1
|
Liu S, Li Y, Yang J, Zhang L, Yan J. An in situ-activated and chemi-excited photooxygenation system based on G-poly(thioacetal) for Aβ 1-42 aggregates. J Mater Chem B 2024; 12:10850-10860. [PMID: 39417544 DOI: 10.1039/d4tb01147c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
The abnormal aggregation of Aβ proteins, inflammatory responses, and mitochondrial dysfunction have been reported as major targets in Alzheimer's disease (AD). Photooxygenation of the amyloid-β peptide (Aβ) is viewed as a promising therapeutic intervention for AD treatment. However, the limitations of the depth of the external light source passing through the brain and the toxic side effects on healthy tissues are two significant challenges in the photooxidation of Aβ aggregates. We proposed a method to initiate the chemical stimulation of Aβ1-42 aggregate oxidation through H2O2 and correct the abnormal microenvironment of the lesions by eliminating the cascading reactions of oxidative stress. The degradable G-poly(thioacetal) undergoes cascade release of cinnamaldehyde (CA) and thioacetal triggered by endogenous H2O2, with CA in turn amplifying degradation by generating more H2O2 through mitochondrial dysfunction. A series of novel photosensitizers have been prepared and synthesized for use in the photodynamic oxidation of Aβ1-42 aggregates under white light activation. The nanoparticles (BD-6-QM/NPs) self-assembled from BD-6-QM, bis[2,4,5-trichloro-6-(pentoxycarbonyl) phenyl] ester (CPPO), and G-poly(thioacetal) not only exhibit H2O2-stimulated controlled release but also can be chemically triggered by H2O2 to generate singlet oxygen to inhibit Aβ1-42 aggregates, reducing the Aβ1-42-induced neurotoxicity.
Collapse
Affiliation(s)
- Shasha Liu
- MOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, P. R. China.
| | - Yanping Li
- School of Medicine, Foshan University, Foshan 528225, P. R. China
| | - Jinrong Yang
- MOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, P. R. China.
| | - Lei Zhang
- MOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, P. R. China.
| | - Jinwu Yan
- MOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, P. R. China.
| |
Collapse
|
2
|
Dhauria M, Mondal R, Deb S, Shome G, Chowdhury D, Sarkar S, Benito-León J. Blood-Based Biomarkers in Alzheimer's Disease: Advancing Non-Invasive Diagnostics and Prognostics. Int J Mol Sci 2024; 25:10911. [PMID: 39456697 PMCID: PMC11507237 DOI: 10.3390/ijms252010911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 10/06/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Alzheimer's disease (AD), the most prevalent form of dementia, is expected to rise dramatically in incidence due to the global population aging. Traditional diagnostic approaches, such as cerebrospinal fluid analysis and positron emission tomography, are expensive and invasive, limiting their routine clinical use. Recent advances in blood-based biomarkers, including amyloid-beta, phosphorylated tau, and neurofilament light, offer promising non-invasive alternatives for early AD detection and disease monitoring. This review synthesizes current research on these blood-based biomarkers, highlighting their potential to track AD pathology and enhance diagnostic accuracy. Furthermore, this review uniquely integrates recent findings on protein-protein interaction networks and microRNA pathways, exploring novel combinations of proteomic, genomic, and epigenomic biomarkers that provide new insights into AD's molecular mechanisms. Additionally, we discuss the integration of these biomarkers with advanced neuroimaging techniques, emphasizing their potential to revolutionize AD diagnostics. Although large-scale validation is still needed, these biomarkers represent a critical advancement toward more accessible, cost-effective, and early diagnostic tools for AD.
Collapse
Affiliation(s)
| | - Ritwick Mondal
- Department of Clinical Pharmacology and Therapeutic Medicine, IPGMER and SSKM Hospital, Kolkata 700020, India;
| | - Shramana Deb
- Department of Stroke Medicine, Institute of Neuroscience, Kolkata 700017, India;
| | - Gourav Shome
- Department of Biological Sciences, Bose Institute, Kolkata 700054, India;
| | - Dipanjan Chowdhury
- Department of Internal Medicine, IPGMER and SSKM Hospital, Kolkata 700020, India; (D.C.); (S.S.)
| | - Shramana Sarkar
- Department of Internal Medicine, IPGMER and SSKM Hospital, Kolkata 700020, India; (D.C.); (S.S.)
| | - Julián Benito-León
- Department of Neurology, University Hospital “12 de Octubre”, ES-28041 Madrid, Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), ES-28041 Madrid, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), ES-28029 Madrid, Spain
- Department of Medicine, Complutense University, ES-28040 Madrid, Spain
| |
Collapse
|
3
|
Sultana R, Butterfield DA. Protein Oxidation in Aging and Alzheimer's Disease Brain. Antioxidants (Basel) 2024; 13:574. [PMID: 38790679 PMCID: PMC11117785 DOI: 10.3390/antiox13050574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 04/28/2024] [Accepted: 05/02/2024] [Indexed: 05/26/2024] Open
Abstract
Proteins are essential molecules that play crucial roles in maintaining cellular homeostasis and carrying out biological functions such as catalyzing biochemical reactions, structural proteins, immune response, etc. However, proteins also are highly susceptible to damage by reactive oxygen species (ROS) and reactive nitrogen species (RNS). In this review, we summarize the role of protein oxidation in normal aging and Alzheimer's disease (AD). The major emphasis of this review article is on the carbonylation and nitration of proteins in AD and mild cognitive impairment (MCI). The oxidatively modified proteins showed a strong correlation with the reported changes in brain structure, carbohydrate metabolism, synaptic transmission, cellular energetics, etc., of both MCI and AD brains compared to the controls. Some proteins were found to be common targets of oxidation and were observed during the early stages of AD, suggesting that those changes might be critical in the onset of symptoms and/or formation of the pathological hallmarks of AD. Further studies are required to fully elucidate the role of protein oxidation and nitration in the progression and pathogenesis of AD.
Collapse
Affiliation(s)
- Rukhsana Sultana
- Department of Neuroscience, School of Behavioral and Brain Sciences, The University of Texas at Dallas, 800 West Campbell Rd., Richardson, TX 75080, USA;
| | - D. Allan Butterfield
- Department of Chemistry, and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506, USA
| |
Collapse
|
4
|
Urati A, Angati A, Singh Gautam A, Dey M, Pandey SK, Singh RK. Neuroprotective responses of quercetin in regulation of biochemical, structural, and neurobehavioral effects in 28-day oral exposure of iron in rats. Toxicol Mech Methods 2024; 34:57-71. [PMID: 37680063 DOI: 10.1080/15376516.2023.2256840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/01/2023] [Accepted: 09/04/2023] [Indexed: 09/09/2023]
Abstract
BACKGROUND Iron is one of the essential metals that functions as a cofactor in various biological cascades in the brain. However, excessive iron accumulation in the brain may lead to neurodegeneration and may show toxic effects. Quercetin, a pigment flavonoid compound, has been proven to be a potent antioxidant and anti-inflammatory that can inhibit lipid peroxidation during metal-induced neurotoxicity. Although iron-induced neuroinflammation and neurodegeneration have been reported in many studies, but the proof for its exact mechanisms needs to be explored. PURPOSE The key target of the study was to explore the neuroprotective effect of quercetin after oral exposure of iron in rats and explore its underlying molecular mechanisms. RESULTS The outcomes of the study have shown that oral exposure to ferrous sulfate may modulate behavioral paradigms such as locomotor activity, neuromuscular coordination, and increased anxiety level. The pro-inflammatory cytokines (TNF-α, IL-1β and IL-6), apoptotic protein (caspase 3), beta-amyloid and phosphorylated tau were found to be increased on iron exposure. Also, the expressions of ferritin heavy and light chain, BACE-1 and GFAP expressions were altered. These behavioral, structural, and biochemical alterations in the brain were significantly and dose-dependently reversed by treatment with quercetin. CONCLUSION The current study provides a fundamental understanding of molecular signaling pathways, and structural proteins implicated in iron-induced neurotoxicity along with the ameliorative effects of quercetin.
Collapse
Affiliation(s)
- Anuradha Urati
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Lucknow, Uttar Pradesh, India
| | - Anok Angati
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Lucknow, Uttar Pradesh, India
| | - Avtar Singh Gautam
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Lucknow, Uttar Pradesh, India
| | - Mangaldeep Dey
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Lucknow, Uttar Pradesh, India
| | - Shivam Kumar Pandey
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Lucknow, Uttar Pradesh, India
| | - Rakesh Kumar Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Lucknow, Uttar Pradesh, India
| |
Collapse
|
5
|
Coria-Lucero C, Castro A, Ledezma C, Leporatti J, Ramirez D, Ghersi M, Delgado SM, Anzulovich AC, Navigatore-Fonzo L. An intracerebroventricular injection of AΒ (1-42) modifies temporal profiles of spatial memory performance and oxidative status in the temporal cortex rat. Brain Res 2023; 1804:148242. [PMID: 36646367 DOI: 10.1016/j.brainres.2023.148242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 12/26/2022] [Accepted: 01/11/2023] [Indexed: 01/15/2023]
Abstract
Alzheimer's dementia (AD) is a neurodegenerative disorder that causes memory loss and dementia in older adults. Intracellular accumulation of Aβ causes an imbalance in the oxidative status and cognitive dysfunctions. Besides oxidative stress and loss of memory, Alzheimer's patients show dysfunction of the circadian rhythms. The objective of this work was to evaluate the consequences of an intracerebroventricular injection of Aβ (1-42) on temporal patterns of cognitive performance, as well as on lipid peroxidation, protein oxidation and total antioxidant capacity levels, in the rat temporal cortex. Holtzman male rats from control and Aβ-injected groups were used in this study. We found that MDA, protein carbonyls and total antioxidant capacity levels displayed day-night oscillations in the rat temporal cortex and spatial memory performance also varied rhythmically. An intracerebroventricular injection of Aβ (1-42) modified temporal patterns of cognitive performance as well as daily profiles of parameters of oxidative stress. Thus, elevated levels of Aβ aggregates induces alterations in daily rhythmicity of parameters of oxidative stress and, consequently, would affect cellular clock activity, affecting the spatial memory performance in the AD.
Collapse
Affiliation(s)
- Cinthia Coria-Lucero
- Chronobiology Laboratory, Faculty of Chemistry, Biochemistry and Pharmacy, National University of San Luis. Multidisciplinary Institute of Biological Research of San Luis (IMIBIO-SL), CONICET, Ejército de Los Andes 950, CP D5700HHW San Luis, Argentina
| | - Andrea Castro
- Chronobiology Laboratory, Faculty of Chemistry, Biochemistry and Pharmacy, National University of San Luis. Multidisciplinary Institute of Biological Research of San Luis (IMIBIO-SL), CONICET, Ejército de Los Andes 950, CP D5700HHW San Luis, Argentina
| | - Carina Ledezma
- Chronobiology Laboratory, Faculty of Chemistry, Biochemistry and Pharmacy, National University of San Luis. Multidisciplinary Institute of Biological Research of San Luis (IMIBIO-SL), CONICET, Ejército de Los Andes 950, CP D5700HHW San Luis, Argentina
| | - Jorge Leporatti
- Faculty of Economic, Legal and Social Sciences, National University of San Luis, Campus Universitario, Ruta Prov. N° 55 (Ex. 148) Extremo Norte, D5700HHW San Luis, Argentina
| | - Darío Ramirez
- Laboratory of Experimental & Translational Medicine (LME&T), Faculty of Chemistry, Biochemistry and Pharmacy, National University of San Luis. Multidisciplinary Institute of Biological Research of San Luis (IMIBIO-SL), CONICET, Ejército de Los Andes 950, CP D5700HHW San Luis, Argentina
| | - Marisa Ghersi
- Institute of Experimental Pharmacology of Córdoba, Faculty of Chemical Sciences, National University of Córdoba (IFEC), CONICET, Haya De La Torre y Medina Allende S/N, CP D5000HHW Córdoba, Argentina
| | - Silvia Marcela Delgado
- Chronobiology Laboratory, Faculty of Chemistry, Biochemistry and Pharmacy, National University of San Luis. Multidisciplinary Institute of Biological Research of San Luis (IMIBIO-SL), CONICET, Ejército de Los Andes 950, CP D5700HHW San Luis, Argentina.
| | - Ana Cecilia Anzulovich
- Chronobiology Laboratory, Faculty of Chemistry, Biochemistry and Pharmacy, National University of San Luis. Multidisciplinary Institute of Biological Research of San Luis (IMIBIO-SL), CONICET, Ejército de Los Andes 950, CP D5700HHW San Luis, Argentina.
| | - Lorena Navigatore-Fonzo
- Chronobiology Laboratory, Faculty of Chemistry, Biochemistry and Pharmacy, National University of San Luis. Multidisciplinary Institute of Biological Research of San Luis (IMIBIO-SL), CONICET, Ejército de Los Andes 950, CP D5700HHW San Luis, Argentina.
| |
Collapse
|
6
|
The Dynamics of β-Amyloid Proteoforms Accumulation in the Brain of a 5xFAD Mouse Model of Alzheimer’s Disease. Int J Mol Sci 2021; 23:ijms23010027. [PMID: 35008451 PMCID: PMC8745018 DOI: 10.3390/ijms23010027] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/16/2021] [Accepted: 12/19/2021] [Indexed: 12/15/2022] Open
Abstract
Alzheimer’s disease (AD) is the leading cause of dementia among the elderly. Neuropathologically, AD is characterized by the deposition of a 39- to 42-amino acid long β-amyloid (Aβ) peptide in the form of senile plaques. Several post-translational modifications (PTMs) in the N-terminal domain have been shown to increase the aggregation and cytotoxicity of Aβ, and specific Aβ proteoforms (e.g., Aβ with isomerized D7 (isoD7-Aβ)) are abundant in the senile plaques of AD patients. Animal models are indispensable tools for the study of disease pathogenesis, as well as preclinical testing. In the presented work, the accumulation dynamics of Aβ proteoforms in the brain of one of the most widely used amyloid-based mouse models (the 5xFAD line) was monitored. Mass spectrometry (MS) approaches, based on ion mobility separation and the characteristic fragment ion formation, were applied. The results indicated a gradual increase in the Aβ fraction of isoD7-Aβ, starting from approximately 8% at 7 months to approximately 30% by 23 months of age. Other specific PTMs, in particular, pyroglutamylation, deamidation, and oxidation, as well as phosphorylation, were also monitored. The results for mice of different ages demonstrated that the accumulation of Aβ proteoforms correlate with the formation of Aβ deposits. Although the mouse model cannot be a complete analogue of the processes occurring in the human brain in AD, and several of the observed parameters differ significantly from human values supposedly due to the limited lifespan of the model animals, this dynamic study provides evidence on at least one of the possible mechanisms that can trigger amyloidosis in AD, i.e., the hypothesis on the relationship between the accumulation of isoD7-Aβ and the progression of AD-like pathology.
Collapse
|
7
|
Nguyen CD, Lee G. Neuroprotective Activity of Melittin-The Main Component of Bee Venom-Against Oxidative Stress Induced by Aβ 25-35 in In Vitro and In Vivo Models. Antioxidants (Basel) 2021; 10:antiox10111654. [PMID: 34829525 PMCID: PMC8614890 DOI: 10.3390/antiox10111654] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/09/2021] [Accepted: 10/18/2021] [Indexed: 11/23/2022] Open
Abstract
Melittin, a 26-amino acid peptide, is the main component of the venom of four honeybee species and exhibits neuroprotective actions. However, it is unclear how melittin ameliorates neuronal cells in oxidative stress and how it affects memory impairment in an in vivo model. We evaluated the neuroprotective effect of melittin on Aβ25–35-induced neuro-oxidative stress in both in vitro HT22 cells and in vivo animal model. Melittin effectively protected against HT22 cell viability and significantly deregulated the Aβ25–35-induced overproduction of intracellular reactive oxygen species. Western blot analysis showed that melittin suppressed cell apoptosis and regulated Bax/Bcl-2 ratio, as well as the expression of proapoptotic related factors: Apoptosis-inducing factor (AIF), Calpain, Cytochrome c (CytoC), Cleaved caspase-3 (Cleacas3). Additionally, melittin enhanced the antioxidant defense pathway by regulating the nuclear translocation of nuclear factor erythroid 2-like 2 (Nrf2) thus upregulated the production of the heme oxygenase-1 (HO-1), a major cellular antioxidant enzyme combating neuronal oxidative stress. Furthermore, melittin treatment activated the Tropomyosin-related kinase receptor B (TrkB)/cAMP Response Element-Binding (CREB)/Brain-derived neurotrophic factor (BDNF), contributing to neuronal neurogenesis, and regulating the normal function of synapses in the brain. In our in vivo experiment, melittin was shown to enhance the depleted learning and memory ability, a novel finding. A mouse model with cognitive deficits induced by Aβ25–35 intracerebroventricular injection was used. Melittin had dose-dependently enhanced neural-disrupted animal behavior and enhanced neurogenesis in the dentate gyrus hippocampal region. Further analysis of mouse brain tissue and serum confirmed that melittin enhanced oxidant–antioxidant balance, cholinergic system activity, and intercellular neurotrophic factors regulation, which were all negatively altered by Aβ25–35. Our study shows that melittin exerts antioxidant and neuroprotective actions against neural oxidative stress. Melittin can be a potential therapeutic agent for neurodegenerative disorders.
Collapse
|
8
|
Bennett JP, Onyango IG. Energy, Entropy and Quantum Tunneling of Protons and Electrons in Brain Mitochondria: Relation to Mitochondrial Impairment in Aging-Related Human Brain Diseases and Therapeutic Measures. Biomedicines 2021; 9:225. [PMID: 33671585 PMCID: PMC7927033 DOI: 10.3390/biomedicines9020225] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/18/2021] [Accepted: 02/18/2021] [Indexed: 11/16/2022] Open
Abstract
Adult human brains consume a disproportionate amount of energy substrates (2-3% of body weight; 20-25% of total glucose and oxygen). Adenosine triphosphate (ATP) is a universal energy currency in brains and is produced by oxidative phosphorylation (OXPHOS) using ATP synthase, a nano-rotor powered by the proton gradient generated from proton-coupled electron transfer (PCET) in the multi-complex electron transport chain (ETC). ETC catalysis rates are reduced in brains from humans with neurodegenerative diseases (NDDs). Declines of ETC function in NDDs may result from combinations of nitrative stress (NS)-oxidative stress (OS) damage; mitochondrial and/or nuclear genomic mutations of ETC/OXPHOS genes; epigenetic modifications of ETC/OXPHOS genes; or defects in importation or assembly of ETC/OXPHOS proteins or complexes, respectively; or alterations in mitochondrial dynamics (fusion, fission, mitophagy). Substantial free energy is gained by direct O2-mediated oxidation of NADH. Traditional ETC mechanisms require separation between O2 and electrons flowing from NADH/FADH2 through the ETC. Quantum tunneling of electrons and much larger protons may facilitate this separation. Neuronal death may be viewed as a local increase in entropy requiring constant energy input to avoid. The ATP requirement of the brain may partially be used for avoidance of local entropy increase. Mitochondrial therapeutics seeks to correct deficiencies in ETC and OXPHOS.
Collapse
Affiliation(s)
| | - Isaac G. Onyango
- International Clinical Research Center, St. Anne’s University Hospital, CZ-65691 Brno, Czech Republic;
| |
Collapse
|
9
|
Sokolova SV, Sozarukova MM, Khannanova AN, Grishina NK, Portnova GV, Proskurnina EV. [Antioxidant status in patients with paranoid schizophrenia and Alzheimer disease]. Zh Nevrol Psikhiatr Im S S Korsakova 2020; 120:82-87. [PMID: 32678552 DOI: 10.17116/jnevro202012006182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
OBJECTIVE To study the antioxidant profile of blood plasma in patients with paranoid schizophrenia and Alzheimer disease (AD). MATERIAL AND METHODS Thirty-three patients with paranoid schizophrenia and 18 patients with AD were included in the study. Patients with schizophrenia were stratified into two subgroups by response to therapy. The indicators of the antioxidant profile were determined using methods based on chemiluminometry and spectrofluorimetry. RESULTS Systemic oxidative stress due to insufficiency of low molecular weight plasma antioxidants is not determined neither in AD nor in treatment resistant schizophrenia. At the same time, a «thiol» oxidative stress, which indirectly indicates a deficiency of the glutathione system, is present in both groups. In patients with paranoid schizophrenia responsive to treatment, systemic oxidative stress is more pronounced and «thiol» oxidative stress is less significant. Among the antipsychotics studied, haloperidol, zuclopenthixol, risperidone and ziprasidone do not exhibit antioxidant properties, but periciazine, clozapine and especially chlorpromazine exhibit strong antioxidant properties, but they unlikely affect the antioxidant potential of blood plasma. CONCLUSIONS The glutathione part of the antioxidant system is mostly affected, but systemic oxidative stress is not significant in patients with treatment resistant paranoid schizophrenia and AD. Oxidative disorders are more pronounced in treatment responsive paranoid schizophrenia.
Collapse
Affiliation(s)
- S V Sokolova
- Lomonosov Moscow State University, Medical Research and Educational Center, Moscow, Russia
| | - M M Sozarukova
- Kurnakov Institute of General and Inorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - A N Khannanova
- Gilyarovsky Psychiatric Hospital, the branch of the Psychiatric Clinical Hospital No. 4 of the Moscow Health Department, Moscow, Russia
| | - N K Grishina
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, Russia
| | - G V Portnova
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, Russia
| | | |
Collapse
|
10
|
Wang S, Yao H, Xu Y, Hao R, Zhang W, Liu H, Huang Y, Guo W, Lu B. Therapeutic potential of a TrkB agonistic antibody for Alzheimer's disease. Theranostics 2020; 10:6854-6874. [PMID: 32550908 PMCID: PMC7295064 DOI: 10.7150/thno.44165] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 05/11/2020] [Indexed: 12/26/2022] Open
Abstract
Repeated failures of "Aβ-lowering" therapies call for new targets and therapeutic approaches for Alzheimer's disease (AD). We propose to treat AD by halting neuronal death and repairing synapses using a BDNF-based therapy. To overcome the poor druggability of BDNF, we have developed an agonistic antibody AS86 to mimic the function of BDNF, and evaluate its therapeutic potential for AD. Method: Biochemical, electrophysiological and behavioral techniques were used to investigate the effects of AS86 in vitro and in vivo. Results: AS86 specifically activated the BDNF receptor TrkB and its downstream signaling, without affecting its other receptor p75NTR. It promoted neurite outgrowth, enhanced spine growth and prevented Aβ-induced cell death in cultured neurons, and facilitated Long-Term Potentiation (LTP) in hippocampal slices. A single-dose tail-vein injection of AS86 activated TrkB signaling in the brain, with a half-life of 6 days in the blood and brain. Bi-weekly peripheral administration of AS86 rescued the deficits in object-recognition memory in the APP/PS1 mouse model. AS86 also reversed spatial memory deficits in the 11-month, but not 14-month old AD mouse model. Conclusion: These results demonstrate the potential of AS86 in AD therapy, suggesting that neuronal and/or synaptic repair as an alternative therapeutic strategy for AD.
Collapse
Affiliation(s)
- Shudan Wang
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China, 100084
- Beijing Tiantan Hospital, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China, 100070
- R&D Center for the Diagnosis and Treatment of Major Brain Diseases, Research Institute of Tsinghua University in Shenzhen, Shenzhen, Guangdong, China, 518057
| | - Hongyang Yao
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China, 100084
| | - Yihua Xu
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China, 100084
- R&D Center for the Diagnosis and Treatment of Major Brain Diseases, Research Institute of Tsinghua University in Shenzhen, Shenzhen, Guangdong, China, 518057
| | - Rui Hao
- Center of Translational Medicine, Tongji Hospital, Tongji University School of Medicine, Shanghai, China, 200065
| | - Wen Zhang
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China, 100084
| | - Hang Liu
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China, 100084
- R&D Center for the Diagnosis and Treatment of Major Brain Diseases, Research Institute of Tsinghua University in Shenzhen, Shenzhen, Guangdong, China, 518057
| | - Ying Huang
- Center of Translational Medicine, Tongji Hospital, Tongji University School of Medicine, Shanghai, China, 200065
| | - Wei Guo
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China, 100084
- Beijing Tiantan Hospital, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China, 100070
- R&D Center for the Diagnosis and Treatment of Major Brain Diseases, Research Institute of Tsinghua University in Shenzhen, Shenzhen, Guangdong, China, 518057
| | - Bai Lu
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China, 100084
- Beijing Tiantan Hospital, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China, 100070
- R&D Center for the Diagnosis and Treatment of Major Brain Diseases, Research Institute of Tsinghua University in Shenzhen, Shenzhen, Guangdong, China, 518057
| |
Collapse
|
11
|
Moore Z, Mobilio F, Walker FR, Taylor JM, Crack PJ. Abrogation of type-I interferon signalling alters the microglial response to Aβ 1-42. Sci Rep 2020; 10:3153. [PMID: 32081950 PMCID: PMC7035268 DOI: 10.1038/s41598-020-59917-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 01/31/2020] [Indexed: 11/08/2022] Open
Abstract
Neuroinflammation and accompanying microglial dysfunction are now appreciated to be involved in Alzheimer's disease (AD) pathogenesis. Critical to the process of neuroinflammation are the type-I interferon (IFN) family of cytokines. Efforts to phenotypically characterize microglia within AD identify distinct populations associated with type-I IFN signalling, yet how this affects underlying microglial function is yet to be fully elucidated. Here we demonstrate that Aβ1-42 exposure increases bioactive levels of type-I IFN produced by primary microglia alongside increased expression of type-I IFN related genes. Primary microglia isolated from brains of APPswePS1ΔE9 mice with ablated type-I IFN signalling show an increased phagocytic ability to uptake FITC-Aβ1-42. Correlative assessment of plaque sizes in aged APPswePS1ΔE9 mice with abrogated type-I IFN signalling show unchanged deposition levels. Microglia from these mice did however show alterations in morphology. This data further highlights the role of type-I IFN signalling within microglia and identifies a role in phagocytosis. As such, targeting both microglial and global type-I IFN signalling presents as a novel therapeutic strategy for AD management.
Collapse
Affiliation(s)
- Zachery Moore
- Department of Pharmacology and Therapeutics, The University of Melbourne, Melbourne, Australia
| | - Frank Mobilio
- Department of Pharmacology and Therapeutics, The University of Melbourne, Melbourne, Australia
| | - Frederick R Walker
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, Australia
| | - Juliet M Taylor
- Department of Pharmacology and Therapeutics, The University of Melbourne, Melbourne, Australia.
| | - Peter J Crack
- Department of Pharmacology and Therapeutics, The University of Melbourne, Melbourne, Australia.
| |
Collapse
|
12
|
Fatafta H, Poojari C, Sayyed-Ahmad A, Strodel B, Owen MC. Role of Oxidized Gly25, Gly29, and Gly33 Residues on the Interactions of Aβ 1-42 with Lipid Membranes. ACS Chem Neurosci 2020; 11:535-548. [PMID: 31939658 DOI: 10.1021/acschemneuro.9b00558] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Oxidative stress is known to play an important role in the pathogenesis of Alzheimer's disease. Moreover, it is becoming increasingly evident that the plasma membrane of neurons plays a role in modulating the aggregation and toxicity of Alzheimer's amyloid-β peptide (Aβ). In this study, the combined and interdependent effects of oxidation and membrane interactions on the 42 residues long Aβ isoform are investigated using molecular simulations. Hamiltonian replica exchange molecular dynamics simulations are utilized to elucidate the impact of selected oxidized glycine residues of Aβ42 on the interactions of the peptide with a model membrane comprised of 70% POPC, 25% cholesterol, and 5% of the ganglioside GM1. The main findings are that, independent of the oxidation state, Aβ prefers binding to GM1 over POPC, which is further enhanced by the oxidation of Gly29 and Gly33 and reduced the formation of β-sheet. Our results suggest that the differences observed in Aβ42 conformations and its interaction with a lipid bilayer upon oxidation originate from the position of the oxidized Gly residue with respect to the hydrophobic sequence of Aβ42 involving the Gly29-XXX-Gly33-XXX-Gly37 motif and from specific interactions between the peptide and the terminal sugar groups of GM1.
Collapse
Affiliation(s)
- Hebah Fatafta
- Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Chetan Poojari
- Department of Physics, University of Helsinki, P.O. Box 64, FI-00014 Helsinki, Finland
- Theoretical Physics and Center for Biophysics, Saarland University, Campus E2 6, 66123 Saarbrücken, Germany
| | | | - Birgit Strodel
- Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, 52425 Jülich, Germany
- Institute of Theoretical and Computational Chemistry, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Michael C. Owen
- CEITEC − Central European Institute of Technology, Masaryk University, Kamenice 753/5, Brno 625 00, Czech Republic
| |
Collapse
|
13
|
Cuevas E, Rosas-Hernandez H, Burks SM, Ramirez-Lee MA, Guzman A, Imam SZ, Ali SF, Sarkar S. Amyloid Beta 25-35 induces blood-brain barrier disruption in vitro. Metab Brain Dis 2019; 34:1365-1374. [PMID: 31267346 DOI: 10.1007/s11011-019-00447-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 06/05/2019] [Indexed: 11/27/2022]
Abstract
The amyloid β-peptide (Aβ) is transported across the blood-brain barrier (BBB) by binding with the receptor for advanced glycation end products (RAGE). Previously, we demonstrated that the Aβ fraction 25-35 (Aβ25-35) increases RAGE expression in the rat hippocampus, likely contributing to its neurotoxic effects. However, it is still debated if the interaction of Aβ with RAGE compromises the BBB function in Alzheimer' disease (AD). Here, we evaluated the effects of Aβ25-35 in an established in vitro model of the BBB. Rat brain microvascular endothelial cells (rBMVECs) were treated with 20 μM active Aβ25-35 or the inactive Aβ35-25 (control), for 24 h. Exposure to Aβ25-35 significantly decreased cell viability, increased cellular necrosis, and increased the production of reactive oxygen species (ROS), which triggered a decrease in the enzyme glutathione peroxidase when compared to the control condition. Aβ25-35 also increased BBB permeability by altering the expression of tight junction proteins (decreasing zonula occludens-1 and increasing occludin). Aβ25-35 induced monolayer disruption and cellular disarrangement of the BBB, with RAGE being highly expressed in the zones of disarrangement. Together, these data suggest that Aβ25-35-induces toxicity by compromising the functionality and integrity of the BBB in vitro. Graphical abstract Aβ25-35 induces BBB dysfunction in vitro, wich is likely mediated by OS and ultimately leads to disruption of BBB integrity and cell death.
Collapse
Affiliation(s)
- Elvis Cuevas
- Division of Neurotoxicology, National Center for Toxicological Research/U.S. Food and Drug Administration, Jefferson, AR, 72079, USA.
| | - Hector Rosas-Hernandez
- Division of Neurotoxicology, National Center for Toxicological Research/U.S. Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Susan M Burks
- Division of Neurotoxicology, National Center for Toxicological Research/U.S. Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Manuel A Ramirez-Lee
- Division of Neurotoxicology, National Center for Toxicological Research/U.S. Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Aida Guzman
- Escuela Nacional Preparatoria-UNAM, Mexico, Mexico
| | - Syed Z Imam
- Division of Neurotoxicology, National Center for Toxicological Research/U.S. Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Syed F Ali
- Division of Neurotoxicology, National Center for Toxicological Research/U.S. Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Sumit Sarkar
- Division of Neurotoxicology, National Center for Toxicological Research/U.S. Food and Drug Administration, Jefferson, AR, 72079, USA
| |
Collapse
|
14
|
Hatziagapiou K, Kakouri E, Lambrou GI, Bethanis K, Tarantilis PA. Antioxidant Properties of Crocus Sativus L. and Its Constituents and Relevance to Neurodegenerative Diseases; Focus on Alzheimer's and Parkinson's Disease. Curr Neuropharmacol 2019; 17:377-402. [PMID: 29564976 PMCID: PMC6482475 DOI: 10.2174/1570159x16666180321095705] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Revised: 01/03/2018] [Accepted: 03/19/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Reactive oxygen species and reactive nitrogen species, which are collectively called reactive oxygen-nitrogen species, are the inevitable by-products of cellular metabolic redox reactions, such as oxidative phosphorylation in the mitochondrial respiratory chain, phagocytosis, reactions of biotransformation of exogenous and endogenous substrata in endoplasmic reticulum, eicosanoid synthesis, and redox reactions in the presence of metal with variable valence. Among medicinal plants, there is growing interest in Crocus Sativus L. It is a perennial, stemless herb, belonging to Iridaceae family, cultivated in various countries such as Greece, Italy, Spain, Israel, Morocco, Turkey, Iran, India, China, Egypt and Mexico. OBJECTIVE The present study aims to address the protective role of Crocus Sativus L. in neurodegeneration with an emphasis in Parkinson's and Alzheimer's disease. MATERIALS AND METHODS An electronic literature search was conducted by two of the authors from 1993 to August 2017. Original articles and systematic reviews (with or without meta-analysis), as well as case reports were selected. Titles and abstracts of papers were screened by a third reviewer to determine whether they met the eligibility criteria, and full texts of the selected articles were retrieved. RESULTS Hence, the authors focused on the literature concerning the role of Crocus Sativus L. on its anti-oxidant and neuroprotective properties. CONCLUSION Literature findings represented in current review herald promising results for using Crocus Sativus L. and/or its active constituents as antioxidants, anti-inflammatory, and neuroprotective agents.
Collapse
Affiliation(s)
- Kyriaki Hatziagapiou
- First Department of Pediatrics, National and Kapodistrian University of Athens, Choremeio Research Laboratory, Hematology/Oncology Unit, Athens, Greece
| | - Eleni Kakouri
- Laboratory of Chemistry, Department of Food Science & Human Nutrition, School of Food Biotechnology and Development, Agricultural University of Athens, Athens, Greece
| | - George I Lambrou
- First Department of Pediatrics, National and Kapodistrian University of Athens, Choremeio Research Laboratory, Hematology/Oncology Unit, Athens, Greece
| | - Kostas Bethanis
- Physics Laboratory, Department of Biotechnology, School of Food Biotechnology and Development, Agricultural University of Athens, Greece
| | - Petros A Tarantilis
- Laboratory of Chemistry, Department of Food Science & Human Nutrition, School of Food Biotechnology and Development, Agricultural University of Athens, Athens, Greece
| |
Collapse
|
15
|
Pilkington AW, Donohoe GC, Akhmedov NG, Ferrebee T, Valentine SJ, Legleiter J. Hydrogen Peroxide Modifies Aβ-Membrane Interactions with Implications for Aβ 40 Aggregation. Biochemistry 2019; 58:2893-2905. [PMID: 31187978 DOI: 10.1021/acs.biochem.9b00233] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Alzheimer's disease (AD) is pathologically characterized by the formation of extracellular senile plaques, predominately comprised of aggregated β-amyloid (Aβ), deposited in the brain. Aβ aggregation can result in a myriad of distinct aggregate species, from soluble oligomers to insoluble fibrils. Aβ strongly interacts with membranes, which can be linked to a variety of potential toxic mechanisms associated with AD. Oxidative damage accompanies the formation of Aβ aggregates, with a 10-50% proportion of Aβ aggregates being oxidized in vivo. Hydrogen peroxide (H2O2) is a reactive oxygen species implicated in a number of neurodegenerative diseases. Recent evidence has demonstrated that the H2O2 concentration fluctuates rapidly in the brain, resulting in large concentration spikes, especially in the synaptic cleft. Here, the impact of environmental H2O2 on Aβ aggregation in the presence and absence of lipid membranes is investigated. Aβ40 was exposed to H2O2, resulting in the selective oxidation of methionine 35 (Met35) to produce Aβ40Met35[O]. While oxidation mildly reduced the rate of Aβ aggregation and produced a distinct fibril morphology at high H2O2 concentrations, H2O2 had a much more pronounced impact on Aβ aggregation in the presence of total brain lipid extract vesicles. The impact of H2O2 on Aβ aggregation in the presence of lipids was associated with a reduced affinity of Aβ for the vesicle surface. However, this reduced vesicle affinity was predominately associated with lipid peroxidation rather than Aβ oxidation.
Collapse
Affiliation(s)
- Albert W Pilkington
- The C. Eugene Bennett Department of Chemistry , West Virginia University , 217 Clark Hall , Morgantown , West Virginia 26506 , United States
| | - Gregory C Donohoe
- The C. Eugene Bennett Department of Chemistry , West Virginia University , 217 Clark Hall , Morgantown , West Virginia 26506 , United States
| | - Novruz G Akhmedov
- The C. Eugene Bennett Department of Chemistry , West Virginia University , 217 Clark Hall , Morgantown , West Virginia 26506 , United States
| | - Timothy Ferrebee
- The C. Eugene Bennett Department of Chemistry , West Virginia University , 217 Clark Hall , Morgantown , West Virginia 26506 , United States
| | - Stephen J Valentine
- The C. Eugene Bennett Department of Chemistry , West Virginia University , 217 Clark Hall , Morgantown , West Virginia 26506 , United States
| | - Justin Legleiter
- The C. Eugene Bennett Department of Chemistry , West Virginia University , 217 Clark Hall , Morgantown , West Virginia 26506 , United States.,Blanchette Rockefeller Neurosciences Institutes , West Virginia University , 1 Medical Center Drive , P.O. Box 9303, Morgantown , West Virginia 26505 , United States.,Department of Neuroscience , West Virginia University , 1 Medical Center Drive , P.O. Box 9303, Morgantown , West Virginia 26505 , United States
| |
Collapse
|
16
|
Leal MS, Briones X, Villalobos V, Queneau Y, Leiva A, Ríos HE, Pavez J, Silva CP, Carrasco C, Neira-Carrillo A, Roth AD, Tamayo L, Urzúa MD. Amino Acid-Functionalized Polyelectrolyte Films as Bioactive Surfaces for Cell Adhesion. ACS APPLIED MATERIALS & INTERFACES 2019; 11:19751-19762. [PMID: 31074956 DOI: 10.1021/acsami.9b02503] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Surfaces were prepared with polyelectrolyte derivatives of poly(styrene- alt-maleic anhydride) (PSMA) functionalized with amino acids of different hydropathy indices, with the aim of evaluating the effect of the chemical functionality of polyelectrolytes on SH-SY5Y neuroblastoma cell adhesion. Functionalizing PSMA derivatives with l-glutamine, l-methionine, and l-tyrosine yielded PSMA-Gln, PSMA-Met, and PSMA-Tyr polyelectrolytes, respectively. We first studied the adsorption behavior of PSMA functionalized with amino acids on silicon wafer surfaces modified with 3-aminopropyltriethoxysilane at pH 4.0 and 7.0 and at low and high ionic strengths. The highest rate of polyelectrolyte adsorption was at pH 4.0 and high ionic strength and was higher with the glutamine and tyrosine films. The advance contact angles (θA) of the polyelectrolyte surfaces showed a moderate effect of ionic strength and pH on polyelectrolyte film wettability, with PSMA-Tyr being slightly more hydrophobic. Atomic force microscopy images of the polyelectrolyte surfaces showed two types of morphology: the well-defined globular nanostructure of PSMA-Met and PSMA-Tyr and densely packed nanofibrous-like structure of PSMA-Gln. The highest level of ionic strength caused a slight decrease in the size of the nanostructure that formed the surface domains, which was reflected in the degree of surface roughness. Cell adhesion assays with the polyelectrolyte film showed that SH-SY5Y neuroblastoma cells cultured on PSMA-Met present a well-extended morphology characterized by a stellate shape, with five or more actin-rich thin processes, whereas SH-SY5Y cells that were seeded on PSMA-Gln and PSMA-Tyr have a round morphology, with fewer and shorter processes. These results indicate that it is possible to modulate the surface characteristics of polyelectrolyte films based on their chemical functionality and environmental parameters such as pH and ionic strength in order to evaluate their effect on cell adhesion. Thus, surfaces prepared from polyelectrolytes functionalized with amino acids are an attractive and simple platform for cell adhesion, which can be used in developing biomaterials with modulated surface properties.
Collapse
Affiliation(s)
- M S Leal
- Departamento de Química, Facultad de Ciencias , Universidad de Chile , Las Palmeras , 3425 Santiago , Chile
| | - X Briones
- Departamento de Química, Facultad de Ciencias , Universidad de Chile , Las Palmeras , 3425 Santiago , Chile
| | - V Villalobos
- Instituto de Ciencias Químicas Aplicadas, Facultad de Ingeniería , Universidad Autónoma de Chile , El Llano Subercaseaux , 2801 San Miguel , Chile
| | - Y Queneau
- Université de Lyon, ICBMS, UMR 5246, CNRS, UCBL, INSA Lyon, CPE Lyon, Bât. Lederer , 1 Rue Victor Grignard , 69622 Villeurbanne Cedex , France
| | - A Leiva
- Departamento Química Física, Facultad de Química , Pontificia Universidad Católica de Chile , Macul , 7820436 Santiago , Chile
| | - H E Ríos
- Departamento de Química, Facultad de Ciencias , Universidad de Chile , Las Palmeras , 3425 Santiago , Chile
| | - J Pavez
- Departamento de Química de los Materiales, Fac. de Química-Biología , Universidad de Santiago de Chile , Av. B. O'Higgins , 3363 Santiago , Chile
| | - C P Silva
- Departamento de Química de los Materiales, Fac. de Química-Biología , Universidad de Santiago de Chile , Av. B. O'Higgins , 3363 Santiago , Chile
| | - C Carrasco
- Departamento de Biología, Facultad de Ciencias , Universidad de Chile , P. C. 780-0023 Santiago , Chile
| | - Andrónico Neira-Carrillo
- Faculty de Ciencias Veterinarias y Pecuarias , Universidad de Chile , Av. Sta. Rosa , 11735 Santiago , Chile
| | - A D Roth
- Departamento de Biología, Facultad de Ciencias , Universidad de Chile , P. C. 780-0023 Santiago , Chile
| | - L Tamayo
- Departamento de Química, Facultad de Ciencias , Universidad de Chile , Las Palmeras , 3425 Santiago , Chile
| | - M D Urzúa
- Departamento de Química, Facultad de Ciencias , Universidad de Chile , Las Palmeras , 3425 Santiago , Chile
| |
Collapse
|
17
|
Sibiya SG, Mbandla MV, Govender T, Shobo A, Daniels WMU. Poly-N-methylated Aβ-Peptide C-Terminal fragments (MEPTIDES) reverse the deleterious effects of amyloid-β in rats. Metab Brain Dis 2018; 33:387-396. [PMID: 28993949 DOI: 10.1007/s11011-017-0118-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 09/26/2017] [Indexed: 01/11/2023]
Abstract
Alzheimer's disease (AD) is characterized by extracellular deposition of amyloid-β (Aβ) plaques. These protein deposits impair synaptic plasticity thereby producing a progressive decline in cognitive function. Current therapies are merely palliative and only slow cognitive decline. Poly-N-methylated Aβ-Peptide C-Terminal Fragments (MEPTIDES) were recently shown to reduce Aβ toxicity in vitro and in Drosophila melanogaster, however whether these novel compounds are effective in inhibiting Aβ-induced toxicity in the mammalian brain remains unclear. We therefore investigated whether MEPTIDES have the ability to reduce the neurotoxic effects of Aβ in male Sprague-Dawley (SD) rats. Aβ42 (100 μg, 2 mM) or vehicle (0.15 M Tris buffer) was stereotaxically injected bilaterally into the dorsal hippocampus at a rate of 1 μl/min for 10 min. The effects on hippocampal-mediated learning were subsequently assessed using the Morris water maze (MWM). The presence of apoptotic activity was also assessed by determining the expression levels of active caspase-3 using real-time polymerase chain reaction and Western Blot techniques. In addition, half of the animals (n = 20) received an intraperitoneal (i.p.) injection of MEPTIDES (2 mg/kg) 48 h after intrahippocampal injection of Aβ42. Matrix-assisted laser desorption/ionization-time-of-flight (MALDI -TOF) mass spectrometry (MS) showed that MEPTIDES crossed the blood brain barrier (BBB) and revealed their distribution in the rat brain. Rats treated with Aβ42 displayed spatial learning deficits and increased hippocampal caspase-3 gene (CASP-3) expression which was reversed by subsequent injection of MEPTIDES. The present results show that MEPTIDES have the potential to reverse the toxic effects of Aβ42 in vivo.
Collapse
Affiliation(s)
- Siya G Sibiya
- College of Health Sciences, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Musa V Mbandla
- College of Health Sciences, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Thavi Govender
- Catalysis and Peptide Research Unit, School of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Adeola Shobo
- Catalysis and Peptide Research Unit, School of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - William M U Daniels
- College of Health Sciences, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa.
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
| |
Collapse
|
18
|
Butterfield DA, Boyd-Kimball D. Oxidative Stress, Amyloid-β Peptide, and Altered Key Molecular Pathways in the Pathogenesis and Progression of Alzheimer's Disease. J Alzheimers Dis 2018; 62:1345-1367. [PMID: 29562527 PMCID: PMC5870019 DOI: 10.3233/jad-170543] [Citation(s) in RCA: 281] [Impact Index Per Article: 40.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2017] [Indexed: 12/12/2022]
Abstract
Oxidative stress is implicated in the pathogenesis and progression of Alzheimer's disease (AD) and its earlier stage, amnestic mild cognitive impairment (aMCI). One source of oxidative stress in AD and aMCI brains is that associated with amyloid-β peptide, Aβ1-42 oligomers. Our laboratory first showed in AD elevated oxidative stress occurred in brain regions rich in Aβ1-42, but not in Aβ1-42-poor regions, and was among the first to demonstrate Aβ peptides led to lipid peroxidation (indexed by HNE) in AD and aMCI brains. Oxidatively modified proteins have decreased function and contribute to damaged key biochemical and metabolic pathways in which these proteins normally play a role. Identification of oxidatively modified brain proteins by the methods of redox proteomics was pioneered in the Butterfield laboratory. Four recurring altered pathways secondary to oxidative damage in brain from persons with AD, aMCI, or Down syndrome with AD are interrelated and contribute to neuronal death. This "Quadrilateral of Neuronal Death" includes altered: glucose metabolism, mTOR activation, proteostasis network, and protein phosphorylation. Some of these pathways are altered even in brains of persons with preclinical AD. We opine that targeting these pathways pharmacologically and with lifestyle changes potentially may provide strategies to slow or perhaps one day, prevent, progression or development of this devastating dementing disorder. This invited review outlines both in vitro and in vivo studies from the Butterfield laboratory related to Aβ1-42 and AD and discusses the importance and implications of some of the major achievements of the Butterfield laboratory in AD research.
Collapse
Affiliation(s)
- D. Allan Butterfield
- Department of Chemistry and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Debra Boyd-Kimball
- Department of Chemistry and Biochemistry, University of Mount Union, Alliance, OH, USA
| |
Collapse
|
19
|
Roher AE, Kokjohn TA, Clarke SG, Sierks MR, Maarouf CL, Serrano GE, Sabbagh MS, Beach TG. APP/Aβ structural diversity and Alzheimer's disease pathogenesis. Neurochem Int 2017; 110:1-13. [PMID: 28811267 PMCID: PMC5688956 DOI: 10.1016/j.neuint.2017.08.007] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 07/25/2017] [Accepted: 08/11/2017] [Indexed: 02/01/2023]
Abstract
The amyloid cascade hypothesis of Alzheimer's disease (AD) proposes amyloid- β (Aβ) is a chief pathological element of dementia. AD therapies have targeted monomeric and oligomeric Aβ 1-40 and 1-42 peptides. However, alternative APP proteolytic processing produces a complex roster of Aβ species. In addition, Aβ peptides are subject to extensive posttranslational modification (PTM). We propose that amplified production of some APP/Aβ species, perhaps exacerbated by differential gene expression and reduced peptide degradation, creates a diverse spectrum of modified species which disrupt brain homeostasis and accelerate AD neurodegeneration. We surveyed the literature to catalog Aβ PTM including species with isoAsp at positions 7 and 23 which may phenocopy the Tottori and Iowa Aβ mutations that result in early onset AD. We speculate that accumulation of these alterations induce changes in secondary and tertiary structure of Aβ that favor increased toxicity, and seeding and propagation in sporadic AD. Additionally, amyloid-β peptides with a pyroglutamate modification at position 3 and oxidation of Met35 make up a substantial portion of sporadic AD amyloid deposits. The intrinsic physical properties of these species, including resistance to degradation, an enhanced aggregation rate, increased neurotoxicity, and association with behavioral deficits, suggest their emergence is linked to dementia. The generation of specific 3D-molecular conformations of Aβ impart unique biophysical properties and a capacity to seed the prion-like global transmission of amyloid through the brain. The accumulation of rogue Aβ ultimately contributes to the destruction of vascular walls, neurons and glial cells culminating in dementia. A systematic examination of Aβ PTM and the analysis of the toxicity that they induced may help create essential biomarkers to more precisely stage AD pathology, design countermeasures and gauge the impacts of interventions.
Collapse
Affiliation(s)
- Alex E Roher
- Division of Neurobiology, Barrow Neurological Institute, Phoenix, AZ 85013, USA; Division of Clinical Education, Midwestern University, Glendale, AZ 85308, USA.
| | - Tyler A Kokjohn
- Department of Microbiology, Midwestern University, Glendale, AZ 85308, USA
| | - Steven G Clarke
- Department of Chemistry and Biochemistry and the Molecular Biology Institute, University of California, Los Angeles, Los Angeles CA 90095-1569, USA
| | - Michael R Sierks
- Department of Chemical Engineering, Arizona State University, Tempe, AZ 85287-6106, USA
| | - Chera L Maarouf
- Laboratory of Neuropathology, Banner Sun Health Research Institute, Sun City, AZ 85351, USA
| | - Geidy E Serrano
- Laboratory of Neuropathology, Banner Sun Health Research Institute, Sun City, AZ 85351, USA
| | - Marwan S Sabbagh
- Alzheimer's and Memory Disorders Division, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Thomas G Beach
- Laboratory of Neuropathology, Banner Sun Health Research Institute, Sun City, AZ 85351, USA
| |
Collapse
|
20
|
Membrane-Accelerated Amyloid-β Aggregation and Formation of Cross-β Sheets. MEMBRANES 2017; 7:membranes7030049. [PMID: 28858214 PMCID: PMC5618134 DOI: 10.3390/membranes7030049] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 07/26/2017] [Accepted: 08/23/2017] [Indexed: 11/17/2022]
Abstract
Amyloid- β aggregates play a causative role in Alzheimer's disease. These aggregates are a product of the physical environment provided by the basic neuronal membrane, composed of a lipid bilayer. The intrinsic properties of the lipid bilayer allow amyloid- β peptides to nucleate and form well-ordered cross- β sheets within the membrane. Here, we correlate the aggregation of the hydrophobic fragment of the amyloid- β protein, A β 25 - 35 , with the hydrophobicity, fluidity, and charge density of a lipid bilayer. We summarize recent biophysical studies of model membranes and relate these to the process of aggregation in physiological systems.
Collapse
|
21
|
Lozano L, Guevara J, Lefebvre T, Ramos-Martinez I, Limón D, Díaz A, Cerón E, Zenteno E. Effect of amyloid-Β (25-35) in hyperglycemic and hyperinsulinemic rats, effects on phosphorylation and O-GlcNAcylation of tau protein. Neuropeptides 2017; 63:18-27. [PMID: 28427866 DOI: 10.1016/j.npep.2017.04.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 03/31/2017] [Accepted: 04/03/2017] [Indexed: 01/09/2023]
Abstract
Aggregation of the amyloid beta (Aβ) peptide and hyperphosphorylation of tau protein, which are markers of Alzheimer's disease (AD), have been reported also in diabetes mellitus (DM). One regulator of tau phosphorylation is O-GlcNAcylation, whereas for hyperphosphorylation it could be GSK3beta, which is activated in hyperglycemic conditions. With this in mind, both O-GlcNAcylation and phosphorylation of tau protein were evaluated in the brain of rats with streptozotocin (STZ)-induced hyperglycemia and hyperinsulinemia and treated with the Aß25-35 peptide in the hippocampal region CA1. Weight, glycated hemoglobin, glucose, and insulin were determined. Male Wistar rats were divided in groups (N=20): a) control, b) treated only with the Aβ25-35 peptide, c) treated with Aβ25-35 and STZ, and d) treated only with STZ. Results showed statistically significant differences in the mean weight, glucose levels, insulin concentration, and HbA1c percentage, between C- and D-treated groups and not STZ-treated A and B (P<0.05). Interestingly, our results showed diminution of O-GlcNAcylation and increase in P-tau-Ser-396 in the hippocampal area of the Aβ25-35- and STZ-treated groups; moreover, enhanced expression of GSK3beta was observed in this last group. Our results suggest that hyperinsulinemia-Aβ25-35-hyperglycemia is relevant for the down regulation of O-GlcNAcylation and up-regulation of the glycogen synthase kinase-3 beta (GSK3beta), favoring Aβ25-35-induced neurotoxicity in the brain of rats.
Collapse
Affiliation(s)
- Liliana Lozano
- Departamento de Bioquímica, Facultad de Medicina Universidad Nacional Autónoma de México, Av. Ciudad Universitaria 3000, C.P. 04510 Coyoacán, CDMX, Mexico; Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Av. Ciudad Universitaria 3000, C.P. 04510 Coyoacán, CDMX, Mexico
| | - Jorge Guevara
- Departamento de Bioquímica, Facultad de Medicina Universidad Nacional Autónoma de México, Av. Ciudad Universitaria 3000, C.P. 04510 Coyoacán, CDMX, Mexico
| | - Tony Lefebvre
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F 59000 Lille, France
| | - Ivan Ramos-Martinez
- Departamento de Bioquímica, Facultad de Medicina Universidad Nacional Autónoma de México, Av. Ciudad Universitaria 3000, C.P. 04510 Coyoacán, CDMX, Mexico
| | - Daniel Limón
- Laboratorio de Neurofarmacología, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Alfonso Díaz
- Departamento de Farmacia, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Eduarda Cerón
- Instituto Nacional de Enfermedades Respiratorias, "Ismael Cosío Villegas", Department of Biochemistry, Calz. de Tlalpan 4502, C.P. 14080 CDMX, Mexico
| | - Edgar Zenteno
- Departamento de Bioquímica, Facultad de Medicina Universidad Nacional Autónoma de México, Av. Ciudad Universitaria 3000, C.P. 04510 Coyoacán, CDMX, Mexico.
| |
Collapse
|
22
|
Carelli-Alinovi C, Misiti F. Methionine 35 sulphoxide reduces toxicity of Aβ in red blood cell. Eur J Clin Invest 2017; 47:314-321. [PMID: 28177519 DOI: 10.1111/eci.12735] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 02/04/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND The oxidation of methionine residue in position 35 of Ab to sulphoxide (Ab-sulphoxide) has the ability to deeply modify wild-type Ab 1-42 (Ab) neurotoxic action. Our previous studies suggest that in nucleated cells, lower toxicity of Ab-sulphoxide might result not from structural alteration, but from elevation of methionine sulphoxide reductase A (MsrA) activity and mRNA levels. DESIGN On this basis, we hypothesised that red blood cell (RBC), a cell devoid almost completely of MsrA activity, shares with nucleated cells an antioxidant system induced by methionine 35 sulphoxide, responsible for the lower toxicity of Ab-sulphoxide in RBC. (Results) Supporting this hypothesis, we found that the low toxicity of Ab-sulphoxide in RBC correlated with pentose phosphate pathway (PPP) flux increase, and this event was associated with a low level of methionine oxidation in total proteins. None of these effects were observed when cells were exposed to Ab native. DISCUSSION These results outline the importance of the redox state of methionine 35 in the modulation of Ab-mediated events and suggest an important protective role for PPP in RBC of patients affected by Alzheimer's disease.
Collapse
Affiliation(s)
- Cristiana Carelli-Alinovi
- School of Medicine, Biochemistry and Clinical Biochemistry Institute, Catholic University, Rome, Italy
| | - Francesco Misiti
- Human Sciences, Society and Health Department, University of Cassino and Southern Lazio, Cassino, Italy
| |
Collapse
|
23
|
Athari Nik Azm S, Vafa M, Sharifzadeh M, Safa M, Barati A, Mirshafiey A. Effects of M2000 (D-Mannuronic Acid) on Learning, Memory Retrieval, and Associated Determinants in a Rat Model of Alzheimer's Disease. Am J Alzheimers Dis Other Demen 2017; 32:12-21. [PMID: 28100077 PMCID: PMC10852923 DOI: 10.1177/1533317516678086] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2024]
Abstract
The d-mannuronic acid (M2000) is a novel nonsteroidal anti-inflammatory drug that has immunosuppressive effects together with antioxidant property. M2000 has shown a notable efficacy in experimental models of multiple sclerosis, rheumatoid arthritis, and nephrotic syndrome. In this work, the effect of M2000 on the treatment of Alzheimer's disease (AD) was performed by Morris water maze experiment, and the immunological assessments were carried out by Western blot, apoptosis (procaspase-3, Bax/Bcl2, P53), enzymatic (superoxide dismutase [SOD]), and nonenzymatic oxidative stress (malondialdehyde [MDA]) tests. We found that pretreatment of AD in the rat model by M2000 had a potent efficacy on rat behavior and also it led to a significant inhibition of amyloid plaque production. Moreover, our data showed that M2000 can reduce the amount of Bax/Bcl2, P53, MDA, and SOD, as well as it normalized the level of procaspase-3. Our results suggest M2000 is a potential therapeutic agent for the treatment of AD.
Collapse
Affiliation(s)
- Somayeh Athari Nik Azm
- Department of Cellular and Molecular Nutrition, School of Nutritional Science and Dietetics, Tehran University of Medical Science, Tehran, Iran
| | - Mohammadreza Vafa
- Department of Nutrition, School of Public Health, Iran University of Medical Science, Tehran, Iran
| | - Mohammad Sharifzadeh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Science, Tehran, Iran
| | - Majid Safa
- Cellular and Molecular Research Centre, School of Allied Medical Science, Iran University of Medical Science, Tehran, Iran
| | - Anis Barati
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Abbas Mirshafiey
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
24
|
Murugavelu M, Karthikeyan B. Synthesis, characterization of Ag-Au core-shell bimetal nanoparticles and its application for electrocatalytic oxidation/sensing of l-methionine. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 70:656-664. [PMID: 27770939 DOI: 10.1016/j.msec.2016.09.046] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2016] [Revised: 08/30/2016] [Accepted: 09/21/2016] [Indexed: 10/21/2022]
|
25
|
Barbieri A, De Carlo Chimienti R, Del Giacco T, Di Stefano S, Lanzalunga O, Lapi A, Mazzonna M, Olivo G, Salamone M. Oxidation of Aryl Diphenylmethyl Sulfides Promoted by a Nonheme Iron(IV)-Oxo Complex: Evidence for an Electron Transfer-Oxygen Transfer Mechanism. J Org Chem 2016; 81:2513-20. [PMID: 26886491 DOI: 10.1021/acs.joc.6b00099] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The oxidation of a series of aryl diphenylmethyl sulfides (4-X-C6H4SCH(C6H5)2, where X = OCH3 (1), X = CH3 (2), X = H (3), and X = CF3 (4)) promoted by the nonheme iron(IV)-oxo complex [(N4Py)Fe(IV)═O](2+) occurs by an electron transfer-oxygen transfer (ET-OT) mechanism as supported by the observation of products (diphenylmethanol, benzophenone, and diaryl disulfides) deriving from α-C-S and α-C-H fragmentation of radical cations 1(+•)-4(+•), formed besides the S-oxidation products (aryl diphenylmethyl sulfoxides). The fragmentation/S-oxidation product ratios regularly increase through a decrease in the electron-donating power of the aryl substituents, that is, by increasing the fragmentation rate constants of the radical cations as indicated by a laser flash photolysis (LFP) study of the photochemical oxidation of 1-4 carried out in the presence of N-methoxyphenanthridinium hexafluorophosphate (MeOP(+)PF6(-)).
Collapse
Affiliation(s)
- Alessia Barbieri
- Dipartimento di Chimica and Istituto CNR di Metodologie Chimiche-IMC, Sezione Meccanismi di Reazione c/o Dipartimento di Chimica, Università degli Studi di Roma "La Sapienza" , P. le A. Moro 5, 00185 Rome, Italy
| | - Rosemilia De Carlo Chimienti
- Dipartimento di Chimica and Istituto CNR di Metodologie Chimiche-IMC, Sezione Meccanismi di Reazione c/o Dipartimento di Chimica, Università degli Studi di Roma "La Sapienza" , P. le A. Moro 5, 00185 Rome, Italy
| | - Tiziana Del Giacco
- Dipartimento di Chimica, Biologia e Biotecnologie and Centro di Eccellenza Materiali Innovativi Nanostrutturati, Università di Perugia , Via Elce di Sotto 8, 06123 Perugia, Italy
| | - Stefano Di Stefano
- Dipartimento di Chimica and Istituto CNR di Metodologie Chimiche-IMC, Sezione Meccanismi di Reazione c/o Dipartimento di Chimica, Università degli Studi di Roma "La Sapienza" , P. le A. Moro 5, 00185 Rome, Italy
| | - Osvaldo Lanzalunga
- Dipartimento di Chimica and Istituto CNR di Metodologie Chimiche-IMC, Sezione Meccanismi di Reazione c/o Dipartimento di Chimica, Università degli Studi di Roma "La Sapienza" , P. le A. Moro 5, 00185 Rome, Italy
| | - Andrea Lapi
- Dipartimento di Chimica and Istituto CNR di Metodologie Chimiche-IMC, Sezione Meccanismi di Reazione c/o Dipartimento di Chimica, Università degli Studi di Roma "La Sapienza" , P. le A. Moro 5, 00185 Rome, Italy
| | - Marco Mazzonna
- Dipartimento di Chimica and Istituto CNR di Metodologie Chimiche-IMC, Sezione Meccanismi di Reazione c/o Dipartimento di Chimica, Università degli Studi di Roma "La Sapienza" , P. le A. Moro 5, 00185 Rome, Italy
| | - Giorgio Olivo
- Dipartimento di Chimica and Istituto CNR di Metodologie Chimiche-IMC, Sezione Meccanismi di Reazione c/o Dipartimento di Chimica, Università degli Studi di Roma "La Sapienza" , P. le A. Moro 5, 00185 Rome, Italy
| | - Michela Salamone
- Dipartimento di Scienze e Tecnologie Chimiche, Università "Tor Vergata" , Via della Ricerca Scientifica 1, I-00133 Rome, Italy
| |
Collapse
|
26
|
Roher AE, Maarouf CL, Kokjohn TA. Familial Presenilin Mutations and Sporadic Alzheimer’s Disease Pathology: Is the Assumption of Biochemical Equivalence Justified? J Alzheimers Dis 2016; 50:645-58. [DOI: 10.3233/jad-150757] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- Alex E. Roher
- Longtine Center for Neurodegenerative Biochemistry, Banner Sun Health Research Institute, Sun City, AZ, USA
| | - Chera L. Maarouf
- Longtine Center for Neurodegenerative Biochemistry, Banner Sun Health Research Institute, Sun City, AZ, USA
| | - Tyler A. Kokjohn
- Department of Microbiology, Midwestern University School of Medicine, Glendale, AZ, USA
| |
Collapse
|
27
|
Triplett JC, Swomley AM, Cai J, Klein JB, Butterfield DA. Quantitative phosphoproteomic analyses of the inferior parietal lobule from three different pathological stages of Alzheimer's disease. J Alzheimers Dis 2016; 49:45-62. [PMID: 26444780 DOI: 10.3233/jad-150417] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Alzheimer's disease (AD), the most common age-related neurodegenerative disorder, is clinically characterized by progressive neuronal loss resulting in loss of memory and dementia. AD is histopathologically characterized by the extensive distribution of senile plaques and neurofibrillary tangles, and synapse loss. Amnestic mild cognitive impairment (MCI) is generally accepted to be an early stage of AD. MCI subjects have pathology and symptoms that fall on the scale intermediately between 'normal' cognition with little or no pathology and AD. A rare number of individuals, who exhibit normal cognition on psychometric tests but whose brains show widespread postmortem AD pathology, are classified as 'asymptomatic' or 'preclinical' AD (PCAD). In this study, we evaluated changes in protein phosphorylation states in the inferior parietal lobule of subjects with AD, MCI, PCAD, and control brain using a 2-D PAGE proteomics approach in conjunction with Pro-Q Diamond phosphoprotein staining. Statistically significant changes in phosphorylation levels were found in 19 proteins involved in energy metabolism, neuronal plasticity, signal transduction, and oxidative stress response. Changes in the disease state phosphoproteome may provide insights into underlying mechanisms for the preservation of memory with expansive AD pathology in PCAD and the progressive memory loss in amnestic MCI that escalates to the dementia and the characteristic pathology of AD brain.
Collapse
Affiliation(s)
- Judy C Triplett
- Department of Chemistry, University of Kentucky, Lexington, KY, USA
| | - Aaron M Swomley
- Department of Chemistry, University of Kentucky, Lexington, KY, USA
| | - Jian Cai
- Department of Nephrology and Proteomics Center, University of Louisville, Louisville, KY, USA
| | - Jon B Klein
- Department of Nephrology and Proteomics Center, University of Louisville, Louisville, KY, USA
| | - D Allan Butterfield
- Department of Chemistry, University of Kentucky, Lexington, KY, USA
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
28
|
Bettoni M, Del Giacco T, Stradiotto M, Elisei F. Photoinduced One-Electron Oxidation of Benzyl Methyl Sulfides in Acetonitrile: Time-Resolved Spectroscopic Evidence for a Thionium Ion Intermediate. J Org Chem 2015; 80:8001-8. [DOI: 10.1021/acs.joc.5b01052] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Marta Bettoni
- Dipartimento
di Ingegneria Civile ed Ambientale, ‡Dipartimento di Chimica, Biologia
e Biotecnologie, and §Centro di Eccellenza Materiali Innovativi Nanostrutturati (CEMIN), Università di Perugia, 06123 Perugia, Italy
| | - Tiziana Del Giacco
- Dipartimento
di Ingegneria Civile ed Ambientale, ‡Dipartimento di Chimica, Biologia
e Biotecnologie, and §Centro di Eccellenza Materiali Innovativi Nanostrutturati (CEMIN), Università di Perugia, 06123 Perugia, Italy
| | - Marina Stradiotto
- Dipartimento
di Ingegneria Civile ed Ambientale, ‡Dipartimento di Chimica, Biologia
e Biotecnologie, and §Centro di Eccellenza Materiali Innovativi Nanostrutturati (CEMIN), Università di Perugia, 06123 Perugia, Italy
| | - Fausto Elisei
- Dipartimento
di Ingegneria Civile ed Ambientale, ‡Dipartimento di Chimica, Biologia
e Biotecnologie, and §Centro di Eccellenza Materiali Innovativi Nanostrutturati (CEMIN), Università di Perugia, 06123 Perugia, Italy
| |
Collapse
|
29
|
Friedemann M, Helk E, Tiiman A, Zovo K, Palumaa P, Tõugu V. Effect of methionine-35 oxidation on the aggregation of amyloid-β peptide. Biochem Biophys Rep 2015; 3:94-99. [PMID: 29124171 PMCID: PMC5668694 DOI: 10.1016/j.bbrep.2015.07.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 07/22/2015] [Accepted: 07/28/2015] [Indexed: 01/12/2023] Open
Abstract
Aggregation of Aβ peptides into amyloid plaques is considered to trigger the Alzheimer’s disease (AD), however the mechanism behind the AD onset has remained elusive. It is assumed that the insoluble Aβ aggregates enhance oxidative stress (OS) by generating free radicals with the assistance of bound copper ions. The aim of our study was to establish the role of Met35 residue in the oxidation and peptide aggregation processes. Met35 can be readily oxidized by H2O2. The fibrillization of Aβ with Met35 oxidized to sulfoxide was three times slower compared to that of the regular peptide. The fibrils of regular and oxidized peptides looked similar under transmission electron microscopy. The relatively small inhibitory effect of methionine oxidation on the fibrillization suggests that the possible variation in the Met oxidation state should not affect the in vivo plaque formation. The peptide oxidation pattern was more complex when copper ions were present: addition of one oxygen atom was still the fastest process, however, it was accompanied by multiple unspecific modifications of peptide residues. Addition of copper ions to the Aβ with oxidized Met35 in the presence of H2O2, resulted a similar pattern of nonspecific modifications, suggesting that the one-electron oxidation processes in the peptide molecule do not depend on the oxidation state of Met35 residue. Thus, it can be concluded that Met35 residue is not a part of the radical generating mechanism of Aβ–Cu(II) complex. Aβ peptides with oxidized Met35 residue fibrillize three times slower than the reduced peptide. Met35 is the only residue in Aβ peptide that is oxidized by H2O2 in the absent of copper ions. In the presence of copper ions as catalyst multiple unspecific oxidative processes occur in Aβ. Previous oxidation of Met35 does not affect the unspecific oxidation in the presence of copper ions.
Collapse
Key Words
- AD, Alzheimer's disease
- Alzheimer's disease
- Aβ, Alzheimer's amyloid peptide
- Copper(II)ion
- HFIP, 1,1,1,3,3,3-hexafluoro-2-propanol
- Methionine oxidation
- OS, oxidative stress
- ROS, reactive oxygen species
- ThT, Thioflavin T
- β-amyloid
Collapse
Affiliation(s)
- Merlin Friedemann
- Department of Gene Technology, Tallinn University of Technology, Akadeemia tee 15, 12618 Tallinn, Estonia
| | - Eneken Helk
- Department of Gene Technology, Tallinn University of Technology, Akadeemia tee 15, 12618 Tallinn, Estonia
| | - Ann Tiiman
- Department of Gene Technology, Tallinn University of Technology, Akadeemia tee 15, 12618 Tallinn, Estonia
| | - Kairit Zovo
- Department of Gene Technology, Tallinn University of Technology, Akadeemia tee 15, 12618 Tallinn, Estonia
| | - Peep Palumaa
- Department of Gene Technology, Tallinn University of Technology, Akadeemia tee 15, 12618 Tallinn, Estonia
| | - Vello Tõugu
- Department of Gene Technology, Tallinn University of Technology, Akadeemia tee 15, 12618 Tallinn, Estonia
| |
Collapse
|
30
|
Ferreira MES, de Vasconcelos AS, da Costa Vilhena T, da Silva TL, da Silva Barbosa A, Gomes ARQ, Dolabela MF, Percário S. Oxidative Stress in Alzheimer's Disease: Should We Keep Trying Antioxidant Therapies? Cell Mol Neurobiol 2015; 35:595-614. [PMID: 25616523 PMCID: PMC11486210 DOI: 10.1007/s10571-015-0157-y] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 01/13/2015] [Indexed: 11/28/2022]
Abstract
The risk of chronic diseases such as Alzheimer's disease is growing as a result of the continuous increasing average life span of the world population, a syndrome characterized by the presence of intraneural neurofibrillary tangles and senile plaques composed mainly by beta-amyloid protein, changes that may cause a number of progressive disorders in the elderly, causing, in its most advanced stage, difficulty in performing normal daily activities, among other manifestations. Therefore, it is important to understand the underlying pathogenic mechanisms of this syndrome. Nevertheless, despite intensive effort to access the physiopathological pathways of the disease, it remains poorly understood. In that context, some hypotheses have arisen, including the recent oxidative stress hypothesis, theory supported by the involvement of oxidative stress in aging, and the vulnerability of neurons to oxidative attack. In the present revision, oxidative changes and redox mechanisms in Alzheimer's disease will be further stressed, as well as the grounds for antioxidant supplementation as adjuvant therapy for the disease will be addressed.
Collapse
Affiliation(s)
- Michelli Erica Souza Ferreira
- Oxidative Stress Research Lab, Institute of Biological Sciences (LAPEO – ICB), Federal University of Pará, Av. Augusto Correa, 01, Belém, PA 66075-110 Brazil
| | - Amanda Soares de Vasconcelos
- Oxidative Stress Research Lab, Institute of Biological Sciences (LAPEO – ICB), Federal University of Pará, Av. Augusto Correa, 01, Belém, PA 66075-110 Brazil
| | - Thyago da Costa Vilhena
- Pharmacy Department, Institute of Health Sciences, Federal University of Pará, Av. Augusto Correa, 01, Belém, PA 66075-110 Brazil
| | - Thiago Leite da Silva
- Pharmacy Department, Institute of Health Sciences, Federal University of Pará, Av. Augusto Correa, 01, Belém, PA 66075-110 Brazil
| | - Aline da Silva Barbosa
- Oxidative Stress Research Lab, Institute of Biological Sciences (LAPEO – ICB), Federal University of Pará, Av. Augusto Correa, 01, Belém, PA 66075-110 Brazil
| | - Antonio Rafael Quadros Gomes
- Oxidative Stress Research Lab, Institute of Biological Sciences (LAPEO – ICB), Federal University of Pará, Av. Augusto Correa, 01, Belém, PA 66075-110 Brazil
| | - Maria Fani Dolabela
- Pharmacy Department, Institute of Health Sciences, Federal University of Pará, Av. Augusto Correa, 01, Belém, PA 66075-110 Brazil
| | - Sandro Percário
- Oxidative Stress Research Lab, Institute of Biological Sciences (LAPEO – ICB), Federal University of Pará, Av. Augusto Correa, 01, Belém, PA 66075-110 Brazil
| |
Collapse
|
31
|
|
32
|
Thummayot S, Tocharus C, Pinkaew D, Viwatpinyo K, Sringarm K, Tocharus J. Neuroprotective effect of purple rice extract and its constituent against amyloid beta-induced neuronal cell death in SK-N-SH cells. Neurotoxicology 2014; 45:149-58. [PMID: 25451968 DOI: 10.1016/j.neuro.2014.10.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 09/28/2014] [Accepted: 10/22/2014] [Indexed: 10/24/2022]
Abstract
This study evaluated the protective effects of purple rice (Oryza sativa L.) extract (PRE) and its major constituent, cyanidin, and their underlying mechanisms against Aβ 25-35-induced neuronal cell death in SK-N-SH cells. Aβ 25-35-induced neuronal toxicity is characterized by decrease in cell viability, the release of lactate dehydrogenase (LDH), decrease superoxide dismutase (SOD) activity, increase in reactive oxygen species (ROS) production, morphological alteration, and activation of mitochondrial death pathway. Pretreatment with PRE and cyanidin significantly attenuated Aβ 25-35-induced loss of cell viability, apoptosis, and increase in ROS and RNS production in a dose-dependent manner. In addition, PRE and cyanidin also helped to bring about the downregulation of the expression of Bax, cytochrome c, cleavage caspase-9, and cleavage caspase-3 proteins, and the upregulation of the Bcl-XL protein in cascade. Therefore, it is evident that PRE and its major constituent, cyanidin, were successful in protecting from the cytotoxic effect of Aβ 25-35 through attenuation ROS and RNS production and modulation of mitochondrial death pathway in SK-N-SH cells. This result suggests that PRE and its major constituent, cyanidin, might be beneficial as potential therapeutic agents in preventing neurodegenerative diseases.
Collapse
Affiliation(s)
- Sarinthorn Thummayot
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Chainarong Tocharus
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Decha Pinkaew
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Kittikun Viwatpinyo
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Korawan Sringarm
- Department of Animal and Aquatic Science, Faculty of Agriculture, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Jiraporn Tocharus
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand.
| |
Collapse
|
33
|
Butterfield DA. The 2013 SFRBM discovery award: selected discoveries from the butterfield laboratory of oxidative stress and its sequela in brain in cognitive disorders exemplified by Alzheimer disease and chemotherapy induced cognitive impairment. Free Radic Biol Med 2014; 74:157-74. [PMID: 24996204 PMCID: PMC4146642 DOI: 10.1016/j.freeradbiomed.2014.06.006] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 06/05/2014] [Accepted: 06/10/2014] [Indexed: 12/21/2022]
Abstract
This retrospective review on discoveries of the roles of oxidative stress in brain of subjects with Alzheimer disease (AD) and animal models thereof as well as brain from animal models of chemotherapy-induced cognitive impairment (CICI) results from the author receiving the 2013 Discovery Award from the Society for Free Radical Biology and Medicine. The paper reviews our laboratory's discovery of protein oxidation and lipid peroxidation in AD brain regions rich in amyloid β-peptide (Aβ) but not in Aβ-poor cerebellum; redox proteomics as a means to identify oxidatively modified brain proteins in AD and its earlier forms that are consistent with the pathology, biochemistry, and clinical presentation of these disorders; how Aβ in in vivo, ex vivo, and in vitro studies can lead to oxidative modification of key proteins that also are oxidatively modified in AD brain; the role of the single methionine residue of Aβ(1-42) in these processes; and some of the potential mechanisms in the pathogenesis and progression of AD. CICI affects a significant fraction of the 14 million American cancer survivors, and due to diminished cognitive function, reduced quality of life of the persons with CICI (called "chemobrain" by patients) often results. A proposed mechanism for CICI employed the prototypical ROS-generating and non-blood brain barrier (BBB)-penetrating chemotherapeutic agent doxorubicin (Dox, also called adriamycin, ADR). Because of the quinone moiety within the structure of Dox, this agent undergoes redox cycling to produce superoxide free radical peripherally. This, in turn, leads to oxidative modification of the key plasma protein, apolipoprotein A1 (ApoA1). Oxidized ApoA1 leads to elevated peripheral TNFα, a proinflammatory cytokine that crosses the BBB to induce oxidative stress in brain parenchyma that affects negatively brain mitochondria. This subsequently leads to apoptotic cell death resulting in CICI. This review outlines aspects of CICI consistent with the clinical presentation, biochemistry, and pathology of this disorder. To the author's knowledge this is the only plausible and self-consistent mechanism to explain CICI. These two different disorders of the CNS affect millions of persons worldwide. Both AD and CICI share free radical-mediated oxidative stress in brain, but the source of oxidative stress is not the same. Continued research is necessary to better understand both AD and CICI. The discoveries about these disorders from the Butterfield Laboratory that led to the 2013 Discovery Award from the Society of Free Radical and Medicine provide a significant foundation from which this future research can be launched.
Collapse
Affiliation(s)
- D Allan Butterfield
- Department of Chemistry, Center of Membrane Sciences, Free Radical Biology in Cancer, Shared Resource Facility of the Markey Cancer Center, Spinal Cord and Brain Injury Research Center, and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506, USA.
| |
Collapse
|
34
|
Swomley AM, Förster S, Keeney JT, Triplett J, Zhang Z, Sultana R, Butterfield DA. Abeta, oxidative stress in Alzheimer disease: evidence based on proteomics studies. BIOCHIMICA ET BIOPHYSICA ACTA 2014; 1842:1248-57. [PMID: 24120836 PMCID: PMC3981962 DOI: 10.1016/j.bbadis.2013.09.015] [Citation(s) in RCA: 142] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Revised: 09/27/2013] [Accepted: 09/28/2013] [Indexed: 01/01/2023]
Abstract
The initiation and progression of Alzheimer disease (AD) is a complex process not yet fully understood. While many hypotheses have been provided as to the cause of the disease, the exact mechanisms remain elusive and difficult to verify. Proteomic applications in disease models of AD have provided valuable insights into the molecular basis of this disorder, demonstrating that on a protein level, disease progression impacts numerous cellular processes such as energy production, cellular structure, signal transduction, synaptic function, mitochondrial function, cell cycle progression, and proteasome function. Each of these cellular functions contributes to the overall health of the cell, and the dysregulation of one or more could contribute to the pathology and clinical presentation in AD. In this review, foci reside primarily on the amyloid β-peptide (Aβ) induced oxidative stress hypothesis and the proteomic studies that have been conducted by our laboratory and others that contribute to the overall understanding of this devastating neurodegenerative disease.
Collapse
Affiliation(s)
- Aaron M Swomley
- Department of Chemistry, Center of Membrane Sciences, University of Kentucky, Lexington, KY 40506, USA; Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506, USA
| | - Sarah Förster
- Department of Biochemistry, Institute of Animal Sciences, University of Bonn, Bonn, Germany
| | - Jierel T Keeney
- Department of Chemistry, Center of Membrane Sciences, University of Kentucky, Lexington, KY 40506, USA; Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506, USA
| | - Judy Triplett
- Department of Chemistry, Center of Membrane Sciences, University of Kentucky, Lexington, KY 40506, USA; Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506, USA
| | - Zhaoshu Zhang
- Department of Chemistry, Center of Membrane Sciences, University of Kentucky, Lexington, KY 40506, USA; Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506, USA
| | - Rukhsana Sultana
- Department of Chemistry, Center of Membrane Sciences, University of Kentucky, Lexington, KY 40506, USA; Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506, USA
| | - D Allan Butterfield
- Department of Chemistry, Center of Membrane Sciences, University of Kentucky, Lexington, KY 40506, USA; Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506, USA.
| |
Collapse
|
35
|
Oxidative stress and metabolic syndrome: cause or consequence of Alzheimer's disease? OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014; 2014:497802. [PMID: 24683436 PMCID: PMC3941786 DOI: 10.1155/2014/497802] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Revised: 12/02/2013] [Accepted: 12/18/2013] [Indexed: 12/30/2022]
Abstract
Alzheimer's disease (AD) is a major neurodegenerative disease affecting the elderly. Clinically, it is characterized by a progressive loss of memory and cognitive function. Neuropathologically, it is characterized by the presence of extracellular β-amyloid (Aβ) deposited as neuritic plaques (NP) and neurofibrillary tangles (NFT) made of abnormal and hyperphosphorylated tau protein. These lesions are capable of generating the neuronal damage that leads to cell death and cognitive failure through the generation of reactive oxygen species (ROS). Evidence indicates the critical role of Aβ metabolism in prompting the oxidative stress observed in AD patients. However, it has also been proposed that oxidative damage precedes the onset of clinical and pathological AD symptoms, including amyloid-β deposition, neurofibrillary tangle formation, vascular malfunction, metabolic syndrome, and cognitive decline. This paper provides a brief description of the three main proteins associated with the development of the disease (Aβ, tau, and ApoE) and describes their role in the generation of oxidative stress. Finally, we describe the mitochondrial alterations that are generated by Aβ and examine the relationship of vascular damage which is a potential prognostic tool of metabolic syndrome. In addition, new therapeutic approaches targeting ROS sources and metabolic support were reported.
Collapse
|
36
|
Butterfield DA, Swomley AM, Sultana R. Amyloid β-peptide (1-42)-induced oxidative stress in Alzheimer disease: importance in disease pathogenesis and progression. Antioxid Redox Signal 2013; 19:823-35. [PMID: 23249141 PMCID: PMC3749710 DOI: 10.1089/ars.2012.5027] [Citation(s) in RCA: 395] [Impact Index Per Article: 32.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Revised: 12/05/2012] [Accepted: 12/17/2012] [Indexed: 12/14/2022]
Abstract
SIGNIFICANCE Alzheimer disease (AD) is an age-related neurodegenerative disease. AD is characterized by progressive cognitive impairment. One of the main histopathological hallmarks of AD brain is the presence of senile plaques (SPs) and another is elevated oxidative stress. The main component of SPs is amyloid beta-peptide (Aβ) that is derived from the proteolytic cleavage of amyloid precursor protein. RECENT ADVANCES Recent studies are consistent with the notion that methionine present at 35 position of Aβ is critical to Aβ-induced oxidative stress and neurotoxicity. Further, we also discuss the signatures of oxidatively modified brain proteins, identified using redox proteomics approaches, during the progression of AD. CRITICAL ISSUES The exact relationships of the specifically oxidatively modified proteins in AD pathogenesis require additional investigation. FUTURE DIRECTIONS Further studies are needed to address whether the therapies directed toward brain oxidative stress and oxidatively modified key brain proteins might help delay or prevent the progression of AD.
Collapse
Affiliation(s)
- D Allan Butterfield
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506-0055, USA.
| | | | | |
Collapse
|
37
|
Affiliation(s)
- Osvaldo Lanzalunga
- a Dipartimento di Chimica, Università di Roma “La Sapienza” and Istituto CNR di Metodologie Chimiche (IMC-CNR) , Sezione Meccanismi di Reazione , P.le Aldo Moro 5, Rome , Italy
| |
Collapse
|
38
|
Xipsiti C, Nicolaides AV. A computational study on the possible role of oxygen in the oxidation of methionine and dimethylsulfide initiated by OH radicals. COMPUT THEOR CHEM 2013. [DOI: 10.1016/j.comptc.2012.12.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
39
|
Fiori J, Naldi M, Bartolini M, Andrisano V. Disclosure of a fundamental clue for the elucidation of the myricetin mechanism of action as amyloid aggregation inhibitor by mass spectrometry. Electrophoresis 2012; 33:3380-6. [PMID: 22961751 DOI: 10.1002/elps.201200186] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Revised: 05/18/2012] [Accepted: 06/06/2012] [Indexed: 12/27/2022]
Abstract
Progression of Alzheimer's disease involves aggregation of amyloid-beta (Aβ) peptides, a complex process that involves the formation of several soluble intermediates and ends up with the deposition of ordered fibrillar architectures. The determination of the Aβ42 self-assembly species targeted by an inhibitor is a key step in the identification of new inhibitors endowed with a suitable profile. In this context, the subtle characterization of myricetin (Myr) mode of action at a molecular level was performed by using different MS techniques, which allowed the monitoring of the modifications induced by Myr on the first occurring Aβ42 self-assembly species. Results showed a persistent level of monomer and decreased formation of ordered Aβ42 aggregates in the presence of Myr, further, nano-LC-nano-ESI-QTOF, MALDI-TOF, and ESI-IT highlighted the formation of a new oxidized Aβ42 species, which is less prone to aggregation, in the presence of Myr. Coupling tryptic digestion and nano-LC-nano-ESI-QTOF also allowed the identification of Met(35) as the specific site of oxidation along the Aβ aminoacid chain. Therefore, the detailed investigation by different MS techniques allowed better understanding of the molecular modification at the basis of the antiaggregating properties of Myr and highlighted its oxidizing action on the residue Met(35) in Aβ monomers.
Collapse
Affiliation(s)
- Jessica Fiori
- Department of Pharmaceutical Sciences, University of Bologna, Bologna, Italy
| | | | | | | |
Collapse
|
40
|
Effects of triptolide on the synaptophysin expression of hippocampal neurons in the AD cellular model. Int Immunopharmacol 2012; 13:175-80. [DOI: 10.1016/j.intimp.2012.03.021] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2012] [Revised: 03/10/2012] [Accepted: 03/21/2012] [Indexed: 11/22/2022]
|
41
|
Moore BD, Chakrabarty P, Levites Y, Kukar TL, Baine AM, Moroni T, Ladd TB, Das P, Dickson DW, Golde TE. Overlapping profiles of Aβ peptides in the Alzheimer's disease and pathological aging brains. ALZHEIMERS RESEARCH & THERAPY 2012; 4:18. [PMID: 22621179 PMCID: PMC3506932 DOI: 10.1186/alzrt121] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Revised: 04/17/2012] [Accepted: 05/23/2012] [Indexed: 12/11/2022]
Abstract
INTRODUCTION A hallmark of Alzheimer's disease (AD) is the presence of senile plaques composed of aggregated amyloid β (Aβ) peptides. Pathological aging (PA) is a postmortem classification that has been used to describe brains with plaque pathology similar in extent to AD, minimal cortical tau pathology, and no accompanying history of cognitive decline in the brain donor prior to death. PA may represent either a prodromal phase of AD, a benign form of Aβ accumulation, or inherent individual resistance to the toxic effects of Aβ accumulation. To attempt to distinguish between these possibilities we have systematically characterized Aβ peptides in a postmortem series of PA, AD and non-demented control (NDC) brains. METHODS Aβ was sequentially extracted with tris buffered saline (TBS), radioimmunoprecipitation buffer (RIPA), 2% sodium dodecyl sulfate (SDS) and 70% formic acid (FA) from the pre-frontal cortex of 16 AD, eight PA, and six NDC patients. These extracts were analyzed by 1) a panel of Aβ sandwich ELISAs, 2) immunoprecipitation followed by mass spectrometry (IP/MS) and 3) western blotting. These studies enabled us to asses Aβ levels and solubility, peptide profiles and oligomeric assemblies. RESULTS In almost all extracts (TBS, RIPA, 2% SDS and 70% FA) the average levels of Aβ1-40, Aβ1-42, Aβ total, and Aβx-42 were greatest in AD. On average, levels were slightly lower in PA, and there was extensive overlap between Aβ levels in individual PA and AD cases. The profiles of Aβ peptides detected using IP/MS techniques also showed extensive similarity between the PA and AD brain extracts. In select AD brain extracts, we detected more amino-terminally truncated Aβ peptides compared to PA patients, but these peptides represented a minor portion of the Aβ observed. No consistent differences in the Aβ assemblies were observed by western blotting in the PA and AD groups. CONCLUSIONS We found extensive overlap with only subtle quantitative differences between Aβ levels, peptide profiles, solubility, and SDS-stable oligomeric assemblies in the PA and AD brains. These cross-sectional data indicate that Aβ accumulation in PA and AD is remarkably similar. Such data would be consistent with PA representing a prodromal stage of AD or a resistance to the toxic effects of Aβ.
Collapse
Affiliation(s)
- Brenda D Moore
- Center for Translational Research in Neurodegenerative Disease, Department of Neuroscience, McKnight Brain Institute, College of Medicine, University of Florida, 1275 Center Drive, Gainesville, FL, 32610, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Del Giacco T, Lanzalunga O, Mazzonna M, Mencarelli P. Structural and solvent effects on the C-S bond cleavage in aryl triphenylmethyl sulfide radical cations. J Org Chem 2012; 77:1843-52. [PMID: 22242842 DOI: 10.1021/jo202418d] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Steady-state and laser flash photolysis (LFP) studies of a series of aryl triphenylmethyl sulfides [1, 3,4-(CH(3)O)(2)-C(6)H(3)SC(C(6)H(5))(3); 2, 4-CH(3)O-C(6)H(4)SC(C(6)H(5))(3); 3, 4-CH(3)-C(6)H(4)SC(C(6)H(5))(3); 4, C(6)H(5)SC(C(6)H(5))(3); and 5, 4-Br-C(6)H(4)SC(C(6)H(5))(3)] has been carried out in the presence of N-methoxyphenanthridinium hexafluorophosphate in CH(3)CN, CH(2)Cl(2), CH(2)Cl(2)/CH(3)CN, and CH(2)Cl(2)/CH(3)OH mixtures. Products deriving from the C-S bond cleavage in the radical cations 1(•+)-5(•+) have been observed in the steady-state photolysis experiments. Time-resolved LFP showed first-order decay of the radical cations accompanied by formation of the triphenylmethyl cation. A significant decrease of the C-S bond cleavage rate constants was observed by increasing the electron-donating power of the arylsulfenyl substituent, that is, by increasing the stability of the radical cations. DFT calculations showed that, in 2(•+) and 3(•+), charge and spin densities are mainly localized in the ArS group. In the TS of the C-S bond cleavage an increase of the positive charge in the trityl moiety and of the spin density on the ArS group is observed. The higher delocalization of the charge in the TS as compared to the initial state is probably at the origin of the observation that the C-S bond cleavage rates decrease by increasing the polarity of the solvent.
Collapse
Affiliation(s)
- Tiziana Del Giacco
- Dipartimento di Chimica and Centro di Eccellenza Materiali Innovativi Nanostrutturati, Università di Perugia, via Elce di Sotto 8, 06123 Perugia, Italy.
| | | | | | | |
Collapse
|
43
|
Cardinale A, Racaniello M, Saladini S, De Chiara G, Mollinari C, de Stefano MC, Pocchiari M, Garaci E, Merlo D. Sublethal doses of β-amyloid peptide abrogate DNA-dependent protein kinase activity. J Biol Chem 2011; 287:2618-31. [PMID: 22139836 DOI: 10.1074/jbc.m111.276550] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Accumulation of DNA damage and deficiency in DNA repair potentially contribute to the progressive neuronal loss in neurodegenerative disorders, including Alzheimer disease (AD). In multicellular eukaryotes, double strand breaks (DSBs), the most lethal form of DNA damage, are mainly repaired by the nonhomologous end joining pathway, which relies on DNA-PK complex activity. Both the presence of DSBs and a decreased end joining activity have been reported in AD brains, but the molecular player causing DNA repair dysfunction is still undetermined. β-Amyloid (Aβ), a potential proximate effector of neurotoxicity in AD, might exert cytotoxic effects by reactive oxygen species generation and oxidative stress induction, which may then cause DNA damage. Here, we show that in PC12 cells sublethal concentrations of aggregated Aβ(25-35) inhibit DNA-PK kinase activity, compromising DSB repair and sensitizing cells to nonlethal oxidative injury. The inhibition of DNA-PK activity is associated with down-regulation of the catalytic subunit DNA-PK (DNA-PKcs) protein levels, caused by oxidative stress and reversed by antioxidant treatment. Moreover, we show that sublethal doses of Aβ(1-42) oligomers enter the nucleus of PC12 cells, accumulate as insoluble oligomeric species, and reduce DNA-PK kinase activity, although in the absence of oxidative stress. Overall, these findings suggest that Aβ mediates inhibition of the DNA-PK-dependent nonhomologous end joining pathway contributing to the accumulation of DSBs that, if not efficiently repaired, may lead to the neuronal loss observed in AD.
Collapse
Affiliation(s)
- Alessio Cardinale
- Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Pisana, Rome 00166, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Lanzalunga O, Lapi A. Recent photo- and radiation chemical studies of sulfur radical cations. J Sulphur Chem 2011. [DOI: 10.1080/17415993.2011.619536] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- Osvaldo Lanzalunga
- a Dipartimento di Chimica and IMC-CNR, Sezione Meccanismi di Reazione , Università “La Sapienza” , P.le A. Moro, 5 I-00185 , Rome , Italy
| | - Andrea Lapi
- a Dipartimento di Chimica and IMC-CNR, Sezione Meccanismi di Reazione , Università “La Sapienza” , P.le A. Moro, 5 I-00185 , Rome , Italy
| |
Collapse
|
45
|
Alí-Torres J, Maréchal JD, Rodríguez-Santiago L, Sodupe M. Three dimensional models of Cu(2+)-Aβ(1-16) complexes from computational approaches. J Am Chem Soc 2011; 133:15008-14. [PMID: 21846101 DOI: 10.1021/ja203407v] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Elucidation of the coordination of metal ions to Aβ is essential to understand their role in its aggregation and to rationally design new chelators with potential therapeutic applications in Alzheimer disease. Because of that, in the last 10 years several studies have focused their attention in determining the coordination properties of Cu(2+) interacting with Aβ. However, more important than characterizing the first coordination sphere of the metal is the determination of the whole Cu(2+)-Aβ structure. In this study, we combine homology modeling (HM) techniques with quantum mechanics based approaches (QM) to determine plausible three-dimensional models for Cu(2+)-Aβ(1-16) with three histidines in their coordination sphere. We considered both ε and δ coordination of histidines 6, 13, and 14 as well as the coordination of different possible candidates containing oxygen as fourth ligand (Asp1, Glu3, Asp7, Glu11, and CO(Ala2)). Among the 32 models that enclose COO(-), the lowest energy structures correspond to [O(E3),N(δ)(H6),N(ε)(H13),N(ε)(H14)] (1), [O(E3),N(δ)(H6),N(δ)(H13),N(δ)(H14)] (2), and [O(D7),N(ε)(H6),N(δ)(H13),N(δ)(H14)] (3). The most stable model containing CO(Ala2) as fourth ligand in the Cu(2+) coordination sphere is [O(c)(A2),N(ε)(H6),N(δ)(H13),N(ε)(H14)] (4). An estimation of the relative stability between Glu3 (1) and CO(Ala2) (4) coordinated complexes seems to indicate that the preference for the latter coordination may be due to solvent effects. The present results also show the relationship between the peptidic and metallic moieties in defining the overall geometry of the complex and illustrate that the final stability of the complexes results from a balance between the metal coordination site and amyloid folding upon complexation.
Collapse
Affiliation(s)
- Jorge Alí-Torres
- Departament de Química, Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | | | | |
Collapse
|
46
|
Alzheimer's disease and related neurodegenerative disorders: implication and counteracting of melatonin. J Appl Biomed 2011. [DOI: 10.2478/v10136-011-0003-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
|
47
|
Axelsen PH, Komatsu H, Murray IVJ. Oxidative stress and cell membranes in the pathogenesis of Alzheimer's disease. Physiology (Bethesda) 2011; 26:54-69. [PMID: 21357903 DOI: 10.1152/physiol.00024.2010] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Amyloid β proteins and oxidative stress are believed to have central roles in the development of Alzheimer's disease. Lipid membranes are among the most vulnerable cellular components to oxidative stress, and membranes in susceptible regions of the brain are compositionally distinct from those in other tissues. This review considers the evidence that membranes are either a source of neurotoxic lipid oxidation products or the target of pathogenic processes involving amyloid β proteins that cause permeability changes or ion channel formation. Progress toward a comprehensive theory of Alzheimer's disease pathogenesis is discussed in which lipid membranes assume both roles and promote the conversion of monomeric amyloid β proteins into fibrils, the pathognomonic histopathological lesion of the disease.
Collapse
Affiliation(s)
- Paul H Axelsen
- Department of Pharmacology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | | | | |
Collapse
|
48
|
Cuevas E, Lantz SM, Tobón-Velasco JC, Newport GD, Wu Q, Virmani A, Ali SF, Santamaría A. On the in vivo early toxic properties of A-beta 25-35 peptide in the rat hippocampus: involvement of the Receptor-for-Advanced Glycation-End-Products and changes in gene expression. Neurotoxicol Teratol 2011; 33:288-96. [PMID: 21216281 DOI: 10.1016/j.ntt.2010.12.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2010] [Revised: 12/23/2010] [Accepted: 12/23/2010] [Indexed: 11/19/2022]
Abstract
Amyloid-beta peptide (Aβ) deposition is assumed to play a pathogenic role in the brain of Alzheimer's disease patients. To date, the precise mechanisms underlying Aβ toxicity are not fully understood. A recent hypothesis suggesting that the Receptor-for-Advanced-Glycation-End-Products (RAGE)-a trans-membrane protein signaling for oxidative stress-is involved in Aβ toxicity is gaining attention. Early Aβ toxicity could indeed help to explain the deleterious events further produced by this molecule in the brain. In this work, we evaluated the pattern of early expression of RAGE in the toxic model induced by Aß₂₅₋₃₅ in rat CA1 region. Intrahippocampal injections of Aβ₂₅₋₃₅ in rats increased the RAGE expression at 24 h post-injection; this event was accompanied by increased components of RAGE downstream signaling in hippocampal cells, such as enhanced expression of the pro-apoptotic factor NF-κB, increased nitric oxide production, LDH leakage, mitochondrial dysfunction, increased TNF-α expression, antioxidant genes down-regulation, and augmented neurodegeneration. Our findings support an active role of RAGE during the early stages of Aβ₂₅₋₃₅ toxicity in the hippocampus.
Collapse
Affiliation(s)
- Elvis Cuevas
- Neurochemistry Laboratory, Division of Neurotoxicology, National Center for Toxicological Research, Jefferson, AR 72079, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Rational development of a strategy for modifying the aggregatibility of proteins. Proc Natl Acad Sci U S A 2011; 108:4297-302. [PMID: 21368182 DOI: 10.1073/pnas.1100195108] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The conversion of peptide and proteins from their soluble state into well-organized aggregates, together with the accompanied oxidation of methionine residue, presents a significant challenge to human health, to the manufacture of protein therapeutics, and to the synthesis of proteins and glycoproteins. Despite their fundamental importance, little is known about the molecular basis of these two side reactions and their control. Here, using chemical peptide synthesis, we further confirmed the importance of the balance between hydrophobic interactions and electrostatic repulsive forces in inducing and inhibiting aggregation and methionine oxidation. Most importantly, through extending the established principle, we are able to effectively stabilize the problematic peptide fragment through the attachment of cleavable arginine tags. Future applications of our approach are expected to facilitate the synthesis and study of difficult peptides, proteins, and glycoproteins and will provide more opportunities for the optimization of protein biopharmaceuticals and for the development of cell-permeable biomolecules.
Collapse
|
50
|
Effect of chloramine-T on long-term potentiation at synapses between perforant path and dentate gyrus in hippocampus of rats in vivo. Neurotoxicology 2011; 32:199-205. [PMID: 21241739 DOI: 10.1016/j.neuro.2011.01.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2010] [Revised: 12/02/2010] [Accepted: 01/10/2011] [Indexed: 01/14/2023]
Abstract
Reactive oxygen species (ROS), including superoxide, are generally considered as neurotoxic molecules whose effects can be alleviated by antioxidant enzymes. However, ROS also are known to be necessary components of the signal transduction cascades underlying normal synaptic plasticity. The oxidant chloramine-T (Ch-T), a specific oxidant to sulphur-containing residues, can oxidize methionine (Met) residues in proteins to alter protein function. To investigate the effect of Ch-T on the induction of hippocampal long-term potentiation (LTP) in dentate gyrus (DG), in vivo electrophysiological recording was employed. It was found that intracerebroventricular (ICV) injection of 0.1 μM Ch-T in 5 μL enhanced hippocampal LTP of rats slightly, whereas, 20 mM Ch-T in 5 μL greatly attenuated LTP. These effects can be reversed by pretreatment with 0.1 mM dithiothretol (DTT), a special thiol reductant. In addition, 0.1 μM Ch-T elevated LTP-induced increase in phosphorylation of Ca²+/calmodulin (CaM)-dependent protein kinase (CaMKII) and neurogranin (Ng), whereas 2 μM and 20 mM Ch-T reduced LTP-induced increase in phosphorylation status of the two key proteins, especially for 20 mM Ch-T. Pretreatment with DTT significantly prevented these effects. Taken together, these findings demonstrated that Ch-T has concentration-dependent effects on the induction of hippocampal LTP in vivo. In brief, low concentration of Ch-T facilitated hippocampal LTP by enhancing LTP-induced increase in p-CaMKII and p-Ng compared to controls, whereas high concentration of Ch-T obviously attenuated LTP accompanied by a decrease in the phosphorylated proteins, and both of these effects can be prevented by DTT. These results indicate that Ch-T modulates hippocampal LTP through regulating phosphorylation status of CaMKII and Ng.
Collapse
|