1051
|
Chauhan PS, Yadav D, Koul B, Mohanta YK, Jin JO. Recent Advances in Nanotechnology: A Novel Therapeutic System for the Treatment of Alzheimer's Disease. Curr Drug Metab 2020; 21:1144-1151. [PMID: 33234100 DOI: 10.2174/1389200221666201124140518] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 07/14/2020] [Accepted: 10/15/2020] [Indexed: 11/22/2022]
Abstract
A amyloid-β (Aβ) plaque formation in the brain is known to be the root cause of Alzheimer's disease (AD), which affects the behavior, memory, and cognitive ability in humans. The brain starts undergoing changes several years before the actual appearance of the symptoms. Nanotechnology could prove to be an alternative strategy for treating the disease effectively. It encompasses the diagnosis as well as the therapeutic aspect using validated biomarkers and nano-based drug delivery systems, respectively. A nano-based therapy may provide an alternate strategy, wherein one targets the protofibrillar amyloid-β (Aβ) structures, and this is followed by their disaggregation as random coils. Conventional/routine drug therapies are inefficient in crossing the blood-brain barrier; however, this hurdle can be overcome with the aid of nanoparticles. The present review highlights the various challenges in the diagnosis and treatment of AD. Meticulous and collaborative research using nanotherapeutic systems could provide remarkable breakthroughs in the early-stage diagnosis and therapy of AD.
Collapse
Affiliation(s)
- Pallavi Singh Chauhan
- Amity Institute of Biotechnology, Amity University Madhya Pradesh, Gwalior, 474005, India
| | - Dhananjay Yadav
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, South Korea
| | - Bhupendra Koul
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara-144411, Punjab, India
| | - Yugal Kishore Mohanta
- Biochemistry Laboratory, Department of Botany, North Orissa University Baripada- 757003, India
| | - Jun-O Jin
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, South Korea
| |
Collapse
|
1052
|
Illig AM, Strodel B. Performance of Markov State Models and Transition Networks on Characterizing Amyloid Aggregation Pathways from MD Data. J Chem Theory Comput 2020; 16:7825-7839. [DOI: 10.1021/acs.jctc.0c00727] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Alexander-Maurice Illig
- Institute of Biological Information Processing: Structural Biochemistry (IBI-7), Forschungszentrum Jülich, 52428 Jülich, Germany
| | - Birgit Strodel
- Institute of Biological Information Processing: Structural Biochemistry (IBI-7), Forschungszentrum Jülich, 52428 Jülich, Germany
- Institute of Theoretical and Computational Chemistry, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| |
Collapse
|
1053
|
Lee HJ, Woo H, Lee HE, Jeon H, Ryu KY, Nam JH, Jeon SG, Park H, Lee JS, Han KM, Lee SM, Kim J, Kang RJ, Lee YH, Kim JI, Hoe HS. The novel DYRK1A inhibitor KVN93 regulates cognitive function, amyloid-beta pathology, and neuroinflammation. Free Radic Biol Med 2020; 160:575-595. [PMID: 32896600 DOI: 10.1016/j.freeradbiomed.2020.08.030] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 07/27/2020] [Accepted: 08/31/2020] [Indexed: 02/08/2023]
Abstract
Regulating amyloid beta (Aβ) pathology and neuroinflammatory responses holds promise for the treatment of Alzheimer's disease (AD) and other neurodegenerative and/or neuroinflammation-related diseases. In this study, the effects of KVN93, an inhibitor of dual-specificity tyrosine phosphorylation-regulated kinase-1A (DYRK1A), on cognitive function and Aβ plaque levels and the underlying mechanism of action were evaluated in 5x FAD mice (a mouse model of AD). KVN93 treatment significantly improved long-term memory by enhancing dendritic synaptic function. In addition, KVN93 significantly reduced Aβ plaque levels in 5x FAD mice by regulating levels of the Aβ degradation enzymes neprilysin (NEP) and insulin-degrading enzyme (IDE). Moreover, Aβ-induced microglial and astrocyte activation were significantly suppressed in the KVN-treated 5xFAD mice. KVN93 altered neuroinflammation induced by LPS in microglial cells but not primary astrocytes by regulating TLR4/AKT/STAT3 signaling, and in wild-type mice injected with LPS, KVN93 treatment reduced microglial and astrocyte activation. Overall, these results suggest that the novel DYRK1A inhibitor KVN93 is a potential therapeutic drug for regulating cognitive/synaptic function, Aβ plaque load, and neuroinflammatory responses in the brain.
Collapse
Affiliation(s)
- Hyun-Ju Lee
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41062, South Korea.
| | - Hanwoong Woo
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41062, South Korea.
| | - Ha-Eun Lee
- School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, South Korea.
| | - Hyongjun Jeon
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41062, South Korea.
| | - Ka-Young Ryu
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41062, South Korea.
| | - Jin Han Nam
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41062, South Korea.
| | - Seong Gak Jeon
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41062, South Korea.
| | - HyunHee Park
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41062, South Korea.
| | - Ji-Soo Lee
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41062, South Korea.
| | - Kyung-Min Han
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41062, South Korea.
| | - Sang Min Lee
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41062, South Korea.
| | - Jeongyeon Kim
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41062, South Korea.
| | - Ri Jin Kang
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41062, South Korea.
| | - Young-Ho Lee
- Research Center of Bioconvergence Analysis, Korea Basic Science Institute (KBSI), Ochang, Cheongju, Chungbuk, 28119, South Korea; Bio-Analytical Science, University of Science and Technology (UST), Gajeong-ro, Yuseong-gu, Daejeon 34113, South Korea; Neurovascular Research Group, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu 41062, South Korea.
| | - Jae-Ick Kim
- School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, South Korea.
| | - Hyang-Sook Hoe
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41062, South Korea; Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science & Technology, Daegu, 42988, South Korea.
| |
Collapse
|
1054
|
Chadha S, Behl T, Sehgal A, Kumar A, Bungau S. Exploring the role of mitochondrial proteins as molecular target in Alzheimer's disease. Mitochondrion 2020; 56:62-72. [PMID: 33221353 DOI: 10.1016/j.mito.2020.11.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/10/2020] [Accepted: 11/11/2020] [Indexed: 02/06/2023]
Abstract
Brain is a fully differentiated organ and is sensitive towards oxidative damage of various compounds including lipids, proteins, and DNA that occurs during process of normal aging and is mainly due to its high energy metabolism and reduced activity of anti-oxidative defense mechanism. Mitochondria are dynamic ATP-generating organelles which constitutes cellular functions such as regulation of intracellular calcium, bio-energetic processes, and reduction-oxidation of cells. Such functioning is negatively affected due to the presence of amyloid β peptide (Aβ) which is involved in pathogenesis of Alzheimer disease (AD). Aβ interacts with mitochondria and leads to mitochondrial dysfunction. Mitochondrial dysfunction, abnormal interactions, oxidative stress, and mis-folding of synaptic proteins inside nervous system are explored and regarded as primary or initial features in insurgence of pathology (AD and other neurological disease). The major histopathological hallmarks of AD are characterized by presence of these hallmarks intracellularly, its further progression and exacerbation which leads to excessive accumulation of oligomeric as well as fibrillar-β-amyloid peptides (present extracellularly) and accumulation of neurofibrillary tangles intracellularly. The current review will focus on alterations and variation in mitochondria/mitochondrial DNA (mtDNA) and the rationale for involvement of related abnormalities in pathogenesis of AD.
Collapse
Affiliation(s)
- Swati Chadha
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Arun Kumar
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Simona Bungau
- Department of Pharmacy, Faculty of Pharmacy, University of Oradea, Romania
| |
Collapse
|
1055
|
Tong Y, Wang S. Not All Stressors Are Equal: Mechanism of Stressors on RPE Cell Degeneration. Front Cell Dev Biol 2020; 8:591067. [PMID: 33330470 PMCID: PMC7710875 DOI: 10.3389/fcell.2020.591067] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 11/02/2020] [Indexed: 12/26/2022] Open
Abstract
Age-related macular degeneration (AMD) is a major cause of irreversible blindness among the elderly population. Dysfunction and degeneration of the retinal pigment epithelial (RPE) layer in the retina underscore the pathogenesis of both dry and wet AMD. Advanced age, cigarette smoke and genetic factors have been found to be the prominent risk factors for AMD, which point to an important role for oxidative stress and aging in AMD pathogenesis. However, the mechanisms whereby oxidative stress and aging lead to RPE cell degeneration are still unclear. As cell senescence and cell death are the major outcomes from oxidative stress and aging, here we review the mechanisms of RPE cell senescence and different kinds of cell death, including apoptosis, necroptosis, pyroptosis, ferroptosis, with an aim to clarify how RPE cell degeneration could occur in response to AMD-related stresses, including H2O2, 4-Hydroxynonenal (4-HNE), N-retinylidene-N-retinyl-ethanolamine (A2E), Alu RNA and amyloid β (Aβ). Besides those, sodium iodate (NaIO3) induced RPE cell degeneration is also discussed in this review. Although NaIO3 itself is not related to AMD, this line of study would help understand the mechanism of RPE degeneration.
Collapse
Affiliation(s)
- Yao Tong
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, United States
| | - Shusheng Wang
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, United States.,Department of Ophthalmology, Tulane University, New Orleans, LA, United States
| |
Collapse
|
1056
|
Karisetty BC, Bhatnagar A, Armour EM, Beaver M, Zhang H, Elefant F. Amyloid-β Peptide Impact on Synaptic Function and Neuroepigenetic Gene Control Reveal New Therapeutic Strategies for Alzheimer's Disease. Front Mol Neurosci 2020; 13:577622. [PMID: 33304239 PMCID: PMC7693454 DOI: 10.3389/fnmol.2020.577622] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 10/22/2020] [Indexed: 01/17/2023] Open
Abstract
Amyloid-β (Aβ) peptides can form protease-resistant aggregates within and outside of neurons. Accumulation of these aggregates is a hallmark of Alzheimer's disease (AD) neuropathology and contributes to devastating cognitive deficits associated with this disorder. The primary etiological factor for Aβ aggregation is either an increase in Aβ production or a decrease in its clearance. Aβ is produced by the sequential activity of β- and γ-secretase on the amyloid precursor protein (APP) and the clearance is mediated by chaperone-mediated mechanisms. The Aβ aggregates vary from soluble monomers and oligomers to insoluble senile plaques. While excess intraneuronal oligomers can transduce neurotoxic signals into neurons causing cellular defects like oxidative stress and neuroepigenetic mediated transcriptional dysregulation, extracellular senile plaques cause neurodegeneration by impairing neural membrane permeabilization and cell signaling pathways. Paradoxically, senile plaque formation is hypothesized to be an adaptive mechanism to sequester excess toxic soluble oligomers while leaving native functional Aβ levels intact. This hypothesis is strengthened by the absence of positive outcomes and side effects from immunotherapy clinical trials aimed at complete Aβ clearance, and support beneficial physiological roles for native Aβ in cellular function. Aβ has been shown to modulate synaptic transmission, consolidate memory, and protect against excitotoxicity. We discuss the current understanding of beneficial and detrimental roles for Aβ in synaptic function and epigenetic gene control and the future promising prospects of early therapeutic interventions aimed at mediating Aβ induced neuroepigenetic and synaptic dysfunctions to delay AD onset.
Collapse
Affiliation(s)
| | | | | | | | | | - Felice Elefant
- Department of Biology, Drexel University, Philadelphia, PA, United States
| |
Collapse
|
1057
|
Koszewicz M, Jaroch J, Brzecka A, Ejma M, Budrewicz S, Mikhaleva LM, Muresanu C, Schield P, Somasundaram SG, Kirkland CE, Avila-Rodriguez M, Aliev G. Dysbiosis is one of the risk factor for stroke and cognitive impairment and potential target for treatment. Pharmacol Res 2020; 164:105277. [PMID: 33166735 DOI: 10.1016/j.phrs.2020.105277] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/25/2020] [Accepted: 10/27/2020] [Indexed: 02/07/2023]
Abstract
More than 50 million people have various forms of cognitive impairment basically caused by neurodegenerative diseases, such as Alzheimer's, Parkinson's, and cerebrovascular diseases as well as stroke. Often these conditions coexist and exacerbate one another. The damaged area in post-stroke dementia may lead to neurodegenerative lesions. Gut microbiome functions like an endocrine organ by generating bioactive metabolites that can directly or indirectly impact human physiology. An alteration in the composition and function of intestinal flora, i.e. gut dysbiosis, is implicated in neurodegenerative and cerebrovascular diseases. Additionally, gut dysbiosis may accelerate the progression of cognitive impairment. Dysbiosis may result from obesity; metabolic disorders, cardiovascular disease, and sleep disorders, Lack of physical activity is associated with dysbiosis as well. These may coexist in various patterns in older people, enhancing the risk, incidence, and progression of cerebrovascular lesions, neurodegenerative disorders, and cognitive impairment, creating a vicious circle. Recently, it has been reported that several metabolites produced by gut microbiota (e.g., trimethylamine/trimethylamine N-oxide, short-chain fatty acids, secondary bile acids) may be linked to neurodegenerative and cerebrovascular diseases. New treatment modalities, including prebiotic and probiotics, may normalize the gut microbiota composition, change the brain-gut barrier, and decrease the risk of the pathology development. Fecal microbiota transplantation, sometimes in combination with other methods, is used for remodeling and replenishing the symbiotic gut microbiome. This promising field of research is associated with basic findings of bidirectional communication between body organs and gut microbiota that creates new possibilities of pharmacological treatments of many clinical conditions. The authors present the role of gut microbiota in physiology, and the novel therapeutic targets in modulation of intestinal microbiota Personalized therapies based on their personal genome make up could offer benefits by modulating microbiota cross-talk with brain and cardiovascular system. A healthy lifestyle, including pre and probiotic nutrition is generally recommended. Prevention may also be enhanced by correcting gut dysbiosis resulting a reduced risk of post-stroke cognitive impairment including dementia.
Collapse
Affiliation(s)
- Magdalena Koszewicz
- Department of Neurology, Wroclaw Medical University, 50-556 Wrocław, Borowska 213, Poland
| | - Joanna Jaroch
- Faculty of Health Sciences, Wroclaw Medical University, 51-618 Wrocław, Bartla 5, Poland; Department of Cardiology, Lower Silesian Specialist Hospital, Fieldorfa 2, 54-049 Wroclaw, Poland
| | - Anna Brzecka
- Department of Pulmonology and Lung Oncology, Wroclaw Medical University, 53-439, Wroclaw, Grabiszynska 105, Poland
| | - Maria Ejma
- Department of Neurology, Wroclaw Medical University, 50-556 Wrocław, Borowska 213, Poland
| | - Slawomir Budrewicz
- Department of Neurology, Wroclaw Medical University, 50-556 Wrocław, Borowska 213, Poland
| | - Liudmila M Mikhaleva
- Federal State Budgetary Institution «Research Institute of Human Morphology», 3, Tsyurupy Str., Moscow, 117418, Russian Federation
| | - Cristian Muresanu
- Research Center for Applied Biotechnology in Diagnosis and Molecular Therapies, Str. Trifoiului nr. 12 G, 400478, Cluj-Napoca, Romania
| | - Pamela Schield
- School of Education & Athletics, Salem University, Salem, WV 26426, United States
| | | | - Cecil E Kirkland
- Department of Biological Sciences, Salem University, Salem, WV, USA
| | - Marco Avila-Rodriguez
- Health Sciences Faculty, Clinic Sciences Department, University of Tolima, 730006 Ibague, Colombia
| | - Gjumrakch Aliev
- Federal State Budgetary Institution «Research Institute of Human Morphology», 3, Tsyurupy Str., Moscow, 117418, Russian Federation; I. M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 8/2 Trubetskaya Str., Moscow, 119991, Russia; Institute of Physiologically Active Compounds, Russian Academy of Sciences, Chernogolovka, 142432, Russia; GALLY International Research Institute, 7733 Louis Pasteur Drive, #330, San Antonio, TX, 78229, USA.
| |
Collapse
|
1058
|
Ibrahim N‘I, Muhammad Ismail Tadj NB, Rahman Sarker MM, Naina Mohamed I. The Potential Mechanisms of the Neuroprotective Actions of Oil Palm Phenolics: Implications for Neurodegenerative Diseases. Molecules 2020; 25:E5159. [PMID: 33167585 PMCID: PMC7664177 DOI: 10.3390/molecules25215159] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/02/2020] [Accepted: 11/02/2020] [Indexed: 12/23/2022] Open
Abstract
Neurodegenerative diseases (ND) can be characterized by degradation and subsequent loss of neurons. ND has been identified as the leading cause of disability-adjusted life years (DALYs) worldwide and is associated with various risk factors such as ageing, certain genetic polymorphisms, inflammation, immune and metabolic conditions that may induce elevated reactive oxygen species (ROS) release and subsequent oxidative stress. Presently, no specific cure or prevention is available for ND patients; the symptoms can be only alleviated via drug treatment or surgery. The existing pharmacological treatments are only available for partial treatment of the symptoms. A natural product known as oil palm phenolics (OPP), which is high in antioxidant, could become a potential supplementary antioxidant for neurodegenerative health. OPP is a water-soluble extract from palm fruit that demonstrated medicinal properties including anti-tumor, anti-diabetic and neuroprotective effects. In this review, OPP was proposed for its neuroprotective effects via several mechanisms including antioxidant and anti-inflammatory properties. Besides, OPP has been found to modulate the genes involved in neurotrophic activity. The evidence and proposed mechanism of OPP on the neuroprotective health may provide a comprehensive natural medicine approach to alleviate the symptoms of neurodegenerative diseases.
Collapse
Affiliation(s)
- Nurul ‘Izzah Ibrahim
- Pharmacoepidemiology and Drug Safety Unit, Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia; (N.I.‘I.); (N.B.M.I.T.)
| | - Nur Balqis Muhammad Ismail Tadj
- Pharmacoepidemiology and Drug Safety Unit, Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia; (N.I.‘I.); (N.B.M.I.T.)
| | - Md. Moklesur Rahman Sarker
- Department of Pharmacy, State University of Bangladesh, 77 Satmasjid Road, Dhanmondi, Dhaka 1205, Bangladesh;
| | - Isa Naina Mohamed
- Pharmacoepidemiology and Drug Safety Unit, Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia; (N.I.‘I.); (N.B.M.I.T.)
| |
Collapse
|
1059
|
Yang AJT, Frendo-Cumbo S, MacPherson REK. Resveratrol and Metformin Recover Prefrontal Cortex AMPK Activation in Diet-Induced Obese Mice but Reduce BDNF and Synaptophysin Protein Content. J Alzheimers Dis 2020; 71:945-956. [PMID: 31450493 DOI: 10.3233/jad-190123] [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] [Indexed: 12/25/2022]
Abstract
BACKGROUND Obesity, insulin resistance, and type 2 diabetes are established risk factors for the development of Alzheimer's disease (AD). Given this connection, two drugs, metformin (MET) and resveratrol (RESV), are considered for the clearance of amyloid-β peptides through AMPK-mediated activation of autophagy. However, overactivation of AMPK observed in late-stage AD brains and relationships between AMPK and neurogenesis (through mTORC1 inhibition), questions treatment with these drugs. OBJECTIVE To examine if MET and/or RESV supplementation activates brain AMPK, regulates markers of autophagy, and affects markers of neuronal health/neurogenesis. METHODS 8-week-old male C57BL/6J mice were fed a low (N = 12; 10% kcal fat; LFD) or high fat diet (N = 40; 60% kcal fat; HFD) for 9 weeks to induce insulin resistance and obesity. HFD mice were then treated with/without MET (250 mg/kg/day), RESV (100 mg/kg/day), or COMBO (MET: 250 mg/kg/day, RESV: 100 mg/kg/day) for 5 weeks. Hippocampus and prefrontal cortex were extracted for western blotting analysis. RESULTS Cortex AMPK (T172) and raptor (S792, the regulatory subunit of mTORC1) phosphorylation were upregulated following RESV, COMBO treatments. mTOR (S2448) and ULK1 (S555) activation was seen following MET, COMBO and RESV, COMBO treatments, respectively, in the cortex and hippocampus. p62 content was decreased following RESV, COMBO, with LC3 content being increased following RESV treatment in the cortex. Brain derived neurotropic factor (BDNF) was significantly decreased following RESV, COMBO, and synaptophysin following all treatment in the cortex. CONCLUSION These results demonstrate that while treatments upregulated markers of autophagy in the prefrontal cortex, reductions in neuronal health markers question the efficacy of AMPK as a therapy for AD.
Collapse
Affiliation(s)
- Alex J T Yang
- Department of Health Sciences, Brock University, St. Catharines, ON, Canada
| | - Scott Frendo-Cumbo
- Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada.,Department of Physiology, University of Toronto, Toronto, ON, Canada
| | | |
Collapse
|
1060
|
Single-molecule studies of amyloid proteins: from biophysical properties to diagnostic perspectives. Q Rev Biophys 2020; 53:e12. [PMID: 33148356 DOI: 10.1017/s0033583520000086] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In neurodegenerative diseases, a wide range of amyloid proteins or peptides such as amyloid-beta and α-synuclein fail to keep native functional conformations, followed by misfolding and self-assembling into a diverse array of aggregates. The aggregates further exert toxicity leading to the dysfunction, degeneration and loss of cells in the affected organs. Due to the disordered structure of the amyloid proteins, endogenous molecules, such as lipids, are prone to interact with amyloid proteins at a low concentration and influence amyloid cytotoxicity. The heterogeneity of amyloid proteinscomplicates the understanding of the amyloid cytotoxicity when relying only on conventional bulk and ensemble techniques. As complementary tools, single-molecule techniques (SMTs) provide novel insights into the different subpopulations of a heterogeneous amyloid mixture as well as the cytotoxicity, in particular as involved in lipid membranes. This review focuses on the recent advances of a series of SMTs, including single-molecule fluorescence imaging, single-molecule force spectroscopy and single-nanopore electrical recording, for the understanding of the amyloid molecular mechanism. The working principles, benefits and limitations of each technique are discussed and compared in amyloid protein related studies.. We also discuss why SMTs show great potential and are worthy of further investigation with feasibility studies as diagnostic tools of neurodegenerative diseases and which limitations are to be addressed.
Collapse
|
1061
|
Fulopova B, Stuart KE, Bennett W, Bindoff A, King AE, Vickers JC, Canty AJ. Regional differences in beta amyloid plaque deposition and variable response to midlife environmental enrichment in the cortex of APP/PS1 mice. J Comp Neurol 2020; 529:1849-1862. [PMID: 33104234 DOI: 10.1002/cne.25060] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 01/21/2023]
Abstract
Environmentally enriched housing conditions can increase performance on cognitive tasks in APP/PS1 mice; however, the potential effects of environmental enrichment (EE) on disease modification in terms of pathological change are inconclusive. We hypothesized that previous contrasting findings may be attributable to regional differences in susceptibility to amyloid beta (Aβ) plaque deposition in cortical regions that are functionally associated with EE. We characterized fibrillar plaque deposition in 6, 12, and 18-22 months old APP/PS1 mice in the prefrontal (PFC), somatosensory (SS2), and primary motor cortex (M1). We found a significant increase in plaque load between 6 and 12 months in all regions. In animals over 12 months, only the PFC region continued to significantly accumulate plaques. Additionally, 12 months old animals subjected to 6 months of EE showed improved spatial navigation and had significantly fewer plaques in M1 and SS2, but not in the PFC. These findings suggest that the PFC region is selectively susceptible to Aβ deposition and less responsive to the attenuating effects of EE. In contrast, M1 and SS2 regions plateau with respect to Aβ deposition by 12 months of age and are susceptible to amyloid pathology modification by midlife EE.
Collapse
Affiliation(s)
- Barbora Fulopova
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| | - Kimberley E Stuart
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| | - William Bennett
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| | - Aidan Bindoff
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| | - Anna E King
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| | - James C Vickers
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| | - Alison J Canty
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| |
Collapse
|
1062
|
Zhang H, Karisetty BC, Bhatnagar A, Armour EM, Beaver M, Roach TV, Mortazavi S, Mandloi S, Elefant F. Tip60 protects against amyloid-β-induced transcriptomic alterations via different modes of action in early versus late stages of neurodegeneration. Mol Cell Neurosci 2020; 109:103570. [PMID: 33160016 DOI: 10.1016/j.mcn.2020.103570] [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: 08/08/2020] [Revised: 10/24/2020] [Accepted: 10/31/2020] [Indexed: 10/23/2022] Open
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disorder hallmarked by amyloid-β (Aβ) plaque accumulation, neuronal cell death, and cognitive deficits that worsen during disease progression. Histone acetylation dysregulation, caused by an imbalance between reduced histone acetyltransferases (HAT) Tip60 and increased histone deacetylase 2 (HDAC2) levels, can directly contribute to AD pathology. However, whether such AD-associated neuroepigenetic alterations occur in response to Aβ peptide production and can be protected against by increasing Tip60 levels over the course of neurodegenerative progression remains unknown. Here we profile Tip60 HAT/HDAC2 dynamics and transcriptome-wide changes across early and late stage AD pathology in the Drosophila brain produced solely by human amyloid-β42. We show that early Aβ42 induction leads to disruption of Tip60 HAT/HDAC2 balance during early neurodegenerative stages preceding Aβ plaque accumulation that persists into late AD stages. Correlative transcriptome-wide studies reveal alterations in biological processes we classified as transient (early-stage only), late-onset (late-stage only), and constant (both). Increasing Tip60 HAT levels in the Aβ42 fly brain protects against AD functional pathologies that include Aβ plaque accumulation, neural cell death, cognitive deficits, and shorter life-span. Strikingly, Tip60 protects against Aβ42-induced transcriptomic alterations via distinct mechanisms during early and late stages of neurodegeneration. Our findings reveal distinct modes of neuroepigenetic gene changes and Tip60 neuroprotection in early versus late stages in AD that can serve as early biomarkers for AD, and support the therapeutic potential of Tip60 over the course of AD progression.
Collapse
Affiliation(s)
- Haolin Zhang
- Department of Biology, Drexel University, Philadelphia, PA, United States
| | | | - Akanksha Bhatnagar
- Department of Biology, Drexel University, Philadelphia, PA, United States
| | - Ellen M Armour
- Department of Biology, Drexel University, Philadelphia, PA, United States
| | - Mariah Beaver
- Department of Biology, Drexel University, Philadelphia, PA, United States
| | - Tiffany V Roach
- Department of Biology, Drexel University, Philadelphia, PA, United States
| | - Sina Mortazavi
- Department of Biology, Drexel University, Philadelphia, PA, United States
| | - Shreya Mandloi
- Department of Biology, Drexel University, Philadelphia, PA, United States
| | - Felice Elefant
- Department of Biology, Drexel University, Philadelphia, PA, United States.
| |
Collapse
|
1063
|
Foster CM, Kennedy KM, Daugherty AM, Rodrigue KM. Contribution of iron and Aβ to age differences in entorhinal and hippocampal subfield volume. Neurology 2020; 95:e2586-e2594. [PMID: 32938781 PMCID: PMC7682827 DOI: 10.1212/wnl.0000000000010868] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Accepted: 06/26/2020] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE To test the hypothesis that the combination of elevated global β-AMYLOID (Aβ) burden and greater striatal iron content would be associated with smaller entorhinal cortex (ERC) volume, but not hippocampal subfield volumes, we measured volume and iron content using high-resolution MRI and Aβ using PET imaging in a cross-sectional sample of 70 cognitively normal older adults. METHODS Participants were scanned with florbetapir 18F PET to obtain Aβ standardized uptake value ratios. Susceptibility-weighted MRI was collected and processed to yield R2* images, and striatal regions of interest (ROIs) were manually placed to obtain a measure of striatal iron burden. Ultra-high resolution T2/PD-weighted MRIs were segmented to measure medial temporal lobe (MTL) volumes. Analyses were conducted using mixed-effects models with MTL ROI as a within-participant factor; age, iron content, and Aβ as between-participant factors; and MTL volumes (ERC and 3 hippocampal subfield regions) as the dependent variable. RESULTS The model indicated a significant 4-way interaction among age, iron, Aβ, and MTL region. Post hoc analyses indicated that the 3-way interaction among age, Aβ, and iron content was selective to the ERC (β = -3.34, standard error = 1.33, 95% confidence interval -5.95 to -0.72), whereas a significant negative association between age and ERC volume was present only in individuals with both elevated iron content and Aβ. CONCLUSIONS These findings highlight the importance of studying Aβ in the context of other, potentially synergistic age-related brain factors such as iron accumulation and the potential role for iron as an important contributor to the earliest, preclinical stages of pathologic aging.
Collapse
Affiliation(s)
- Chris M Foster
- From the School of Behavioral and Brain Sciences (C.M.F., K.M.K., K.M.R.), Center for Vital Longevity, University of Texas at Dallas; and Department of Psychology (A.M.D.) and Department of Psychiatry and Behavioral Neurosciences, Institute of Gerontology, Wayne State University, Detroit, MI
| | - Kristen M Kennedy
- From the School of Behavioral and Brain Sciences (C.M.F., K.M.K., K.M.R.), Center for Vital Longevity, University of Texas at Dallas; and Department of Psychology (A.M.D.) and Department of Psychiatry and Behavioral Neurosciences, Institute of Gerontology, Wayne State University, Detroit, MI
| | - Ana M Daugherty
- From the School of Behavioral and Brain Sciences (C.M.F., K.M.K., K.M.R.), Center for Vital Longevity, University of Texas at Dallas; and Department of Psychology (A.M.D.) and Department of Psychiatry and Behavioral Neurosciences, Institute of Gerontology, Wayne State University, Detroit, MI
| | - Karen M Rodrigue
- From the School of Behavioral and Brain Sciences (C.M.F., K.M.K., K.M.R.), Center for Vital Longevity, University of Texas at Dallas; and Department of Psychology (A.M.D.) and Department of Psychiatry and Behavioral Neurosciences, Institute of Gerontology, Wayne State University, Detroit, MI.
| |
Collapse
|
1064
|
Lauer AA, Mett J, Janitschke D, Thiel A, Stahlmann CP, Bachmann CM, Ritzmann F, Schrul B, Müller UC, Stein R, Riemenschneider M, Grimm HS, Hartmann T, Grimm MOW. Regulatory feedback cycle of the insulin-degrading enzyme and the amyloid precursor protein intracellular domain: Implications for Alzheimer's disease. Aging Cell 2020; 19:e13264. [PMID: 33128835 PMCID: PMC7681056 DOI: 10.1111/acel.13264] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 09/16/2020] [Accepted: 10/02/2020] [Indexed: 12/04/2022] Open
Abstract
One of the major pathological hallmarks of Alzheimer´s disease (AD) is an accumulation of amyloid‐β (Aβ) in brain tissue leading to formation of toxic oligomers and senile plaques. Under physiological conditions, a tightly balanced equilibrium between Aβ‐production and ‐degradation is necessary to prevent pathological Aβ‐accumulation. Here, we investigate the molecular mechanism how insulin‐degrading enzyme (IDE), one of the major Aβ‐degrading enzymes, is regulated and how amyloid precursor protein (APP) processing and Aβ‐degradation is linked in a regulatory cycle to achieve this balance. In absence of Aβ‐production caused by APP or Presenilin deficiency, IDE‐mediated Aβ‐degradation was decreased, accompanied by a decreased IDE activity, protein level, and expression. Similar results were obtained in cells only expressing a truncated APP, lacking the APP intracellular domain (AICD) suggesting that AICD promotes IDE expression. In return, APP overexpression mediated an increased IDE expression, comparable results were obtained with cells overexpressing C50, a truncated APP representing AICD. Beside these genetic approaches, also AICD peptide incubation and pharmacological inhibition of the γ‐secretase preventing AICD production regulated IDE expression and promoter activity. By utilizing CRISPR/Cas9 APP and Presenilin knockout SH‐SY5Y cells results were confirmed in a second cell line in addition to mouse embryonic fibroblasts. In vivo, IDE expression was decreased in mouse brains devoid of APP or AICD, which was in line with a significant correlation of APP expression level and IDE expression in human postmortem AD brains. Our results show a tight link between Aβ‐production and Aβ‐degradation forming a regulatory cycle in which AICD promotes Aβ‐degradation via IDE and IDE itself limits its own production by degrading AICD.
Collapse
Affiliation(s)
- Anna A. Lauer
- Experimental Neurology Saarland University Homburg/Saar Germany
| | - Janine Mett
- Experimental Neurology Saarland University Homburg/Saar Germany
- Biosciences Zoology/Physiology‐Neurobiology Faculty NT‐Natural Science and Technology Saarland University Saarbrücken Germany
| | | | - Andrea Thiel
- Experimental Neurology Saarland University Homburg/Saar Germany
| | | | | | - Felix Ritzmann
- Department of Internal Medicine V – Pulmonology Allergology, Respiratory Intensive Care Medicine Saarland University Hospital Homburg/Saar Germany
| | - Bianca Schrul
- Medical Biochemistry and Molecular Biology Center for Molecular Signaling (PZMS) Faculty of Medicine Saarland University Homburg/Saar Germany
| | - Ulrike C. Müller
- Department of Functional Genomics Institute of Pharmacy and Molecular Biotechnology Heidelberg University Germany
| | - Reuven Stein
- Department of Neurology George S. Wise Faculty of Life Sciences Tel Aviv University Ramat Aviv Israel
| | | | - Heike S. Grimm
- Experimental Neurology Saarland University Homburg/Saar Germany
| | - Tobias Hartmann
- Experimental Neurology Saarland University Homburg/Saar Germany
- Deutsches Institut für DemenzPrävention (DIDP) Saarland University Homburg/Saar Germany
| | - Marcus O. W. Grimm
- Experimental Neurology Saarland University Homburg/Saar Germany
- Deutsches Institut für DemenzPrävention (DIDP) Saarland University Homburg/Saar Germany
| |
Collapse
|
1065
|
Seol W, Kim H, Son I. Urinary Biomarkers for Neurodegenerative Diseases. Exp Neurobiol 2020; 29:325-333. [PMID: 33154195 PMCID: PMC7649089 DOI: 10.5607/en20042] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 10/12/2020] [Accepted: 10/19/2020] [Indexed: 12/13/2022] Open
Abstract
Global incidence of neurodegenerative diseases (NDDs) such as Alzheimer's disease (AD) and Parkinson's disease (PD) is rapidly increasing, but the diagnosis of these diseases at their early stage is challenging. Therefore, the availability of reproducible and reliable biomarkers to diagnose such diseases is more critical than ever. In addition, biomarkers could be used not only to diagnose diseases but also to monitor the development of disease therapeutics. Urine is an excellent biofluid that can be utilized as a source of biomarker to diagnose not only several renal diseases but also other diseases because of its abundance in invasive sampling. However, urine was conventionally regarded as inappropriate as a source of biomarker for neurodegenerative diseases because it is anatomically distant from the central nervous system (CNS), a major pathologic site of NDD, in comparison to other biofluids such as cerebrospinal fluid (CSF) and plasma. However, recent studies have suggested that urine could be utilized as a source of NDD biomarker if an appropriate marker is predetermined by metabolomic and proteomic approaches in urine and other samples. In this review, we summarize such studies related to NDD.
Collapse
Affiliation(s)
- Wongi Seol
- InAm Neuroscience Research Center, Gunpo 15865, Korea
| | - Hyejung Kim
- InAm Neuroscience Research Center, Gunpo 15865, Korea
| | - Ilhong Son
- InAm Neuroscience Research Center, Gunpo 15865, Korea
- Department of Neurology, Sanbon Medical Center, College of Medicine, Wonkwang University, Gunpo 15865, Korea
| |
Collapse
|
1066
|
Camarillo-López RH, Hernández Rodríguez M, Torres-Ramos MA, Arciniega-Martínez IM, García-Marín ID, Correa Basurto J, Méndez Méndez JV, Rosales-Hernández MC. Tert-butyl-(4-hydroxy-3-((3-(2-methylpiperidin-yl)propyl)carbamoyl)phenyl)carbamate Has Moderated Protective Activity in Astrocytes Stimulated with Amyloid Beta 1-42 and in a Scopolamine Model. Molecules 2020; 25:molecules25215009. [PMID: 33137907 PMCID: PMC7672627 DOI: 10.3390/molecules25215009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 10/21/2020] [Accepted: 10/22/2020] [Indexed: 12/17/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease with no cure nowadays; there is no treatment either to prevent or to stop its progression. In vitro studies suggested that tert-butyl-(4-hydroxy-3-((3-(2-methylpiperidin-yl)propyl)carbamoyl)phenyl) carbamate named the M4 compound can act as both β-secretase and an acetylcholinesterase inhibitor, preventing the amyloid beta peptide (Aβ) aggregation and the formation of fibrils (fAβ) from Aβ1-42. This work first aimed to assess in in vitro studies to see whether the death of astrocyte cells promoted by Aβ1-42 could be prevented. Second, our work investigated the ability of the M4 compound to inhibit amyloidogenesis using an in vivo model after scopolamine administration. The results showed that M4 possesses a moderate protective effect in astrocytes against Aβ1-42 due to a reduction in the TNF-α and free radicals observed in cell cultures. In the in vivo studies, however, no significant effect of M4 was observed in comparison with a galantamine model employed in rats, in which case this outcome was attributed to the bioavailability of M4 in the brain of the rats.
Collapse
Affiliation(s)
- Raúl Horacio Camarillo-López
- Laboratorio de Biofísica y biocatálisis, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Diaz Mirón s/n, 11340 Ciudad de México, Mexico; (R.H.C.-L.); (M.H.R.); (I.D.G.-M.)
| | - Maricarmen Hernández Rodríguez
- Laboratorio de Biofísica y biocatálisis, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Diaz Mirón s/n, 11340 Ciudad de México, Mexico; (R.H.C.-L.); (M.H.R.); (I.D.G.-M.)
| | - Mónica Adriana Torres-Ramos
- Unidad Periférica de Neurociencias, Facultad de Medicina UNAM-Instituto Nacional de Neurología y Neurocirugía, MVS-SSA, Insurgentes sur 3877, La Fama, Tlalpan, 14269 Ciudad de México, Mexico;
| | - Ivonne Maciel Arciniega-Martínez
- Laboratorio de Inmunidad de Mucosas, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, 11340 Ciudad de México, Mexico;
| | - Iohanan Daniel García-Marín
- Laboratorio de Biofísica y biocatálisis, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Diaz Mirón s/n, 11340 Ciudad de México, Mexico; (R.H.C.-L.); (M.H.R.); (I.D.G.-M.)
| | - José Correa Basurto
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotecnológica, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón, 11340 Ciudad de México, Mexico;
| | - Juan Vicente Méndez Méndez
- Centro de Nanociencias y Micro y Nanotecnologías, Instituto Politécnico Nacional. Av. Luis Enrique Erro s/n, Nueva Industrial Vallejo, Gustavo A. Madero, 07738 Ciudad de México, Mexico;
| | - Martha Cecilia Rosales-Hernández
- Laboratorio de Biofísica y biocatálisis, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Diaz Mirón s/n, 11340 Ciudad de México, Mexico; (R.H.C.-L.); (M.H.R.); (I.D.G.-M.)
- Correspondence:
| |
Collapse
|
1067
|
Verheggen ICM, de Jong JJA, van Boxtel MPJ, Postma AA, Verhey FRJ, Jansen JFA, Backes WH. Permeability of the windows of the brain: feasibility of dynamic contrast-enhanced MRI of the circumventricular organs. Fluids Barriers CNS 2020; 17:66. [PMID: 33115484 PMCID: PMC7594295 DOI: 10.1186/s12987-020-00228-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 10/17/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Circumventricular organs (CVOs) are small structures without a blood-brain barrier surrounding the brain ventricles that serve homeostasic functions and facilitate communication between the blood, cerebrospinal fluid and brain. Secretory CVOs release peptides and sensory CVOs regulate signal transmission. However, pathogens may enter the brain through the CVOs and trigger neuroinflammation and neurodegeneration. We investigated the feasibility of dynamic contrast-enhanced (DCE) MRI to assess the CVO permeability characteristics in vivo, and expected significant contrast uptake in these regions, due to blood-brain barrier absence. METHODS Twenty healthy, middle-aged to older males underwent brain DCE MRI. Pharmacokinetic modeling was applied to contrast concentration time-courses of CVOs, and in reference to white and gray matter. We investigated whether a significant and positive transfer from blood to brain could be measured in the CVOs, and whether this differed between secretory and sensory CVOs or from normal-appearing brain matter. RESULTS In both the secretory and sensory CVOs, the transfer constants were significantly positive, and all secretory CVOs had significantly higher transfer than each sensory CVO. The transfer constants in both the secretory and sensory CVOs were higher than in the white and gray matter. CONCLUSIONS Current measurements confirm the often-held assumption of highly permeable CVOs, of which the secretory types have the strongest blood-to-brain transfer. The current study suggests that DCE MRI could be a promising technique to further assess the function of the CVOs and how pathogens can potentially enter the brain via these structures. TRIAL REGISTRATION Netherlands Trial Register number: NL6358, date of registration: 2017-03-24.
Collapse
Affiliation(s)
- Inge C M Verheggen
- Department of Psychiatry and Neuropsychology, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands.
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands.
- Alzheimer Center Limburg, Maastricht, The Netherlands.
| | - Joost J A de Jong
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Martin P J van Boxtel
- Department of Psychiatry and Neuropsychology, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
- Alzheimer Center Limburg, Maastricht, The Netherlands
| | - Alida A Postma
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Frans R J Verhey
- Department of Psychiatry and Neuropsychology, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
- Alzheimer Center Limburg, Maastricht, The Netherlands
| | - Jacobus F A Jansen
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Walter H Backes
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- School for Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
1068
|
Pietroboni AM, Colombi A, Carandini T, Scarpini E, Galimberti D, Bozzali M. The Role of Amyloid-β in White Matter Damage: Possible Common Pathogenetic Mechanisms in Neurodegenerative and Demyelinating Diseases. J Alzheimers Dis 2020; 78:13-22. [PMID: 32925075 DOI: 10.3233/jad-200868] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Just as multiple sclerosis (MS) has long been primarily considered a white matter (WM) disease, Alzheimer's disease (AD) has for decades been regarded only as a grey matter disorder. However, convergent evidences have suggested that WM abnormalities are also important components of AD, at the same extent as axonal and neuronal loss is critically involved in MS pathophysiology since early clinical stages. These observations have motivated a more thorough investigation about the possible mechanisms that could link neuroinflammation and neurodegeneration, focusing on amyloid-β (Aβ). Neuroimaging studies have found that patients with AD have widespread WM abnormalities already at the earliest disease stages and prior to the presence of Aβ plaques. Moreover, a correlation between cerebrospinal fluid (CSF) Aβ levels and WM lesion load was found. On the other hand, recent studies suggest a predictive role for CSF Aβ levels in MS, possibly due in the first instance to the reduced capacity for remyelination, consequently to a higher risk of WM damage progression, and ultimately to neuronal loss. We undertook a review of the recent findings concerning the involvement of CSF Aβ levels in the MS disease course and of the latest evidence of AD related WM abnormalities, with the aim to discuss the potential causes that may connect WM damage and amyloid pathology.
Collapse
Affiliation(s)
- Anna M Pietroboni
- Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy.,University of Milan, Dino Ferrari Centre, Milan, Italy
| | | | - Tiziana Carandini
- Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy.,University of Milan, Dino Ferrari Centre, Milan, Italy
| | - Elio Scarpini
- Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy.,University of Milan, Dino Ferrari Centre, Milan, Italy
| | - Daniela Galimberti
- Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy.,University of Milan, Dino Ferrari Centre, Milan, Italy
| | - Marco Bozzali
- Department of Neuroscience 'Rita Levi Montalcini', University of Torino, Turin, Italy.,Department of Neuroscience, Brighton and Sussex Medical School, University of Sussex, Brighton, UK
| |
Collapse
|
1069
|
Atlante A, Amadoro G, Bobba A, Latina V. Functional Foods: An Approach to Modulate Molecular Mechanisms of Alzheimer's Disease. Cells 2020; 9:E2347. [PMID: 33114170 PMCID: PMC7690784 DOI: 10.3390/cells9112347] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/15/2020] [Accepted: 10/21/2020] [Indexed: 12/13/2022] Open
Abstract
A new epoch is emerging with intense research on nutraceuticals, i.e., "food or food product that provides medical or health benefits including the prevention and treatment of diseases", such as Alzheimer's disease. Nutraceuticals act at different biochemical and metabolic levels and much evidence shows their neuroprotective effects; in particular, they are able to provide protection against mitochondrial damage, oxidative stress, toxicity of β-amyloid and Tau and cell death. They have been shown to influence the composition of the intestinal microbiota significantly contributing to the discovery that differential microorganisms composition is associated with the formation and aggregation of cerebral toxic proteins. Further, the routes of interaction between epigenetic mechanisms and the microbiota-gut-brain axis have been elucidated, thus establishing a modulatory role of diet-induced epigenetic changes of gut microbiota in shaping the brain. This review examines recent scientific literature addressing the beneficial effects of some natural products for which mechanistic evidence to prevent or slowdown AD are available. Even if the road is still long, the results are already exceptional.
Collapse
Affiliation(s)
- Anna Atlante
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM)-CNR, Via G. Amendola 122/O, 70126 Bari, Italy;
| | - Giuseppina Amadoro
- Institute of Translational Pharmacology (IFT)-CNR, Via Fosso del Cavaliere 100, 00133 Rome, Italy;
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy;
| | - Antonella Bobba
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM)-CNR, Via G. Amendola 122/O, 70126 Bari, Italy;
| | - Valentina Latina
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy;
| |
Collapse
|
1070
|
Rivera-Marrero S, Bencomo-Martínez A, Orta Salazar E, Sablón-Carrazana M, García-Pupo L, Zoppolo F, Arredondo F, Dapueto R, Daniela Santi M, Kreimerman I, Pardo T, Reyes L, Galán L, León-Chaviano S, Espinosa-Rodríguez LA, Menéndez-Soto Del Valle R, Savio E, Díaz Cintra S, Rodríguez-Tanty C. A new naphthalene derivative with anti-amyloidogenic activity as potential therapeutic agent for Alzheimer's disease. Bioorg Med Chem 2020; 28:115700. [PMID: 33069076 DOI: 10.1016/j.bmc.2020.115700] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 08/04/2020] [Indexed: 11/24/2022]
Abstract
The aggregation of β-amyloid peptides is associated to neurodegeneration in Alzheimer's disease (AD) patients. Consequently, the inhibition of both oligomerization and fibrillation of β-amyloid peptides is considered a plausible therapeutic approach for AD. Herein, the synthesis of new naphthalene derivatives and their evaluation as anti-β-amyloidogenic agents are presented. Molecular dynamic simulations predicted the formation of thermodynamically stable complexes between the compounds, the Aβ1-42 peptide and fibrils. In human microglia cells, these compounds inhibited the aggregation of Aβ1-42 peptide. The lead compound 8 showed a high affinity to amyloid plaques in mice brain ex vivo assays and an adequate log Poct/PBS value. Compound 8 also improved the cognitive function and decreased hippocampal β-amyloid burden in the brain of 3xTg-AD female mice. Altogether, our results suggest that 8 could be a novel therapeutic agent for AD.
Collapse
Affiliation(s)
- Suchitil Rivera-Marrero
- Department of Neurochemistry, Cuban Center for Neurosciences, Street. 190 e/ 25 and 27, Cubanacan, Playa, Havana, CP 11600, Cuba
| | - Alberto Bencomo-Martínez
- Department of Neurochemistry, Cuban Center for Neurosciences, Street. 190 e/ 25 and 27, Cubanacan, Playa, Havana, CP 11600, Cuba
| | - Erika Orta Salazar
- Institute of Neurobiology (INB), Developmental Neurobiology and Neurophysiology, UNAM Juriquilla Querétaro, Mexico
| | - Marquiza Sablón-Carrazana
- Department of Neurochemistry, Cuban Center for Neurosciences, Street. 190 e/ 25 and 27, Cubanacan, Playa, Havana, CP 11600, Cuba
| | - Laura García-Pupo
- Department of Neurochemistry, Cuban Center for Neurosciences, Street. 190 e/ 25 and 27, Cubanacan, Playa, Havana, CP 11600, Cuba
| | - Florencia Zoppolo
- Biomedical and Pharmaceutical Chemistry, Uruguayan Centre of Molecular Imaging (CUDIM), Montevideo, Uruguay
| | - Florencia Arredondo
- Biomedical and Pharmaceutical Chemistry, Uruguayan Centre of Molecular Imaging (CUDIM), Montevideo, Uruguay
| | - Rosina Dapueto
- Biomedical and Pharmaceutical Chemistry, Uruguayan Centre of Molecular Imaging (CUDIM), Montevideo, Uruguay
| | - María Daniela Santi
- Biomedical and Pharmaceutical Chemistry, Uruguayan Centre of Molecular Imaging (CUDIM), Montevideo, Uruguay
| | - Ingrid Kreimerman
- Biomedical and Pharmaceutical Chemistry, Uruguayan Centre of Molecular Imaging (CUDIM), Montevideo, Uruguay
| | - Tania Pardo
- Biomedical and Pharmaceutical Chemistry, Uruguayan Centre of Molecular Imaging (CUDIM), Montevideo, Uruguay
| | - Laura Reyes
- Biomedical and Pharmaceutical Chemistry, Uruguayan Centre of Molecular Imaging (CUDIM), Montevideo, Uruguay
| | - Lídice Galán
- Department of Neurochemistry, Cuban Center for Neurosciences, Street. 190 e/ 25 and 27, Cubanacan, Playa, Havana, CP 11600, Cuba
| | - Samila León-Chaviano
- Department of Neurochemistry, Cuban Center for Neurosciences, Street. 190 e/ 25 and 27, Cubanacan, Playa, Havana, CP 11600, Cuba
| | - Luis A Espinosa-Rodríguez
- Center of Genetic Engineering and Biotechnology (CIGB), Ave 31 e/ 158 and 190, Havana, CP10600, Cuba
| | - Roberto Menéndez-Soto Del Valle
- Department of Neurochemistry, Cuban Center for Neurosciences, Street. 190 e/ 25 and 27, Cubanacan, Playa, Havana, CP 11600, Cuba
| | - Eduardo Savio
- Biomedical and Pharmaceutical Chemistry, Uruguayan Centre of Molecular Imaging (CUDIM), Montevideo, Uruguay
| | - Sofía Díaz Cintra
- Institute of Neurobiology (INB), Developmental Neurobiology and Neurophysiology, UNAM Juriquilla Querétaro, Mexico.
| | - Chryslaine Rodríguez-Tanty
- Department of Neurochemistry, Cuban Center for Neurosciences, Street. 190 e/ 25 and 27, Cubanacan, Playa, Havana, CP 11600, Cuba.
| |
Collapse
|
1071
|
Blank M, Hopf C. Spatially resolved mass spectrometry analysis of amyloid plaque-associated lipids. J Neurochem 2020; 159:330-342. [PMID: 33048341 DOI: 10.1111/jnc.15216] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/06/2020] [Accepted: 10/04/2020] [Indexed: 12/18/2022]
Abstract
Over the last 10 years, considerable technical advances in mass spectrometry (MS)-based bioanalysis have enabled the investigation of lipid signatures in neuropathological structures. In Alzheimer´s Disease (AD) research, it is now well accepted that lipid dysregulation plays a key role in AD pathogenesis and progression. This review summarizes current MS-based strategies, notably MALDI and ToF-SIMS imaging as well as laser capture microdissection combined with LC-ESI-MS. It also presents recent advances to assess lipid alterations associated with Amyloid-β plaques, one of the hallmarks of AD. Collectively, these methodologies offer new opportunities for the study of lipids, thus pushing forward our understanding of their role in such a complex and still untreatable disease as AD.
Collapse
Affiliation(s)
- Martina Blank
- Center for Mass Spectrometry and Optical Spectroscopy (CeMOS), Mannheim University of Applied Sciences, Mannheim, Germany.,Center for Structural Molecular Biology (CEBIME/PROPESQ), Federal University of Santa Catarina, Florianópolis, Brazil
| | - Carsten Hopf
- Center for Mass Spectrometry and Optical Spectroscopy (CeMOS), Mannheim University of Applied Sciences, Mannheim, Germany
| |
Collapse
|
1072
|
Bukke VN, Archana M, Villani R, Romano AD, Wawrzyniak A, Balawender K, Orkisz S, Beggiato S, Serviddio G, Cassano T. The Dual Role of Glutamatergic Neurotransmission in Alzheimer's Disease: From Pathophysiology to Pharmacotherapy. Int J Mol Sci 2020; 21:ijms21207452. [PMID: 33050345 PMCID: PMC7589203 DOI: 10.3390/ijms21207452] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/28/2020] [Accepted: 10/05/2020] [Indexed: 12/19/2022] Open
Abstract
Alzheimer's disease (AD) is an age-related dementia and neurodegenerative disorder, characterized by Aβ and tau protein deposition impairing learning, memory and suppressing synaptic plasticity of neurons. Increasing evidence suggests that there is a link between the glucose and glutamate alterations with age that down-regulates glucose utilization reducing glutamate levels in AD patients. Deviations in brain energy metabolism reinforce the development of AD by hampering glutamate levels in the brain. Glutamate is a nonessential amino acid and the major excitatory neurotransmitter synthesized from glucose. Alterations in cerebral glucose and glutamate levels precede the deposition of Aβ plaques. In the brain, over 40% of neuronal synapses are glutamatergic and disturbances in glutamatergic function have been implicated in pathophysiology of AD. Nevertheless, targeting the glutamatergic system seems to be a promising strategy to develop novel, improved therapeutics for AD. Here, we review data supporting the involvement of the glutamatergic system in AD pathophysiology as well as the efficacy of glutamatergic agents in this neurodegenerative disorder. We also discuss exciting new prospects for the development of improved therapeutics for this devastating disorder.
Collapse
Affiliation(s)
- Vidyasagar Naik Bukke
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy;
| | - Moola Archana
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (M.A.); (R.V.); (A.D.R.); (G.S.)
| | - Rosanna Villani
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (M.A.); (R.V.); (A.D.R.); (G.S.)
| | - Antonino Davide Romano
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (M.A.); (R.V.); (A.D.R.); (G.S.)
| | - Agata Wawrzyniak
- Morphological Science Department of Human Anatomy, Medical Faculty University of Rzeszów, 35-310 Rzeszów, Poland; (A.W.); (K.B.); (S.O.)
| | - Krzysztof Balawender
- Morphological Science Department of Human Anatomy, Medical Faculty University of Rzeszów, 35-310 Rzeszów, Poland; (A.W.); (K.B.); (S.O.)
| | - Stanislaw Orkisz
- Morphological Science Department of Human Anatomy, Medical Faculty University of Rzeszów, 35-310 Rzeszów, Poland; (A.W.); (K.B.); (S.O.)
| | - Sarah Beggiato
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, 66100 Chieti, Italy;
| | - Gaetano Serviddio
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (M.A.); (R.V.); (A.D.R.); (G.S.)
| | - Tommaso Cassano
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy;
- Correspondence:
| |
Collapse
|
1073
|
Bonfili L, Cecarini V, Gogoi O, Gong C, Cuccioloni M, Angeletti M, Rossi G, Eleuteri AM. Microbiota modulation as preventative and therapeutic approach in Alzheimer's disease. FEBS J 2020; 288:2836-2855. [PMID: 32969566 DOI: 10.1111/febs.15571] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 08/27/2020] [Accepted: 09/17/2020] [Indexed: 12/23/2022]
Abstract
The gut microbiota coevolves with its host, and numerous factors like diet, lifestyle, drug intake and geographical location continuously modify its composition, deeply influencing host health. Recent studies demonstrated that gut dysbiosis can alter normal brain function through the so-called gut-brain axis, a bidirectional communication network between the central nervous system and the gastrointestinal tract, thus playing a key role in the pathogenesis of neurodegenerative disorders, such as Alzheimer's disease (AD). In this perspective, in the constant search for novel treatments in AD, the rational modulation of gut microbiota composition could represent a promising approach to prevent or delay AD onset or to counteract its progression. Preclinical and human studies on microbiota modulation through oral bacteriotherapy and faecal transplantation showed anti-inflammatory and antioxidant effects, upregulation of plasma concentration of neuroprotective hormones, restoration of impaired proteolytic pathways, amelioration of energy homeostasis with consequent decrease of AD molecular hallmarks and improvement of behavioural and cognitive performances. In this review, we dissect the role of gut microbiota in AD and highlight recent advances in the development of new multitarget strategies for microbiota modulation to be used as possible preventative and therapeutic approaches in AD.
Collapse
Affiliation(s)
- Laura Bonfili
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Valentina Cecarini
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Olee Gogoi
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Chunmei Gong
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | | | - Mauro Angeletti
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Giacomo Rossi
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Anna Maria Eleuteri
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| |
Collapse
|
1074
|
Sadeghmousavi S, Eskian M, Rahmani F, Rezaei N. The effect of insomnia on development of Alzheimer's disease. J Neuroinflammation 2020; 17:289. [PMID: 33023629 PMCID: PMC7542374 DOI: 10.1186/s12974-020-01960-9] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 09/23/2020] [Indexed: 02/07/2023] Open
Abstract
Alzheimer's disease (AD) is the most common type of dementia and a neurodegenerative disorder characterized by memory deficits especially forgetting recent information, recall ability impairment, and loss of time tracking, problem-solving, language, and recognition difficulties. AD is also a globally important health issue but despite all scientific efforts, the treatment of AD is still a challenge. Sleep has important roles in learning and memory consolidation. Studies have shown that sleep deprivation (SD) and insomnia are associated with the pathogenesis of Alzheimer's disease and may have an impact on the symptoms and development. Thus, sleep disorders have decisive effects on AD; this association deserves more attention in research, diagnostics, and treatment, and knowing this relation also can help to prevent AD through screening and proper management of sleep disorders. This study aimed to show the potential role of SD and insomnia in the pathogenesis and progression of AD.
Collapse
Affiliation(s)
- Shaghayegh Sadeghmousavi
- Neuroimaging Network (NIN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahsa Eskian
- Neuroimaging Network (NIN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Farzaneh Rahmani
- Neuroimaging Network (NIN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Radiology, Washington University in St. Louis, St. Louis, MO, USA
| | - Nima Rezaei
- Neuroimaging Network (NIN), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
1075
|
Romanucci V, García-Viñuales S, Tempra C, Bernini R, Zarrelli A, Lolicato F, Milardi D, Di Fabio G. Modulating Aβ aggregation by tyrosol-based ligands: The crucial role of the catechol moiety. Biophys Chem 2020; 265:106434. [DOI: 10.1016/j.bpc.2020.106434] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 06/30/2020] [Accepted: 07/12/2020] [Indexed: 01/19/2023]
|
1076
|
Pandey G, Ramakrishnan V. Invasive and non-invasive therapies for Alzheimer's disease and other amyloidosis. Biophys Rev 2020; 12:1175-1186. [PMID: 32930962 PMCID: PMC7575678 DOI: 10.1007/s12551-020-00752-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 08/31/2020] [Indexed: 12/12/2022] Open
Abstract
Advancements in medical science have facilitated in extending human lives. The increased life expectancy, though, has come at a cost. The cases of an aging population suffering from degenerative diseases like Alzheimer's disease (AD) are presently at its all-time high. Amyloidosis disorders such as AD are triggered by an abnormal transition of soluble proteins into their highly ordered aggregated forms. The landscape of amyloidosis treatment remains unchanged, and there is no cure for such disorders. However, an increased understanding of the mechanism of amyloid self-assembly has given hope for a possible therapeutic solution. In this review, we will discuss the current state of molecular and non-molecular options for therapeutic intervention of amyloidosis. We highlight the efficacy of non-invasive physical therapies as possible alternatives to their molecular counterparts. Graphical abstract.
Collapse
Affiliation(s)
- Gaurav Pandey
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, India
| | - Vibin Ramakrishnan
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, India.
| |
Collapse
|
1077
|
Jaunmuktane Z, Brandner S. Invited Review: The role of prion-like mechanisms in neurodegenerative diseases. Neuropathol Appl Neurobiol 2020; 46:522-545. [PMID: 31868945 PMCID: PMC7687189 DOI: 10.1111/nan.12592] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 11/30/2019] [Accepted: 12/17/2019] [Indexed: 12/12/2022]
Abstract
The prototype of transmissible neurodegenerative proteinopathies is prion diseases, characterized by aggregation of abnormally folded conformers of the native prion protein. A wealth of mechanisms has been proposed to explain the conformational conversion from physiological protein into misfolded, pathological form, mode of toxicity, propagation from cell-to-cell and regional spread. There is increasing evidence that other neurodegenerative diseases, most notably Alzheimer's disease (Aβ and tau), Parkinson's disease (α-synuclein), frontotemporal dementia (TDP43, tau or FUS) and motor neurone disease (TDP43), exhibit at least some of the misfolded prion protein properties. In this review, we will discuss to what extent each of the properties of misfolded prion protein is known to occur for Aβ, tau, α-synuclein and TDP43, with particular focus on self-propagation through seeding, conformational strains, selective cellular and regional vulnerability, stability and resistance to inactivation, oligomers, toxicity and summarize the most recent literature on transmissibility of neurodegenerative disorders.
Collapse
Affiliation(s)
- Z. Jaunmuktane
- Division of NeuropathologyNational Hospital for Neurology and NeurosurgeryUniversity College London NHS Foundation Trust
- Department of Clinical and Movement Neurosciences and Queen Square Brain Bank for Neurological Disorders
| | - S. Brandner
- Division of NeuropathologyNational Hospital for Neurology and NeurosurgeryUniversity College London NHS Foundation Trust
- Department of Neurodegenerative diseaseQueen Square Institute of NeurologyUniversity College LondonLondonUK
| |
Collapse
|
1078
|
Rasgado LAV, Urbieta AT, Jiménez JMM. Affected albumin endocytosis as a new neurotoxicity mechanism of amyloid beta. AIMS Neurosci 2020; 7:344-359. [PMID: 32995492 PMCID: PMC7519963 DOI: 10.3934/neuroscience.2020021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 09/17/2020] [Indexed: 11/18/2022] Open
Abstract
Senile plaques, a hallmark of Alzheimer's disease, are composed by Amyloid-Beta (Aβ). Aβ 25-35 toxicity is caused mainly by increasing reactive oxygen species (ROS), which is reversed by albumin preventing Aβ internalization. In addition, key cellular processes and basic cell functions require of endocytosis, particularly relevant in neurons. To understand the protective effect of albumin and the toxicity mechanism of Aβ, the need of albumin uptake for neurons protection as well as the possible influence of Aβ on albumin endocytosis were investigated. With this aim the influence of lectin from soybeans (LEC), which prevents albumin endocytosis, on the effects of Aβ 25-35 on cellular morphology and viability, ROS generation and Aβ uptake with and without albumin in neurons in primary culture was investigated. Influence of Aβ on albumin endocytosis was studied using FITC-labelled albumin. LEC did not modify Aβ effects with or without albumin on neuronal morphology, but increased cell viability. LEC increased ROS generation with and without Aβ in the same magnitude. Diminished Aβ internalization observed with albumin was not affected by LEC. In presence of Aβ albumin is internalized, but endosomes did not deliver their cargo to the lysosomes for degradation. It is concluded that formation of Aβ-albumin complex does not require of albumin internalization, thus is extracellular. Aβ affects albumin endocytosis preventing late endosomes and lysosomes degradation, probably caused by changes in albumin structure or deregulation in vesicular transport. Considering the consequences such as its osmotic effects, the inability to exert its antioxidant properties, its effects on neuronal plasticity and excitability albumin affected endocytosis induced by Aβ is proposed as a new physiopathology mechanism in AD. It is hypothesized that there is critical intraneuronal level above which albumin becomes toxic.
Collapse
Affiliation(s)
- Lourdes A Vega Rasgado
- Laboratorio de Neuroquímica, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, México
| | - Arantxa Tabernero Urbieta
- Instituto de Neurociencias de Castilla y León (INCYL), c/Pintor Fernando Gallego 1, 37007 Salamanca, Spain
| | - José María Medina Jiménez
- Instituto de Neurociencias de Castilla y León (INCYL), c/Pintor Fernando Gallego 1, 37007 Salamanca, Spain
| |
Collapse
|
1079
|
Qiang W, Doherty KE, Klees LM, Tobin-Miyaji Y. Time-Dependent Lipid Dynamics, Organization and Peptide-Lipid Interaction in Phospholipid Bilayers with Incorporated β-Amyloid Oligomers. J Phys Chem Lett 2020; 11:8329-8336. [PMID: 32931283 PMCID: PMC7647725 DOI: 10.1021/acs.jpclett.0c01967] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Nonfibrillar β-amyloid (Aβ) oligomers are considered as major neurotoxic species in the pathology of Alzheimer's disease. The presence of Aβ oligomers was shown to cause membrane disruptions in a broad range of model systems. However, the molecular basis of such a disruption process remains unknown. We previously demonstrated that membrane-incorporated 40-residue Aβ (Aβ40) oligomers could form coaggregates with phospholipids. This process occurred more rapidly than the fibrillization of Aβ40 and led to more severe membrane disruption. The present study probes the time-dependent changes in lipid dynamics, bilayer structures, and peptide-lipid interactions along the time course of the oligomer-induced membrane disruption, using solid-state NMR spectroscopy. Our results suggest the presence of certain intermediate states with phospholipid molecules entering the C-terminal hydrogen-bonding networks of the Aβ40 oligomeric cores. This work provides insights on the molecular mechanisms of Aβ40-oligomer-induced membrane disruption.
Collapse
Affiliation(s)
- Wei Qiang
- Department of Chemistry, Binghamton University, State University of New York, Binghamton, New York 13902
| | - Katelynne E. Doherty
- Department of Chemistry, Binghamton University, State University of New York, Binghamton, New York 13902
| | - Lukas M. Klees
- Department of Chemistry, Binghamton University, State University of New York, Binghamton, New York 13902
| | - Yuto Tobin-Miyaji
- Department of Chemistry, Binghamton University, State University of New York, Binghamton, New York 13902
| |
Collapse
|
1080
|
Graykowski DR, Wang YZ, Upadhyay A, Savas JN. The Dichotomous Role of Extracellular Vesicles in the Central Nervous System. iScience 2020; 23:101456. [PMID: 32835924 PMCID: PMC7452271 DOI: 10.1016/j.isci.2020.101456] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 06/20/2020] [Accepted: 08/10/2020] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs) are important mediators of intercellular communication. Interest in the role of central nervous system (CNS)-derived EVs has been increasing; however, some skepticism of their importance has persisted because many aspects of their biology remain elusive. This ambiguity is largely due to technical barriers that hamper our ability to achieve a comprehensive understanding of their molecular components and mechanisms responsible for their transmission and uptake. However, accumulating evidence supports the notion that EVs play important roles in basic physiological processes within the CNS during neurodevelopment and synaptic plasticity. Interestingly, EVs also act to spread toxic polypeptides in neurodegenerative diseases. Developing a more profound understanding of the role that EVs play in the CNS could lead to the identification of biomarkers and potential vehicles for drug delivery. Here we highlight our current understanding of CNS EVs and summarize our current understanding of their complex role in the CNS.
Collapse
Affiliation(s)
- David R. Graykowski
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Yi-Zhi Wang
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Arun Upadhyay
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Jeffrey N. Savas
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
1081
|
Chaudhary A, Singh V, Varadwaj PK, Mani A. Screening natural inhibitors against upregulated G-protein coupled receptors as potential therapeutics of Alzheimer's disease. J Biomol Struct Dyn 2020; 40:673-684. [PMID: 32900274 DOI: 10.1080/07391102.2020.1817784] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Computational approaches have been helpful in high throughput screening of drug libraries and designing ligands against receptors. Alzheimer's disease is a complex neurological disorder, which causes dementia. In this disease neurons are damaged due to formation of Amyloid-beta plaques and neurofibrillary tangles, which along with some other factors contributes to disease development and progression. The objective of this study was to predict tertiary structures of five G-protein coulped neurotransmitter receptors; CHRM5, CYSLTR2, DRD5, GALR1 and HTR2C, that are upregulated in Alzheimer's disease, and to screen potential inhibitors for against these receptors. In this study, Comparative modelling, molecular docking, MMGBSA analysis, ADMET screening and molecular dynamics simulation were performed. Tertiary structures of the five GPCRs were predicted and further subjected to molecular docking against natural compounds. Pharmacokinetic studies of natural compounds were also conducted for assessing drug-likeness properties. Molecular dynamics simulations were performed to investigate the structural stability and binding affinities of each complex. Finally, the results suggested that ZINC04098704, ZINC31170017, ZINC05998597, ZINC67911229, and ZINC67910690 had better binding affinity with CHRM5, CYSLTR2, DRD5, GALR1, and HTR2C (5-HT2C) proteins, respectively.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Amit Chaudhary
- Department of Biotechnology, Motilal Nehru National Institute of Technology, Allahabad Prayagraj (Allahabad), Uttar Pradesh, India
| | - Vishal Singh
- Department of Applied Sciences, Indian Institute of Information Technology, Devghat, Jhalwa Allahabad, Uttar Pradesh, India
| | - Pritish Kumar Varadwaj
- Department of Applied Sciences, Indian Institute of Information Technology, Devghat, Jhalwa Allahabad, Uttar Pradesh, India
| | - Ashutosh Mani
- Department of Biotechnology, Motilal Nehru National Institute of Technology, Allahabad Prayagraj (Allahabad), Uttar Pradesh, India
| |
Collapse
|
1082
|
Rudajev V, Novotny J. The Role of Lipid Environment in Ganglioside GM1-Induced Amyloid β Aggregation. MEMBRANES 2020; 10:membranes10090226. [PMID: 32916822 PMCID: PMC7558528 DOI: 10.3390/membranes10090226] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 09/04/2020] [Accepted: 09/07/2020] [Indexed: 01/14/2023]
Abstract
Ganglioside GM1 is the most common brain ganglioside enriched in plasma membrane regions known as lipid rafts or membrane microdomains. GM1 participates in many modulatory and communication functions associated with the development, differentiation, and protection of neuronal tissue. It has, however, been demonstrated that GM1 plays a negative role in the pathophysiology of Alzheimer's disease (AD). The two features of AD are the formation of intracellular neurofibrillary bodies and the accumulation of extracellular amyloid β (Aβ). Aβ is a peptide characterized by intrinsic conformational flexibility. Depending on its partners, Aβ can adopt different spatial arrangements. GM1 has been shown to induce specific changes in the spatial organization of Aβ, which lead to enhanced peptide accumulation and deleterious effect especially on neuronal membranes containing clusters of this ganglioside. Changes in GM1 levels and distribution during the development of AD may contribute to the aggravation of the disease.
Collapse
|
1083
|
López-Laguna H, Sánchez J, Unzueta U, Mangues R, Vázquez E, Villaverde A. Divalent Cations: A Molecular Glue for Protein Materials. Trends Biochem Sci 2020; 45:992-1003. [PMID: 32891514 DOI: 10.1016/j.tibs.2020.08.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 07/30/2020] [Accepted: 08/06/2020] [Indexed: 02/06/2023]
Abstract
Among inorganic materials, divalent cations modulate thousands of physiological processes that support life. Their roles in protein assembly and aggregation are less known, although they are progressively being brought to light. We review the structural roles of divalent cations here, as well as the novel protein materials that are under development, in which they are used as glue-like agents. More specifically, we discuss how mechanically stable nanoparticles, fibers, matrices, and hydrogels are generated through their coordination with histidine-rich proteins. We also describe how the rational use of divalent cations combined with simple protein engineering offers unexpected and very simple biochemical approaches to biomaterial design that might address unmet clinical needs in precision medicine.
Collapse
Affiliation(s)
- Hèctor López-Laguna
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, 08193, Barcelona, Spain; Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, 08193, Barcelona, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain
| | - Julieta Sánchez
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, 08193, Barcelona, Spain; Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, 08193, Barcelona, Spain; Instituto de Investigaciones Biológicas y Tecnológicas (IIBYT) (CONICET-Universidad Nacional de Córdoba), ICTA & Cátedra de Química Biológica, Departamento de Química, FCEFyN, X 5016GCA, Córdoba, Argentina
| | - Ugutz Unzueta
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, 08193, Barcelona, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain; Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; Josep Carreras Research Institute, 08041 Barcelona, Spain.
| | - Ramón Mangues
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain; Biomedical Research Institute Sant Pau (IIB-Sant Pau), Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; Josep Carreras Research Institute, 08041 Barcelona, Spain
| | - Esther Vázquez
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, 08193, Barcelona, Spain; Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, 08193, Barcelona, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain
| | - Antonio Villaverde
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, 08193, Barcelona, Spain; Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, 08193, Barcelona, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain.
| |
Collapse
|
1084
|
Gupta VB, Chitranshi N, den Haan J, Mirzaei M, You Y, Lim JK, Basavarajappa D, Godinez A, Di Angelantonio S, Sachdev P, Salekdeh GH, Bouwman F, Graham S, Gupta V. Retinal changes in Alzheimer's disease- integrated prospects of imaging, functional and molecular advances. Prog Retin Eye Res 2020; 82:100899. [PMID: 32890742 DOI: 10.1016/j.preteyeres.2020.100899] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 08/24/2020] [Accepted: 08/27/2020] [Indexed: 12/31/2022]
Abstract
Alzheimer's Disease (AD) is a devastating neurodegenerative disorder of the brain, clinically characterised by cognitive deficits that gradually worsen over time. There is, at present, no established cure, or disease-modifying treatments for AD. As life expectancy increases globally, the number of individuals suffering from the disease is projected to increase substantially. Cumulative evidence indicates that AD neuropathological process is initiated several years, if not decades, before clinical signs are evident in patients, and diagnosis made. While several imaging, cognitive, CSF and blood-based biomarkers have been proposed for the early detection of AD; their sensitivity and specificity in the symptomatic stages is highly variable and it is difficult to justify their use in even earlier, pre-clinical stages of the disease. Research has identified potentially measurable functional, structural, metabolic and vascular changes in the retina during early stages of AD. Retina offers a distinctively accessible insight into brain pathology and current and developing ophthalmic technologies have provided us with the possibility of detecting and characterising subtle, disease-related changes. Recent human and animal model studies have further provided mechanistic insights into the biochemical pathways that are altered in the retina in disease, including amyloid and tau deposition. This information coupled with advances in molecular imaging has allowed attempts to monitor biochemical changes and protein aggregation pathology in the retina in AD. This review summarises the existing knowledge that informs our understanding of the impact of AD on the retina and highlights some of the gaps that need to be addressed. Future research will integrate molecular imaging innovation with functional and structural changes to enhance our knowledge of the AD pathophysiological mechanisms and establish the utility of monitoring retinal changes as a potential biomarker for AD.
Collapse
Affiliation(s)
- Veer B Gupta
- School of Medicine, Deakin University, VIC, Australia
| | - Nitin Chitranshi
- Faculty of Medicine Health and Human Sciences, Macquarie University, North Ryde, NSW, 2109, Australia
| | - Jurre den Haan
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands
| | - Mehdi Mirzaei
- Faculty of Medicine Health and Human Sciences, Macquarie University, North Ryde, NSW, 2109, Australia
| | - Yuyi You
- Faculty of Medicine Health and Human Sciences, Macquarie University, North Ryde, NSW, 2109, Australia
| | - Jeremiah Kh Lim
- Optometry and Vision Science, College of Nursing and Health Sciences, Bedford Park, South Australia, 5042, Australia
| | - Devaraj Basavarajappa
- Faculty of Medicine Health and Human Sciences, Macquarie University, North Ryde, NSW, 2109, Australia
| | - Angela Godinez
- Faculty of Medicine Health and Human Sciences, Macquarie University, North Ryde, NSW, 2109, Australia
| | - Silvia Di Angelantonio
- Center for Life Nanoscience, Istituto Italiano di Tecnologia, Rome, Italy; Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Perminder Sachdev
- Centre for Healthy Brain and Ageing (CHeBA), School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia; Neuropsychiatric Institute, Prince of Wales Hospital, Sydney, New South Wales, Australia
| | - Ghasem H Salekdeh
- Department of Molecular Systems Biology, Cell Science Research Center, Royan, Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Femke Bouwman
- Department of Neurology, Alzheimer Center Amsterdam, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, the Netherlands
| | - Stuart Graham
- Faculty of Medicine Health and Human Sciences, Macquarie University, North Ryde, NSW, 2109, Australia; Save Sight Institute, Sydney University, Sydney, NSW, 2000, Australia.
| | - Vivek Gupta
- Faculty of Medicine Health and Human Sciences, Macquarie University, North Ryde, NSW, 2109, Australia.
| |
Collapse
|
1085
|
Schrauben M, Dempster E, Lunnon K. Applying gene-editing technology to elucidate the functional consequence of genetic and epigenetic variation in Alzheimer's disease. Brain Pathol 2020; 30:992-1004. [PMID: 32654206 PMCID: PMC8018012 DOI: 10.1111/bpa.12881] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 04/05/2020] [Indexed: 12/15/2022] Open
Abstract
Recent studies have highlighted a potential role of genetic and epigenetic variation in the development of Alzheimer's disease. Application of the CRISPR-Cas genome-editing platform has enabled investigation of the functional impact that Alzheimer's disease-associated gene mutations have on gene expression. Moreover, recent advances in the technology have led to the generation of CRISPR-Cas-based tools that allow for high-throughput interrogation of different risk variants to elucidate the interplay between genomic regulatory features, epigenetic modifications, and chromatin structure. In this review, we examine the various iterations of the CRISPR-Cas system and their potential application for exploring the complex interactions and disruptions in gene regulatory circuits that contribute to Alzheimer's disease.
Collapse
Affiliation(s)
| | - Emma Dempster
- University of Exeter Medical SchoolExeter UniversityExeterUK
| | - Katie Lunnon
- University of Exeter Medical SchoolExeter UniversityExeterUK
| |
Collapse
|
1086
|
Ennerfelt HE, Lukens JR. The role of innate immunity in Alzheimer's disease. Immunol Rev 2020; 297:225-246. [PMID: 32588460 PMCID: PMC7783860 DOI: 10.1111/imr.12896] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 05/23/2020] [Accepted: 05/28/2020] [Indexed: 12/12/2022]
Abstract
The amyloid hypothesis has dominated Alzheimer's disease (AD) research for almost 30 years. This hypothesis hinges on the predominant clinical role of the amyloid beta (Aβ) peptide in propagating neurofibrillary tangles (NFTs) and eventual cognitive impairment in AD. Recent research in the AD field has identified the brain-resident macrophages, known as microglia, and their receptors as integral regulators of both the initiation and propagation of inflammation, Aβ accumulation, neuronal loss, and memory decline in AD. Emerging studies have also begun to reveal critical roles for distinct innate immune pathways in AD pathogenesis, which has led to great interest in harnessing the innate immune response as a therapeutic strategy to treat AD. In this review, we will highlight recent advancements in our understanding of innate immunity and inflammation in AD onset and progression. Additionally, there has been mounting evidence suggesting pivotal contributions of environmental factors and lifestyle choices in AD pathogenesis. Therefore, we will also discuss recent findings, suggesting that many of these AD risk factors influence AD progression via modulation of microglia and immune responses.
Collapse
Affiliation(s)
- Hannah E. Ennerfelt
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA
- Neuroscience Graduate Program, University of Virginia, Charlottesville, VA 22908, USA
- Cell and Molecular Biology Training Program, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - John R. Lukens
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia, Charlottesville, VA 22908, USA
- Neuroscience Graduate Program, University of Virginia, Charlottesville, VA 22908, USA
- Cell and Molecular Biology Training Program, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
1087
|
Abdullah A, Mohd Murshid N, Makpol S. Antioxidant Modulation of mTOR and Sirtuin Pathways in Age-Related Neurodegenerative Diseases. Mol Neurobiol 2020; 57:5193-5207. [PMID: 32865663 DOI: 10.1007/s12035-020-02083-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 08/19/2020] [Indexed: 12/11/2022]
Abstract
In the human body, cell division and metabolism are expected to transpire uneventfully for approximately 25 years. Then, secondary metabolism and cell damage products accumulate, and ageing phenotypes are acquired, causing the progression of disease. Among these age-related diseases, neurodegenerative diseases have attracted considerable attention because of their irreversibility, the absence of effective treatment and their relationship with social and economic pressures. Mechanistic (formerly mammalian) target of rapamycin (mTOR), sirtuin (SIRT) and insulin/insulin growth factor 1 (IGF1) signalling pathways are among the most important pathways in ageing-associated conditions, such as neurodegeneration. These longevity-related pathways are associated with a diversity of various processes, including metabolism, cognition, stress reaction and brain plasticity. In this review, we discuss the roles of sirtuin and mTOR in ageing and neurodegeneration, with an emphasis on their regulation of autophagy, apoptosis and mitochondrial energy metabolism. The intervention of neurodegeneration using potential antioxidants, including vitamins, phytochemicals, resveratrol, herbals, curcumin, coenzyme Q10 and minerals, specifically aimed at retaining mitochondrial function in the treatment of Alzheimer's disease, Parkinson's disease and Huntington's disease is highlighted.
Collapse
Affiliation(s)
- Asmaa Abdullah
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Level 17, Preclinical Building, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, 56000, Kuala Lumpur, Malaysia
| | - Nuraqila Mohd Murshid
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Level 17, Preclinical Building, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, 56000, Kuala Lumpur, Malaysia
| | - Suzana Makpol
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Level 17, Preclinical Building, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, 56000, Kuala Lumpur, Malaysia.
| |
Collapse
|
1088
|
Gasecka A, Siwik D, Gajewska M, Jaguszewski MJ, Mazurek T, Filipiak KJ, Postuła M, Eyileten C. Early Biomarkers of Neurodegenerative and Neurovascular Disorders in Diabetes. J Clin Med 2020; 9:E2807. [PMID: 32872672 PMCID: PMC7564566 DOI: 10.3390/jcm9092807] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/23/2020] [Accepted: 08/28/2020] [Indexed: 12/13/2022] Open
Abstract
Diabetes mellitus (DM) is a common disease worldwide. There is a strong association between DM and neurovascular and neurodegenerative disorders. The first group mainly consists of diabetic retinopathy, diabetic neuropathy and stroke, whereas, the second group includes Alzheimer's disease, Parkinson's disease, mild cognitive impairment and dementia. The aforementioned diseases have a common pathophysiological background including insulin resistance, oxidative stress, atherosclerosis and vascular injury. The increasing prevalence of neurovascular and neurodegenerative disorders among diabetic patients has resulted in an urgent need to develop biomarkers for their prediction and/or early detection. The aim of this review is to present the potential application of the most promising biomarkers of diabetes-related neurodegenerative and neurovascular disorders, including amylin, β-amyloid, C-reactive protein (CRP), dopamine, gamma-glutamyl transferase (GGT), glycogen synthase kinase 3β, homocysteine, microRNAs (mi-RNAs), paraoxonase 1, phosphoinositide 3-kinases, tau protein and various growth factors. The most clinically promising biomarkers of neurovascular and neurodegenerative complications in DM are hsCRP, GGT, homocysteine and miRNAs. However, all biomarkers discussed in this review could become a part of the potential multi-biomarker screening panel for diabetic patients at risk of neurovascular and neurodegenerative complications.
Collapse
Affiliation(s)
- Aleksandra Gasecka
- 1st Chair and Department of Cardiology, Medical University of Warsaw, 02-097 Warsaw, Poland; (D.S.); (M.G.); (T.M.); (K.J.F.)
| | - Dominika Siwik
- 1st Chair and Department of Cardiology, Medical University of Warsaw, 02-097 Warsaw, Poland; (D.S.); (M.G.); (T.M.); (K.J.F.)
| | - Magdalena Gajewska
- 1st Chair and Department of Cardiology, Medical University of Warsaw, 02-097 Warsaw, Poland; (D.S.); (M.G.); (T.M.); (K.J.F.)
| | | | - Tomasz Mazurek
- 1st Chair and Department of Cardiology, Medical University of Warsaw, 02-097 Warsaw, Poland; (D.S.); (M.G.); (T.M.); (K.J.F.)
| | - Krzysztof J. Filipiak
- 1st Chair and Department of Cardiology, Medical University of Warsaw, 02-097 Warsaw, Poland; (D.S.); (M.G.); (T.M.); (K.J.F.)
| | - Marek Postuła
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology, Medical University of Warsaw, 80-211 Warsaw, Poland; (M.P.); (C.E.)
| | - Ceren Eyileten
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology, Medical University of Warsaw, 80-211 Warsaw, Poland; (M.P.); (C.E.)
| |
Collapse
|
1089
|
Kaur A, New EJ, Sunde M. Strategies for the Molecular Imaging of Amyloid and the Value of a Multimodal Approach. ACS Sens 2020; 5:2268-2282. [PMID: 32627533 DOI: 10.1021/acssensors.0c01101] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Protein aggregation has been widely implicated in neurodegenerative diseases such as Alzheimer's disease, frontotemporal dementia, Parkinson's disease, and Huntington disease, as well as in systemic amyloidoses and conditions associated with localized amyloid deposits, such as type-II diabetes. The pressing need for a better understanding of the factors governing protein assembly has driven research for the development of molecular sensors for amyloidogenic proteins. To date, a number of sensors have been developed that report on the presence of protein aggregates utilizing various modalities, and their utility demonstrated for imaging protein aggregation in vitro and in vivo. Analysis of these sensors highlights the various advantages and disadvantages of the different imaging modalities and makes clear that multimodal sensors with properties amenable to more than one imaging technique need to be developed. This critical review highlights the key molecular scaffolds reported for molecular imaging modalities such as fluorescence, positron emission tomography, single photon emission computed tomography, and magnetic resonance imaging and includes discussion of the advantages and disadvantages of each modality, and future directions for the design of amyloid sensors. We also discuss the recent efforts focused on the design and development of multimodal sensors and the value of cross-validation across multiple modalities.
Collapse
Affiliation(s)
- Amandeep Kaur
- The University of Sydney, School of Medical Sciences, Faculty of Medicine and Health, Sydney, New South Wales 2006, Australia
- The University of Sydney, Nano Institute (Sydney Nano), The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Elizabeth J. New
- The University of Sydney, Nano Institute (Sydney Nano), The University of Sydney, Sydney, New South Wales 2006, Australia
- The University of Sydney, School of Chemistry, Faculty of Science, Sydney, New South Wales 2006, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Margaret Sunde
- The University of Sydney, School of Medical Sciences, Faculty of Medicine and Health, Sydney, New South Wales 2006, Australia
- The University of Sydney, Nano Institute (Sydney Nano), The University of Sydney, Sydney, New South Wales 2006, Australia
| |
Collapse
|
1090
|
Toyos-Rodríguez C, García-Alonso FJ, de la Escosura-Muñiz A. Electrochemical Biosensors Based on Nanomaterials for Early Detection of Alzheimer's Disease. SENSORS (BASEL, SWITZERLAND) 2020; 20:E4748. [PMID: 32842632 PMCID: PMC7506792 DOI: 10.3390/s20174748] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/18/2020] [Accepted: 08/19/2020] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is an untreatable neurodegenerative disease that initially manifests as difficulty to remember recent events and gradually progresses to cognitive impairment. The incidence of AD is growing yearly as life expectancy increases, thus early detection is essential to ensure a better quality of life for diagnosed patients. To reach that purpose, electrochemical biosensing has emerged as a cost-effective alternative to traditional diagnostic techniques, due to its high sensitivity and selectivity. Of special relevance is the incorporation of nanomaterials in biosensors, as they contribute to enhance electron transfer while promoting the immobilization of biological recognition elements. Moreover, nanomaterials have also been employed as labels, due to their unique electroactive and electrocatalytic properties. The aim of this review is to add value in the advances achieved in the detection of AD biomarkers, the strategies followed for the incorporation of nanomaterials and its effect in biosensors performance.
Collapse
Affiliation(s)
- Celia Toyos-Rodríguez
- NanoBioAnalysis Group-Department of Physical and Analytical Chemistry, University of Oviedo, Julián Clavería 8, 33006 Oviedo, Spain;
- Biotechnology Institute of Asturias, University of Oviedo, Santiago Gascon Building, 33006 Oviedo, Spain;
| | - Francisco Javier García-Alonso
- Biotechnology Institute of Asturias, University of Oviedo, Santiago Gascon Building, 33006 Oviedo, Spain;
- NanoBioAnalysis Group-Department of Organic and Inorganic Chemistry, University of Oviedo, Julián Clavería 8, 33006 Oviedo, Spain
| | - Alfredo de la Escosura-Muñiz
- NanoBioAnalysis Group-Department of Physical and Analytical Chemistry, University of Oviedo, Julián Clavería 8, 33006 Oviedo, Spain;
- Biotechnology Institute of Asturias, University of Oviedo, Santiago Gascon Building, 33006 Oviedo, Spain;
| |
Collapse
|
1091
|
Rezabakhsh A, Rahbarghazi R, Fathi F. Surface plasmon resonance biosensors for detection of Alzheimer's biomarkers; an effective step in early and accurate diagnosis. Biosens Bioelectron 2020; 167:112511. [PMID: 32858422 DOI: 10.1016/j.bios.2020.112511] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 08/06/2020] [Accepted: 08/10/2020] [Indexed: 12/17/2022]
Abstract
The rapid and direct detection of biomarkers in biofluids at clinically relevant concentrations faces serious limitations to develop diagnostic criteria for neurodegenerative diseases such as Alzheimer's disease (AD). In this regard, the early detection of biomarkers correlated with AD using novel modalities and instruments is at the center of attention. Recently, some newly invented optical-based biosensors namely Surface Plasmon Resonance (SPR) has been extensively investigated for the detection of biomarkers using a label-free method or by checking interaction between ligand and analyte. These approaches can sense a very small amount of target molecules in the blood and cerebrospinal fluids samples. In this review, the different hypothesis related to AD, and the structural properties of AD biomarkers was introduced. Also, we aim to highlight the specific role of available SPR-based sensing methods for early detection of AD biomarkers such as aggregated β-amyloid and tau proteins. Efforts to better understand the accuracy and efficiency of optical-based biosensors in the field of neurodegenerative disease enable us to accelerate the advent of novel modalities in the clinical setting for therapeutic and diagnostic purposes.
Collapse
Affiliation(s)
- Aysa Rezabakhsh
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Emergency Medicine Research Team, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farzaneh Fathi
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran; Biosensor Sciences and Technologies Research Center (BSTRC), Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
1092
|
Kumar A, Bagri K, Nimbhal M, Kumar P. In silico exploration of the fingerprints triggering modulation of glutaminyl cyclase inhibition for the treatment of Alzheimer's disease using SMILES based attributes in Monte Carlo optimization. J Biomol Struct Dyn 2020; 39:7181-7193. [PMID: 32795153 DOI: 10.1080/07391102.2020.1806111] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Alzheimer's disease is the most common neurodegenerative disorder and being a social burden Alzheimer's has become an economic liability on developing countries. With limited understanding regarding the cause of disease, it is commonly identified by extracellular deposit of amyloid β (Aβ) peptides as senile plaques. Pyroglutamated Aβ is identified from the brain of AD patients and constituted the majority of total Aβ present. The formation of Pyroglutamated Aβ could be hindered by the use of Glutaminyl cyclase inhibitors and could efficiently improve the symptoms of Alzheimer's. The literature revealed the competence of quantitative structure activity/property relationship studies in drug discovery. The present work explores the efficiency of Monte Carlo based QSAR modelling studies on a dataset of 125 Glutaminyl cyclase inhibitors with pKi taken as the endpoint for QSAR analysis. The dataset is divided into training, subtraining, calibration and validation sets resulting in the generation of five random splits. The validation is performed in accordance with the Organization of Economic Corporation and Development principles. The values of R2, Q2, index of ideality of correlation, concordance correlation coefficient, av. rm2 and delta rm2 of calibration set of the best split are found to be 0.9012, 0.8775, 0.9479, 0.9435, 0.8347 and 0.0847, respectively. The structural features responsible for increasing the inhibitory activity are identified. These structural features are added to a base compound from the dataset to design six novel molecules. These new molecules possess improved inhibitory activity as compare to the base compound. The results are further supported by docking studies.Communicated by Vsevolod Makeev.
Collapse
Affiliation(s)
- Ashwani Kumar
- Department of Pharmaceutical Sciences, Guru Jambheshwar University of Science & Technology, Hisar, India
| | - Kiran Bagri
- Department of Pharmaceutical Sciences, Guru Jambheshwar University of Science & Technology, Hisar, India
| | - Manisha Nimbhal
- Department of Pharmaceutical Sciences, Guru Jambheshwar University of Science & Technology, Hisar, India
| | - Parvin Kumar
- Department of Chemistry, Kurukshetra University, Kurukshetra, India
| |
Collapse
|
1093
|
3D In Vitro Human Organ Mimicry Devices for Drug Discovery, Development, and Assessment. ADVANCES IN POLYMER TECHNOLOGY 2020. [DOI: 10.1155/2020/6187048] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The past few decades have shown significant advancement as complex in vitro humanized systems have substituted animal trials and 2D in vitro studies. 3D humanized platforms mimic the organs of interest with their stimulations (physical, electrical, chemical, and mechanical). Organ-on-chip devices, including in vitro modelling of 3D organoids, 3D microfabrication, and 3D bioprinted platforms, play an essential role in drug discovery, testing, and assessment. In this article, a thorough review is provided of the latest advancements in the area of organ-on-chip devices targeting liver, kidney, lung, gut, heart, skin, and brain mimicry devices for drug discovery, development, and/or assessment. The current strategies, fabrication methods, and the specific application of each device, as well as the advantages and disadvantages, are presented for each reported platform. This comprehensive review also provides some insights on the challenges and future perspectives for the further advancement of each organ-on-chip device.
Collapse
|
1094
|
Muralidar S, Ambi SV, Sekaran S, Thirumalai D, Palaniappan B. Role of tau protein in Alzheimer's disease: The prime pathological player. Int J Biol Macromol 2020; 163:1599-1617. [PMID: 32784025 DOI: 10.1016/j.ijbiomac.2020.07.327] [Citation(s) in RCA: 137] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 07/07/2020] [Accepted: 07/31/2020] [Indexed: 01/11/2023]
Abstract
Alzheimer's disease (AD) is a prevalently found tauopathy characterized by memory loss and cognitive insufficiency. AD is an age-related neurodegenerative disease with two major hallmarks which includes extracellular amyloid plaques made of amyloid-β (Aβ) and intracellular neurofibrillary tangles of hyperphosphorylated tau. With population aging worldwide, there is an indispensable need for treatment strategies that can potentially manage this developing dementia. Despite broad researches on targeting Aβ in the past two decades, research findings on Aβ targeted therapeutics failed to prove efficacy in the treatment of AD. Tau protein with its extensive pathological role in several neurodegenerative diseases can be considered as a promising target candidate for developing therapeutic interventions. The abnormal hyperphosphorylation of tau plays detrimental pathological functions which ultimately lead to neurodegeneration. This review will divulge the importance of tau in AD pathogenesis, the interplay of Aβ and tau, the pathological functions of tau, and potential therapeutic strategies for an effective management of neuronal disorders.
Collapse
Affiliation(s)
- Shibi Muralidar
- School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur 613401, Tamil Nadu, India
| | - Senthil Visaga Ambi
- School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur 613401, Tamil Nadu, India.
| | - Saravanan Sekaran
- School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur 613401, Tamil Nadu, India; Centre for Nanotechnology & Advanced Biomaterials (CeNTAB), School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur 613401, Tamil Nadu, India
| | - Diraviyam Thirumalai
- School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur 613401, Tamil Nadu, India
| | - Balamurugan Palaniappan
- School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur 613401, Tamil Nadu, India
| |
Collapse
|
1095
|
Pradhan K, Das G, Kar C, Mukherjee N, Khan J, Mahata T, Barman S, Ghosh S. Rhodamine-Based Metal Chelator: A Potent Inhibitor of Metal-Catalyzed Amyloid Toxicity. ACS OMEGA 2020; 5:18958-18967. [PMID: 32775897 PMCID: PMC7408195 DOI: 10.1021/acsomega.0c02235] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 06/19/2020] [Indexed: 05/28/2023]
Abstract
Alzheimer's disease (AD) exhibits a multitude of syndromes which add up to its complex nature. In AD, amyloid plaques are deposited along with abnormal accumulation of transition-metal ions. These transition-metal ions are redox-active and help to induce the formation of various polymorphic forms of amyloid-β. Amyloid oligomeric and fibrillar aggregates are the main cause for neuronal toxicity. Another reason for neuronal toxicity arises from generation of reactive oxygen species (ROS) catalyzed by redox-active metal ions through Fenton's reaction. In this direction, an Aβ inhibitor possessing the metal chelation property will be the most promising approach against multifaceted AD. Herein, a rhodamine-B-based compound (Rh-BT) has been designed and synthesized. Rhodamine was attached with benzothiazole as a recognition unit for amyloid-β aggregates. The molecule can effectively capture redox metal ions from the Aβ-Cu2+ complex as well as inhibit Aβ self-assembly such as toxic oligomeric and fibrillar aggregates. Various biophysical assays show that Rh-BT interacts with the Aβ peptide, is capable of decreasing metal-induced ROS generation, and inhibits Aβ-Cu2+-induced cytotoxicity. All these results support the multifunctional nature of Rh-BT, which has an Aβ-specific recognition unit. In addition to the above properties, Rh-BT also exhibits good serum stability in vivo and blood-brain barrier permeability. Therefore, Rh-BT can be considered as a potent multifunctional therapeutic for the treatment of AD.
Collapse
Affiliation(s)
- Krishnangsu Pradhan
- Organic
and Medicinal Chemistry and Structural Biology and Bioinformatics
Division, CSIR-Indian Institute of Chemical
Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata 700 032, West Bengal, India
| | - Gaurav Das
- Organic
and Medicinal Chemistry and Structural Biology and Bioinformatics
Division, CSIR-Indian Institute of Chemical
Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata 700 032, West Bengal, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Chirantan Kar
- Organic
and Medicinal Chemistry and Structural Biology and Bioinformatics
Division, CSIR-Indian Institute of Chemical
Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata 700 032, West Bengal, India
| | - Nabanita Mukherjee
- Department
of Bioscience & Bioengineering, Indian
Institute of Technology Jodhpur, NH 65, Surpura Bypass Road, Karwar, Rajasthan 342037, India
| | - Juhee Khan
- Organic
and Medicinal Chemistry and Structural Biology and Bioinformatics
Division, CSIR-Indian Institute of Chemical
Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata 700 032, West Bengal, India
| | - Tanushree Mahata
- Organic
and Medicinal Chemistry and Structural Biology and Bioinformatics
Division, CSIR-Indian Institute of Chemical
Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata 700 032, West Bengal, India
| | - Surajit Barman
- Organic
and Medicinal Chemistry and Structural Biology and Bioinformatics
Division, CSIR-Indian Institute of Chemical
Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata 700 032, West Bengal, India
| | - Surajit Ghosh
- Organic
and Medicinal Chemistry and Structural Biology and Bioinformatics
Division, CSIR-Indian Institute of Chemical
Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata 700 032, West Bengal, India
- Department
of Bioscience & Bioengineering, Indian
Institute of Technology Jodhpur, NH 65, Surpura Bypass Road, Karwar, Rajasthan 342037, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
1096
|
Bécot A, Volgers C, van Niel G. Transmissible Endosomal Intoxication: A Balance between Exosomes and Lysosomes at the Basis of Intercellular Amyloid Propagation. Biomedicines 2020; 8:biomedicines8080272. [PMID: 32759666 PMCID: PMC7459801 DOI: 10.3390/biomedicines8080272] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 07/28/2020] [Accepted: 07/31/2020] [Indexed: 12/16/2022] Open
Abstract
In Alzheimer′s disease (AD), endolysosomal dysfunctions are amongst the earliest cellular features to appear. Each organelle of the endolysosomal system, from the multivesicular body (MVB) to the lysosome, contributes to the homeostasis of amyloid precursor protein (APP) cleavage products including β-amyloid (Aβ) peptides. Hence, this review will attempt to disentangle how changes in the endolysosomal system cumulate to the generation of toxic amyloid species and hamper their degradation. We highlight that the formation of MVBs and the generation of amyloid species are closely linked and describe how the molecular machineries acting at MVBs determine the generation and sorting of APP cleavage products towards their degradation or release in association with exosomes. In particular, we will focus on AD-related distortions of the endolysomal system that divert it from its degradative function to favour the release of exosomes and associated amyloid species. We propose here that such an imbalance transposed at the brain scale poses a novel concept of transmissible endosomal intoxication (TEI). This TEI would initiate a self-perpetuating transmission of endosomal dysfunction between cells that would support the propagation of amyloid species in neurodegenerative diseases.
Collapse
|
1097
|
Jamerlan A, An SSA, Hulme J. Advances in amyloid beta oligomer detection applications in Alzheimer's disease. Trends Analyt Chem 2020. [DOI: 10.1016/j.trac.2020.115919] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
1098
|
The Viral Hypothesis in Alzheimer's Disease: Novel Insights and Pathogen-Based Biomarkers. J Pers Med 2020; 10:jpm10030074. [PMID: 32751069 PMCID: PMC7563893 DOI: 10.3390/jpm10030074] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/27/2020] [Accepted: 07/28/2020] [Indexed: 02/07/2023] Open
Abstract
Early diagnosis of Alzheimer’s disease (AD) and the identification of significant risk factors are necessary to better understand disease progression, and to develop intervention-based therapies prior to significant neurodegeneration. There is thus a critical need to establish biomarkers which can predict the risk of developing AD before the onset of cognitive decline. A number of studies have indicated that exposure to various microbial pathogens can accelerate AD pathology. Additionally, several studies have indicated that amyloid-β possess antimicrobial properties and may act in response to infection as a part of the innate immune system. These findings have led some to speculate that certain types of infections may play a significant role in AD pathogenesis. In this review, we will provide an overview of studies which suggest pathogen involvement in AD. Additionally, we will discuss a number of pathogen-associated biomarkers which may be effective in establishing AD risk. Infections that increase the risk of AD represent a modifiable risk factor which can be treated with therapeutic intervention. Pathogen-based biomarkers may thus be a valuable tool for evaluating and decreasing AD risk across the population.
Collapse
|
1099
|
Mihardja M, Roy J, Wong KY, Aquili L, Heng BC, Chan YS, Fung ML, Lim LW. Therapeutic potential of neurogenesis and melatonin regulation in Alzheimer's disease. Ann N Y Acad Sci 2020; 1478:43-62. [PMID: 32700392 DOI: 10.1111/nyas.14436] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 06/09/2020] [Accepted: 06/18/2020] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disorder characterized by the hallmark pathologies of amyloid-beta plaques and neurofibrillary tangles. Symptoms of this devastating disease include behavioral changes and deterioration of higher cognitive functions. Impairment of neurogenesis has also been shown to occur in AD, which adversely impacts new neuronal cell growth, differentiation, and survival. This impairment possibly results from the cumulative effects of the various pathologies of AD. Preclinical studies have suggested that the administration of melatonin-the pineal hormone primarily responsible for the regulation of the circadian rhythm-targets the effects of AD pathologies and improves cognitive impairment. It is postulated that by mitigating the effect of these pathologies, melatonin can also rescue neurogenesis impairment. This review aims to explore the effect of AD pathologies on neurogenesis, as well as the mechanisms by which melatonin is able to ameliorate AD pathologies to potentially promote neurogenesis.
Collapse
Affiliation(s)
- Mazel Mihardja
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Jaydeep Roy
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Kan Yin Wong
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Luca Aquili
- Division of Psychology, College of Health and Human Sciences, Charles Darwin University, Darwin, Australia
| | - Boon Chin Heng
- Department of Biological Sciences, Sunway University, Bandar Sunway, Malaysia.,Peking University School of Stomatology, Beijing, China
| | - Ying-Shing Chan
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Man Lung Fung
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Lee Wei Lim
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.,Department of Biological Sciences, Sunway University, Bandar Sunway, Malaysia
| |
Collapse
|
1100
|
Iqbal G, Braidy N, Ahmed T. Blood-Based Biomarkers for Predictive Diagnosis of Cognitive Impairment in a Pakistani Population. Front Aging Neurosci 2020; 12:223. [PMID: 32848704 PMCID: PMC7396488 DOI: 10.3389/fnagi.2020.00223] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 06/22/2020] [Indexed: 12/27/2022] Open
Abstract
Numerous studies have identified an association between age-related cognitive impairment (CI) and oxidative damage, accumulation of metals, amyloid levels, tau, and deranged lipid profile. There is a concerted effort to establish the reliability of these blood-based biomarkers for predictive diagnosis of CI and its progression. We assessed the serum levels of high-density lipoprotein (HDL) cholesterol, low-density lipoprotein (LDL) cholesterol, triglycerides, total cholesterol, selected metals (Cu, Al, Zn, Pb, Mn, Cad), and total-tau and amyloid beta-42 protein in mild (n = 71), moderate (n = 86) and severe (n = 25) cognitively impaired patients and compared them with age-matched healthy controls (n = 90) from Pakistan. We found that a decrease in HDL cholesterol (correlation coefficient r = 0.467) and amyloid beta-42 (r = 0.451) were associated with increased severity of CI. On the other hand, an increase in cholesterol ratio (r = -0.562), LDL cholesterol (r = -0.428), triglycerides, and total-tau (r = -0.443) were associated with increased severity of CI. Increases in cholesterol ratio showed the strongest association and correlated with increases in tau concentration (r = 0.368), and increased triglycerides were associated with decreased amyloid beta-42 (r = -0.345). Increased Cu levels showed the strongest association with tau increase and increased Zn and Pb levels showed the strongest association with reduced amyloid beta-42 levels. Receiver Operating Characteristic (ROC) showed the cutoff values of blood metals (Al, Pb, Cu, Cad, Zn, and Mn), total-tau, and amyloid beta-42 with sensitivity and specificity. Our data show for the first time that blood lipids, metals (particularly Cu, Zn, Pb, and Al), serum amyloid-beta-42/tau proteins modulate each other's levels and can be collectively used as a predictive marker for CI.
Collapse
Affiliation(s)
- Ghazala Iqbal
- Neurobiology Laboratory, Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences & Technology (NUST), Islamabad, Pakistan
| | - Nady Braidy
- Centre for Healthy Ageing, School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Touqeer Ahmed
- Neurobiology Laboratory, Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences & Technology (NUST), Islamabad, Pakistan
| |
Collapse
|