences, University of Beira Interior, Av. Infante D. Henrique, 6200-506, Covilhã, Portugal.
| |
Collapse
|
46
|
Killing Me Softly: Connotations to Unfolded Protein Response and Oxidative Stress in Alzheimer's Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:1805304. [PMID: 26881014 PMCID: PMC4736771 DOI: 10.1155/2016/1805304] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 11/28/2015] [Accepted: 12/07/2015] [Indexed: 11/18/2022]
Abstract
This review is focused on the possible causes of mitochondrial dysfunction in AD, underlying molecular mechanisms of this malfunction, possible causes and known consequences of APP, Aβ, and hyperphosphorylated tau presence in mitochondria, and the contribution of altered lipid metabolism (nonsterol isoprenoids) to pathological processes leading to increased formation and accumulation of the aforementioned hallmarks of AD. Abnormal protein folding and unfolded protein response seem to be the outcomes of impaired glycosylation due to metabolic disturbances in geranylgeraniol intermediary metabolism. The origin and consecutive fate of APP, Aβ, and tau are emphasized on intracellular trafficking apparently influenced by inaccurate posttranslational modifications. We hypothesize that incorrect intracellular processing of APP determines protein translocation to mitochondria in AD. Similarly, without obvious reasons, the passage of Aβ and tau to mitochondria is observed. APP targeted to mitochondria blocks the activity of protein translocase complex resulting in poor import of proteins central to oxidative phosphorylation. Besides, APP, Aβ, and neurofibrillary tangles of tau directly or indirectly impair mitochondrial biochemistry and bioenergetics, with concomitant generation of oxidative/nitrosative stress. Limited protective mechanisms are inadequate to prevent the free radical-mediated lesions. Finally, neuronal loss is observed in AD-affected brains typically by pathologic apoptosis.
Collapse
|
47
|
Dong W, Wang F, Guo W, Zheng X, Chen Y, Zhang W, Shi H. Aβ25-35 Suppresses Mitochondrial Biogenesis in Primary Hippocampal Neurons. Cell Mol Neurobiol 2016; 36:83-91. [PMID: 26055049 PMCID: PMC11482447 DOI: 10.1007/s10571-015-0222-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Accepted: 06/02/2015] [Indexed: 12/11/2022]
Abstract
Mitochondrial biogenesis is involved in the regulation of mitochondrial content, morphology, and function. Impaired mitochondrial biogenesis has been observed in Alzheimer's disease. Amyloid-β (Aβ) has been shown to cause mitochondrial dysfunction in cultured neurons, but its role in mitochondrial biogenesis in neurons remains poorly defined. AMP-activated protein kinase (AMPK) and sirtuin 1 (SIRT1) are key energy-sensing molecules regulating mitochondrial biogenesis. In addition, peroxisome proliferator-activated receptor-γ coactivator 1-alpha (PGC-1α), the master regulator of mitochondrial biogenesis, is a target for SIRT1 deacetylase activity. In this study, we investigated the effects of Aβ25-35 on mitochondrial biogenesis in cultured hippocampal neurons and the underlying mechanisms. In primary hippocampal neurons, we found that 24-h incubation with Aβ25-35 suppressed both phosphorylations of AMPK and SIRT1 expression and increased PGC-1α acetylation expression. In addition, Aβ25-35 also resulted in a decrease in mitochondrial DNA copy number, as well as decreases in the expression of mitochondrial biogenesis factors (PGC-1α, NRF 1, NRF 2, and Tfam). Taken together, these data show that Aβ25-35 suppresses mitochondrial biogenesis in hippocampal neurons. Aβ25-35-induced impairment of mitochondrial biogenesis may be associated with the inhibition of the AMPK-SIRT1-PGC-1α pathway.
Collapse
Affiliation(s)
- Weiguo Dong
- Department of Integrated Traditional Chinese and Western Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, People's Republic of China.
| | - Feng Wang
- Department of Integrated Traditional Chinese and Western Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, People's Republic of China
| | - Wanqing Guo
- The Third People's Hospital of Fujian Province, Fuzhou, 350122, Fujian, People's Republic of China
| | - Xuehua Zheng
- Department of Integrated Traditional Chinese and Western Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, People's Republic of China
| | - Yue Chen
- Department of Integrated Traditional Chinese and Western Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, People's Republic of China
| | - Wenguang Zhang
- Department of Integrated Traditional Chinese and Western Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, People's Republic of China
| | - Hong Shi
- Department of Integrated Traditional Chinese and Western Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, People's Republic of China
| |
Collapse
|
48
|
Findlay JA, Hamilton DL, Ashford MLJ. BACE1 activity impairs neuronal glucose oxidation: rescue by beta-hydroxybutyrate and lipoic acid. Front Cell Neurosci 2015; 9:382. [PMID: 26483636 PMCID: PMC4589671 DOI: 10.3389/fncel.2015.00382] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 09/14/2015] [Indexed: 11/13/2022] Open
Abstract
Glucose hypometabolism and impaired mitochondrial function in neurons have been suggested to play early and perhaps causative roles in Alzheimer's disease (AD) pathogenesis. Activity of the aspartic acid protease, beta-site amyloid precursor protein (APP) cleaving enzyme 1 (BACE1), responsible for beta amyloid peptide generation, has recently been demonstrated to modify glucose metabolism. We therefore examined, using a human neuroblastoma (SH-SY5Y) cell line, whether increased BACE1 activity is responsible for a reduction in cellular glucose metabolism. Overexpression of active BACE1, but not a protease-dead mutant BACE1, protein in SH-SY5Y cells reduced glucose oxidation and the basal oxygen consumption rate, which was associated with a compensatory increase in glycolysis. Increased BACE1 activity had no effect on the mitochondrial electron transfer process but was found to diminish substrate delivery to the mitochondria by inhibition of key mitochondrial decarboxylation reaction enzymes. This BACE1 activity-dependent deficit in glucose oxidation was alleviated by the presence of beta hydroxybutyrate or α-lipoic acid. Consequently our data indicate that raised cellular BACE1 activity drives reduced glucose oxidation in a human neuronal cell line through impairments in the activity of specific tricarboxylic acid cycle enzymes. Because this bioenergetic deficit is recoverable by neutraceutical compounds we suggest that such agents, perhaps in conjunction with BACE1 inhibitors, may be an effective therapeutic strategy in the early-stage management or treatment of AD.
Collapse
Affiliation(s)
- John A Findlay
- Division of Cardiovascular and Diabetes Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee Dundee, UK
| | - David L Hamilton
- Division of Cardiovascular and Diabetes Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee Dundee, UK
| | - Michael L J Ashford
- Division of Cardiovascular and Diabetes Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee Dundee, UK
| |
Collapse
|
49
|
Controlled and Impaired Mitochondrial Quality in Neurons: Molecular Physiology and Prospective Pharmacology. Pharmacol Res 2015; 99:410-24. [DOI: 10.1016/j.phrs.2015.03.021] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 03/27/2015] [Accepted: 03/27/2015] [Indexed: 01/08/2023]
|
50
|
Neuroprotection by Cocktails of Dietary Antioxidants under Conditions of Nerve Growth Factor Deprivation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:217258. [PMID: 26236423 PMCID: PMC4510258 DOI: 10.1155/2015/217258] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 06/09/2015] [Accepted: 06/10/2015] [Indexed: 12/12/2022]
Abstract
Dietary antioxidants may be useful in counteracting the chronic inflammatory status in neurodegenerative diseases by reducing oxidative stress due to accumulation of reactive oxygen species (ROS). In this study, we newly described the efficacy of a number of dietary antioxidants (polyphenols, carotenoids, thiolic compounds, and oligoelements) on viability of neuronal PC12 cells following Nerve Growth Factor (NGF) deprivation, a model of age-related decrease of neurotrophic support that triggers neuronal loss. Neuroprotection by antioxidants during NGF deprivation for 24 h was largely dependent on their concentrations: all dietary antioxidants were able to efficiently support cell viability by reducing ROS levels and restoring mitochondrial function, while preserving the neuronal morphology. Moreover, ROS reduction and neuroprotection during NGF withdrawal were also achieved with defined cocktails of 3-6 different antioxidants at concentrations 5-60 times lower than those used in single treatments, suggesting that their antioxidant activity was preserved also at very low concentrations. Overall, these data indicate the beneficial effects of antioxidants against oxidative stress induced by decreased NGF availability and suggest that defined cocktails of dietary factors at low concentrations might be a suitable strategy to reduce oxidative damage in neurodegenerative diseases, while limiting possible side effects.
Collapse
|