1
|
Reddi Sree R, Kalyan M, Anand N, Mani S, Gorantla VR, Sakharkar MK, Song BJ, Chidambaram SB. Newer Therapeutic Approaches in Treating Alzheimer's Disease: A Comprehensive Review. ACS OMEGA 2025; 10:5148-5171. [PMID: 39989768 PMCID: PMC11840625 DOI: 10.1021/acsomega.4c05527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 10/04/2024] [Accepted: 10/09/2024] [Indexed: 02/25/2025]
Abstract
Alzheimer's disease (AD) is an aging-related irreversible neurodegenerative disease affecting mostly the elderly population. The main pathological features of AD are the extracellular Aβ plaques generated by APP cleavage through the amyloidogenic pathway, the intracellular neurofibrillary tangles (NFT) resulting from the hyperphosphorylated tau proteins, and cholinergic neurodegeneration. However, the actual causes of AD are unknown, but several studies suggest hereditary mutations in PSEN1 and -2, APOE4, APP, and the TAU genes are the major perpetrators. In order to understand the etiology and pathogenesis of AD, various hypotheses are proposed. These include the following hypotheses: amyloid accumulation, tauopathy, inflammation, oxidative stress, mitochondrial dysfunction, glutamate/excitotoxicity, cholinergic deficiency, and gut dysbiosis. Currently approved therapeutic interventions are donepezil, galantamine, and rivastigmine, which are cholinesterase inhibitors (ChEIs), and memantine, which is an N-methyl-d-aspartate (NMDA) antagonist. These treatment strategies focus on only symptomatic management of AD by attenuating symptoms but not regeneration of neurons or clearance of Aβ plaques and hyperphosphorylated Tau. This review focuses on the pathophysiology, novel therapeutic targets, and disease-altering treatments such as α-secretase modulators, active immunotherapy, passive immunotherapy, natural antioxidant products, nanomaterials, antiamyloid therapy, tau aggregation inhibitors, transplantation of fecal microbiota or stem cells, and microtubule stabilizers that are in clinical trials or still under investigation.
Collapse
Affiliation(s)
- Radhakrishna Reddi Sree
- Department
of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
| | - Manjunath Kalyan
- Department
of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
- Centre
for Experimental Pharmacology & Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
| | - Nikhilesh Anand
- Department
of Pharmacology, American University of
Antigua College of Medicine, University Park, Jabberwock Beach Road, Coolidge, Antigua, Barbuda
| | - Sangeetha Mani
- Department
of Pharmacognosy, Sri Ramachandra Faculty of Pharmacy, Sri Ramachandra Institute of Higher Education and
Research, Porur, Chennai 600116, India
| | - Vasavi Rakesh Gorantla
- Department
of Anatomical Sciences, St. George’s University School of Medicine, St. George’s University, Saint George, Grenada
| | - Meena Kishore Sakharkar
- College
of
Pharmacy and Nutrition, University of Saskatchewan, 107 Wiggins Road, Saskatoon, Saskatchewan S7N 5C9, Canada
| | - Byoung-Joon Song
- Section
of Molecular Pharmacology and Toxicology, Laboratory of Membrane Biochemistry
and Biophysics, National Institute on Alcohol
Abuse and Alcoholism, National Institutes of Health, Rockville, Maryland 20892, United States
| | - Saravana Babu Chidambaram
- Department
of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
- Centre
for Experimental Pharmacology & Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
| |
Collapse
|
2
|
Huang M, Sui R, Zhang L, Zhu Y, Yuan X, Jiang H, Mao X. Rosavin thwarts amyloid-β-induced macromolecular damages and neurotoxicity, exhibiting anti-Alzheimer's disease activity in Wister rat model. Inflammopharmacology 2024; 32:1461-1474. [PMID: 37758932 DOI: 10.1007/s10787-023-01320-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 08/04/2023] [Indexed: 09/29/2023]
Abstract
Lately, interest surrounding the utilization of plant-derived compounds as a viable beneficial approach for treating Alzheimer's disease (AD) has significantly increased. This study aimed to assess the defensive properties of rosavin against Alzheimer's disease induced by amyloid-β, utilizing experimental models. We found that rosavin exhibited anti-aggregation and disaggregation properties, suggesting its potential to prevent the gathering of Aβ-aggregates. In vitro experiments revealed that rosavin effectively mitigated the neurotoxicity induced by Aβ in Neuro-2a cells, showcasing its protective potential. Rosavin significantly improved the Aβ-induced cognitive deficits in Wistar rats, particularly in spatial memory. Which the pathophysiology of AD includes oxidative damage, which negatively impacts biological macromolecules. Triggers the apoptotic process, causing macromolecular destruction. Interestingly, rosavin attenuated Aβ-induced macromolecular damages, thereby preserving neuronal integrity. Furthermore, the activation of antioxidative defense enzymes by rosavin inhibited oxidative damage. The positive outcomes associated with rosavin were primarily attributed to its capacity to enhance acetylcholine-mediated effects. Finally, rosavin has the potential to alleviate Aβ-induced neurotoxicity and macromolecular damages, ultimately resulting in enhanced memorial and reasoning function in Wistar rats, offering promising prospects for the treatment of AD.
Collapse
Affiliation(s)
- Meiyi Huang
- Department of Neurology, The First Affiliated Hospital of Jinzhou Medical University, No 2, Section 5, Renmin Street, Jinzhou, 121099, China
| | - Rubo Sui
- Department of Neurology, The First Affiliated Hospital of Jinzhou Medical University, No 2, Section 5, Renmin Street, Jinzhou, 121099, China.
| | - Lei Zhang
- School of Nursing, Jinzhou Medical University, Jinzhou, 121099, China
| | - Yue Zhu
- Department of Neurology, The First Affiliated Hospital of Jinzhou Medical University, No 2, Section 5, Renmin Street, Jinzhou, 121099, China
| | - Xueling Yuan
- Department of Neurology, The First Affiliated Hospital of Jinzhou Medical University, No 2, Section 5, Renmin Street, Jinzhou, 121099, China
| | - Hongxin Jiang
- Department of Radiology, Gucheng County Hospital, Gucheng, 253809, China
| | - Xin Mao
- Department of Neurology, The First Affiliated Hospital of Jinzhou Medical University, No 2, Section 5, Renmin Street, Jinzhou, 121099, China.
| |
Collapse
|
3
|
Pappolla MA, Martins RN, Poeggeler B, Omar RA, Perry G. Oxidative Stress in Alzheimer's Disease: The Shortcomings of Antioxidant Therapies. J Alzheimers Dis 2024; 101:S155-S178. [PMID: 39422961 DOI: 10.3233/jad-240659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by gradual and progressive cognitive decline leading to dementia. At its core, the neuropathological features of AD include hallmark accumulations of amyloid-β and hyperphosphorylated tau proteins. Other harmful processes, such as oxidative stress and inflammation, contribute to the disease's neuropathological progression. This review evaluates the role of oxidative stress in AD, placing a spotlight on the disappointing outcomes of various antioxidant clinical trials. Several hypotheses are discussed that might elucidate the failures of these therapies in AD. Specifically: 1) The paradoxical and overlooked harmful implications of prooxidant intermediates, particularly stemming from conventional antioxidants like vitamins E and C; 2) The challenges and failure to appreciate the issue of bioavailability-epitomized by the dictum "no on-site protection, no protection"-and the preeminent, yet often ignored, role played by endogenous antioxidant enzymes in combating oxidative stress; 3) The influence of unrecognized etiologies, such as latent infectious agents and others, as foundational drivers of oxidative stress in AD; 4) The underestimation of the complexity of oxidative mechanisms and the necessity of multi-targeted therapeutic approaches, such as those provided by various diets; and 5) The limitations of clinical trial designs in fully capturing the effects of antioxidants on AD progression. This article also examines the outcomes of select clinical trials while highlighting the challenges and barriers these therapies pose, offering insights into potential mechanisms to overcome their marginal success.
Collapse
Affiliation(s)
- Miguel A Pappolla
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
| | - Ralph N Martins
- Aging and Alzheimer's Disease Centre, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Australia
| | - Burkhard Poeggeler
- Johann-Friedrich-Blumenbach-Institute for Zoology and Anthropology, Faculty of Biology and Psychology, Georg August University, Gottingen, Germany
| | - Rawhi A Omar
- Department of Pathology, University of Louisville, Louisville, KY, USA
| | - George Perry
- Department of Neuroscience, Developmental and Regenerative Biology, University of Texas at San Antonio, San Antonio, TX, USA
| |
Collapse
|
4
|
Cruciani F, Aparo A, Brusini L, Combi C, Storti SF, Giugno R, Menegaz G, Boscolo Galazzo I. Identifying the joint signature of brain atrophy and gene variant scores in Alzheimer's Disease. J Biomed Inform 2024; 149:104569. [PMID: 38104851 DOI: 10.1016/j.jbi.2023.104569] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 11/20/2023] [Accepted: 12/07/2023] [Indexed: 12/19/2023]
Abstract
The joint modeling of genetic data and brain imaging information allows for determining the pathophysiological pathways of neurodegenerative diseases such as Alzheimer's disease (AD). This task has typically been approached using mass-univariate methods that rely on a complete set of Single Nucleotide Polymorphisms (SNPs) to assess their association with selected image-derived phenotypes (IDPs). However, such methods are prone to multiple comparisons bias and, most importantly, fail to account for potential cross-feature interactions, resulting in insufficient detection of significant associations. Ways to overcome these limitations while reducing the number of traits aim at conveying genetic information at the gene level and capturing the integrated genetic effects of a set of genetic variants, rather than looking at each SNP individually. Their associations with brain IDPs are still largely unexplored in the current literature, though they can uncover new potential genetic determinants for brain modulations in the AD continuum. In this work, we explored an explainable multivariate model to analyze the genetic basis of the grey matter modulations, relying on the AD Neuroimaging Initiative (ADNI) phase 3 dataset. Cortical thicknesses and subcortical volumes derived from T1-weighted Magnetic Resonance were considered to describe the imaging phenotypes. At the same time the genetic counterpart was represented by gene variant scores extracted by the Sequence Kernel Association Test (SKAT) filtering model. Moreover, transcriptomic analysis was carried on to assess the expression of the resulting genes in the main brain structures as a form of validation. Results highlighted meaningful genotype-phenotype interactionsas defined by three latent components showing a significant difference in the projection scores between patients and controls. Among the significant associations, the model highlighted EPHX1 and BCAS1 gene variant scores involved in neurodegenerative and myelination processes, hence relevant for AD. In particular, the first was associated with decreased subcortical volumes and the second with decreasedtemporal lobe thickness. Noteworthy, BCAS1 is particularly expressed in the dentate gyrus. Overall, the proposed approach allowed capturing genotype-phenotype interactions in a restricted study cohort that was confirmed by transcriptomic analysis, offering insights into the underlying mechanisms of neurodegeneration in AD in line with previous findings and suggesting new potential disease biomarkers.
Collapse
Affiliation(s)
- Federica Cruciani
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy.
| | - Antonino Aparo
- Department of Computer Science, University of Verona, Verona, Italy
| | - Lorenza Brusini
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | - Carlo Combi
- Department of Computer Science, University of Verona, Verona, Italy
| | - Silvia F Storti
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | - Rosalba Giugno
- Department of Computer Science, University of Verona, Verona, Italy
| | - Gloria Menegaz
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | | |
Collapse
|
5
|
Perluigi M, Di Domenico F, Butterfield DA. Oxidative damage in neurodegeneration: roles in the pathogenesis and progression of Alzheimer disease. Physiol Rev 2024; 104:103-197. [PMID: 37843394 PMCID: PMC11281823 DOI: 10.1152/physrev.00030.2022] [Citation(s) in RCA: 39] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 03/30/2023] [Accepted: 05/24/2023] [Indexed: 10/17/2023] Open
Abstract
Alzheimer disease (AD) is associated with multiple etiologies and pathological mechanisms, among which oxidative stress (OS) appears as a major determinant. Intriguingly, OS arises in various pathways regulating brain functions, and it seems to link different hypotheses and mechanisms of AD neuropathology with high fidelity. The brain is particularly vulnerable to oxidative damage, mainly because of its unique lipid composition, resulting in an amplified cascade of redox reactions that target several cellular components/functions ultimately leading to neurodegeneration. The present review highlights the "OS hypothesis of AD," including amyloid beta-peptide-associated mechanisms, the role of lipid and protein oxidation unraveled by redox proteomics, and the antioxidant strategies that have been investigated to modulate the progression of AD. Collected studies from our groups and others have contributed to unraveling the close relationships between perturbation of redox homeostasis in the brain and AD neuropathology by elucidating redox-regulated events potentially involved in both the pathogenesis and progression of AD. However, the complexity of AD pathological mechanisms requires an in-depth understanding of several major intracellular pathways affecting redox homeostasis and relevant for brain functions. This understanding is crucial to developing pharmacological strategies targeting OS-mediated toxicity that may potentially contribute to slow AD progression as well as improve the quality of life of persons with this severe dementing disorder.
Collapse
Affiliation(s)
- Marzia Perluigi
- Department of Biochemical Sciences "A. Rossi Fanelli," Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Fabio Di Domenico
- Department of Biochemical Sciences "A. Rossi Fanelli," Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - D Allan Butterfield
- Department of Chemistry and Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, United States
| |
Collapse
|
6
|
Zhang Y, Kiryu H. Identification of oxidative stress-related genes differentially expressed in Alzheimer's disease and construction of a hub gene-based diagnostic model. Sci Rep 2023; 13:6817. [PMID: 37100862 PMCID: PMC10133299 DOI: 10.1038/s41598-023-34021-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 04/22/2023] [Indexed: 04/28/2023] Open
Abstract
Alzheimer's disease (AD) is the most prevalent dementia disorder globally, and there are still no effective interventions for slowing or stopping the underlying pathogenic mechanisms. There is strong evidence implicating neural oxidative stress (OS) and ensuing neuroinflammation in the progressive neurodegeneration observed in the AD brain both during and prior to symptom emergence. Thus, OS-related biomarkers may be valuable for prognosis and provide clues to therapeutic targets during the early presymptomatic phase. In the current study, we gathered brain RNA-seq data of AD patients and matched controls from the Gene Expression Omnibus (GEO) to identify differentially expressed OS-related genes (OSRGs). These OSRGs were analyzed for cellular functions using the Gene Ontology (GO) database and used to construct a weighted gene co-expression network (WGCN) and protein-protein interaction (PPI) network. Receiver operating characteristic (ROC) curves were then constructed to identify network hub genes. A diagnostic model was established based on these hub genes using Least Absolute Shrinkage and Selection Operator (LASSO) and ROC analyses. Immune-related functions were examined by assessing correlations between hub gene expression and immune cell brain infiltration scores. Further, target drugs were predicted using the Drug-Gene Interaction database, while regulatory miRNAs and transcription factors were predicted using miRNet. In total, 156 candidate genes were identified among 11046 differentially expressed genes, 7098 genes in WGCN modules, and 446 OSRGs, and 5 hub genes (MAPK9, FOXO1, BCL2, ETS1, and SP1) were identified by ROC curve analyses. These hub genes were enriched in GO annotations "Alzheimer's disease pathway," "Parkinson's Disease," "Ribosome," and "Chronic myeloid leukemia." In addition, 78 drugs were predicted to target FOXO1, SP1, MAPK9, and BCL2, including fluorouracil, cyclophosphamide, and epirubicin. A hub gene-miRNA regulatory network with 43 miRNAs and hub gene-transcription factor (TF) network with 36 TFs were also generated. These hub genes may serve as biomarkers for AD diagnosis and provide clues to novel potential treatment targets.
Collapse
Affiliation(s)
- Yanting Zhang
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.
| | - Hisanori Kiryu
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| |
Collapse
|
7
|
Ngum JA, Tatang FJ, Toumeni MH, Nguengo SN, Simo USF, Mezajou CF, Kameni C, Ngongang NN, Tchinda MF, Dongho Dongmo FF, Akami M, Ngane Ngono AR, Tamgue O. An overview of natural products that modulate the expression of non-coding RNAs involved in oxidative stress and inflammation-associated disorders. Front Pharmacol 2023; 14:1144836. [PMID: 37168992 PMCID: PMC10165025 DOI: 10.3389/fphar.2023.1144836] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 03/24/2023] [Indexed: 05/13/2023] Open
Abstract
Oxidative stress is a state in which oxidants are produced in excess in the body's tissues and cells, resulting in a biological imbalance amid the generation of reactive oxygen and nitrogen species (RONS) from redox reactions. In case of insufficient antioxidants to balance, the immune system triggers signaling cascades to mount inflammatory responses. Oxidative stress can have deleterious effects on major macromolecules such as lipids, proteins, and nucleic acids, hence, Oxidative stress and inflammation are among the multiple factors contributing to the etiology of several disorders such as diabetes, cancers, and cardiovascular diseases. Non-coding RNAs (ncRNAs) which were once referred to as dark matter have been found to function as key regulators of gene expression through different mechanisms. They have dynamic roles in the onset and development of inflammatory and oxidative stress-related diseases, therefore, are potential targets for the control of those diseases. One way of controlling those diseases is through the use of natural products, a rich source of antioxidants that have drawn attention with several studies showing their involvement in combating chronic diseases given their enormous gains, low side effects, and toxicity. In this review, we highlighted the natural products that have been reported to target ncRNAs as mediators of their biological effects on oxidative stress and several inflammation-associated disorders. Those natural products include Baicalein, Tanshinone IIA, Geniposide, Carvacrol/Thymol, Triptolide, Oleacein, Curcumin, Resveratrol, Solarmargine, Allicin, aqueous extract or pulp of Açai, Quercetin, and Genistein. We also draw attention to some other compounds including Zanthoxylum bungeanum, Canna genus rhizome, Fuzi-ganjiang herb pair, Aronia melanocarpa, Peppermint, and Gingerol that are effective against oxidative stress and inflammation-related disorders, however, have no known effect on ncRNAs. Lastly, we touched on the many ncRNAs that were found to play a role in oxidative stress and inflammation-related disorders but have not yet been investigated as targets of a natural product. Shedding more light into these two last points of shadow will be of great interest in the valorization of natural compounds in the control and therapy of oxidative stress- and inflammation-associated disorders.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Ousman Tamgue
- Department of Biochemistry, Faculty of Sciences, University of Douala, Douala, Cameroon
| |
Collapse
|
8
|
Crosstalk between Oxidative Stress and Aging in Neurodegeneration Disorders. Cells 2023; 12:cells12050753. [PMID: 36899889 PMCID: PMC10001353 DOI: 10.3390/cells12050753] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/11/2023] [Accepted: 02/22/2023] [Indexed: 03/02/2023] Open
Abstract
The world population is aging rapidly, and increasing lifespan exacerbates the burden of age-related health issues. On the other hand, premature aging has begun to be a problem, with increasing numbers of younger people suffering aging-related symptoms. Advanced aging is caused by a combination of factors: lifestyle, diet, external and internal factors, as well as oxidative stress (OS). Although OS is the most researched aging factor, it is also the least understood. OS is important not only in relation to aging but also due to its strong impact on neurodegenerative diseases such as amyotrophic lateral sclerosis (ALS), frontotemporal dementia (FTD), Alzheimer's disease (AD), and Parkinson's disease (PD). In this review, we will discuss the aging process in relation to OS, the function of OS in neurodegenerative disorders, and prospective therapeutics capable of relieving neurodegenerative symptoms associated with the pro-oxidative condition.
Collapse
|
9
|
Almikhlafi MA, Karami MM, Jana A, Alqurashi TM, Majrashi M, Alghamdi BS, Ashraf GM. Mitochondrial Medicine: A Promising Therapeutic Option Against Various Neurodegenerative Disorders. Curr Neuropharmacol 2023; 21:1165-1183. [PMID: 36043795 PMCID: PMC10286591 DOI: 10.2174/1570159x20666220830112408] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/05/2022] [Accepted: 07/14/2022] [Indexed: 11/22/2022] Open
Abstract
Abnormal mitochondrial morphology and metabolic dysfunction have been observed in many neurodegenerative disorders (NDDs). Mitochondrial dysfunction can be caused by aberrant mitochondrial DNA, mutant nuclear proteins that interact with mitochondria directly or indirectly, or for unknown reasons. Since mitochondria play a significant role in neurodegeneration, mitochondriatargeted therapies represent a prosperous direction for the development of novel drug compounds that can be used to treat NDDs. This review gives a brief description of how mitochondrial abnormalities lead to various NDDs such as Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis. We further explore the promising therapeutic effectiveness of mitochondria- directed antioxidants, MitoQ, MitoVitE, MitoPBN, and dimebon. We have also discussed the possibility of mitochondrial gene therapy as a therapeutic option for these NDDs.
Collapse
Affiliation(s)
- Mohannad A. Almikhlafi
- Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, Madinah, Saudi Arabia
| | - Mohammed M. Karami
- Department of Physiology, Neuroscience Unit, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ankit Jana
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Campus-11, Patia, Bhubaneswar, Odisha, 751024, India
| | - Thamer M. Alqurashi
- Department of Pharmacology, Faculty of Medicine, Rabigh, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammed Majrashi
- Department of Pharmacology, Faculty of Medicine, University of Jeddah, Jeddah, Saudi Arabia
| | - Badrah S. Alghamdi
- Department of Physiology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- The Neuroscience Research Unit, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ghulam Md. Ashraf
- Department of Medical Laboratory Sciences, College of Health Sciences, University of Sharjah, University City, Sharjah 27272, United Arab Emirates
| |
Collapse
|
10
|
Zhang H, Liu X, Liu Y, Liu J, Gong X, Li G, Tang M. Crosstalk between regulatory non-coding RNAs and oxidative stress in Parkinson’s disease. Front Aging Neurosci 2022; 14:975248. [PMID: 36016854 PMCID: PMC9396353 DOI: 10.3389/fnagi.2022.975248] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
Parkinson’s disease is the second most common neurodegenerative disease after Alzheimer’s disease, which imposes an ever-increasing burden on society. Many studies have indicated that oxidative stress may play an important role in Parkinson’s disease through multiple processes related to dysfunction or loss of neurons. Besides, several subtypes of non-coding RNAs are found to be involved in this neurodegenerative disorder. However, the interplay between oxidative stress and regulatory non-coding RNAs in Parkinson’s disease remains to be clarified. In this article, we comprehensively survey and overview the role of regulatory ncRNAs in combination with oxidative stress in Parkinson’s disease. The interaction between them is also summarized. We aim to provide readers with a relatively novel insight into the pathogenesis of Parkinson’s disease, which would contribute to the development of pre-clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Hantao Zhang
- School of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Xiaoyan Liu
- School of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Yi Liu
- School of Life Sciences, Jiangsu University, Zhenjiang, China
- Institute of Animal Husbandry, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Junlin Liu
- School of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Xun Gong
- Department of Rheumatology & Immunology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Gang Li
- Department of Vascular Surgery, The Second Affiliated Hospital of Shandong First Medical University, Taian, China
- *Correspondence: Gang Li Min Tang
| | - Min Tang
- School of Life Sciences, Jiangsu University, Zhenjiang, China
- *Correspondence: Gang Li Min Tang
| |
Collapse
|
11
|
Zhou Y, Kandel N, Bartoli M, Serafim LF, ElMetwally AE, Falkenberg SM, Paredes XE, Nelson CJ, Smith N, Padovano E, Zhang W, Mintz KJ, Ferreira BC, Cilingir EK, Chen J, Shah SK, Prabhakar R, Tagliaferro A, Wang C, Leblanc RM. Structure-Activity Relationship of Carbon Nitride Dots in Inhibiting Tau Aggregation. CARBON 2022; 193:1-16. [PMID: 35463198 PMCID: PMC9030089 DOI: 10.1016/j.carbon.2022.03.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Due to the numerous failed clinical trials of anti-amyloid drugs, microtubule associated protein tau (MAPT) now stands out as one of the most promising targets for AD therapy. In this study, we report for the first time the structure-dependent MAPT aggregation inhibition of carbon nitride dots (CNDs). CNDs have exhibited great promise as a potential treatment of Alzheimer's disease (AD) by inhibiting the aggregation of MAPT. In order to elucidate its structure-activity relationship, CNDs were separated via column chromatography and five fractions with different structures were obtained that were characterized by multiple spectroscopy methods. The increase of surface hydrophilic functional groups is consistent with the increase of polarity from fraction 1 to 5. Particle sizes (1-2 nm) and zeta potentials (~-20 mV) are similar among five fractions. With the increase of polarity from fraction 1 to 5, their MAPT aggregation inhibition capacity was weakened. This suggests hydrophobic interactions between CNDs and MAPT, validated via molecular dynamics simulations. With a zebrafish blood-brain barrier (BBB) model, CNDs were observed to cross the BBB through passive diffusion. CNDs were also found to inhibit the generation of multiple reactive oxygen species, which is an important contributor to AD pathogenesis.
Collapse
Affiliation(s)
- Yiqun Zhou
- Department of Chemistry, University of Miami, Coral Gables, FL 33146, USA
- C-Dots, LLC, Miami, FL 33136, USA
| | - Nabin Kandel
- Department of Biological Sciences, Rensselaer Polytechnic Institute, NY 12180, USA
| | - Mattia Bartoli
- Center for Sustainable Future, Italian Institute of Technology, Via Livorno 60, Turin 10144, Italy
| | | | | | | | - Xavier E. Paredes
- Department of Chemistry, University of Miami, Coral Gables, FL 33146, USA
| | | | - Nathan Smith
- Department of Biological Sciences, Rensselaer Polytechnic Institute, NY 12180, USA
| | - Elisa Padovano
- Department of Applied Science and Technology, Politecnico di Torino, Italy
| | - Wei Zhang
- Department of Chemistry, University of Miami, Coral Gables, FL 33146, USA
| | - Keenan J. Mintz
- Department of Chemistry, University of Miami, Coral Gables, FL 33146, USA
- School of Materials Science and Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | | | | | - Jiuyan Chen
- Department of Chemistry, University of Miami, Coral Gables, FL 33146, USA
| | - Sujit K. Shah
- Department of Chemistry, University of Miami, Coral Gables, FL 33146, USA
- Department of Chemistry, Mahendra Morang Adarsh Multiple Campus, Tribhuvan University, Biratnagar 56613, Nepal
| | - Rajeev Prabhakar
- Department of Chemistry, University of Miami, Coral Gables, FL 33146, USA
| | | | - Chunyu Wang
- Department of Biological Sciences, Rensselaer Polytechnic Institute, NY 12180, USA
| | - Roger M. Leblanc
- Department of Chemistry, University of Miami, Coral Gables, FL 33146, USA
| |
Collapse
|
12
|
Scuto M, Modafferi S, Rampulla F, Zimbone V, Tomasello M, Spano’ S, Ontario M, Palmeri A, Trovato Salinaro A, Siracusa R, Di Paola R, Cuzzocrea S, Calabrese E, Wenzel U, Calabrese V. Redox modulation of stress resilience by Crocus Sativus L. for potential neuroprotective and anti-neuroinflammatory applications in brain disorders: From molecular basis to therapy. Mech Ageing Dev 2022; 205:111686. [DOI: 10.1016/j.mad.2022.111686] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/18/2022] [Accepted: 05/18/2022] [Indexed: 12/13/2022]
|
13
|
Oyeleke MB, Owoyele BV. Saponins and flavonoids from Bacopa floribunda plant extract exhibit antioxidant and anti-inflammatory effects on amyloid beta 1-42-induced Alzheimer's disease in BALB/c mice. JOURNAL OF ETHNOPHARMACOLOGY 2022; 288:114997. [PMID: 35033624 DOI: 10.1016/j.jep.2022.114997] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/30/2021] [Accepted: 01/11/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Bacopa floribunda (BF), a locally available plant has been employed traditionally as memory enhancer in Southwestern, Nigeria. It has been utilized in traditional and Ayurvedic medicine as brain tonic for enhancing memory, anti-aging and forestalling series of psychological disorders. However, there is a dearth of scientific information on the mechanism(s) of action of important phytochemicals from BF extract on dementia. AIM OF THE STUDY Alzheimer's disease, the commonest form of dementia has been postulated to triple by 2050 as a result of increase in life expectancy. This study therefore assessed and compared the possible mechanism(s) of action of flavonoids and saponins from BF on Amyloid beta (Aβ1-42)-induced dementia in male BALB/c mice. MATERIALS AND METHODS Eighty (80) healthy BALB/c mice divided into 10 groups (n = 8) were given a single bilateral ICV injection of Aβ1-42 or normal saline. Graded doses of Saponins and flavonoids (50, 100 and 200 mg/kg) were used as treatment for 21 days. Hippocampal homogenates were assayed for the levels of antioxidants, oxidative stress and neuroinflammatory markers. In vitro antioxidant activity of flavonoids and saponins were equally assessed using standard procedures. The extent of microglial activation was quantified through immunohistochemistry procedure. RESULTS Aβ1-42 successfully caused a spike in hippocampal levels of MDA, IL1β, TNF-α including MPO levels and invariably decreased antioxidant activities. Likewise an increase in reactive microglia (microgliosis) was observed. However, crude saponins and flavonoids from BF were able to suppress microgliosis, oxidative stress and neuroinflammation induced by Aβ1- 42 and were observed to be more effective at higher doses of saponins (100 mg/kg and 200 mg/kg) and flavonoid (100 mg/kg). CONCLUSIONS Phytochemicals from BF efficiently exhibited dose dependent alleviation of some symptoms associated with Alzheimer's disease.
Collapse
Affiliation(s)
- Mosunmola Busayo Oyeleke
- Department of Physiology, Faculty of Basic Medical Sciences, College of Medicine and Health Sciences, Afe Babalola University, P.M.B, 5454, Ado-Ekiti, Nigeria; Department of Physiology, Neuroscience and Inflammation Unit, Faculty of Basic Medical Sciences, University of Ilorin, P.M.B, 1515, Ilorin, Nigeria.
| | - Bamidele Victor Owoyele
- Department of Physiology, Neuroscience and Inflammation Unit, Faculty of Basic Medical Sciences, University of Ilorin, P.M.B, 1515, Ilorin, Nigeria.
| |
Collapse
|
14
|
The Role of Melatonin on Behavioral Changes and Concomitant Oxidative Stress in icvAβ 1-42 Rat Model with Pinealectomy. Int J Mol Sci 2021; 22:ijms222312763. [PMID: 34884567 PMCID: PMC8657444 DOI: 10.3390/ijms222312763] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/16/2021] [Accepted: 11/22/2021] [Indexed: 02/06/2023] Open
Abstract
One of the pathological hallmarks of Alzheimer’s disease (AD) associated with its progression that contributes to β-amyloid (Aβ) generation is oxidative stress (OS). Clinical data suggest that melatonin is a potent antioxidant that might be effective in the adjunctive therapy of this neurodegenerative disease. The present study aimed to explore the role of melatonin on behavioral changes and markers of OS in three rat models, namely, pinealectomy (pin) model of melatonin deficit, intracerebroventricular (icv)Aβ1-42 model of AD, and combination of both pin and Aβ1-42 model (pin+icvAβ1-42). The chronic injection with vehicle/melatonin (50 mg/kg, i.p. for 40 days) started on the same day of sham/pin and icv vehicle/Aβ1-42 infusion procedures. Anxiety in the open field and the elevated plus-maze test and cognitive responses in the object recognition test were tested between the 30th–35th day after the surgical procedures. Markers of OS in the frontal cortex (FC) and hippocampus were detected by the ELISA method. Melatonin treatment corrected the exacerbated anxiety response only in the pin+icvAβ1-42 model while it alleviated the cognitive impairment in the three models. Pinealectomy disturbed the antioxidant system via enhanced SOD activity and decreased GSH levels both in the FC and hippocampus. The Aβ1-42 model decreased the SOD activity in the FC and elevated the MDA level in the two brain structures. The pin+icvAβ1-42 model impaired the antioxidant system and elevated lipid peroxidation. Melatonin supplementation restored only the elevated MDA level of icvAβ1-42 and pin+icvAβ1-42 model in the hippocampus. In conclusion, our study reveals that the pin+icvAβ1-42 rat model triggers more pronounced anxiety and alterations in markers of OS that may be associated with melatonin deficit concomitant to icvAβ1-42-induced AD pathology.
Collapse
|
15
|
Shen L, Wang P, Ke Y. DNA Nanotechnology-Based Biosensors and Therapeutics. Adv Healthc Mater 2021; 10:e2002205. [PMID: 34085411 DOI: 10.1002/adhm.202002205] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/19/2021] [Indexed: 12/19/2022]
Abstract
Over the past few decades, DNA nanotechnology engenders a vast variety of programmable nanostructures utilizing Watson-Crick base pairing. Due to their precise engineering, unprecedented programmability, and intrinsic biocompatibility, DNA nanostructures cannot only interact with small molecules, nucleic acids, proteins, viruses, and cancer cells, but also can serve as nanocarriers to deliver different therapeutic agents. Such addressability innate to DNA nanostructures enables their use in various fields of biomedical applications such as biosensors and cancer therapy. This review is begun with a brief introduction of the development of DNA nanotechnology, followed by a summary of recent applications of DNA nanostructures in biosensors and therapeutics. Finally, challenges and opportunities for practical applications of DNA nanotechnology are discussed.
Collapse
Affiliation(s)
- Luyao Shen
- Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology and Emory University Atlanta GA 30322 USA
- Institute of Molecular Medicine Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine State Key Laboratory of Oncogenes and Related Genes Renji Hospital School of Medicine Shanghai Jiao Tong University Shanghai 200127 China
| | - Pengfei Wang
- Institute of Molecular Medicine Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine State Key Laboratory of Oncogenes and Related Genes Renji Hospital School of Medicine Shanghai Jiao Tong University Shanghai 200127 China
| | - Yonggang Ke
- Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology and Emory University Atlanta GA 30322 USA
| |
Collapse
|
16
|
Balendra V, Singh SK. Therapeutic potential of astaxanthin and superoxide dismutase in Alzheimer's disease. Open Biol 2021; 11:210013. [PMID: 34186009 PMCID: PMC8241491 DOI: 10.1098/rsob.210013] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Oxidative stress, the imbalance of the antioxidant system, results in an accumulation of neurotoxic proteins in Alzheimer's disease (AD). The antioxidant system is composed of exogenous and endogenous antioxidants to maintain homeostasis. Superoxide dismutase (SOD) is an endogenous enzymatic antioxidant that converts superoxide ions to hydrogen peroxide in cells. SOD supplementation in mice prevented cognitive decline in stress-induced cells by reducing lipid peroxidation and maintaining neurogenesis in the hippocampus. Furthermore, SOD decreased expression of BACE1 while reducing plaque burden in the brain. Additionally, Astaxanthin (AST), a potent exogenous carotenoid, scavenges superoxide anion radicals. Mice treated with AST showed slower memory decline and decreased depositions of amyloid-beta (Aβ) and tau protein. Currently, the neuroprotective potential of these supplements has only been examined separately in studies. However, a single antioxidant cannot sufficiently resist oxidative damage to the brain, therefore, a combinatory approach is proposed as a relevant therapy for ameliorating pathological changes in AD.
Collapse
Affiliation(s)
| | - Sandeep Kumar Singh
- Indian Scientific Education and Technology (ISET) Foundation, Lucknow 226002, India
| |
Collapse
|
17
|
Chubarev VN, Beeraka NM, Sinelnikov MY, Bulygin KV, Nikolenko VN, Mihaylenko E, Tarasov VV, Mikhaleva LM, Poltronieri P, Viswanadha VP, Somasundaram SG, Kirkland CE, Chen K, Liu J, Fan R, Kamal MA, Mironov AA, Madhunapantula SV, Pretorius E, Dindyaev SV, Muresanu C, Sukocheva OA. Health Science Community Will Miss This Bright and Uniting Star: In Memory of Professor Gjumrakch Aliev, M.D, Ph.D. Cancers (Basel) 2021; 13:1965. [PMID: 33921833 PMCID: PMC8072812 DOI: 10.3390/cancers13081965] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 04/15/2021] [Indexed: 02/05/2023] Open
Abstract
It is with deep sadness that we offer our memorial on the unexpected demise of our dear colleague, Professor Gjumrakch Aliev [...].
Collapse
Affiliation(s)
- Vladimir N. Chubarev
- Faculty of Pharmacology, Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, bld. 2, 119991 Moscow, Russia; (V.N.C.); (N.M.B.); (M.Y.S.); (K.V.B.); (V.N.N.); (E.M.); (V.V.T.)
| | - Narasimha M. Beeraka
- Faculty of Pharmacology, Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, bld. 2, 119991 Moscow, Russia; (V.N.C.); (N.M.B.); (M.Y.S.); (K.V.B.); (V.N.N.); (E.M.); (V.V.T.)
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education and Research (JSS AHER), Bannimantapa, Sri Shivarathreeshwara Nagar, Mysuru, Karnataka 570 015, India;
| | - Mikhail Y. Sinelnikov
- Faculty of Pharmacology, Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, bld. 2, 119991 Moscow, Russia; (V.N.C.); (N.M.B.); (M.Y.S.); (K.V.B.); (V.N.N.); (E.M.); (V.V.T.)
| | - Kirill V. Bulygin
- Faculty of Pharmacology, Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, bld. 2, 119991 Moscow, Russia; (V.N.C.); (N.M.B.); (M.Y.S.); (K.V.B.); (V.N.N.); (E.M.); (V.V.T.)
- Faculty of Medicine, M.V. Lomonosov Moscow State University, 117192 Moscow, Russia
| | - Vladimir N. Nikolenko
- Faculty of Pharmacology, Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, bld. 2, 119991 Moscow, Russia; (V.N.C.); (N.M.B.); (M.Y.S.); (K.V.B.); (V.N.N.); (E.M.); (V.V.T.)
- Faculty of Medicine, M.V. Lomonosov Moscow State University, 117192 Moscow, Russia
| | - Elizaveta Mihaylenko
- Faculty of Pharmacology, Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, bld. 2, 119991 Moscow, Russia; (V.N.C.); (N.M.B.); (M.Y.S.); (K.V.B.); (V.N.N.); (E.M.); (V.V.T.)
| | - Vadim V. Tarasov
- Faculty of Pharmacology, Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, bld. 2, 119991 Moscow, Russia; (V.N.C.); (N.M.B.); (M.Y.S.); (K.V.B.); (V.N.N.); (E.M.); (V.V.T.)
| | | | - Palmiro Poltronieri
- Institute of Sciences of Food Productions, National Research Council of Italy, via Monteroni km 7, 73100 Lecce, Italy;
| | | | - Siva G. Somasundaram
- Department of Biological Sciences, Salem University, Salem, WV 26426, USA; (S.G.S.); (C.E.K.)
| | - Cecil E. Kirkland
- Department of Biological Sciences, Salem University, Salem, WV 26426, USA; (S.G.S.); (C.E.K.)
| | - Kuo Chen
- Cancer Center, Department of Radiation Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China; (K.C.); (J.L.); (R.F.)
| | - Junqi Liu
- Cancer Center, Department of Radiation Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China; (K.C.); (J.L.); (R.F.)
| | - Ruitai Fan
- Cancer Center, Department of Radiation Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China; (K.C.); (J.L.); (R.F.)
| | - Mohammad Amjad Kamal
- West China School of Nursing/Institutes for Systems Genetics, The Frontier Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China;
- King Fahd Medical Research Center, King Abdulaziz University, P. O. Box 80216, Jeddah 21589, Saudi Arabia
- Enzymoics, 7 Peterlee Place, Novel Global Community Educational Foundation, Hebersham, NSW 2770, Australia
| | - Alexander A. Mironov
- Laboratory of Electron Microscopy, The FIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy;
| | - SubbaRao V. Madhunapantula
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education and Research (JSS AHER), Bannimantapa, Sri Shivarathreeshwara Nagar, Mysuru, Karnataka 570 015, India;
| | - Etheresia Pretorius
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Arcadia 0007, South Africa;
| | - Sergey V. Dindyaev
- Department of Histology, Embryology & Cytology, Pediatric Faculty, Federal State Budgetary Educational Institution of Higher Education “Ivanovo State Medical Academy” of the Ministry of Healthcare of the Russian Federation (FSBEI HE IvSMA MOH Russia), 8 Sheremetyevsky Ave., 153012 Ivanovo, Russia;
| | - Cristian Muresanu
- Research Center for Applied Biotechnology in Diagnosis and Molecular Therapies, Str. Trifoiului nr. 12 G, 400478 Cluj-Napoca, Romania;
| | - Olga A. Sukocheva
- Discipline of Health Sciences, College of Nursing and Health Sciences, Flinders University of South Australia, Adelaide 5001, Australia
| |
Collapse
|
18
|
Chen K, Shen W, Gao Z, Luo C. Stress response capacity analysis during aging and possible new insights into aging studies. Curr Genet 2021; 67:417-420. [PMID: 33580302 DOI: 10.1007/s00294-021-01159-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 01/26/2021] [Accepted: 01/29/2021] [Indexed: 01/08/2023]
Abstract
The causes and consequences of aging have always been a concern. In recent studies, changes in the stress response capacity of cells during aging were quantitatively analyzed. It was found that aging was accompanied by a decline in response capacity. When the response capacity decreased to a critical value, which we assumed was the internal noise level, the cell soon died. To survive, the response capacity should be, at minimum, sufficiently strong to resist intracellular noise. Here, we discuss the role of stress response capacity in aging and conjecture that lifespan might be extended by enhancing stress response capacity.
Collapse
Affiliation(s)
- Kaiyue Chen
- The State Key Laboratory for Artificial Microstructures and Mesoscopic Physics, School of Physics, Peking University, Beijing, China.,Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Wenting Shen
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Ziqing Gao
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Chunxiong Luo
- The State Key Laboratory for Artificial Microstructures and Mesoscopic Physics, School of Physics, Peking University, Beijing, China. .,Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China.
| |
Collapse
|
19
|
Malfatti MC, Antoniali G, Codrich M, Burra S, Mangiapane G, Dalla E, Tell G. New perspectives in cancer biology from a study of canonical and non-canonical functions of base excision repair proteins with a focus on early steps. Mutagenesis 2021; 35:129-149. [PMID: 31858150 DOI: 10.1093/mutage/gez051] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 12/05/2019] [Indexed: 12/15/2022] Open
Abstract
Alterations of DNA repair enzymes and consequential triggering of aberrant DNA damage response (DDR) pathways are thought to play a pivotal role in genomic instabilities associated with cancer development, and are further thought to be important predictive biomarkers for therapy using the synthetic lethality paradigm. However, novel unpredicted perspectives are emerging from the identification of several non-canonical roles of DNA repair enzymes, particularly in gene expression regulation, by different molecular mechanisms, such as (i) non-coding RNA regulation of tumour suppressors, (ii) epigenetic and transcriptional regulation of genes involved in genotoxic responses and (iii) paracrine effects of secreted DNA repair enzymes triggering the cell senescence phenotype. The base excision repair (BER) pathway, canonically involved in the repair of non-distorting DNA lesions generated by oxidative stress, ionising radiation, alkylation damage and spontaneous or enzymatic deamination of nucleotide bases, represents a paradigm for the multifaceted roles of complex DDR in human cells. This review will focus on what is known about the canonical and non-canonical functions of BER enzymes related to cancer development, highlighting novel opportunities to understand the biology of cancer and representing future perspectives for designing new anticancer strategies. We will specifically focus on APE1 as an example of a pleiotropic and multifunctional BER protein.
Collapse
Affiliation(s)
- Matilde Clarissa Malfatti
- Laboratory of Molecular Biology and DNA repair, Department of Medicine (DAME), University of Udine, Udine, Italy
| | - Giulia Antoniali
- Laboratory of Molecular Biology and DNA repair, Department of Medicine (DAME), University of Udine, Udine, Italy
| | - Marta Codrich
- Laboratory of Molecular Biology and DNA repair, Department of Medicine (DAME), University of Udine, Udine, Italy
| | - Silvia Burra
- Laboratory of Molecular Biology and DNA repair, Department of Medicine (DAME), University of Udine, Udine, Italy
| | - Giovanna Mangiapane
- Laboratory of Molecular Biology and DNA repair, Department of Medicine (DAME), University of Udine, Udine, Italy
| | - Emiliano Dalla
- Laboratory of Molecular Biology and DNA repair, Department of Medicine (DAME), University of Udine, Udine, Italy
| | - Gianluca Tell
- Laboratory of Molecular Biology and DNA repair, Department of Medicine (DAME), University of Udine, Udine, Italy
| |
Collapse
|
20
|
Leszek J, Mikhaylenko EV, Belousov DM, Koutsouraki E, Szczechowiak K, Kobusiak-Prokopowicz M, Mysiak A, Diniz BS, Somasundaram SG, Kirkland CE, Aliev G. The Links between Cardiovascular Diseases and Alzheimer's Disease. Curr Neuropharmacol 2021; 19:152-169. [PMID: 32727331 PMCID: PMC8033981 DOI: 10.2174/1570159x18666200729093724] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 06/02/2020] [Accepted: 07/16/2020] [Indexed: 12/17/2022] Open
Abstract
The root cause of non-inherited Alzheimer's disease (AD) remains unknown despite hundreds of research studies performed to attempt to solve this problem. Since proper prophylaxis remains the best strategy, many scientists have studied the risk factors that may affect AD development. There is robust evidence supporting the hypothesis that cardiovascular diseases (CVD) may contribute to AD progression, as the diseases often coexist. Therefore, a lack of well-defined diagnostic criteria makes studying the relationship between AD and CVD complicated. Additionally, inflammation accompanies the pathogenesis of AD and CVD, and is not only a consequence but also implicated as a significant contributor to the course of the diseases. Of note, АроЕε4 is found to be one of the major risk factors affecting both the cardiovascular and nervous systems. According to genome wide association and epidemiological studies, numerous common risk factors have been associated with the development of AD-related pathology. Furthermore, the risk of developing AD and CVDs appears to be increased by a wide range of conditions and lifestyle factors: hypertension, dyslipidemia, hypercholesterolemia, hyperhomocysteinemia, gut/oral microbiota, physical activity, and diet. This review summarizes the literature and provides possible mechanistic links between CVDs and AD.
Collapse
Affiliation(s)
- Jerzy Leszek
- Address correspondence to these authors at the Department of Psychiatry, Wrocław Medical University, Ul. Pasteura 10, 50-367, Wroclaw, Poland;, E-mail: and GALLY International Research Institute, 7733 Louis Pasteur Drive, #330, San Antonio, TX 78229, USA; Tel: +1-210-442-8625 or +1-440-263-7461; E-mails: ,
| | | | | | | | | | | | | | | | | | | | - Gjumrakch Aliev
- Address correspondence to these authors at the Department of Psychiatry, Wrocław Medical University, Ul. Pasteura 10, 50-367, Wroclaw, Poland;, E-mail: and GALLY International Research Institute, 7733 Louis Pasteur Drive, #330, San Antonio, TX 78229, USA; Tel: +1-210-442-8625 or +1-440-263-7461; E-mails: ,
| |
Collapse
|
21
|
Belousov DM, Mikhaylenko EV, Somasundaram SG, Kirkland CE, Aliev G. The Dawn of Mitophagy: What Do We Know by Now? Curr Neuropharmacol 2021; 19:170-192. [PMID: 32442087 PMCID: PMC8033973 DOI: 10.2174/1570159x18666200522202319] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 05/10/2020] [Accepted: 05/17/2020] [Indexed: 01/31/2023] Open
Abstract
Mitochondria are essential organelles for healthy eukaryotic cells. They produce energyrich phosphate bond molecules (ATP) through oxidative phosphorylation using ionic gradients. The presence of mitophagy pathways in healthy cells enhances cell protection during mitochondrial damage. The PTEN-induced putative kinase 1 (PINK1)/Parkin-dependent pathway is the most studied for mitophage. In addition, there are other mechanisms leading to mitophagy (FKBP8, NIX, BNIP3, FUNDC1, BCL2L13). Each of these provides tethering of a mitochondrion to an autophagy apparatus via the interaction between receptor proteins (Optineurin, p62, NDP52, NBR1) or the proteins of the outer mitochondrial membrane with ATG9-like proteins (LC3A, LC3B, GABARAP, GABARAPL1, GATE16). Another pathogenesis of mitochondrial damage is mitochondrial depolarization. Reactive oxygen species (ROS) antioxidant responsive elements (AREs) along with antioxidant genes, including pro-autophagic genes, are all involved in mitochondrial depolarization. On the other hand, mammalian Target of Rapamycin Complex 1 (mTORC1) and AMP-dependent kinase (AMPK) are the major regulatory factors modulating mitophagy at the post-translational level. Protein-protein interactions are involved in controlling other mitophagy processes. The objective of the present review is to analyze research findings regarding the main pathways of mitophagy induction, recruitment of the autophagy machinery, and their regulations at the levels of transcription, post-translational modification and protein-protein interaction that appeared to be the main target during the development and maturation of neurodegenerative disorders.
Collapse
Affiliation(s)
| | | | | | - Cecil E. Kirkland
- Address correspondence to this author at the Department of Biological Sciences, Salem University, Salem, WV, 26426, USA & GALLY International Research Institute, San Antonio, TX 78229, USA;, E-mails: ,
| | - Gjumrakch Aliev
- Address correspondence to this author at the Department of Biological Sciences, Salem University, Salem, WV, 26426, USA & GALLY International Research Institute, San Antonio, TX 78229, USA;, E-mails: ,
| |
Collapse
|
22
|
RNA and Oxidative Stress in Alzheimer's Disease: Focus on microRNAs. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:2638130. [PMID: 33312335 PMCID: PMC7721489 DOI: 10.1155/2020/2638130] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 10/21/2020] [Accepted: 10/29/2020] [Indexed: 01/31/2023]
Abstract
Oxidative stress (OS) is one of the major pathomechanisms of Alzheimer's disease (AD), which is closely associated with other key events in neurodegeneration such as mitochondrial dysfunction, inflammation, metal dysregulation, and protein misfolding. Oxidized RNAs are identified in brains of AD patients at the prodromal stage. Indeed, oxidized mRNA, rRNA, and tRNA lead to retarded or aberrant protein synthesis. OS interferes with not only these translational machineries but also regulatory mechanisms of noncoding RNAs, especially microRNAs (miRNAs). MiRNAs can be oxidized, which causes misrecognizing target mRNAs. Moreover, OS affects the expression of multiple miRNAs, and conversely, miRNAs regulate many genes involved in the OS response. Intriguingly, several miRNAs embedded in upstream regulators or downstream targets of OS are involved also in neurodegenerative pathways in AD. Specifically, seven upregulated miRNAs (miR-125b, miR-146a, miR-200c, miR-26b, miR-30e, miR-34a, miR-34c) and three downregulated miRNAs (miR-107, miR-210, miR-485), all of which are associated with OS, are found in vulnerable brain regions of AD at the prodromal stage. Growing evidence suggests that altered miRNAs may serve as targets for developing diagnostic or therapeutic tools for early-stage AD. Focusing on a neuroprotective transcriptional repressor, REST, and the concept of hormesis that are relevant to the OS response may provide clues to help us understand the role of the miRNA system in cellular and organismal adaptive mechanisms to OS.
Collapse
|
23
|
Arrigoni F, Rizza F, Tisi R, De Gioia L, Zampella G, Bertini L. On the propagation of the OH radical produced by Cu-amyloid beta peptide model complexes. Insight from molecular modelling. Metallomics 2020; 12:1765-1780. [PMID: 33052996 DOI: 10.1039/d0mt00113a] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Oxidative stress and metal dyshomeostasis are considered as crucial factors in the pathogenesis of Alzheimer's disease (AD). Indeed, transition metal ions such as Cu(ii) can generate Reactive Oxygen Species (ROS) via O2 Fenton-like reduction, catalyzed by Cu(ii) coordinated to the Amyloid beta (Aβ) peptide. Despite intensive effort, the mechanisms of ROS-induced molecular damage remain poorly understood. In the present paper, we investigate on the basis of molecular modelling computations the mechanism of OH radical propagation toward the Aβ peptide, starting from the end-product of OH radical generation by Cu(ii)·Aβ. We evaluate (i) the OH oxidative capacity, as well as the energetics of the possible Aβ oxidation target residues, by quantum chemistry Density Functional Theory (DFT) on coordination models of Cu(ii)/OH/Aβ and (ii) the motion of the OH˙ approaching the Aβ target residues by classical Molecular Dynamics (MD) on the full peptide Cu(ii)/OH/Aβ(1-16). The results show that the oxidative capacity of OH coordinated Cu(ii)Aβ is significantly lower than that of the free OH radical and that propagation toward Aβ Asp and His residues is favoured over Tyr residues. These results are discussed on the basis of the recent literature on in vitro Aβ metal-catalyzed oxidation and on the possible implications for the AD oxidative stress mechanism.
Collapse
Affiliation(s)
- Federica Arrigoni
- Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy.
| | | | | | | | | | | |
Collapse
|
24
|
Zhang X, Li L. The Significance of 8-oxoGsn in Aging-Related Diseases. Aging Dis 2020; 11:1329-1338. [PMID: 33014540 PMCID: PMC7505272 DOI: 10.14336/ad.2019.1021] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 10/21/2019] [Indexed: 01/10/2023] Open
Abstract
Aging is a common risk factor for the occurrence and development of many diseases, such as Parkinson’s disease, Alzheimer’s disease, diabetes, hypertension, atherosclerosis and coronary heart disease, and cancer, among others, and is a key problem threatening the health and life expectancy of the elderly. Oxidative damage is an important mechanism involved in aging. The latest discovery pertaining to oxidative damage is that 8-oxoGsn (8-oxo-7,8-dihydroguanosine), an oxidative damage product of RNA, can represent the level of oxidative stress. The significance of RNA oxidative damage to aging has not been fully explained, but the relationship between the accumulation of 8-oxoGsn, a marker of RNA oxidative damage, and the occurrence of diseases has been confirmed in many aging-related diseases. Studying the aging mechanism, monitoring the aging level of the body and exploring the corresponding countermeasures are of great significance for achieving healthy aging and promoting public health and social development. This article reviews the progress of research on 8-oxoGsn in aging-related diseases.
Collapse
Affiliation(s)
- Xinmu Zhang
- Department of Medical Oncology, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Lin Li
- Department of Medical Oncology, Beijing Hospital, National Center of Gerontology, Beijing, China
| |
Collapse
|
25
|
Uddin MS, Al Mamun A, Kabir MT, Ashraf GM, Bin-Jumah MN, Abdel-Daim MM. Multi-Target Drug Candidates for Multifactorial Alzheimer's Disease: AChE and NMDAR as Molecular Targets. Mol Neurobiol 2020; 58:281-303. [PMID: 32935230 DOI: 10.1007/s12035-020-02116-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 09/02/2020] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is one of the most common forms of dementia among elder people, which is a progressive neurodegenerative disease that results from a chronic loss of cognitive activities. It has been observed that AD is multifactorial, hence diverse pharmacological targets that could be followed for the treatment of AD. The Food and Drug Administration has approved two types of medications for AD treatment such as cholinesterase inhibitors (ChEIs) and N-methyl-D-aspartic acid receptor (NMDAR) antagonists. Rivastigmine, donepezil, and galantamine are the ChEIs that have been approved to treat AD. On the other hand, memantine is the only non-competitive NMDAR antagonist approved in AD treatment. As compared with placebo, it has been revealed through clinical studies that many single-target therapies are unsuccessful to treat multifactorial Alzheimer's symptoms or disease progression. Therefore, due to the complex nature of AD pathophysiology, diverse pharmacological targets can be hunted. In this article, based on the entwined link of acetylcholinesterase (AChE) and NMDAR, we represent several multifunctional compounds in the rational design of new potential AD medications. This review focus on the significance of privileged scaffolds in the generation of the multi-target lead compound for treating AD, investigating the idea and challenges of multi-target drug design. Furthermore, the most auspicious elementary units for designing as well as synthesizing hybrid drugs are demonstrated as pharmacological probes in the rational design of new potential AD therapeutics.
Collapse
Affiliation(s)
- Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh.
- Pharmakon Neuroscience Research Network, Dhaka, Bangladesh.
| | - Abdullah Al Mamun
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh
- Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | | | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - May N Bin-Jumah
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh 11474, Saudi Arabia
| | - Mohamed M Abdel-Daim
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| |
Collapse
|
26
|
Synthesis of new ferulic/lipoic/comenic acid-melatonin hybrids as antioxidants and Nrf2 activators via Ugi reaction. Future Med Chem 2020; 11:3097-3108. [PMID: 31838896 DOI: 10.4155/fmc-2019-0191] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Aim: Oxidative stress has been implicated in the pathogenesis of many neurodegenerative diseases, and particularly in Alzheimer's disease. Results: This work describes the Ugi multicomponent synthesis, antioxidant power and Nrf2 pathway induction in antioxidant response element cells of (E)-N-(2-((2-(1H-indol-3-yl)ethyl)amino)-2-oxoethyl)-N-(2-(5-(benzyloxy)-1H-indol-3-yl)ethyl)-3-(4-hydroxy-3-methoxyphenyl)acryl amides 8a-d, N-(2-((2-(1H-indol-3-yl)ethyl)amino)-2-oxoethyl)-N-(2-(5-(benzyloxy)-1H-indol-3-yl)ethyl)-5-(1,2-dithiolan-3-yl)pentanamides 8e-h and N-(2-((2-(1H-indol-3-yl)ethyl)amino)-2-oxoethyl)-N-(2-(5-(benzyloxy)-1H-indol-3-yl)ethyl)-5-hydroxy-4-oxo-4H-pyran-2-carboxamides 8i,j. Conclusion: We have identified compounds 8e and 8g, showing a potent antioxidant capacity, a remarkable neuroprotective effect against the cell death induced by H2O2 in SH-SY5Y cells, and a performing activation of the Nrf2 signaling pathway, as very interesting new antioxidant agents for pathologies that curse with oxidative stress.
Collapse
|
27
|
Uddin MS, Tewari D, Mamun AA, Kabir MT, Niaz K, Wahed MII, Barreto GE, Ashraf GM. Circadian and sleep dysfunction in Alzheimer's disease. Ageing Res Rev 2020; 60:101046. [PMID: 32171783 DOI: 10.1016/j.arr.2020.101046] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 02/05/2020] [Accepted: 03/10/2020] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is a devastating and irreversible cognitive impairment and the most common type of dementia. Along with progressive cognitive impairment, dysfunction of the circadian rhythms also plays a pivotal role in the progression of AD. A mutual relationship among circadian rhythms, sleep, and AD has been well-recommended. The etiopathogenesis of the disturbances of the circadian system and AD share some general features that also unlock the outlook of observing them as a mutually dependent pathway. Indeed, the burden of amyloid β (Aβ), neurofibrillary tangles (NFTs), neuroinflammation, oxidative stress, and dysfunction of circadian rhythms may lead to AD. Aging can alter both sleep timings and quality that can be strongly disrupted in AD. Increased production of Aβ and reduced Aβ clearance are caused by a close interplay of Aβ, sleep disturbance and raised wakefulness. Besides Aβ, the impact of tau pathology is possibly noteworthy to the sleep deprivation found in AD. Hence, this review is focused on the primary mechanistic complexities linked to disruption of circadian rhythms, sleep deprivation, and AD. Furthermore, this review also highlights the potential therapeutic strategies to abate AD pathogenesis.
Collapse
|
28
|
Kim JW, Byun MS, Yi D, Lee JH, Jeon SY, Ko K, Jung G, Lee HN, Lee JY, Sohn CH, Lee YS, Shin SA, Kim YK, Lee DY. Serum Uric Acid, Alzheimer-Related Brain Changes, and Cognitive Impairment. Front Aging Neurosci 2020; 12:160. [PMID: 32581770 PMCID: PMC7291838 DOI: 10.3389/fnagi.2020.00160] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 05/08/2020] [Indexed: 12/27/2022] Open
Abstract
Background Despite known associations of lower serum uric acid (UA) with Alzheimer’s disease (AD) dementia or AD-related cognitive impairment, little is known regarding the underlying patho-mechanisms. We aimed to examine the relationships of serum UA with in vivo AD pathologies including cerebral beta-amyloid (Aβ) and tau deposition, AD-signature region cerebral glucose metabolism (AD-CM), and white matter hyperintensities (WMH). We also investigated the association between serum UA and cognitive performance, and then assessed whether such an association is mediated by the brain pathologies. Methods A total of 430 non-demented older adults underwent comprehensive clinical assessments, measurement of serum UA level, and multimodal brain imaging, including Pittsburgh compound B-positron emission tomography (PET), AV-1451 PET, fluorodeoxyglucose (FDG)-PET, and magnetic resonance imaging scans. Mini-Mental State Examination (MMSE) and word list recall (WLR) test scores were used to measure cognitive performance. Results Serum UA level was significantly associated with AD-CM, but not with Aβ deposition, tau deposition, or WMH volume. Serum UA levels also had significant association with WLR and marginal association with MMSE; such associations disappeared when AD-CM was controlled as a covariate, indicating that AD-CM has a mediating effect. Conclusion The findings of the present study indicate that there is an association of low serum UA with AD-related cerebral hypometabolism, and whether this represents a causal relationship remains to be determined.
Collapse
Affiliation(s)
- Jee Wook Kim
- Department of Neuropsychiatry, Hallym University Dongtan Sacred Heart Hospital, Hwaseong, South Korea.,Department of Psychiatry, Hallym University College of Medicine, Chuncheon, South Korea
| | - Min Soo Byun
- Institute of Human Behavioral Medicine, Medical Research Center Seoul National University, Seoul, South Korea
| | - Dahyun Yi
- Institute of Human Behavioral Medicine, Medical Research Center Seoul National University, Seoul, South Korea
| | - Jun Ho Lee
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, South Korea
| | - So Yeon Jeon
- Department of Neuropsychiatry, Chungnam National University Hospital, Daejeon, South Korea
| | - Kang Ko
- Department of Geriatric Psychiatry, National Center for Mental Health, Seoul, South Korea
| | - Gijung Jung
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, South Korea
| | - Han Na Lee
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul, South Korea
| | - Jun-Young Lee
- Department of Neuropsychiatry, SMG-SNU Boramae Medical Center, Seoul, South Korea.,Department of Psychiatry, Seoul National University College of Medicine, Seoul, South Korea
| | - Chul-Ho Sohn
- Department of Radiology, Seoul National University Hospital, Seoul, South Korea
| | - Yun-Sang Lee
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Seong A Shin
- Department of Nuclear Medicine, SMG-SNU Boramae Medical Center, Seoul, South Korea
| | - Yu Kyeong Kim
- Department of Nuclear Medicine, SMG-SNU Boramae Medical Center, Seoul, South Korea
| | - Dong Young Lee
- Institute of Human Behavioral Medicine, Medical Research Center Seoul National University, Seoul, South Korea.,Department of Neuropsychiatry, Seoul National University Hospital, Seoul, South Korea.,Department of Psychiatry, Seoul National University College of Medicine, Seoul, South Korea
| | | |
Collapse
|
29
|
Strobel S, Grünblatt E, Heinsen H, Riederer P, Espach T, Meder M, Monoranu CM. Astrocyte- and Microglia-Specific Mitochondrial DNA Deletions Levels in Sporadic Alzheimer's Disease. J Alzheimers Dis 2020; 67:149-157. [PMID: 30475765 DOI: 10.3233/jad-180661] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Oxidative stress is implicated in the pathogenesis of neurodegenerative diseases, including sporadic Alzheimer's disease (AD). Mitochondrial DNA (mtDNA) deletions are markers of oxidative damage with an age-dependent accumulation. In a previous study, we analyzed mtDNA levels in diverse neuronal cell types in order to unravel the impact of oxidative stress in brains of AD patients. The aim of this study was to identify possible correlations between mtDNA deletion levels of selected astrocytes and microglia from three brain regions with different vulnerability to AD pathology and different stages of disease compared to controls. Our results reflect a higher vulnerability of hippocampal astrocytes and microglia to oxidative stress compared to other brain regions, such as cerebellum and brainstem.
Collapse
Affiliation(s)
- Sabrina Strobel
- Institute of Pathology, University of Wuerzburg, Germany.,Comprehensive Cancer Center (CCC) Mainfranken Wuerzburg, Germany
| | - Edna Grünblatt
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital of Psychiatry Zurich, University of Zurich, Zürich, Switzerland.,Neuroscience Center Zurich, University of Zurich and the ETH Zurich, Zürich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Switzerland
| | - Helmut Heinsen
- Department of Psychiatry, Morphological Brain Research Unit, University of Wuerzburg, Wuerzburg, Germany.,Department of Pathology, University of São Paulo, São Paulo, Brazil
| | - Peter Riederer
- Center of Mental Health, Department of Psychiatry, Psychosomatics and Psychotherapy, University of Wuerzburg, Germany.,Department and Research Unit of Psychiatry, University of Southern Denmark Odense, Denmark
| | - Thomas Espach
- Institute of Pathology, Department of Neuropathology, University of Wuerzburg, Germany
| | - Michael Meder
- Institute of Pathology, University of Wuerzburg, Germany.,Comprehensive Cancer Center (CCC) Mainfranken Wuerzburg, Germany
| | - Camelia-Maria Monoranu
- Institute of Pathology, Department of Neuropathology, University of Wuerzburg, Germany.,Comprehensive Cancer Center (CCC) Mainfranken Wuerzburg, Germany
| |
Collapse
|
30
|
Architecture of The Human Ape1 Interactome Defines Novel Cancers Signatures. Sci Rep 2020; 10:28. [PMID: 31913336 PMCID: PMC6949240 DOI: 10.1038/s41598-019-56981-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 12/16/2019] [Indexed: 12/19/2022] Open
Abstract
APE1 is essential in cancer cells due to its central role in the Base Excision Repair pathway of DNA lesions and in the transcriptional regulation of genes involved in tumor progression/chemoresistance. Indeed, APE1 overexpression correlates with chemoresistance in more aggressive cancers, and APE1 protein-protein interactions (PPIs) specifically modulate different protein functions in cancer cells. Although important, a detailed investigation on the nature and function of protein interactors regulating APE1 role in tumor progression and chemoresistance is still lacking. The present work was aimed at analyzing the APE1-PPI network with the goal of defining bad prognosis signatures through systematic bioinformatics analysis. By using a well-characterized HeLa cell model stably expressing a flagged APE1 form, which was subjected to extensive proteomics analyses for immunocaptured complexes from different subcellular compartments, we here demonstrate that APE1 is a central hub connecting different subnetworks largely composed of proteins belonging to cancer-associated communities and/or involved in RNA- and DNA-metabolism. When we performed survival analysis in real cancer datasets, we observed that more than 80% of these APE1-PPI network elements is associated with bad prognosis. Our findings, which are hypothesis generating, strongly support the possibility to infer APE1-interactomic signatures associated with bad prognosis of different cancers; they will be of general interest for the future definition of novel predictive disease biomarkers. Future studies will be needed to assess the function of APE1 in the protein complexes we discovered. Data are available via ProteomeXchange with identifier PXD013368.
Collapse
|
31
|
An Z, Yan J, Zhang Y, Pei R. Applications of nanomaterials for scavenging reactive oxygen species in the treatment of central nervous system diseases. J Mater Chem B 2020; 8:8748-8767. [DOI: 10.1039/d0tb01380c] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Nanomaterials with excellent ROS-scavenging ability and biodistribution are considered as promising candidates in alleviating oxidative stress and restoring redox balance in CNS diseases, further facilitating the function recovery of the CNS.
Collapse
Affiliation(s)
- Zhen An
- CAS Key Laboratory for Nano-Bio Interface
- Suzhou Institute of Nano-Tech and Nano-Bionics
- Chinese Academy of Sciences
- Suzhou
- China
| | - Jincong Yan
- CAS Key Laboratory for Nano-Bio Interface
- Suzhou Institute of Nano-Tech and Nano-Bionics
- Chinese Academy of Sciences
- Suzhou
- China
| | - Ye Zhang
- CAS Key Laboratory for Nano-Bio Interface
- Suzhou Institute of Nano-Tech and Nano-Bionics
- Chinese Academy of Sciences
- Suzhou
- China
| | - Renjun Pei
- CAS Key Laboratory for Nano-Bio Interface
- Suzhou Institute of Nano-Tech and Nano-Bionics
- Chinese Academy of Sciences
- Suzhou
- China
| |
Collapse
|
32
|
Milanese C, Bombardieri CR, Sepe S, Barnhoorn S, Payán-Goméz C, Caruso D, Audano M, Pedretti S, Vermeij WP, Brandt RMC, Gyenis A, Wamelink MM, de Wit AS, Janssens RC, Leen R, van Kuilenburg ABP, Mitro N, Hoeijmakers JHJ, Mastroberardino PG. DNA damage and transcription stress cause ATP-mediated redesign of metabolism and potentiation of anti-oxidant buffering. Nat Commun 2019; 10:4887. [PMID: 31653834 PMCID: PMC6814737 DOI: 10.1038/s41467-019-12640-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 09/22/2019] [Indexed: 12/13/2022] Open
Abstract
Accumulation of DNA lesions causing transcription stress is associated with natural and accelerated aging and culminates with profound metabolic alterations. Our understanding of the mechanisms governing metabolic redesign upon genomic instability, however, is highly rudimentary. Using Ercc1-defective mice and Xpg knock-out mice, we demonstrate that combined defects in transcription-coupled DNA repair (TCR) and in nucleotide excision repair (NER) directly affect bioenergetics due to declined transcription, leading to increased ATP levels. This in turn inhibits glycolysis allosterically and favors glucose rerouting through the pentose phosphate shunt, eventually enhancing production of NADPH-reducing equivalents. In NER/TCR-defective mutants, augmented NADPH is not counterbalanced by increased production of pro-oxidants and thus pentose phosphate potentiation culminates in an over-reduced redox state. Skin fibroblasts from the TCR disease Cockayne syndrome confirm results in animal models. Overall, these findings unravel a mechanism connecting DNA damage and transcriptional stress to metabolic redesign and protective antioxidant defenses. ERCC1 is involved in a number of DNA repair pathways including nucleotide excision repair. Here the authors showed that reduced transcription in Ercc1-deficient mouse livers and cells increases ATP levels, suppressing glycolysis and rerouting glucose into the pentose phosphate shunt that generates reductive stress.
Collapse
Affiliation(s)
- Chiara Milanese
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Cíntia R Bombardieri
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Sara Sepe
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Sander Barnhoorn
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - César Payán-Goméz
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands.,Facultad de Ciencias Naturales y Matemáticas, Universidad del Rosario, Bogotá, Colombia
| | - Donatella Caruso
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Matteo Audano
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Silvia Pedretti
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Wilbert P Vermeij
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Renata M C Brandt
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Akos Gyenis
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands.,Cologne Excellence Cluster for Cellular Stress Responses in Ageing-Associated Diseases (CECAD) and Systems Biology of Ageing Cologne, University of Cologne, Cologne, Germany
| | - Mirjam M Wamelink
- Department of Clinical Chemistry, VU University Medical Center, Amsterdam, the Netherlands
| | - Annelieke S de Wit
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Roel C Janssens
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - René Leen
- Laboratory of Genetic Metabolic Diseases, Academic Medical Center, Amsterdam, the Netherlands
| | | | - Nico Mitro
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Jan H J Hoeijmakers
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands.,Cologne Excellence Cluster for Cellular Stress Responses in Ageing-Associated Diseases (CECAD) and Systems Biology of Ageing Cologne, University of Cologne, Cologne, Germany.,Oncode Institute, Princess Máxima Center, Utrecht, Netherlands
| | - Pier G Mastroberardino
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands. .,Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy.
| |
Collapse
|
33
|
Karwowski BT. The Influence of (5' R)- and (5' S)-5',8-Cyclo-2'-Deoxyadenosine on UDG and hAPE1 Activity. Tandem Lesions are the Base Excision Repair System's Nightmare. Cells 2019; 8:cells8111303. [PMID: 31652769 PMCID: PMC6912673 DOI: 10.3390/cells8111303] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 10/21/2019] [Accepted: 10/22/2019] [Indexed: 12/30/2022] Open
Abstract
DNA lesions are formed continuously in each living cell as a result of environmental factors, ionisation radiation, metabolic processes, etc. Most lesions are removed from the genome by the base excision repair system (BER). The activation of the BER protein cascade starts with DNA damage recognition by glycosylases. Uracil-DNA glycosylase (UDG) is one of the most evolutionary preserved glycosylases which remove the frequently occurring 2′-deoxyuridine from single (ss) and double-stranded (ds) oligonucleotides. Conversely, the unique tandem lesions (5′R)- and (5′S)-5′,8-cyclo-2′-deoxyadenosine (cdA) are not suitable substrates for BER machinery and are released from the genome by the nucleotide excision repair (NER) system. However, the cyclopurines appearing in a clustered DNA damage structure can influence the BER process of other lesions like dU. In this article, UDG inhibition by 5′S- and 5′R-cdA is shown and discussed in an experimental and theoretical manner. This phenomenon was observed when a tandem lesion appears in single or double-stranded oligonucleotides next to dU, on its 3′-end side. The cdA shift to the 5′-end side of dU in ss-DNA stops this effect in both cdA diastereomers. Surprisingly, in the case of ds-DNA, 5′S-cdA completely blocks uracil excision by UDG. Conversely, 5′R-cdA allows glycosylase for uracil removal, but the subsequently formed apurinic/apyrimidinic (AP) site is not suitable for human AP-site endonuclease 1 (hAPE1) activity. In conclusion, the appearance of the discussed tandem lesion in the structure of single or double-stranded DNA can stop the entire base repair process at its beginning, which due to UDG and hAPE1 inhibition can lead to mutagenesis. On the other hand, the presented results can cast some light on the UDG or hAPE1 inhibitors being used as a potential treatment.
Collapse
Affiliation(s)
- Bolesław T Karwowski
- DNA Damage Laboratory of the Food Science Department, Faculty of Pharmacy, Medical University of Lodz, ul. Muszynskiego 1, 90-151 Lodz, Poland.
| |
Collapse
|
34
|
Yumoto S, Kakimi S, Ishikawa A. Colocalization of Aluminum and Iron in Nuclei of Nerve Cells in Brains of Patients with Alzheimer's Disease. J Alzheimers Dis 2019; 65:1267-1281. [PMID: 30149443 PMCID: PMC6218123 DOI: 10.3233/jad-171108] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Increasing evidence indicates that metal-induced oxidative stress plays a pivotal role in the pathogenesis of Alzheimer's disease (AD). Recently, the presence of 8-hydroxydeoxyguanosine, a biomarker of oxidative DNA damage, was demonstrated in nuclear DNA (nDNA) in the AD brain. Iron (Fe) is a pro-oxidant metal capable of generating hydroxyl radicals that can oxidize DNA, and aluminum (Al) has been reported to facilitate Fe-mediated oxidation. In the present study, we examined the elements contained in the nuclei of nerve cells in AD brains using scanning electron microscopy coupled with energy-dispersive X-ray spectroscopy (SEM-EDS). Our results demonstrated that Al and Fe were colocalized in the nuclei of nerve cells in the AD brain. Within the nuclei, the highest levels of both Al and Fe were measured in the nucleolus. The SEM-EDS analysis also revealed the colocalization of Al and Fe in the heterochromatin and euchromatin in neuronal nuclei in the AD brain. Notably, the levels of Al and Fe in the nucleus of nerve cells in the AD brain were markedly higher than those in age-matched control brains. We hypothesize that the colocalization of Al and Fe in the nucleus of nerve cells might induce oxidative damage to nDNA and concurrently inhibit the repair of oxidatively damaged nDNA. An imbalance caused by the increase in DNA damage and the decrease in DNA repair activities might lead to the accumulation of unrepaired damaged DNA, eventually causing neurodegeneration and the development of AD.
Collapse
Affiliation(s)
- Sakae Yumoto
- Yumoto Institute of Neurology, Kawadacho, Shinjuku-ku, Tokyo, Japan
| | - Shigeo Kakimi
- Department of Functional Morphology, Nihon University School of Medicine, Ohyaguchiuemachi, Itabashi-ku, Tokyo, Japan
| | - Akira Ishikawa
- Department of Physics, College of Humanities and Sciences, Nihon University, Sakurajousui, Setagaya-ku, Tokyo, Japan
| |
Collapse
|
35
|
Noriega L, Díaz A, Limón D, Castro ME, Caballero NA, Ramírez RE, Perez-Aguilar JM, Melendez FJ. Inhibitory mechanism of 17β-aminoestrogens in the formation of Aβ aggregates. J Mol Model 2019; 25:229. [PMID: 31321557 DOI: 10.1007/s00894-019-4128-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 07/07/2019] [Indexed: 12/01/2022]
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disorder associated with the aggregation of the amyloid-beta peptide (Aβ) into large oligomers and fibrils that damage healthy brain cells. The predominant peptide fragments in the plaques are mainly formed by the Aβ1-40 and Aβ1-42 peptides, albeit the eleven-residue Aβ25-35 segment is largely used in biological studies because it retains the neurotoxic properties of the longer Aβ peptides. Recent studies indicate that treatment with therapeutic steroid hormones reduces the progress of the disease in AD models. Particularly, treatment with 17β-aminoestrogens (AEs) has shown a significant alleviation of the AD development by inhibiting oxidative stress and neuronal death. Yet, the mechanism by which the AE molecules exhibit their beneficial effects remains speculative. To shed light into the molecular mechanism of inhibition of the AD development by AEs, we investigated the possibility of direct interaction with the Aβ25-35 peptide. First, we calculate various interacting electronic properties of three AE derivatives as follows: prolame, butolame, and pentolame by performing DFT calculations. To account for the polymorphic nature of the Aβ aggregates, we considered four different Aβ25-35 systems extracted from AD relevant fibril structures. From the calculation of different electron density properties, specific interacting loci were identified that guided the construction and optimization of various complexes. Interestingly, the results suggest a similar inhibitory mechanism based on the direct interaction between the AEs and the M35 residue that seems to be general and independent of the polymorphic properties of the Aβ aggregates. Our analysis of the complex formation provides a structural framework for understanding the AE therapeutic properties in the molecular inhibitory mechanism of Aβ aggregation.
Collapse
Affiliation(s)
- Lisset Noriega
- Laboratorio de Química Teórica, Centro de Investigación. Depto. de Fisicoquímica, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Edif 105-I, San Claudio y 22 Sur, Ciudad Universitaria, Col. San Manuel, 72570, Puebla, Mexico
| | - Alfonso Díaz
- Departamento de Farmacia, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Av. San Claudio y 14 Sur, Col. San Manuel, 72570, Puebla, Mexico
| | - Daniel Limón
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Av. San Claudio y 14 Sur, Col. San Manuel, 72570, Puebla, Mexico
| | - María Eugenia Castro
- Centro de Química, Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla, Complejo de Ciencias, ICUAP, Edif. IC8, 22 Sur y San Claudio, Ciudad Universitaria, 72570, Puebla, Mexico
| | - Norma A Caballero
- Facultad de Ciencias Biológicas, Benemérita Universidad Autónoma de Puebla, San Claudio y 14 Sur, Ciudad Universitaria, Col. San Manuel, 72570, Puebla, Mexico
| | - Ramsés E Ramírez
- Departamento de Fisicomatemáticas, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Edif 105-I, San Claudio y 22 Sur, Ciudad Universitaria, Col. San Manuel, 72570, Puebla, Mexico
| | - Jose Manuel Perez-Aguilar
- Laboratorio de Química Teórica, Centro de Investigación. Depto. de Fisicoquímica, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Edif 105-I, San Claudio y 22 Sur, Ciudad Universitaria, Col. San Manuel, 72570, Puebla, Mexico.
| | - Francisco J Melendez
- Laboratorio de Química Teórica, Centro de Investigación. Depto. de Fisicoquímica, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Edif 105-I, San Claudio y 22 Sur, Ciudad Universitaria, Col. San Manuel, 72570, Puebla, Mexico.
| |
Collapse
|
36
|
Malfatti MC, Henneke G, Balachander S, Koh KD, Newnam G, Uehara R, Crouch RJ, Storici F, Tell G. Unlike the Escherichia coli counterpart, archaeal RNase HII cannot process ribose monophosphate abasic sites and oxidized ribonucleotides embedded in DNA. J Biol Chem 2019; 294:13061-13072. [PMID: 31300556 DOI: 10.1074/jbc.ra119.009493] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 07/05/2019] [Indexed: 12/12/2022] Open
Abstract
The presence of ribonucleoside monophosphates (rNMPs) in nuclear DNA decreases genome stability. To ensure survival despite rNMP insertions, cells have evolved a complex network of DNA repair mechanisms, in which the ribonucleotide excision repair pathway, initiated by type 2 RNase H (RNase HII/2), plays a major role. We recently demonstrated that eukaryotic RNase H2 cannot repair damage, that is, ribose monophosphate abasic (both apurinic or apyrimidinic) site (rAP) or oxidized rNMP embedded in DNA. Currently, it remains unclear why RNase H2 is unable to repair these modified nucleic acids having either only a sugar moiety or an oxidized base. Here, we compared the endoribonuclease specificity of the RNase HII enzymes from the archaeon Pyrococcus abyssi and the bacterium Escherichia coli, examining their ability to process damaged rNMPs embedded in DNA in vitro We found that E. coli RNase HII cleaves both rAP and oxidized rNMP sites. In contrast, like the eukaryotic RNase H2, P. abyssi RNase HII did not display any rAP or oxidized rNMP incision activities, even though it recognized them. Notably, the archaeal enzyme was also inactive on a mismatched rNMP, whereas the E. coli enzyme displayed a strong preference for the mispaired rNMP over the paired rNMP in DNA. On the basis of our biochemical findings and also structural modeling analyses of RNase HII/2 proteins from organisms belonging to all three domains of life, we propose that RNases HII/2's dual roles in ribonucleotide excision repair and RNA/DNA hydrolysis result in limited acceptance of modified rNMPs embedded in DNA.
Collapse
Affiliation(s)
- Matilde Clarissa Malfatti
- Laboratory of Molecular Biology and DNA Repair, Department of Medicine, University of Udine, 33100 Udine, Italy
| | - Ghislaine Henneke
- Ifremer, Univ Brest, CNRS, Laboratoire de Microbiologie des Environnements Extrêmes, F-29280 Plouzané, France
| | - Sathya Balachander
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia 30332
| | - Kyung Duk Koh
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia 30332
| | - Gary Newnam
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia 30332
| | - Ryo Uehara
- Ritsumeikan Global Innovation Research Organization, Ritsumeikan University, 1-1-1 Noji-higashi, Kusatsu, Shiga 525-8577, Japan
| | - Robert J Crouch
- Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892
| | - Francesca Storici
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia 30332
| | - Gianluca Tell
- Laboratory of Molecular Biology and DNA Repair, Department of Medicine, University of Udine, 33100 Udine, Italy.
| |
Collapse
|
37
|
DNA repair and neurological disease: From molecular understanding to the development of diagnostics and model organisms. DNA Repair (Amst) 2019; 81:102669. [PMID: 31331820 DOI: 10.1016/j.dnarep.2019.102669] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In both replicating and non-replicating cells, the maintenance of genomic stability is of utmost importance. Dividing cells can repair DNA damage during cell division, tolerate the damage by employing potentially mutagenic DNA polymerases or die via apoptosis. However, the options for accurate DNA repair are more limited in non-replicating neuronal cells. If DNA damage is left unresolved, neuronal cells die causing neurodegenerative disorders. A number of pathogenic variants of DNA repair proteins have been linked to multiple neurological diseases. The current challenge is to harness our knowledge of fundamental properties of DNA repair to improve diagnosis, prognosis and treatment of such debilitating disorders. In this perspective, we will focus on recent efforts in identifying novel DNA repair biomarkers for the diagnosis of neurological disorders and their use in monitoring the patient response to therapy. These efforts are greatly facilitated by the development of model organisms such as zebrafish, which will also be summarised.
Collapse
|
38
|
Fattoretti P, Malavolta M, Fabbietti P, Papa R, Giacconi R, Costarelli L, Galeazzi R, Paoloni C, Postacchini D, Lattanzio F, Giuli C. Oxidative Stress in Elderly with Different Cognitive Status: My Mind Project. J Alzheimers Dis 2019; 63:1405-1414. [PMID: 29843238 DOI: 10.3233/jad-171117] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND Biomarkers of oxidative stress have been associated with cognitive status in humans and have been proposed to guide prognosis/treatment in Alzheimer's disease (AD) and mild cognitive impairment (MCI). OBJECTIVE The aim of this study was to compare oxidative stress status in the plasma of mild-moderate AD, MCI, and healthy elderly with normal cognition (HE) undergoing a non-pharmacological intervention including multi-modal cognitive training ("My Mind Project"). METHODS A prospective randomized trial involving 321 elderly people enrolled in Marche Region, Italy. Each subject was randomly assigned to an experimental (cognitive training) or to a control group. Cognitive performances and biomarkers have been analyzed before intervention (baseline), immediately after termination (follow-up 1), after 6 months (follow-up 2), and after 2 years (follow-up 3). The biological antioxidant potential (BAP) to Diacron reactive oxygen metabolites (d-ROM) ratio has been used as an indicator of oxidative stress status and as outcome variable. RESULTS We have found no differences in the oxidative status among AD, MCI, and HE. Neither did we find a significant effect of the intervention within experimental groups. Gender was the sole factor with a strong significant effect on BAP/d-ROM. CONCLUSIONS Based on these results, the utility of biomarkers of oxidative stress to guide prognosis/treatment in AD or MCI seems to be limited by lack of specificity, large interindividual variability, and gender bias.
Collapse
Affiliation(s)
- Patrizia Fattoretti
- Cellular Bioenergetics Laboratory, Center for Neurobiology of Aging, INRCA-IRCCS, Ancona, Italy
| | - Marco Malavolta
- Advanced Technology Center for Aging Research, INRCA-IRCCS, Ancona, Italy
| | - Paolo Fabbietti
- Unit of Geriatric Pharmacoepidemiology, INRCA-IRCCS, Cosenza, Italy
| | - Roberta Papa
- Centre of Socio-Economic Gerontological Research, INRCA-IRCCS, Ancona, Italy
| | - Robertina Giacconi
- Advanced Technology Center for Aging Research, INRCA-IRCCS, Ancona, Italy
| | - Laura Costarelli
- Clinical Laboratory and Molecular Diagnostics, INRCA-IRCCS, Ancona, Italy
| | - Roberta Galeazzi
- Clinical Laboratory and Molecular Diagnostics, INRCA-IRCCS, Ancona, Italy
| | | | | | | | - Cinzia Giuli
- Geriatrics Operative Unit, INRCA-IRCCS, Fermo, Italy
| |
Collapse
|
39
|
Fluorescent protein nanoparticles: Synthesis and recognition of cellular oxidation damage. Colloids Surf B Biointerfaces 2019; 177:219-227. [PMID: 30743069 DOI: 10.1016/j.colsurfb.2019.01.065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 01/25/2019] [Accepted: 01/31/2019] [Indexed: 12/19/2022]
Abstract
Intracellular reactive oxygen species (ROS) generation are associated with many diseases. Lots of studies focus on the detection of intracellular ROS by small fluorescent molecules. However, ROS recognized by biocompatible nanoparticles are relatively less reported. It is widely known that albumin-based nanomaterials possess unique advantages in biomedical applications because they are biodegradable and biocompatible. Herein, fluorescent protein nanoparticles (PNPs) were prepared using BSA as a starting material without introducing extra fluorescent molecules. The blue fluorescent PNPs were well characterized by FL, FTIR, CD, TEM, DLS, etc. It was revealed that the PNPs exhibited two types of emissive centers through FL spectra and the fluorescence lifetimes. Further mechanism study indicated that the fluorescence of the PNPs was mainly derived from three kinds of aromatic amino acids, namely tryptophan, tyrosine and phenylalanine. Moreover, the fluorescence properties of the PNPs were tightly related to pH. The PNPs displayed excellent stabilities under harsh conditions as well as physiological conditions. In addition, the PNPs (200 μg/mL) were nontoxic to HeLa and GES-1 cell lines, showing good biocompatibility. The cellular uptake of PNPs was occurred only when the cells were stressed with glucose oxidase or H2O2, thereafter the bright blue fluorescence was observed, indicating that it could be utilized for the recognition of cellular oxidation damage. These findings will offer novel clues for the future synthesis of even brighter protein nanoparticles and their biomedical applications.
Collapse
|
40
|
Timpano S, Guild BD, Specker EJ, Melanson G, Medeiros PJ, Sproul SLJ, Uniacke J. Physioxic human cell culture improves viability, metabolism, and mitochondrial morphology while reducing DNA damage. FASEB J 2019; 33:5716-5728. [PMID: 30649960 DOI: 10.1096/fj.201802279r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Multicellular organisms balance oxygen delivery and toxicity by having oxygen pass through several barriers before cellular delivery. In human cell culture, these physiologic barriers are removed, exposing cells to higher oxygen levels. Human cells cultured in ambient air may appear normal, but this is difficult to assess without a comparison at physiologic oxygen. Here, we examined the effects of culturing human cells throughout the spectrum of oxygen availability on oxidative damage to macromolecules, viability, proliferation, the antioxidant and DNA damage responses, metabolism, and mitochondrial fusion and morphology. We surveyed 4 human cell lines cultured for 3 d at 7 oxygen conditions between 1 and 21% O2. We show that oxygen levels and cellular benefit are not inversely proportional, but the benefit peaks within the physioxic range. Normoxic cells are in a perpetual state of responding to damaged macromolecules and mitochondrial networks relative to physioxic cells, which could compromise an investigation. These data contribute to the concept of an optimal oxygen availability for cell culture in the physioxic range where the oxygen is not too high to reduce oxidative damage, and not too low for efficient oxidative metabolism, but just right: the Goldiloxygen zone.-Timpano, S., Guild, B. D., Specker, E. J., Melanson, G., Medeiros, P. J., Sproul, S. L. J., Uniacke, J. Physioxic human cell culture improves viability, metabolism, and mitochondrial morphology while reducing DNA damage.
Collapse
Affiliation(s)
- Sara Timpano
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Brianna D Guild
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Erin J Specker
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Gaelan Melanson
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Philip J Medeiros
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Shannon L J Sproul
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - James Uniacke
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
41
|
Laser ablation ICP-MS for simultaneous quantitative imaging of iron and ferroportin in hippocampus of human brain tissues with Alzheimer's disease. Talanta 2019; 197:413-421. [PMID: 30771955 DOI: 10.1016/j.talanta.2019.01.056] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 01/11/2019] [Accepted: 01/14/2019] [Indexed: 11/23/2022]
Abstract
Laser ablation inductively coupled plasma - mass spectrometry (LA-ICP-MS) is proposed for a better understanding of metals and proteins distribution in micrometre structures of human brain tissues. Simultaneous absolute quantitative imaging of Fe and ferroportin (FPN), in 5 µm thick tissue sections of the stratum pyramidale of hippocampus CA1 region, was carried out for Alzheimer disease (AD) patients and healthy controls (HC). For the imaging of FPN by LA-ICP-MS, antibodies were labelled via carbodiimide crosslinking with fluorescent gold nanoclusters (AuNCs) of 2.2 nm diameter, enabling a high amplification (314 gold atoms per NC). Laboratory made gelatin standards containing Fe and Au were used for LA-ICP-MS calibration. Results showed that iron presents an increased concentration in AD donors compared with HC donors, whereas similar concentrations of FPN in AD donors with respect to HC donors were obtained. The average absolute FPN concentrations in selected areas obtained with the proposed AuNCs method were compared with the levels obtained by densitometric analysis with a traditional IHC approach, observing a similar trend in all cases.
Collapse
|
42
|
Yusuf M, Khan M, Robaian MA, Khan RA. Biomechanistic insights into the roles of oxidative stress in generating complex neurological disorders. Biol Chem 2018; 399:305-319. [PMID: 29261511 DOI: 10.1515/hsz-2017-0250] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Accepted: 12/07/2017] [Indexed: 12/13/2022]
Abstract
Neurological diseases like Alzheimer's disease, epilepsy, parkinsonism, depression, Huntington's disease and amyotrophic lateral sclerosis prevailing globally are considered to be deeply influenced by oxidative stress-based changes in the biochemical settings of the organs. The excess oxygen concentration triggers the production of reactive oxygen species, and even the intrinsic antioxidant enzyme system, i.e. SOD, CAT and GSHPx, fails to manage their levels and keep them under desirable limits. This consequently leads to oxidation of protein, lipids and nucleic acids in the brain resulting in apoptosis, proteopathy, proteasomes and mitochondrion dysfunction, glial cell activation as well as neuroinflammation. The present exploration deals with the evidence-based mechanism of oxidative stress towards development of key neurological diseases along with the involved biomechanistics and biomaterials.
Collapse
Affiliation(s)
- Mohammad Yusuf
- College of Pharmacy, Taif University, Taif-Al-Haweiah 21974, Saudi Arabia
| | - Maria Khan
- College of Pharmacy, Taif University, Taif-Al-Haweiah 21974, Saudi Arabia
| | - Majed A Robaian
- College of Pharmacy, Taif University, Taif-Al-Haweiah 21974, Saudi Arabia
| | - Riaz A Khan
- Medicinal Chemistry Department, Qassim University, Qassim 51452, Saudi Arabia
- Department of Chemistry, MRIU, Faridabad, HR 121 001, India
| |
Collapse
|
43
|
Peña-Bautista C, Vigor C, Galano JM, Oger C, Durand T, Ferrer I, Cuevas A, López-Cuevas R, Baquero M, López-Nogueroles M, Vento M, Hervás D, García-Blanco A, Cháfer-Pericás C. Plasma lipid peroxidation biomarkers for early and non-invasive Alzheimer Disease detection. Free Radic Biol Med 2018; 124:388-394. [PMID: 29969716 DOI: 10.1016/j.freeradbiomed.2018.06.038] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 06/28/2018] [Accepted: 06/29/2018] [Indexed: 01/21/2023]
Abstract
INTRODUCTION Alzheimer Disease (AD) standard diagnosis is based on evaluations and biomarkers that are non-specific, expensive, or requires invasive sampling. Therefore, an early, and non-invasive diagnosis is required. As regards molecular mechanisms, recent research has shown that lipid peroxidation plays an important role. METHODS Well-defined participants groups were recruited. Lipid peroxidation compounds were determined in plasma using a validated analytical method. Statistical studies consisted of an elastic-net-penalized logistic regression adjustment. RESULTS The regression model fitted to the data included six variables (lipid peroxidation biomarkers) as potential predictors of early AD. This model achieved an apparent area under the receiver operating characteristics (AUC-ROCs) of 0.883 and a bootstrap-validated AUC-ROC of 0.817. Calibration of the model showed very low deviations from real probabilities. CONCLUSION A satisfactory early diagnostic model has been obtained from plasma levels of 6 lipid peroxidation compounds, indicating the individual probability of suffering from early AD.
Collapse
Affiliation(s)
| | - Claire Vigor
- Institut des Biomolécules Max Mousseron, IBMM, University of Montpellier, CNRS ENSCM, Montpellier, France
| | - Jean-Marie Galano
- Institut des Biomolécules Max Mousseron, IBMM, University of Montpellier, CNRS ENSCM, Montpellier, France
| | - Camille Oger
- Institut des Biomolécules Max Mousseron, IBMM, University of Montpellier, CNRS ENSCM, Montpellier, France
| | - Thierry Durand
- Institut des Biomolécules Max Mousseron, IBMM, University of Montpellier, CNRS ENSCM, Montpellier, France
| | - Inés Ferrer
- Neurology Unit, University and Polytechnic Hospital La Fe, Valencia, Spain
| | - Ana Cuevas
- Neurology Unit, University and Polytechnic Hospital La Fe, Valencia, Spain
| | | | - Miguel Baquero
- Neurology Unit, University and Polytechnic Hospital La Fe, Valencia, Spain
| | | | - Máximo Vento
- Neonatal Research Unit, Health Research Institute La Fe, Valencia, Spain
| | - David Hervás
- Biostatistical Unit, Health Research Institute La Fe, Valencia, Spain
| | - Ana García-Blanco
- Neonatal Research Unit, Health Research Institute La Fe, Valencia, Spain.
| | | |
Collapse
|
44
|
Circadian Rhythm and Alzheimer's Disease. Med Sci (Basel) 2018; 6:medsci6030052. [PMID: 29933646 PMCID: PMC6164904 DOI: 10.3390/medsci6030052] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 06/19/2018] [Accepted: 06/19/2018] [Indexed: 12/14/2022] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disorder with a growing epidemiological importance characterized by significant disease burden. Sleep-related pathological symptomatology often accompanies AD. The etiology and pathogenesis of disrupted circadian rhythm and AD share common factors, which also opens the perspective of viewing them as a mutually dependent process. This article focuses on the bi-directional relationship between these processes, discussing the pathophysiological links and clinical aspects. Common mechanisms linking both processes include neuroinflammation, neurodegeneration, and circadian rhythm desynchronization. Timely recognition of sleep-specific symptoms as components of AD could lead to an earlier and correct diagnosis with an opportunity of offering treatments at an earlier stage. Likewise, proper sleep hygiene and related treatments ought to be one of the priorities in the management of the patient population affected by AD. This narrative review brings a comprehensive approach to clearly demonstrate the underlying complexities linking AD and circadian rhythm disruption. Most clinical data are based on interventions including melatonin, but larger-scale research is still scarce. Following a pathophysiological reasoning backed by evidence gained from AD models, novel anti-inflammatory treatments and those targeting metabolic alterations in AD might prove useful for normalizing a disrupted circadian rhythm. By restoring it, benefits would be conferred for immunological, metabolic, and behavioral function in an affected individual. On the other hand, a balanced circadian rhythm should provide greater resilience to AD pathogenesis.
Collapse
|
45
|
Jin YY, Singh P, Chung HJ, Hong ST. Blood Ammonia as a Possible Etiological Agent for Alzheimer's Disease. Nutrients 2018; 10:E564. [PMID: 29734664 PMCID: PMC5986444 DOI: 10.3390/nu10050564] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 04/26/2018] [Accepted: 05/01/2018] [Indexed: 02/07/2023] Open
Abstract
Alzheimer’s disease (AD), characterized by cognitive decline and devastating neurodegeneration, is the most common age-related dementia. Since AD is a typical example of a complex disease that is affected by various genetic and environmental factors, various factors could be involved in preventing and/or treating AD. Extracellular accumulation of beta-amyloid peptide (Aβ) and intracellular accumulation of tau undeniably play essential roles in the etiology of AD. However, interestingly enough, medications targeting Aβ or tau all failed and the only clinically efficient medications for AD are drugs targeting the cholinergic pathway. Also, a very intriguing discovery in AD is that the Mediterranean diet (MeDi), containing an unusually large quantity of Lactobacilli, is very effective in preventing AD. Based on recently emerging findings, it is our opinion that the reduction of blood ammonia levels by Lactobacilli in MeDi is the therapeutic agent of MeDi for AD. The recent evidence of Lactobacilli lowering blood ammonia level not only provides a link between AD and MeDi but also provides a foundation of pharmabiotics for hyperammonemia as well as various neurological diseases.
Collapse
Affiliation(s)
- Yan Yan Jin
- Department of Biomedical Sciences and Institute for Medical Science, Chonbuk National University Medical School, Jeonju, Chonbuk 54907, Korea.
| | - Parul Singh
- Department of Biomedical Sciences and Institute for Medical Science, Chonbuk National University Medical School, Jeonju, Chonbuk 54907, Korea.
| | - Hea-Jong Chung
- Department of Biomedical Sciences and Institute for Medical Science, Chonbuk National University Medical School, Jeonju, Chonbuk 54907, Korea.
| | - Seong-Tschool Hong
- Department of Biomedical Sciences and Institute for Medical Science, Chonbuk National University Medical School, Jeonju, Chonbuk 54907, Korea.
| |
Collapse
|
46
|
Gill I, Kaur S, Kaur N, Dhiman M, Mantha AK. Phytochemical Ginkgolide B Attenuates Amyloid-β1-42 Induced Oxidative Damage and Altered Cellular Responses in Human Neuroblastoma SH-SY5Y Cells. J Alzheimers Dis 2018; 60:S25-S40. [PMID: 28234255 DOI: 10.3233/jad-161086] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Oxidative stress is an upsurge in reactive oxygen/nitrogen species (ROS/RNS), which aggravates damage to cellular components viz. lipids, proteins, and nucleic acids resulting in impaired cellular functions and neurological pathologies including Alzheimer's disease (AD). In the present study, we have examined amyloid-β (Aβ)-induced oxidative stress responses, a major cause for AD, in the undifferentiated and differentiated human neuroblastoma SH-SY5Y cells. Aβ1-42-induced oxidative damage was evaluated on lipids by lipid peroxidation; proteins by protein carbonyls; antioxidant status by SOD and GSH enzyme activities; and DNA and RNA damage levels by evaluating the number of AP sites and 8-OHG base damages produced. In addition, the neuro-protective role of the phytochemical ginkgolide B (GB) in countering Aβ1-42-induced oxidative stress was assessed. We report that the differentiated cells are highly vulnerable to Aβ1-42-induced oxidative stress events as exerted by the deposition of Aβ in AD. Results of the current study suggest that the pre-treatment of GB, followed by Aβ1-42 treatment for 24 h, displayed neuro-protective potential, which countered Aβ1-42-induced oxidative stress responses in both undifferentiated and differentiated SH-SY5Y neuronal cells by: 1) hampering production of ROS and RNS; 2) reducing lipid peroxidation; 3) decreasing protein carbonyl content; 4) restoring antioxidant activities of SOD and GSH enzymes; and 5) maintaining genome integrity by reducing the oxidative DNA and RNA base damages. In conclusion, Aβ1-42 induces oxidative damage to the cellular biomolecules, which are associated with AD pathology, and are protected by the pre-treatment of GB against Aβ-toxicity. Taken together, this study advocates for phytochemical-based therapeutic interventions against AD.
Collapse
Affiliation(s)
- Iqbal Gill
- Centre for Animal Sciences, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, Punjab, India
| | - Sukhchain Kaur
- Centre for Biochemistry and Microbial Sciences, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, Punjab, India
| | - Navrattan Kaur
- Centre for Animal Sciences, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, Punjab, India
| | - Monisha Dhiman
- Centre for Biochemistry and Microbial Sciences, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, Punjab, India
| | - Anil K Mantha
- Centre for Animal Sciences, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, Punjab, India
| |
Collapse
|
47
|
Jensen HLB, Lillenes MS, Rabano A, Günther CC, Riaz T, Kalayou ST, Ulstein ID, Bøhmer T, Tønjum T. Expression of nucleotide excision repair in Alzheimer's disease is higher in brain tissue than in blood. Neurosci Lett 2018; 672:53-58. [PMID: 29474873 DOI: 10.1016/j.neulet.2018.02.043] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 02/08/2018] [Accepted: 02/19/2018] [Indexed: 11/16/2022]
Abstract
Age-related changes are increased in patients with Alzheimer's disease (AD), including oxidative stress and DNA damage. We propose that genotoxic stress and DNA repair responses influence neurodegeneration in the pathogenesis of AD. Here, we focus on nucleotide excision repair (NER). Real-time qPCR and mass spectrometry were employed to determine the expression levels of selected NER components. The mRNA levels of the genes encoding the NER proteins RAD23B, RPA1, ERCC1, PCNA and LIG3 as well as the NER-interacting base excision repair protein MPG in blood and brain tissue from four brain regions in patients with AD or mild cognitive impairment and healthy controls (HC), were assessed. NER mRNA levels were significantly higher in brain tissue than in blood. Further, LIG3 mRNA levels in the frontal cortex was higher in AD versus HC, while mRNA levels of MPG and LIG3 in entorhinal cortex and RPA1 in the cerebellum were lower in AD versus HC. In blood, RPA1 and ERCC1 mRNA levels were lower in AD patients than in HC. Alterations in gene expression of NER components between brain regions were associated with AD, connecting DNA repair to AD pathogenesis and suggesting a distinct role for NER in the brain.
Collapse
Affiliation(s)
| | - Meryl S Lillenes
- Department of Microbiology, University of Oslo, Oslo, Norway; Department of Microbiology, Oslo University Hospital, Oslo, Norway.
| | - Alberto Rabano
- Centro Investigación Enfermedades Neurológicas (CIEN), Spain.
| | | | - Tahira Riaz
- Department of Microbiology, Oslo University Hospital, Oslo, Norway.
| | - Shewit T Kalayou
- Department of Microbiology, Oslo University Hospital, Oslo, Norway.
| | - Ingun D Ulstein
- The Memory Clinic, Department of Geriatric Medicine, Oslo University Hospital, Oslo, Norway.
| | - Thomas Bøhmer
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway.
| | - Tone Tønjum
- Department of Microbiology, University of Oslo, Oslo, Norway; Department of Microbiology, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
48
|
Alhasani RH, Biswas L, Tohari AM, Zhou X, Reilly J, He JF, Shu X. Gypenosides protect retinal pigment epithelium cells from oxidative stress. Food Chem Toxicol 2018; 112:76-85. [DOI: 10.1016/j.fct.2017.12.037] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 12/06/2017] [Accepted: 12/19/2017] [Indexed: 02/07/2023]
|
49
|
Bengoetxea X, de Cerain AL, Azqueta A, Ramirez MJ. Purported Interactions of Amyloid-β and Glucocorticoids in Cytotoxicity and Genotoxicity: Implications in Alzheimer's Disease. J Alzheimers Dis 2018; 54:1085-1094. [PMID: 27589535 DOI: 10.3233/jad-160636] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disorder characterized by the presence of aggregates of the amyloid-β peptide (Aβ) that are believed to be neurotoxic. One of the purposed damaging mechanisms of Aβ is oxidative insult, which eventually could damage the cellular genome. Stress and associated increases in glucocorticoids (GCs) have been described as a risk factor for the development of AD, although the purported genotoxic effects of GCs have not been fully characterized. Therefore, it is possible to speculate about purported synergistic effects of GCs on the Aβ-driven genotoxic damage. This in vitro study addresses the single and combined cyto/genotoxic effects of Aβ and GCs in SH-SY5Y cells. Cytotoxicity was determined by the MTT assay, and the genotoxic effects were studied using the comet assay. A comet assay derivation allows for measuring the presence of the FPG-sensitive sites (mainly 8-oxoguanines) in the DNA, apart from the DNA strand breaks. Treatment with Aβ (10 μM, 72 h) induced cytotoxicity (35% decrease in cell viability) and DNA strand breaks, but had no significant effect on oxidative DNA damage (FPG sites). Corticosterone showed no effect on cell viability, genotoxicity, or reparation processes. Corticosterone was unable to neither reverse nor potentiate Aβ driven effects. The present results suggest the existence of alternative mechanisms for the Aβ driven damage, not involving oxidative damage of DNA. In addition, could be suggested that the interaction between Aβ and GCs in AD does not seem to involve DNA damage.
Collapse
|
50
|
Arredondo M, González M, Latorre M. Copper. TRACE ELEMENTS AND MINERALS IN HEALTH AND LONGEVITY 2018. [DOI: 10.1007/978-3-030-03742-0_2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|