1
|
Mohd Murshid N, Mohd Sahardi NFN, Makpol S. Advancing Alzheimer's Disease Modelling by Developing a Refined Biomimetic Brain Microenvironment for Facilitating High-Throughput Screening of Pharmacological Treatment Strategies. Int J Mol Sci 2024; 26:241. [PMID: 39796097 PMCID: PMC11719782 DOI: 10.3390/ijms26010241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/19/2024] [Accepted: 11/21/2024] [Indexed: 01/13/2025] Open
Abstract
Alzheimer's disease (AD) poses a significant worldwide health challenge, requiring novel approaches for improved models and treatment development. This comprehensive review emphasises the systematic development and improvement of a biomimetic brain environment to address the shortcomings of existing AD models and enhance the efficiency of screening potential drug treatments. We identify drawbacks in traditional models and emphasise the necessity for more physiologically accurate systems through an in-depth analysis of current literature. This review aims to study the development of an advanced AD model that accurately replicates key AD pathophysiological aspects using cutting-edge biomaterials and microenvironment design. Incorporating biomolecular elements like Tau proteins and beta-amyloid (Aβ) plaques improve the accuracy of illustrating disease mechanisms. The expected results involve creating a solid foundation for high-throughput screening with enhanced scalability, translational significance, and the possibility of speeding up drug discovery. Thus, this review fills the gaps in AD modelling and shows potential for creating precise and efficient drug treatments for AD.
Collapse
Affiliation(s)
- Nuraqila Mohd Murshid
- Department of Biochemistry, Faculty of Medicine, Level 17 Preclinical Building, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia;
| | - Nur Fatin Nabilah Mohd Sahardi
- Secretariat of Research and Innovation, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia;
| | - Suzana Makpol
- Department of Biochemistry, Faculty of Medicine, Level 17 Preclinical Building, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia;
| |
Collapse
|
2
|
Luo Y, Yang H, Yan X, Wu Y, Wei G, Wu X, Tian X, Xiong Y, Wu G, Wen H. Transcranial Direct Current Stimulation Alleviates Neurovascular Unit Dysfunction in Mice With Preclinical Alzheimer’s Disease. Front Aging Neurosci 2022; 14:857415. [PMID: 35493946 PMCID: PMC9047023 DOI: 10.3389/fnagi.2022.857415] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 03/24/2022] [Indexed: 12/26/2022] Open
Abstract
Neurons, glial cells and blood vessels are collectively referred to as the neurovascular unit (NVU). In the Alzheimer’s disease (AD) brain, the main components of the NVU undergo pathological changes. Transcranial direct current stimulation (tDCS) can protect neurons, induce changes in glial cells, regulate cerebral blood flow, and exert long-term neuroprotection. However, the mechanism by which tDCS improves NVU function is unclear. In this study, we explored the effect of tDCS on the NVU in mice with preclinical AD and the related mechanisms. 10 sessions of tDCS were given to six-month-old male APP/PS1 mice in the preclinical stage. The model group, sham stimulation group, and control group were made up of APP/PS1 mice and C57 mice of the same age. All mice were histologically evaluated two months after receiving tDCS. Protein content was measured using Western blotting and an enzyme-linked immunosorbent assay (ELISA). The link between glial cells and blood vessels was studied using immunofluorescence staining and lectin staining. The results showed that tDCS affected the metabolism of Aβ; the levels of Aβ, amyloid precursor protein (APP) and BACE1 were significantly reduced, and the levels of ADAM10 were significantly increased in the frontal cortex and hippocampus in the stimulation group. In the stimulation group, tDCS reduced the protein levels of Iba1 and GFAP and increased the protein levels of NeuN, LRP1 and PDGRFβ. This suggests that tDCS can improve NVU function in APP/PS1 mice in the preclinical stage. Increased blood vessel density and blood vessel length, decreased IgG extravasation, and increased the protein levels of occludin and coverage of astrocyte foot processes with blood vessels suggested that tDCS had a protective effect on the blood-brain barrier. Furthermore, the increased numbers of Vimentin, S100 expression and blood vessels (lectin-positive) around Aβ indicated that the effect of tDCS was mediated by astrocytes and blood vessels. There was no significant difference in these parameters between the model group and the sham stimulation group. In conclusion, our results show that tDCS can improve NVU function in APP/PS1 mice in the preclinical stage, providing further support for the use of tDCS as a treatment for AD.
Collapse
Affiliation(s)
- Yinpei Luo
- Chongqing Key Laboratory of Neurobiology, Department of Neurobiology, Army Medical University, Chongqing, China
- Laboratory of Neural Regulation and Rehabilitation Technology, Chongqing Medical Electronics Engineering Technology Research Center, College of Bioengineering, Chongqing University, Chongqing, China
| | - Hong Yang
- Laboratory of Neural Regulation and Rehabilitation Technology, Chongqing Medical Electronics Engineering Technology Research Center, College of Bioengineering, Chongqing University, Chongqing, China
| | - Xiaojing Yan
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing, China
| | - Yaran Wu
- Department of Biochemistry and Molecular Biology, Army Medical University, Chongqing, China
| | - Guoliang Wei
- Laboratory of Neural Regulation and Rehabilitation Technology, Chongqing Medical Electronics Engineering Technology Research Center, College of Bioengineering, Chongqing University, Chongqing, China
| | - Xiaoying Wu
- Laboratory of Neural Regulation and Rehabilitation Technology, Chongqing Medical Electronics Engineering Technology Research Center, College of Bioengineering, Chongqing University, Chongqing, China
| | - Xuelong Tian
- Laboratory of Neural Regulation and Rehabilitation Technology, Chongqing Medical Electronics Engineering Technology Research Center, College of Bioengineering, Chongqing University, Chongqing, China
| | - Ying Xiong
- Chongqing Key Laboratory of Neurobiology, Department of Neurobiology, Army Medical University, Chongqing, China
| | - Guangyan Wu
- Experimental Center of Basic Medicine, Army Medical University, Chongqing, China
- Guangyan Wu,
| | - Huizhong Wen
- Chongqing Key Laboratory of Neurobiology, Department of Neurobiology, Army Medical University, Chongqing, China
- *Correspondence: Huizhong Wen,
| |
Collapse
|
3
|
Mamsa SSA, Meloni BP. Arginine and Arginine-Rich Peptides as Modulators of Protein Aggregation and Cytotoxicity Associated With Alzheimer's Disease. Front Mol Neurosci 2021; 14:759729. [PMID: 34776866 PMCID: PMC8581540 DOI: 10.3389/fnmol.2021.759729] [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: 08/17/2021] [Accepted: 09/29/2021] [Indexed: 01/10/2023] Open
Abstract
A substantial body of evidence indicates cationic, arginine-rich peptides (CARPs) are effective therapeutic compounds for a range of neurodegenerative pathologies, with beneficial effects including the reduction of excitotoxic cell death and mitochondrial dysfunction. CARPs, therefore, represent an emergent class of promising neurotherapeutics with multimodal mechanisms of action. Arginine itself is a known chaotrope, able to prevent misfolding and aggregation of proteins. The putative role of proteopathies in chronic neurodegenerative diseases such as Alzheimer's disease (AD) warrants investigation into whether CARPs could also prevent the aggregation and cytotoxicity of amyloidogenic proteins, particularly amyloid-beta and tau. While monomeric arginine is well-established as an inhibitor of protein aggregation in solution, no studies have comprehensively discussed the anti-aggregatory properties of arginine and CARPs on proteins associated with neurodegenerative disease. Here, we review the structural, physicochemical, and self-associative properties of arginine and the guanidinium moiety, to explore the mechanisms underlying the modulation of protein aggregation by monomeric and multimeric arginine molecules. Arginine-rich peptide-based inhibitors of amyloid-beta and tau aggregation are discussed, as well as further modulatory roles which could reduce proteopathic cytotoxicity, in the context of therapeutic development for AD.
Collapse
Affiliation(s)
- Somayra S A Mamsa
- School of Molecular Sciences, Faculty of Science, The University of Western Australia, Perth, WA, Australia.,Perron Institute for Neurological and Translational Science, QEII Medical Centre, Perth, WA, Australia
| | - Bruno P Meloni
- Perron Institute for Neurological and Translational Science, QEII Medical Centre, Perth, WA, Australia.,Centre for Neuromuscular and Neurological Disorders, The University of Western Australia, Crawley, WA, Australia.,Department of Neurology, Sir Charles Gairdner Hospital, QEII Medical Centre, Perth, WA, Australia
| |
Collapse
|
4
|
Li L, Lv Z, Man Z, Xu Z, Wei Y, Geng H, Fu H. Polarity-active NIR probes with strong two-photon absorption and ultrahigh binding affinity of insulin amyloid fibrils. Chem Sci 2021; 12:3308-3313. [PMID: 34164100 PMCID: PMC8179388 DOI: 10.1039/d0sc03907a] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 01/01/2021] [Indexed: 12/30/2022] Open
Abstract
Amyloid fibrils are associated with many neurodegenerative diseases. In situ and in vivo visualization of amyloid fibrils is important for medical diagnostics and requires fluorescent probes with both excitation and emission wavelengths in the far-red and NIR region, and simultaneously with high binding-affinity to amyloid fibrils and the ability to cross the blood-brain barrier, which, however, remain a challenge. Here, we rationally design and synthesize an excellent polarity-sensitive two-photon excited NIR fluorophore (TZPI) based on a donor (D)-acceptor (A)-ion compound. The electron-rich carbazole group and the ionic pyridinium bromide group, linked by an electron-poor π-conjugated benzothiadiazole group, ensure strong near infrared (NIR) emission. Furthermore, the lipophilic carbazole together with the benzothiadiazole group facilitates docking of the probe in the hydrophobic domains of amyloid aggregates with the dissociation constant K d = 20 nM and 13.5-fold higher binding affinity to insulin fibrils than the commercial probe ThT. On association with the amyloid fibrils, the tiny decrease in polarity leads to a large increase in its NIR emission intensity with an on-off ratio > 10; meanwhile, the TZPI probe exhibits a quantum yield of up to 30% and two-photon absorption cross-section values of up to 467.6 GM at 890 nm. Moreover, the application of TZPI in two-photon imaging is investigated. The ultrahigh binding affinity, the strong NIR emission, the good two-photon absorption properties, the high photo-stability, the appropriate molecular mass of 569 Da and the lipophilicity with log P = 1.66 ± 0.1 to cross the BBB make TZPI promising as an ideal candidate for detecting amyloid plaques in vivo.
Collapse
Affiliation(s)
- Li Li
- Beijing Key Laboratory for Optical Materials and Photonic Devices, Capital Normal University Beijing 100048 China
| | - Zheng Lv
- Beijing Key Laboratory for Optical Materials and Photonic Devices, Capital Normal University Beijing 100048 China
- Key Laboratory of Molecular Optoelectronic Sciences, Institute of Molecular Plus, Tianjin Collaborative Innovation Center of Chemical Science and Engineering, Tianjin University Tianjin 300072 China
| | - Zhongwei Man
- Beijing Key Laboratory for Optical Materials and Photonic Devices, Capital Normal University Beijing 100048 China
| | - Zhenzhen Xu
- Beijing Key Laboratory for Optical Materials and Photonic Devices, Capital Normal University Beijing 100048 China
| | - YuLing Wei
- Beijing Key Laboratory for Optical Materials and Photonic Devices, Capital Normal University Beijing 100048 China
| | - Hua Geng
- Beijing Key Laboratory for Optical Materials and Photonic Devices, Capital Normal University Beijing 100048 China
| | - Hongbing Fu
- Beijing Key Laboratory for Optical Materials and Photonic Devices, Capital Normal University Beijing 100048 China
- Key Laboratory of Molecular Optoelectronic Sciences, Institute of Molecular Plus, Tianjin Collaborative Innovation Center of Chemical Science and Engineering, Tianjin University Tianjin 300072 China
| |
Collapse
|
5
|
Vasefi M, Ghaboolian-Zare E, Abedelwahab H, Osu A. Environmental toxins and Alzheimer's disease progression. Neurochem Int 2020; 141:104852. [PMID: 33010393 DOI: 10.1016/j.neuint.2020.104852] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/05/2020] [Accepted: 09/18/2020] [Indexed: 12/29/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia, which causes progressive memory loss and cognitive decline. Effective strategies to treat or prevent remains one of the most challenging undertakings in the medical field. AD is a complex and multifactorial disease that involves several risk factors. Aging and genetic factors both play important roles in the onset of the AD, however; certain environmental factors have been reported to increase the risk of AD. Chronic exposure to toxins has been seen as an environmental factor that may increase the risk of developing a neurodegenerative disease such as AD. Exposure to metals and biotoxins produced by bacteria, molds, and viruses may contribute to the cognitive decline and pathophysiology associated with AD. Toxins may contribute to the pathology of the disease through various mechanisms such as deposition of amyloid-beta (Aβ) plaques and tangles in the brain, induction of apoptosis, inflammation, or oxidative damage. Here, we will review how toxins affect brain physiology with a focus on mechanisms by which toxins may contribute to the development and progression of AD. A better understanding of these mechanisms may help contribute towards the development of an effective strategy to slow the progression of AD.
Collapse
Affiliation(s)
- Maryam Vasefi
- Department Biology, Lamar University, Beaumont, TX, United States.
| | | | | | - Anthony Osu
- Department Biology, Lamar University, Beaumont, TX, United States
| |
Collapse
|
6
|
Ayata P, Schaefer A. Innate sensing of mechanical properties of brain tissue by microglia. Curr Opin Immunol 2020; 62:123-130. [PMID: 32058296 PMCID: PMC7067639 DOI: 10.1016/j.coi.2020.01.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 12/20/2019] [Accepted: 01/07/2020] [Indexed: 01/09/2023]
Abstract
External organic or inorganic objects (foreign bodies) that are inadvertently or purposefully placed in the human or animal tissues can trigger local tissue responses that aim at the elimination and/or segregation of foreign bodies from the tissue. The foreign body response (FBR) may have major implications for neurodegeneration associated with the formation of aberrant protein-based aggregates or plaques. The distinct physical features of the plaques, including high rigidity and varying surface properties, may trigger microglial mechanosensing of the plaque as a foreign body. The microglial FBR may have a dual function by promoting and/or suppressing the plaque driven neurodegeneration. Microglial contact with the plaque may trigger inflammatory activation of microglia and support microglia-driven neuronal damage. Conversely, persistent microglial activation may trigger the formation of a microglia-supported cell barrier that segregates and compacts the plaques thus preventing further plaque-induced damage to healthy neurons.
Collapse
Affiliation(s)
- Pinar Ayata
- Nash Family Department of Neuroscience, Center for Glial Biology, Ronald M. Loeb Center for Alzheimer’s Disease, Precision Immunology Institute, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Anne Schaefer
- Nash Family Department of Neuroscience, Center for Glial Biology, Ronald M. Loeb Center for Alzheimer’s Disease, Precision Immunology Institute, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY,Corresponding author
| |
Collapse
|
7
|
Al Kahtani MA. Effect of both selenium and biosynthesized nanoselenium particles on cadmium-induced neurotoxicity in albino rats. Hum Exp Toxicol 2019; 39:159-172. [DOI: 10.1177/0960327119880589] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Because cadmium (Cd) is not naturally degradable by ecosystems, it interferes with many types of food chains. Cd accumulates in the kidney, liver and in the nervous tissues, especially the brain. The neurotoxicity of Cd is very high, as it alters the integrity, and increases the permeability, of the blood–brain barrier. Cd penetrates and accumulates in neurons in the brains of rats. This study reveals that Cd decreases antioxidant enzymes and increases oxidative stress in the brain. In addition, Cd increases lipid peroxidation of brain tissues. Cd increases the expression of the Cu/Zn superoxide dismutase gene. It also affects cholinergic, glutamatergic, gamma-Aminobutyric acid (GABAergic), dopamine, serotonin and acetylcholine neurotransmitters in brain tissue. Consequently, Cd increases the formation of amyloid β, a neurotoxic index, and induces apoptosis by changing the quality and the quantity of Bcl-2, Bax and p53 proteins. In conclusion, both selenium and nanoselenium show potential antioxidant activity and promote recovery from the neurotoxic action of Cd.
Collapse
Affiliation(s)
- MA Al Kahtani
- Department of Biology, Science College, King Khalid University, Abha, Saudi Arabia
- Research Centre for Advanced Materials Science (RCAMS), King Khalid University, Abha, Saudi Arabia
| |
Collapse
|
8
|
Concentration-dependent effects of mercury and lead on Aβ42: possible implications for Alzheimer's disease. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2019; 48:173-187. [PMID: 30603762 DOI: 10.1007/s00249-018-1344-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 09/24/2018] [Accepted: 12/10/2018] [Indexed: 02/06/2023]
Abstract
Mercury (Hg) and lead (Pb) are known to be toxic non-radioactive elements, with well-described neurotoxicology. Much evidence supports the implication of metals as potential risk cofactors in Alzheimer's disease (AD). Although the action mechanism of the two metals remains unclear, Hg and Pb toxicity in AD could depend on their ability to favour misfolding and aggregation of amyloid beta proteins (Aβs) that seem to have toxic properties, particularly in their aggregated state. In our study, we evaluated the effect of Hg and Pb both on the Aβ42 ion channel incorporated in a planar lipid membrane made up of phosphatidylcholine containing 30% cholesterol and on the secondary structure of Aβ42 in an aqueous environment. The effects of Hg and Pb on the Aβ42 peptide were observed for its channel incorporated into a membrane as well as for the peptide in solution. A decreasing Aβ42 channel frequency and the formation of large and amorphous aggregates in solution that are prone to precipitate were both dependent on metal concentration. These experimental data suggest that Hg and Pb interact directly with Aβs, strengthening the hypothesis that the two metals may be a risk factor in AD.
Collapse
|
9
|
Giacomazza D, D’Andrea D, Provenzano A, Picone P, Provenzano F, Guarrasi V, Raimondo M, San Biagio PL, Passantino R, Mangione MR, Di Carlo M, Costa MA. The precious content of the olive mill wastewater: the protective effect of the antioxidant fraction in cell cultures. CYTA - JOURNAL OF FOOD 2018. [DOI: 10.1080/19476337.2018.1458752] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Affiliation(s)
- Daniela Giacomazza
- Istituto di BioFisica (IBF – Palermo Unit), Consiglio Nazionale delle Ricerche, Palermo, Italy
| | - Daniela D’Andrea
- Istituto di BioFisica (IBF – Palermo Unit), Consiglio Nazionale delle Ricerche, Palermo, Italy
| | - Alessia Provenzano
- Istituto di BioFisica (IBF – Palermo Unit), Consiglio Nazionale delle Ricerche, Palermo, Italy
| | - Pasquale Picone
- Istituto di Biomedicina e Immunologia Molecolare “A. Monroy” (IBIM), Consiglio Nazionale delle Ricerche, Palermo, Italy
| | - Fiorenza Provenzano
- Istituto di BioFisica (IBF – Palermo Unit), Consiglio Nazionale delle Ricerche, Palermo, Italy
| | - Valeria Guarrasi
- Istituto di BioFisica (IBF – Palermo Unit), Consiglio Nazionale delle Ricerche, Palermo, Italy
| | - Marco Raimondo
- Istituto di BioFisica (IBF – Palermo Unit), Consiglio Nazionale delle Ricerche, Palermo, Italy
| | - Pier Luigi San Biagio
- Istituto di BioFisica (IBF – Palermo Unit), Consiglio Nazionale delle Ricerche, Palermo, Italy
| | - Rosa Passantino
- Istituto di BioFisica (IBF – Palermo Unit), Consiglio Nazionale delle Ricerche, Palermo, Italy
| | - Maria Rosalia Mangione
- Istituto di BioFisica (IBF – Palermo Unit), Consiglio Nazionale delle Ricerche, Palermo, Italy
| | - Marta Di Carlo
- Istituto di Biomedicina e Immunologia Molecolare “A. Monroy” (IBIM), Consiglio Nazionale delle Ricerche, Palermo, Italy
| | - Maria Assunta Costa
- Istituto di BioFisica (IBF – Palermo Unit), Consiglio Nazionale delle Ricerche, Palermo, Italy
| |
Collapse
|
10
|
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
|
11
|
Fricker M, Tolkovsky AM, Borutaite V, Coleman M, Brown GC. Neuronal Cell Death. Physiol Rev 2018; 98:813-880. [PMID: 29488822 PMCID: PMC5966715 DOI: 10.1152/physrev.00011.2017] [Citation(s) in RCA: 770] [Impact Index Per Article: 110.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 05/23/2017] [Accepted: 07/10/2017] [Indexed: 02/07/2023] Open
Abstract
Neuronal cell death occurs extensively during development and pathology, where it is especially important because of the limited capacity of adult neurons to proliferate or be replaced. The concept of cell death used to be simple as there were just two or three types, so we just had to work out which type was involved in our particular pathology and then block it. However, we now know that there are at least a dozen ways for neurons to die, that blocking a particular mechanism of cell death may not prevent the cell from dying, and that non-neuronal cells also contribute to neuronal death. We review here the mechanisms of neuronal death by intrinsic and extrinsic apoptosis, oncosis, necroptosis, parthanatos, ferroptosis, sarmoptosis, autophagic cell death, autosis, autolysis, paraptosis, pyroptosis, phagoptosis, and mitochondrial permeability transition. We next explore the mechanisms of neuronal death during development, and those induced by axotomy, aberrant cell-cycle reentry, glutamate (excitoxicity and oxytosis), loss of connected neurons, aggregated proteins and the unfolded protein response, oxidants, inflammation, and microglia. We then reassess which forms of cell death occur in stroke and Alzheimer's disease, two of the most important pathologies involving neuronal cell death. We also discuss why it has been so difficult to pinpoint the type of neuronal death involved, if and why the mechanism of neuronal death matters, the molecular overlap and interplay between death subroutines, and the therapeutic implications of these multiple overlapping forms of neuronal death.
Collapse
Affiliation(s)
- Michael Fricker
- Hunter Medical Research Institute, University of Newcastle, Callaghan, New South Wales , Australia ; Department of Clinical Neurosciences, University of Cambridge , Cambridge , United Kingdom ; Neuroscience Institute, Lithuanian University of Health Sciences , Kaunas , Lithuania ; and Department of Biochemistry, University of Cambridge , Cambridge , United Kingdom
| | - Aviva M Tolkovsky
- Hunter Medical Research Institute, University of Newcastle, Callaghan, New South Wales , Australia ; Department of Clinical Neurosciences, University of Cambridge , Cambridge , United Kingdom ; Neuroscience Institute, Lithuanian University of Health Sciences , Kaunas , Lithuania ; and Department of Biochemistry, University of Cambridge , Cambridge , United Kingdom
| | - Vilmante Borutaite
- Hunter Medical Research Institute, University of Newcastle, Callaghan, New South Wales , Australia ; Department of Clinical Neurosciences, University of Cambridge , Cambridge , United Kingdom ; Neuroscience Institute, Lithuanian University of Health Sciences , Kaunas , Lithuania ; and Department of Biochemistry, University of Cambridge , Cambridge , United Kingdom
| | - Michael Coleman
- Hunter Medical Research Institute, University of Newcastle, Callaghan, New South Wales , Australia ; Department of Clinical Neurosciences, University of Cambridge , Cambridge , United Kingdom ; Neuroscience Institute, Lithuanian University of Health Sciences , Kaunas , Lithuania ; and Department of Biochemistry, University of Cambridge , Cambridge , United Kingdom
| | - Guy C Brown
- Hunter Medical Research Institute, University of Newcastle, Callaghan, New South Wales , Australia ; Department of Clinical Neurosciences, University of Cambridge , Cambridge , United Kingdom ; Neuroscience Institute, Lithuanian University of Health Sciences , Kaunas , Lithuania ; and Department of Biochemistry, University of Cambridge , Cambridge , United Kingdom
| |
Collapse
|
12
|
Karaballi RA, Merchant S, Power SR, Brosseau CL. Electrochemical surface-enhanced Raman spectroscopy (EC-SERS) study of the interaction between protein aggregates and biomimetic membranes. Phys Chem Chem Phys 2018; 20:4513-4526. [DOI: 10.1039/c7cp06838g] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
EC-SERS is used for the first time to characterize protein aggregate–biomembrane interactions.
Collapse
Affiliation(s)
| | | | - Sasha R. Power
- Department of Chemistry
- Saint Mary's University
- Halifax
- Canada
| | | |
Collapse
|
13
|
Picone P, Sabatino MA, Ditta LA, Amato A, San Biagio PL, Mulè F, Giacomazza D, Dispenza C, Di Carlo M. Nose-to-brain delivery of insulin enhanced by a nanogel carrier. J Control Release 2017; 270:23-36. [PMID: 29196041 DOI: 10.1016/j.jconrel.2017.11.040] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 11/21/2017] [Accepted: 11/25/2017] [Indexed: 02/07/2023]
Abstract
Recent evidences suggest that insulin delivery to the brain can be an important pharmacological therapy for some neurodegenerative pathologies, including Alzheimer disease (AD). Due to the presence of the Blood Brain Barrier, a suitable carrier and an appropriate route of administration are required to increase the efficacy and safety of the treatment. Here, poly(N-vinyl pyrrolidone)-based nanogels (NG), synthetized by e-beam irradiation, alone and with covalently attached insulin (NG-In) were characterized for biocompatibility and brain delivery features in a mouse model. Preliminarily, the biodistribution of the "empty" nanocarrier after intraperitoneal (i.p.) injection was investigated by using a fluorescent-labeled NG. By fluorescence spectroscopy, SEM and dynamic light scattering analyses we established that urine clearance occurs in 24h. Histological liver and kidneys inspections indicated that no morphological alterations of tissues occurred and no immunological response was activated after NG injection. Furthermore, after administration of the insulin-conjugated nanogels (NG-In) through the intranasal route (i.n.) no alteration or immunogenic response of the nasal mucosa was observed, suggesting that the formulation is well tolerated in mouse. Moreover, an enhancement of NG-In delivery to the different brain areas and of its biological activity, measured as Akt activation levels, with reference to free insulin administration was demonstrated. Taken together, these results indicate that the synthesized NG-In enhances brain insulin delivery upon i.n. administration and strongly encourage its further evaluation as therapeutic agent against some neurodegenerative diseases.
Collapse
Affiliation(s)
- Pasquale Picone
- Istituto di Biomedicina e Immunologia Molecolare (IBIM), Consiglio Nazionale Delle Ricerche, Via U. La Malfa 153, 90146 Palermo, Italy
| | - Maria Antonietta Sabatino
- Dipartimento dell'Innovazione Industriale e Digitale (DIID), Università di Palermo, Viale delle Scienze, Edificio 6, 90128 Palermo, Italy
| | - Lorena Anna Ditta
- Dipartimento dell'Innovazione Industriale e Digitale (DIID), Università di Palermo, Viale delle Scienze, Edificio 6, 90128 Palermo, Italy
| | - Antonella Amato
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, Viale delle Scienze, Edificio 16, 90128 Palermo, Italy
| | - Pier Luigi San Biagio
- Istituto di Biofisica (IBF), Consiglio Nazionale Delle Ricerche, Via U. La Malfa 153, 90146 Palermo, Italy
| | - Flavia Mulè
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, Viale delle Scienze, Edificio 16, 90128 Palermo, Italy
| | - Daniela Giacomazza
- Istituto di Biofisica (IBF), Consiglio Nazionale Delle Ricerche, Via U. La Malfa 153, 90146 Palermo, Italy.
| | - Clelia Dispenza
- Dipartimento dell'Innovazione Industriale e Digitale (DIID), Università di Palermo, Viale delle Scienze, Edificio 6, 90128 Palermo, Italy; Istituto di Biofisica (IBF), Consiglio Nazionale Delle Ricerche, Via U. La Malfa 153, 90146 Palermo, Italy.
| | - Marta Di Carlo
- Istituto di Biomedicina e Immunologia Molecolare (IBIM), Consiglio Nazionale Delle Ricerche, Via U. La Malfa 153, 90146 Palermo, Italy.
| |
Collapse
|
14
|
Proximate Mediators of Microvascular Dysfunction at the Blood-Brain Barrier: Neuroinflammatory Pathways to Neurodegeneration. BIOMED RESEARCH INTERNATIONAL 2017; 2017:1549194. [PMID: 28890893 PMCID: PMC5584365 DOI: 10.1155/2017/1549194] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 07/09/2017] [Indexed: 12/14/2022]
Abstract
Current projections are that by 2050 the numbers of people aged 65 and older with Alzheimer's disease (AD) in the US may increase threefold while dementia is projected to double every 20 years reaching ~115 million by 2050. AD is clinically characterized by progressive dementia and neuropathologically by neuronal and synapse loss, accumulation of amyloid plaques, and neurofibrillary tangles (NFTs) in specific brain regions. The preclinical or presymptomatic stage of AD-related brain changes may begin over 20 years before symptoms occur, making development of noninvasive biomarkers essential. Distinct from neuroimaging and cerebrospinal fluid biomarkers, plasma or serum biomarkers can be analyzed to assess (i) the presence/absence of AD, (ii) the risk of developing AD, (iii) the progression of AD, or (iv) AD response to treatment. No unifying theory fully explains the neurodegenerative brain lesions but neuroinflammation (a lethal stressor for healthy neurons) is universally present. Current consensus is that the earlier the diagnosis, the better the chance to develop treatments that influence disease progression. In this article we provide a detailed review and analysis of the role of the blood-brain barrier (BBB) and damage-associated molecular patterns (DAMPs) as well as coagulation molecules in the onset and progression of these neurodegenerative disorders.
Collapse
|
15
|
Manso Y, Comes G, López-Ramos JC, Belfiore M, Molinero A, Giralt M, Carrasco J, Adlard PA, Bush AI, Delgado-García JM, Hidalgo J. Overexpression of Metallothionein-1 Modulates the Phenotype of the Tg2576 Mouse Model of Alzheimer's Disease. J Alzheimers Dis 2016; 51:81-95. [PMID: 26836194 DOI: 10.3233/jad-151025] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Alzheimer's disease (AD) is the most commonly diagnosed dementia, where signs of neuroinflammation and oxidative stress are prominent. In this study we intend to further characterize the roles of the antioxidant, anti-inflammatory, and heavy metal binding protein, metallothionein-1 (MT-1), by crossing Mt1 overexpressing mice with a well-known mouse model of AD, Tg2576 mice, which express the human amyloid-β protein precursor (hAβPP) with the Swedish K670N/M671L mutations. Mt1 overexpression increased overall perinatal survival, but did not affect significantly hAβPP-induced mortality and weight loss in adult mice. Amyloid plaque burden in ∼14-month-old mice was increased by Mt1 overexpression in the hippocampus but not the cortex. Despite full length hAβPP levels and amyloid plaques being increased by Mt1 overexpression in the hippocampus of both sexes, oligomeric and monomeric forms of Aβ, which may contribute more to toxicity, were decreased in the hippocampus of females and increased in males. Several behavioral traits such as exploration, anxiety, and learning were altered in Tg2576 mice to various degrees depending on the age and the sex. Mt1 overexpression ameliorated the effects of hAβPP on exploration in young females, and potentiated those on anxiety in old males, and seemed to improve the rate of spatial learning (Morris water maze) and the learning elicited by a classical conditioning procedure (eye-blink test). These results clearly suggest that MT-1 may be involved in AD pathogenesis.
Collapse
Affiliation(s)
- Yasmina Manso
- Animal Physiology Unit, Department of Cellular Biology, Physiology and Immunology, Faculty of Biosciences Bellaterra, Barcelona, Spain.,Institute of Neurosciences, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Gemma Comes
- Animal Physiology Unit, Department of Cellular Biology, Physiology and Immunology, Faculty of Biosciences Bellaterra, Barcelona, Spain.,Institute of Neurosciences, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | | | - Mónica Belfiore
- Animal Physiology Unit, Department of Cellular Biology, Physiology and Immunology, Faculty of Biosciences Bellaterra, Barcelona, Spain.,Institute of Neurosciences, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Amalia Molinero
- Animal Physiology Unit, Department of Cellular Biology, Physiology and Immunology, Faculty of Biosciences Bellaterra, Barcelona, Spain.,Institute of Neurosciences, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Mercedes Giralt
- Animal Physiology Unit, Department of Cellular Biology, Physiology and Immunology, Faculty of Biosciences Bellaterra, Barcelona, Spain.,Institute of Neurosciences, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Javier Carrasco
- Animal Physiology Unit, Department of Cellular Biology, Physiology and Immunology, Faculty of Biosciences Bellaterra, Barcelona, Spain.,Institute of Neurosciences, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Paul A Adlard
- The Florey Institute of Neuroscience and Mental Health and The University of Melbourne, Parkville, Victoria, Australia
| | - Ashley I Bush
- The Florey Institute of Neuroscience and Mental Health and The University of Melbourne, Parkville, Victoria, Australia
| | | | - Juan Hidalgo
- Animal Physiology Unit, Department of Cellular Biology, Physiology and Immunology, Faculty of Biosciences Bellaterra, Barcelona, Spain.,Institute of Neurosciences, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| |
Collapse
|
16
|
Cheng SB, Sharma S. Preeclampsia and health risks later in life: an immunological link. Semin Immunopathol 2016; 38:699-708. [PMID: 27339196 DOI: 10.1007/s00281-016-0579-8] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 06/16/2016] [Indexed: 12/11/2022]
Abstract
Pregnancy represents a period of physiological stress, and although this stress is experienced for a very modest portion of life, it is now recognized as a window to women's future health, often by unmasking predispositions to conditions that only become symptomatic later in life. In normal pregnancy, the mother experiences mild metabolic syndrome-like condition through week 20 of gestation. A pronounced phenotype of metabolic syndrome may program pregnancy complications such as preeclampsia. Preeclampsia is a serious complication with a myriad of manifestations for mother and offspring. This pregnancy syndrome is a polygenic disease and has been now linked to higher incidence of cardiovascular disease, diabetes, and several other disorders associated with vulnerable organs. Furthermore, the offspring born to preeclamptic mothers also exhibit an elevated risk of cardiovascular disease, stroke, and mental disorders during adulthood. This suggests that preeclampsia not only exposes the mother and the fetus to complications during pregnancy but also programs chronic diseases in later life. The etiology of preeclampsia is thought to be primarily associated with poor placentation and entails excessive maternal inflammation and endothelial dysfunction. It is well established now that the maternal immune system and the placenta are involved in a highly choreographed cross-talk that underlies adequate spiral artery remodeling required for uteroplacental perfusion and free flow of nutrients to the fetus. Since normal pregnancy is associated with a sequence of events represented by temporal events of inflammation (implantation), anti-inflammation (gestation), and inflammation (parturition), it is quite possible that unscheduled alterations in these regulatory responses may lead to pathologic consequences. Although it is not clear whether immunological alterations occur early in pregnancy, it is proposed that dysregulated systemic and placental immunity contribute to impaired angiogenesis and the onset of preeclampsia. This review will focus on important aspects of the immune system that coordinate with placental dysfunction to program preeclampsia and influence health in later life.
Collapse
Affiliation(s)
- Shi-Bin Cheng
- Department of Pediatrics, Women and Infants' Hospital of Rhode Island, Warren Alpert Medical School of Brown University, 101 Dudley Street, Providence, RI, 02905, USA
| | - Surendra Sharma
- Department of Pediatrics, Women and Infants' Hospital of Rhode Island, Warren Alpert Medical School of Brown University, 101 Dudley Street, Providence, RI, 02905, USA.
| |
Collapse
|
17
|
Labour MN, Vigier S, Lerner D, Marcilhac A, Belamie E. 3D compartmented model to study the neurite-related toxicity of Aβ aggregates included in collagen gels of adaptable porosity. Acta Biomater 2016; 37:38-49. [PMID: 27057929 DOI: 10.1016/j.actbio.2016.04.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 02/29/2016] [Accepted: 04/03/2016] [Indexed: 12/19/2022]
Abstract
UNLABELLED Insoluble deposits of β-amyloid (Aβ) are associated to neurodegenerative pathologies, in particular Alzheimer's Disease (AD). The toxicity of synthetic amyloid-like peptides has been largely demonstrated and shown to depend upon their aggregation state. However, standard 2D cell culture conditions are not well suited to study the role of the close vicinity of Aβ aggregates and growing neurites in the degenerative process. Here, we have designed a compartmented set-up where model neural cells are differentiated on the surface of Aβ-containing collagen matrices. The average pore size can be modulated, from below 0.2μm to more than 0.5μm by simple treatment with collagenase, to respectively hamper or permit neurite outgrowth towards the depth of the matrix. Dense Aβ aggregates (Congo red and ThT-positive) were obtained inside the collagen matrix with a homogeneous distribution and dimensions similar to those observed in post-mortem brain slices from Alzheimer's patients. The aggregates are not toxic to cells when the pore size is small, in spite of relatively high concentrations of 0.05-0.62mg of peptide per gram of collagen (equivalent to 11.3-113μM). In contrast, on Aβ-containing matrices with large pores, massive neural death is observed when the cells are seeded in the same conditions. It is the first time to our knowledge that Aβ aggregates with a typical morphology of dense plaques are obtained within a porous biomimetic matrix, and are shown to be toxic only when accessible to differentiating cells. STATEMENT OF SIGNIFICANCE Insoluble deposits of β-amyloid (Aβ) are associated to neurodegenerative pathologies, in particular Alzheimer's Disease (AD). In this study, we have formed Aβ aggregates directly inside a biomimetic collagen matrix loaded with growth factors to induce the differentiation of PC12 or SH-SY6Y cells. For the first time, we show that when the contact between cells and Aβ aggregates is allowed by opening up the matrix porosity, the close vicinity with aggregates induces neurite dystrophy. The compartmented 3D culture model developed and used in this study is a valuable tool to study the cytotoxicity of preformed dense Aβ aggregates and proves that contact between the aggregates and neurons is required to induce neurodegenerative processes.
Collapse
|
18
|
Anastasio TJ. Temporal-logic analysis of microglial phenotypic conversion with exposure to amyloid-β. MOLECULAR BIOSYSTEMS 2016; 11:434-53. [PMID: 25406664 DOI: 10.1039/c4mb00457d] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Alzheimer Disease (AD) remains a leading killer with no adequate treatment. Ongoing research increasingly implicates the brain's immune system as a critical contributor to AD pathogenesis, but the complexity of the immune contribution poses a barrier to understanding. Here I use temporal logic to analyze a computational specification of the immune component of AD. Temporal logic is an extension of logic to propositions expressed in terms of time. It has traditionally been used to analyze computational specifications of complex engineered systems but applications to complex biological systems are now appearing. The inflammatory component of AD involves the responses of microglia to the peptide amyloid-β (Aβ), which is an inflammatory stimulus and a likely causative AD agent. Temporal-logic analysis of the model provides explanations for the puzzling findings that Aβ induces an anti-inflammatory and well as a pro-inflammatory response, and that Aβ is phagocytized by microglia in young but not in old animals. To potentially explain the first puzzle, the model suggests that interferon-γ acts as an "autocrine bridge" over which an Aβ-induced increase in pro-inflammatory cytokines leads to an increase in anti-inflammatory mediators also. To potentially explain the second puzzle, the model identifies a potential instability in signaling via insulin-like growth factor 1 that could explain the failure of old microglia to phagocytize Aβ. The model predicts that augmentation of insulin-like growth factor 1 signaling, and activation of protein kinase C in particular, could move old microglia from a neurotoxic back toward a more neuroprotective and phagocytic phenotype.
Collapse
Affiliation(s)
- Thomas J Anastasio
- Computational Neurobiology Laboratory, Department of Molecular and Integrative Physiology, and Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
19
|
Sáez-Orellana F, Godoy PA, Bastidas CY, Silva-Grecchi T, Guzmán L, Aguayo LG, Fuentealba J. ATP leakage induces P2XR activation and contributes to acute synaptic excitotoxicity induced by soluble oligomers of β-amyloid peptide in hippocampal neurons. Neuropharmacology 2015; 100:116-23. [PMID: 25896766 DOI: 10.1016/j.neuropharm.2015.04.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 03/10/2015] [Accepted: 04/07/2015] [Indexed: 01/06/2023]
Abstract
Recent studies suggest that the toxic effects of Aβ can be attributed to its capability to insert in membranes and form pore-like structures, which are permeable to cations and molecules such as ATP. Our working hypothesis is that Aβ increases extracellular ATP causing activation of P2X receptors and potentiating excitatory synaptic activity. We found that soluble oligomers of β-amyloid peptide increased cytosolic Ca(2+) 4-fold above control (415 ± 28% of control). Also, ATP leakage (157 ± 10% of control) was independent of extracellular Ca(2+), suggesting that ATP traveled from the cytosol through an Aβ pore-mediated efflux and not from exocytotic mechanisms. The subsequent activation of P2XR by ATP can contribute to the cytosolic Ca(2+) increase observed with Aβ. Additionally, we found that β-amyloid oligomers bind preferentially to excitatory neurons inducing an increase in excitatory synaptic current frequency (248.1 ± 32.7%) that was blocked by the use of P2XR antagonists such as PPADS (Aβ + PPADS: 110.9 ± 18.35%) or Apyrase plus DPCPX (Aβ + inhibitors: 98.97 ± 17.4%). Taken together, we suggest that Aβ induces excitotoxicity by binding preferentially to excitatory neuron membranes forming a non-selective pore and by increasing intracellular calcium by itself and through P2XR activation by extracellular ATP leading to an augmention in mEPSC activity. All these effects were blocked with a non-specific P2XR antagonist, indicating that part of the neurotoxicity of Aβ is mediated by P2XR activation and facilitation of excitatory neurotransmitter release. These findings suggest that P2XR can be considered as a potential new target for the development of drugs or pharmacological tools to treat Alzheimer's disease. This article is part of the Special Issue entitled 'Synaptopathy--from Biology to Therapy'.
Collapse
Affiliation(s)
- F Sáez-Orellana
- Laboratory of Screening of Neuroactive Compound, Physiology Department, Faculty of Biological Sciences, Universidad de Concepción, Concepción, Chile
| | - P A Godoy
- Laboratory of Screening of Neuroactive Compound, Physiology Department, Faculty of Biological Sciences, Universidad de Concepción, Concepción, Chile
| | - C Y Bastidas
- Laboratory of Screening of Neuroactive Compound, Physiology Department, Faculty of Biological Sciences, Universidad de Concepción, Concepción, Chile
| | - T Silva-Grecchi
- Laboratory of Screening of Neuroactive Compound, Physiology Department, Faculty of Biological Sciences, Universidad de Concepción, Concepción, Chile
| | - L Guzmán
- Neurophysiology Laboratory, Physiology Department, Faculty of Biological Sciences, Universidad de Concepción, Concepción, Chile
| | - L G Aguayo
- Neurophysiology Laboratory, Physiology Department, Faculty of Biological Sciences, Universidad de Concepción, Concepción, Chile
| | - J Fuentealba
- Laboratory of Screening of Neuroactive Compound, Physiology Department, Faculty of Biological Sciences, Universidad de Concepción, Concepción, Chile; Center for Advanced Research on Biomedicine (CIAB-UdeC), Physiology Department, Faculty of Biological Sciences, Universidad de Concepción, Concepción, Chile.
| |
Collapse
|
20
|
Picone P, Nuzzo D, Caruana L, Messina E, Barera A, Vasto S, Di Carlo M. Metformin increases APP expression and processing via oxidative stress, mitochondrial dysfunction and NF-κB activation: Use of insulin to attenuate metformin's effect. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:1046-59. [PMID: 25667085 DOI: 10.1016/j.bbamcr.2015.01.017] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Revised: 01/21/2015] [Accepted: 01/25/2015] [Indexed: 12/14/2022]
Abstract
Clinical and experimental biomedical studies have shown Type 2 diabetes mellitus (T2DM) to be a risk factor for the development of Alzheimer's disease (AD). This study demonstrates the effect of metformin, a therapeutic biguanide administered for T2DM therapy, on β-amyloid precursor protein (APP) metabolism in in vitro, ex vivo and in vivo models. Furthermore, the protective role of insulin against metformin is also demonstrated. In LAN5 neuroblastoma cells, metformin increases APP and presenilin levels, proteins involved in AD. Overexpression of APP and presenilin 1 (Pres 1) increases APP cleavage and intracellular accumulation of β-amyloid peptide (Aβ), which, in turn, promotes aggregation of Aβ. In the experimental conditions utilized the drug causes oxidative stress, mitochondrial damage, decrease of Hexokinase-II levels and cytochrome C release, all of which lead to cell death. Several changes in oxidative stress-related genes following metformin treatment were detected by PCR arrays specific for the oxidative stress pathway. These effects of metformin were found to be antagonized by the addition of insulin, which reduced Aβ levels, oxidative stress, mitochondrial dysfunction and cell death. Similarly, antioxidant molecules, such as ferulic acid and curcumin, are able to revert metformin's effect. Comparable results were obtained using peripheral blood mononuclear cells. Finally, the involvement of NF-κB transcription factor in regulating APP and Pres 1 expression was investigated. Upon metformin treatment, NF-κB is activated and translocates from the cytoplasm to the nucleus, where it induces increased APP and Pres 1 transcription. The use of Bay11-7085 inhibitor suppressed the effect of metformin on APP and Pres 1 expression.
Collapse
Affiliation(s)
- Pasquale Picone
- Institute of Biomedicine and Molecular Immunology "Alberto Monroy" (IBIM), Consiglio Nazionale delle Ricerche (CNR), 90146 Palermo, Italy
| | - Domenico Nuzzo
- Institute of Biomedicine and Molecular Immunology "Alberto Monroy" (IBIM), Consiglio Nazionale delle Ricerche (CNR), 90146 Palermo, Italy
| | - Luca Caruana
- Institute of Biomedicine and Molecular Immunology "Alberto Monroy" (IBIM), Consiglio Nazionale delle Ricerche (CNR), 90146 Palermo, Italy
| | - Elisa Messina
- Institute of Biomedicine and Molecular Immunology "Alberto Monroy" (IBIM), Consiglio Nazionale delle Ricerche (CNR), 90146 Palermo, Italy
| | - Annalisa Barera
- Department of STEBICEF, University of Palermo, 90100 Palermo, Italy
| | - Sonya Vasto
- Institute of Biomedicine and Molecular Immunology "Alberto Monroy" (IBIM), Consiglio Nazionale delle Ricerche (CNR), 90146 Palermo, Italy; Department of STEBICEF, University of Palermo, 90100 Palermo, Italy
| | - Marta Di Carlo
- Institute of Biomedicine and Molecular Immunology "Alberto Monroy" (IBIM), Consiglio Nazionale delle Ricerche (CNR), 90146 Palermo, Italy.
| |
Collapse
|
21
|
Increased inflammatory response in cytomegalovirus seropositive patients with Alzheimer's disease. PLoS One 2014; 9:e96779. [PMID: 24804776 PMCID: PMC4013077 DOI: 10.1371/journal.pone.0096779] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 04/11/2014] [Indexed: 12/18/2022] Open
Abstract
Alzheimer’s disease (AD) has been associated with increased local inflammation in the affected brain regions, and in some studies also with elevated levels of proinflammatory cytokines in peripheral blood. Cytomegalovirus (CMV) is known to promote a more effector-oriented phenotype in the T-cell compartment, increasing with age. The aim of this study was to investigate the inflammatory response of peripheral blood mononuclear cells (PBMCs) from AD patients and non-demented (ND) controls. Using a multiplex Luminex xMAP assay targeting GM-CSF, IFN-γ, IL-1β, IL-2, IL-4, IL-5, IL-6, IL-8, IP-10 and TNF-α, cytokine profiles from PBMCs were analysed after stimulation with anti-CD3/CD28 beads, CMV pp65 peptide mix or amyloid β (Aβ) protofibrils, respectively. CMV seropositive AD subjects presented with higher IFN-γ levels after anti-CD3/CD28 and CMV pp65 but not after Aβ stimulation, compared to CMV seropositive ND controls. When analysing IFN-γ response to anti-CD3/CD28 stimulation on a subgroup level, CMV seropositive AD subjects presented with higher levels compared to both CMV seronegative AD and CMV seropositive ND subjects. Taken together, our data from patients with clinically manifest AD suggest a possible role of CMV as an inflammatory promoter in AD immunology. Further studies of AD patients at earlier stages of disease, could provide better insight into the pathophysiology.
Collapse
|
22
|
Notarachille G, Arnesano F, Calò V, Meleleo D. Heavy metals toxicity: effect of cadmium ions on amyloid beta protein 1-42. Possible implications for Alzheimer's disease. Biometals 2014; 27:371-88. [PMID: 24557150 DOI: 10.1007/s10534-014-9719-6] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Accepted: 02/12/2014] [Indexed: 02/02/2023]
Abstract
Cadmium (Cd) is an environmental contaminant, highly toxic to humans. This biologically non-essential element accumulates in the body, especially in the kidney, liver, lung and brain and can induce several toxic effects, depending on the concentration and the exposure time. Cd has been linked to Alzheimer's disease (AD) as a probable risk factor, as it shows higher concentrations in brain tissues of AD patients than in healthy people, its implication in the formation of neurofibrillary tangles and in the aggregation process of amyloid beta peptides (AβPs). AβPs seem to have toxic properties, particularly in their aggregated state; insoluble AβP forms, such as small and large aggregates, protofibrils and fibrils, appear to be implicated in the pathogenesis of AD. In our study, we have evaluated the effect of Cd, at different concentrations, both on the AβP1-42 ion channel incorporated in a planar lipid membrane made up of phosphatidylcholine containing 30 % cholesterol and on the secondary structure of AβP1-42 in aqueous environment. Cadmium is able to interact with the AβP1-42 peptide by acting on the channel incorporated into the membrane as well as on the peptide in solution, both decreasing AβP1-42 channel frequency and in solution forming large and amorphous aggregates prone to precipitate. These experimental observations suggesting a toxic role for Cd strengthen the hypothesis that Cd may interact directly with AβPs and may be a risk factor in AD.
Collapse
Affiliation(s)
- Gabriella Notarachille
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari "Aldo Moro", via E. Orabona 4, 70126, Bari, Italy
| | | | | | | |
Collapse
|
23
|
Whiteley CG. Arginine metabolising enzymes as targets against Alzheimers’ disease. Neurochem Int 2014; 67:23-31. [DOI: 10.1016/j.neuint.2014.01.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Revised: 01/26/2014] [Accepted: 01/28/2014] [Indexed: 01/26/2023]
|
24
|
Dao AT, Zagaar MA, Levine AT, Salim S, Eriksen JL, Alkadhi KA. Treadmill exercise prevents learning and memory impairment in Alzheimer's disease-like pathology. Curr Alzheimer Res 2014; 10:507-15. [PMID: 23627709 DOI: 10.2174/1567205011310050006] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Revised: 01/29/2013] [Accepted: 01/30/2013] [Indexed: 12/29/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that is characterized by progressive memory loss. In contrast, accumulating evidence suggests a neuroprotective role of regular exercise in aging associated memory impairment. In this study, we investigated the ability of regular exercise to prevent impairments of short-term memory (STM) and early long-term potentiation (E-LTP) in area CA1 of the hippocampus in a rat model of AD (i.c.v. infusion of 250 pmol/day Aβ1-42 peptides). We utilized behavioral assessment, in vivo electrophysiological recording, and immunoblotting in 4 groups of adult Wistar rats: control, treadmill exercise (Ex), β-amyloid-infused (Aβ), and amyloid-infused/treadmill exercised (Ex/Aβ). Our findings indicated that Aβ rats made significantly more errors in the radial arm water maze (RAWM) compared to all other groups and exhibited suppressed E-LTP in area CA1, which correlated with deleterious alterations in the levels of memory and E-LTP-related signaling molecules including calcineurin (PP2B), brain derivedneurotrophic factor (BDNF) and phosphorylated CaMKII (p-CaMKII). Compared to controls, Ex and Ex/Aβ rats showed a similar behavioral performance and a normal E-LTP with no detrimental changes in the levels of PP2B, BDNF, and p- CaMKII. We conclude that treadmill exercise maybe able to prevent cognitive impairment associated with AD pathology.
Collapse
Affiliation(s)
- An T Dao
- Department of PPS, College of Pharmacy, University of Houston, Houston, TX 77204-5037, USA
| | - Munder A Zagaar
- Texas Southern University Department of Pharmacy Practice and Clinical Health Sciences Houston, TX 77004
| | | | | | | | | |
Collapse
|
25
|
Raychaudhuri S, Raychaudhuri SC. Death ligand concentration and the membrane proximal signaling module regulate the type 1/type 2 choice in apoptotic death signaling. SYSTEMS AND SYNTHETIC BIOLOGY 2013; 8:83-97. [PMID: 24592294 DOI: 10.1007/s11693-013-9124-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Revised: 08/29/2013] [Accepted: 09/02/2013] [Indexed: 01/06/2023]
Abstract
Apoptotic death pathways are frequently activated by death ligand induction and subsequent activation of the membrane proximal signaling module. Death receptors cluster upon binding to death ligands, leading to formation of a membrane proximal death-inducing-signaling-complex (DISC). In this membrane proximal signalosome, initiator caspases (caspase 8) are processed resulting in activation of both type 1 and type 2 pathways of apoptosis signaling. How the type 1/type 2 choice is made is an important question in the systems biology of apoptosis signaling. In this study, we utilize a Monte Carlo based in silico approach to elucidate the role of membrane proximal signaling module in the type 1/type 2 choice of apoptosis signaling. Our results provide crucial mechanistic insights into the formation of DISC signalosome and caspase 8 activation. Increased concentration of death ligands was shown to correlate with increased type 1 activation. We also study the caspase 6 mediated system level feedback activation of apoptosis signaling and its role in the type 1/type 2 choice. Our results clarify the basis of cell-to-cell stochastic variability in apoptosis activation and ramifications of this issue is further discussed in the context of therapies for cancer and neurodegenerative disorders.
Collapse
Affiliation(s)
- Subhadip Raychaudhuri
- Indraprastha Institute of Information Technology, Delhi, 110020 Delhi India ; Department of Chemistry, University of California, Davis, Davis, CA 95776 USA
| | | |
Collapse
|
26
|
Hemmati F, Dargahi L, Nasoohi S, Omidbakhsh R, Mohamed Z, Chik Z, Naidu M, Ahmadiani A. Neurorestorative effect of FTY720 in a rat model of Alzheimer's disease: Comparison with Memantine. Behav Brain Res 2013; 252:415-21. [DOI: 10.1016/j.bbr.2013.06.016] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Revised: 06/03/2013] [Accepted: 06/07/2013] [Indexed: 12/14/2022]
|
27
|
Jose JC, Sengupta N. Molecular dynamics simulation studies of the structural response of an isolated Aβ1–42 monomer localized in the vicinity of the hydrophilic TiO2 surface. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2013; 42:487-94. [DOI: 10.1007/s00249-013-0900-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 02/21/2013] [Accepted: 03/21/2013] [Indexed: 12/12/2022]
|
28
|
Anastasio TJ. Exploring the contribution of estrogen to amyloid-Beta regulation: a novel multifactorial computational modeling approach. Front Pharmacol 2013; 4:16. [PMID: 23459573 PMCID: PMC3585711 DOI: 10.3389/fphar.2013.00016] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Accepted: 01/31/2013] [Indexed: 11/23/2022] Open
Abstract
According to the amyloid hypothesis, Alzheimer Disease results from the accumulation beyond normative levels of the peptide amyloid-β (Aβ). Perhaps because of its pathological potential, Aβ and the enzymes that produce it are heavily regulated by the molecular interactions occurring within cells, including neurons. This regulation involves a highly complex system of intertwined normative and pathological processes, and the sex hormone estrogen contributes to it by influencing the Aβ-regulation system at many different points. Owing to its high complexity, Aβ regulation and the contribution of estrogen are very difficult to reason about. This report describes a computational model of the contribution of estrogen to Aβ regulation that provides new insights and generates experimentally testable and therapeutically relevant predictions. The computational model is written in the declarative programming language known as Maude, which allows not only simulation but also analysis of the system using temporal-logic. The model illustrates how the various effects of estrogen could work together to reduce Aβ levels, or prevent them from rising, in the presence of pathological triggers. The model predicts that estrogen itself should be more effective in reducing Aβ than agonists of estrogen receptor α (ERα), and that agonists of ERβ should be ineffective. The model shows how estrogen itself could dramatically reduce Aβ, and predicts that non-steroidal anti-inflammatory drugs should provide a small additional benefit. It also predicts that certain compounds, but not others, could augment the reduction in Aβ due to estrogen. The model is intended as a starting point for a computational/experimental interaction in which model predictions are tested experimentally, the results are used to confirm, correct, and expand the model, new predictions are generated, and the process continues, producing a model of ever increasing explanatory power and predictive value.
Collapse
Affiliation(s)
- Thomas J Anastasio
- Computational Neurobiology Laboratory, Beckman Institute, Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign Urbana, IL, USA
| |
Collapse
|
29
|
Lee S, Tong M, Hang S, Deochand C, de la Monte S. CSF and Brain Indices of Insulin Resistance, Oxidative Stress and Neuro-Inflammation in Early versus Late Alzheimer's Disease. ACTA ACUST UNITED AC 2013; 3:128. [PMID: 25035815 PMCID: PMC4096626 DOI: 10.4172/2161-0460.1000128] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Alzheimer’s disease (AD) is characterized by progressive impairments in cognitive and behavioral functions with deficits in learning, memory and executive reasoning. Growing evidence points toward brain insulin and insulin-like growth factor (IGF) resistance-mediated metabolic derangements as critical etiologic factors in AD. This suggests that indices of insulin/IGF resistance and their consequences, i.e. oxidative stress, neuro-inflammation, and reduced neuronal plasticity, should be included in biomarker panels for AD. Herein, we examine a range of metabolic, inflammatory, stress, and neuronal plasticity related proteins in early AD, late AD, and aged control postmortem brain, postmortem ventricular fluid (VF), and clinical cerebrospinal fluid (CSF) samples. In AD brain, VF, and CSF samples the trends with respect to alterations in metabolic, neurotrophin, and stress indices were similar, but for pro-inflammatory cytokines, the patterns were discordant. With the greater severities of dementia and neurodegeneration, the differences from control were more pronounced for late AD (VF and brain) than early or moderate AD (brain, VF and CSF). The findings suggest that the inclusion of metabolic, neurotrophin, stress biomarkers in AβPP-Aβ+pTau CSF-based panels could provide more information about the status and progression of neurodegeneration, as well as aid in predicting progression from early- to late-stage AD. Furthermore, standardized multi-targeted molecular assays of neurodegeneration could help streamline postmortem diagnoses, including assessments of AD severity and pathology.
Collapse
Affiliation(s)
- Sarah Lee
- Department of Medicine, Rhode Island Hospital, Warren Alpert Medical School, Providence, RI, USA
| | - Ming Tong
- Department of Medicine, Rhode Island Hospital, Warren Alpert Medical School, Providence, RI, USA
| | - Steven Hang
- Department of Medicine, Warren Alpert Medical School, Providence, RI, USA
| | - Chetram Deochand
- Departments of Medicine, Rhode Island Hospital, Brown University, Providence, RI, USA
| | - Suzanne de la Monte
- Department of Medicine, Pathology (Neuropathology), Neurology and Neurosurgery, Rhode Island Hospital, Warren Alpert Medical School, Brown University, Providence, RI, USA
| |
Collapse
|
30
|
Chami L, Checler F. BACE1 is at the crossroad of a toxic vicious cycle involving cellular stress and β-amyloid production in Alzheimer's disease. Mol Neurodegener 2012; 7:52. [PMID: 23039869 PMCID: PMC3507664 DOI: 10.1186/1750-1326-7-52] [Citation(s) in RCA: 120] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Accepted: 10/03/2012] [Indexed: 02/06/2023] Open
Abstract
Alzheimer’s disease (AD) is a complex age-related pathology, the etiology of which has not been firmly delineated. Among various histological stigmata, AD-affected brains display several cellular dysfunctions reflecting enhanced oxidative stress, inflammation process and calcium homeostasis disturbance. Most of these alterations are directly or indirectly linked to amyloid β-peptides (Aβ), the production, molecular nature and biophysical properties of which likely conditions the degenerative process. It is particularly noticeable that, in a reverse control process, the above-described cellular dysfunctions alter Aβ peptides levels. β-secretase βAPP-cleaving enzyme 1 (BACE1) is a key molecular contributor of this cross-talk. This enzyme is responsible for the primary cleavage generating the N-terminus of “full length” Aβ peptides and is also transcriptionally induced by several cellular stresses. This review summarizes data linking brain insults to AD-like pathology and documents the key role of BACE1 at the cross-road of a vicious cycle contributing to Aβ production.
Collapse
Affiliation(s)
- Linda Chami
- Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275 CNRS/UNSA, 06560 Valbonne, France
| | | |
Collapse
|
31
|
Piccini A, Borghi R, Guglielmotto M, Tamagno E, Cirmena G, Garuti A, Pollero V, Cammarata S, Fornaro M, Messa M, Colombo L, Salmona M, Perry G, Tabaton M. β-Amyloid 1-42 induces physiological transcriptional regulation of BACE1. J Neurochem 2012; 122:1023-31. [DOI: 10.1111/j.1471-4159.2012.07834.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
32
|
Masters CL, Selkoe DJ. Biochemistry of amyloid β-protein and amyloid deposits in Alzheimer disease. Cold Spring Harb Perspect Med 2012; 2:a006262. [PMID: 22675658 PMCID: PMC3367542 DOI: 10.1101/cshperspect.a006262] [Citation(s) in RCA: 419] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Progressive cerebral deposition of the amyloid β-protein (Aβ) in brain regions serving memory and cognition is an invariant and defining feature of Alzheimer disease. A highly similar but less robust process accompanies brain aging in many nondemented humans, lower primates, and some other mammals. The discovery of Aβ as the subunit of the amyloid fibrils in meningocerebral blood vessels and parenchymal plaques has led to innumerable studies of its biochemistry and potential cytotoxic properties. Here we will review the discovery of Aβ, numerous aspects of its complex biochemistry, and current attempts to understand how a range of Aβ assemblies, including soluble oligomers and insoluble fibrils, may precipitate and promote neuronal and glial alterations that underlie the development of dementia. Although the role of Aβ as a key molecular factor in the etiology of Alzheimer disease remains controversial, clinical trials of amyloid-lowering agents, reviewed elsewhere in this book, are poised to resolve the question of its pathogenic primacy.
Collapse
Affiliation(s)
- Colin L Masters
- The Mental Health Research Institute, The University of Melbourne, Parkville 3010, Australia.
| | | |
Collapse
|
33
|
Smid SD, Maag JL, Musgrave IF. Dietary polyphenol-derived protection against neurotoxic β-amyloid protein: from molecular to clinical. Food Funct 2012; 3:1242-50. [DOI: 10.1039/c2fo30075c] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
34
|
Takahashi T, Mihara H. FRET detection of amyloid β-peptide oligomerization using a fluorescent protein probe presenting a pseudo-amyloid structure. Chem Commun (Camb) 2012; 48:1568-70. [DOI: 10.1039/c1cc14552e] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
35
|
Anastasio TJ. Data-driven modeling of Alzheimer Disease pathogenesis. J Theor Biol 2011; 290:60-72. [DOI: 10.1016/j.jtbi.2011.08.038] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2011] [Revised: 08/27/2011] [Accepted: 08/29/2011] [Indexed: 01/28/2023]
|
36
|
Polymorphic structures of Alzheimer's β-amyloid globulomers. PLoS One 2011; 6:e20575. [PMID: 21687730 PMCID: PMC3110195 DOI: 10.1371/journal.pone.0020575] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2011] [Accepted: 05/04/2011] [Indexed: 01/25/2023] Open
Abstract
Background Misfolding and self-assembly of Amyloid-β (Aβ) peptides into amyloid fibrils is pathologically linked to the development of Alzheimer's disease. Polymorphic Aβ structures derived from monomers to intermediate oligomers, protofilaments, and mature fibrils have been often observed in solution. Some aggregates are on-pathway species to amyloid fibrils, while the others are off-pathway species that do not evolve into amyloid fibrils. Both on-pathway and off-pathway species could be biologically relevant species. But, the lack of atomic-level structural information for these Aβ species leads to the difficulty in the understanding of their biological roles in amyloid toxicity and amyloid formation. Methods and Findings Here, we model a series of molecular structures of Aβ globulomers assembled by monomer and dimer building blocks using our peptide-packing program and explicit-solvent molecular dynamics (MD) simulations. Structural and energetic analysis shows that although Aβ globulomers could adopt different energetically favorable but structurally heterogeneous conformations in a rugged energy landscape, they are still preferentially organized by dynamic dimeric subunits with a hydrophobic core formed by the C-terminal residues independence of initial peptide packing and organization. Such structural organizations offer high structural stability by maximizing peptide-peptide association and optimizing peptide-water solvation. Moreover, curved surface, compact size, and less populated β-structure in Aβ globulomers make them difficult to convert into other high-order Aβ aggregates and fibrils with dominant β-structure, suggesting that they are likely to be off-pathway species to amyloid fibrils. These Aβ globulomers are compatible with experimental data in overall size, subunit organization, and molecular weight from AFM images and H/D amide exchange NMR. Conclusions Our computationally modeled Aβ globulomers provide useful insights into structure, dynamics, and polymorphic nature of Aβ globulomers which are completely different from Aβ fibrils, suggesting that these globulomers are likely off-pathway species and explaining the independence of the aggregation kinetics between Aβ globulomers and fibrils.
Collapse
|
37
|
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
|
38
|
Insulin promotes survival of amyloid-beta oligomers neuroblastoma damaged cells via caspase 9 inhibition and Hsp70 upregulation. J Biomed Biotechnol 2010; 2010:147835. [PMID: 20490276 PMCID: PMC2871552 DOI: 10.1155/2010/147835] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2009] [Revised: 02/24/2010] [Accepted: 02/24/2010] [Indexed: 11/21/2022] Open
Abstract
Alzheimer's disease (AD) and type 2 diabetes are connected in a way that is still not completely understood, but insulin resistance has been implicated as a risk factor for developing AD. Here we show an evidence that insulin is capable of reducing cytotoxicity induced by Amyloid-beta peptides (A-beta) in its oligomeric form in a dose-dependent manner. By TUNEL and biochemical assays we demonstrate that the recovery of the cell viability is obtained by inhibition of intrinsic apoptotic program, triggered by A-beta and involving caspase 9 and 3 activation. A protective role of insulin on mitochondrial damage is also shown by using Mito-red vital dye. Furthermore, A-beta activates the stress inducible Hsp70 protein in LAN5 cells and an overexpression is detectable after the addition of insulin, suggesting that this major induction is the necessary condition to activate a cell survival program. Together, these results may provide opportunities for the design of preventive and therapeutic strategies against AD.
Collapse
|
39
|
Tabaton M, Zhu X, Perry G, Smith MA, Giliberto L. Signaling effect of amyloid-beta(42) on the processing of AbetaPP. Exp Neurol 2009; 221:18-25. [PMID: 19747481 DOI: 10.1016/j.expneurol.2009.09.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2009] [Revised: 09/01/2009] [Accepted: 09/02/2009] [Indexed: 12/28/2022]
Abstract
The effects of amyloid-beta are extremely complex. Current work in the field of Alzheimer disease is focusing on discerning the impact between the physiological signaling effects of soluble low molecular weight amyloid-beta species and the more global cellular damage that could derive from highly concentrated and/or aggregated amyloid. Being able to dissect the specific signaling events, to understand how soluble amyloid-beta induces its own production by up-regulating BACE1 expression, could lead to new tools to interrupt the distinctive feedback cycle with potential therapeutic consequences. Here we describe a positive loop that exists between the secretases that are responsible for the generation of the amyloid-beta component of Alzheimer disease. According to our hypothesis, in familial Alzheimer disease, the primary overproduction of amyloid-beta can induce BACE1 transcription and drive a further increase of amyloid-beta precursor protein processing and resultant amyloid-beta production. In sporadic Alzheimer disease, many factors, among them oxidative stress and inflammation, with consequent induction of presenilins and BACE1, would activate a loop and proceed with the generation of amyloid-beta and its signaling role onto BACE1 transcription. This concept of a signaling effect by and feedback on the amyloid-beta precursor protein will likely shed light on how amyloid-beta generation, oxidative stress, and secretase functions are intimately related in sporadic Alzheimer disease.
Collapse
Affiliation(s)
- Massimo Tabaton
- Departments of Neuroscience, Ophthalmology, and Genetics, University of Genova, Genova, Italy.
| | | | | | | | | |
Collapse
|