1
|
Yang X, Yao K, Zhang M, Zhang W, Zu H. New insight into the role of altered brain cholesterol metabolism in the pathogenesis of AD: A unifying cholesterol hypothesis and new therapeutic approach for AD. Brain Res Bull 2025; 224:111321. [PMID: 40164234 DOI: 10.1016/j.brainresbull.2025.111321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/16/2025] [Accepted: 03/24/2025] [Indexed: 04/02/2025]
Abstract
The dysregulation of cholesterol metabolism homeostasis has been universally suggested in the aeotiology of Alzheimer's disease (AD). Initially, studies indicate that alteration of serum cholesterol level might contribute to AD. However, because blood-brain barrier impedes entry of plasma cholesterol, brain cells are not directly influenced by plasma cholesterol. Furthermore, mounting evidences suggest a link between alteration of brain cholesterol metabolism and AD. Interestingly, Amyloid-β proteins (Aβ) can markedly inhibit cellular cholesterol biosynthesis and lower cellular cholesterol content in cultured cells. And Aβ overproduction/overload induces a significant decrease of brain cellular cholesterol content in familial AD (FAD) animals. Importantly, mutations or polymorphisms of genes related to brain cholesterol transportation, such as ApoE4, ATP binding cassette (ABC) transporters, low-density lipoprotein receptor (LDLR) family and Niemann-Pick C disease 1 or 2 (NPC1/2), obviously lead to decreased brain cholesterol transport, resulting in brain cellular cholesterol loss, which could be tightly associated with AD pathological impairments. Additionally, accumulating data show that there are reduction of brain cholesterol biosynthesis and/or disorder of brain cholesterol trafficking in a variety of sporadic AD (SAD) animals and patients. Collectively, compelling evidences indicate that FAD and SAD could share one common and overlapping neurochemical mechanism: brain neuronal/cellular cholesterol deficiency. Therefore, accumulated evidences strongly support a novel hypothesis that deficiency of brain cholesterol contributes to the onset and progression of AD. This review highlights the pivotal role of brain cholesterol deficiency in the pathogenesis of AD. The hypothesis offers valuable insights for the future development of AD treatment.
Collapse
Affiliation(s)
- Xiaobo Yang
- Department of Neurology, Jinshan Hospital affiliated to Fudan University, Shanghai 201508, China; Department of Neurology, Shanghai Xuhui Central Hospital, Fudan University, Shanghai 200237, China
| | - Kai Yao
- Department of Neurology, Jinshan Hospital affiliated to Fudan University, Shanghai 201508, China
| | - Mengqi Zhang
- Department of Neurology, Jinshan Hospital affiliated to Fudan University, Shanghai 201508, China
| | - Wenbin Zhang
- Department of Neurology, Jinshan Hospital affiliated to Fudan University, Shanghai 201508, China
| | - Hengbing Zu
- Department of Neurology, Jinshan Hospital affiliated to Fudan University, Shanghai 201508, China.
| |
Collapse
|
2
|
Yu W, Li Y, Li Y, Hu J, Wu J, Chen X, Huang Y, Shi X. Connexin43 Contributes to Alzheimer's Disease by Promoting the Mitochondria-Associated Membrane-Related Autophagy Inhibition. Mol Neurobiol 2025; 62:4319-4337. [PMID: 39438345 PMCID: PMC11880138 DOI: 10.1007/s12035-024-04536-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 10/05/2024] [Indexed: 10/25/2024]
Abstract
The perturbed structure and function of mitochondria-associated membranes (MAM), instead of the amyloid cascade, have been gradually proposed to play a basic role in the pathogenesis of Alzheimer's disease (AD). Notably, autophagy inhibition is one of the main mechanisms of MAM dysfunction and plays an important role in neuronal injury. However, the upstream molecular mechanism underlying the MAM dysfunctions remains elusive. Here, we defined an unexpected and critical role of connexin43 (Cx43) in regulating the MAM structure. The expression levels of Cx43 and mitofusin-2 (MFN2, the MAM biomarker) increase significantly in 9-month-old APPswe/PS1dE9 double-transgenic AD model mice, and there is an obvious colocalization relationship. Moreover, both AD mice and cells lacking Cx43 exhibit an evident reduction in the MAM contact sites, which subsequently promotes the conversion from microtubule-associated protein 1 light-chain 3B I (LC3B-I) to LC3B-II via inhibition mTOR-dependent pathway and then initiates the generation of autophagosomes. Autophagosome formation ultimately promotes β-amyloid (Aβ) clearance and attenuates Aβ-associated pathological changes in AD, mainly including astrogliosis and neuronal apoptosis. Our findings not only reveal a previously unrecognized effect of Cx43 on MAM upregulation but also highlight the major player of MAM-induced autophagy inhibition in Cx43-facilitated AD pathogenesis, providing a novel insight into the alternative therapeutic strategies for the early treatment of AD.
Collapse
Affiliation(s)
- Weiwei Yu
- Department of Neurology, Peking University Shenzhen Hospital, Futian District, 1120 Lianhua Road, Shenzhen, 518036, China
| | - Yunong Li
- Department of Neurology, Peking University Shenzhen Hospital, Futian District, 1120 Lianhua Road, Shenzhen, 518036, China
| | - Yao Li
- Department of Neurology, Peking University Shenzhen Hospital, Futian District, 1120 Lianhua Road, Shenzhen, 518036, China
| | - Jun Hu
- Department of Neurology, Peking University Shenzhen Hospital, Futian District, 1120 Lianhua Road, Shenzhen, 518036, China
| | - Jun Wu
- Department of Neurology, Peking University Shenzhen Hospital, Futian District, 1120 Lianhua Road, Shenzhen, 518036, China
| | - Xuhui Chen
- Department of Neurology, Peking University Shenzhen Hospital, Futian District, 1120 Lianhua Road, Shenzhen, 518036, China
| | - Yining Huang
- Department of Neurology, Peking University First Hospital, 8 Xishiku Street Xicheng District, Beijing, 100034, China.
| | - Xin Shi
- Department of Neurology, Peking University Shenzhen Hospital, Futian District, 1120 Lianhua Road, Shenzhen, 518036, China.
| |
Collapse
|
3
|
Dai Y, Zhang Q, Gu R, Chen J, Ye P, Zhu H, Tang M, Nie X. Metal ion formulations for diabetic wound healing: Mechanisms and therapeutic potential. Int J Pharm 2024; 667:124889. [PMID: 39481815 DOI: 10.1016/j.ijpharm.2024.124889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 10/09/2024] [Accepted: 10/28/2024] [Indexed: 11/03/2024]
Abstract
Metals are vital in human physiology, which not only act as enzyme catalysts in the processes of superoxide dismutase and glucose phosphorylation, but also affect the redox process, osmotic adjustment, metabolism and neural signals. However, metal imbalances can lead to diseases such as diabetes, which is marked by chronic hyperglycemia and affects wound healing. The hyperglycemic milieu of diabetes impairs wound healing, posing significant challenges to patient quality of life. Wound healing encompasses a complex cascade of hemostasis, inflammation, proliferation, and remodeling phases, which are susceptible to disruption in hyperglycemic conditions. In recent decades, metals have emerged as critical facilitators of wound repair by enhancing antimicrobial properties (e.g., iron and silver), providing angiogenic stimulation (copper), promoting antioxidant activity and growth factor synthesis (zinc), and supporting wound closure (calcium and magnesium). Consequently, research has pivoted towards the development of metal ion-based therapeutics, including innovative formulations such as nano-hydrogels, nano-microneedle dressings, and microneedle patches. Prepared by combining macromolecular materials such as chitosan, hyaluronic acid and sodium alginate with metals, aiming at improving the management of diabetic wounds. This review delineates the roles of key metals in human physiology and evaluates the application of metal ions in diabetic wound management strategies.
Collapse
Affiliation(s)
- Yuhe Dai
- College of Pharmacy, Zunyi Medical University, Zunyi 563006, China; Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China.
| | - Qianbo Zhang
- College of Pharmacy, Zunyi Medical University, Zunyi 563006, China; Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China.
| | - Rifang Gu
- College of Pharmacy, Zunyi Medical University, Zunyi 563006, China; School Medical Office, Zunyi Medical University, Zunyi 563006, China.
| | - Jitao Chen
- College of Pharmacy, Zunyi Medical University, Zunyi 563006, China; Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China.
| | - Penghui Ye
- College of Pharmacy, Zunyi Medical University, Zunyi 563006, China; Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China.
| | - Huan Zhu
- College of Pharmacy, Zunyi Medical University, Zunyi 563006, China; Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China.
| | - Ming Tang
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| | - Xuqiang Nie
- College of Pharmacy, Zunyi Medical University, Zunyi 563006, China; Key Lab of the Basic Pharmacology of the Ministry of Education & Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563006, China.
| |
Collapse
|
4
|
Maggiore A, Latina V, D'Erme M, Amadoro G, Coccurello R. Non-canonical pathways associated to Amyloid beta and tau protein dyshomeostasis in Alzheimer's disease: A narrative review. Ageing Res Rev 2024; 102:102578. [PMID: 39542177 DOI: 10.1016/j.arr.2024.102578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/07/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024]
Abstract
Alzheimer's Disease (AD) is the most common form of dementia among elderly people. This disease imposes a significant burden on the healthcare system, society, and economy due to the increasing global aging population. Current trials with drugs or bioactive compounds aimed at reducing cerebral Amyloid beta (Aβ) plaques and tau protein neurofibrillary tangles, which are the two main hallmarks of this devastating neurodegenerative disease, have not provided significant results in terms of their neuropathological outcomes nor met the expected clinical end-points. Ageing, genetic and environmental risk factors, along with different clinical symptoms suggest that AD is a complex and heterogeneous disorder with multiple interconnected pathological pathways rather than a single disease entity. In the present review, we highlight and discuss various non-canonical, Aβ-independent mechanisms, like gliosis, unhealthy dietary intake, lipid and sugar signaling, and cerebrovascular damage that contribute to the onset and development of AD. We emphasize that challenging the traditional "amyloid cascade hypothesis" may improve our understanding of this age-related complex syndrome and help fight the progressive cognitive decline in AD.
Collapse
Affiliation(s)
- Anna Maggiore
- Department of Biochemical Sciences, Sapienza University, P.le Aldo Moro 5, Rome 00185, Italy; Department of Brain Sciences, Imperial College, London, UK
| | - Valentina Latina
- European Brain Research Institute (EBRI), Viale Regina Elena 295, Rome 00161, Italy; Institute of Translational Pharmacology (IFT) CNR, Via Fosso del Cavaliere 100, Rome 00133, Italy
| | - Maria D'Erme
- Department of Biochemical Sciences, Sapienza University, P.le Aldo Moro 5, Rome 00185, Italy
| | - Giuseppina Amadoro
- European Brain Research Institute (EBRI), Viale Regina Elena 295, Rome 00161, Italy; Institute of Translational Pharmacology (IFT) CNR, Via Fosso del Cavaliere 100, Rome 00133, Italy.
| | - Roberto Coccurello
- Institute for Complex System (ISC) CNR, Via dei Taurini 19, Rome 00185, Italy; IRCSS Santa Lucia Foundation, European Center for Brain Research, Via Fosso del Fiorano 64-65, Rome 00143, Italy.
| |
Collapse
|
5
|
Bruno F, Abondio P, Bruno R, Ceraudo L, Paparazzo E, Citrigno L, Luiselli D, Bruni AC, Passarino G, Colao R, Maletta R, Montesanto A. Alzheimer's disease as a viral disease: Revisiting the infectious hypothesis. Ageing Res Rev 2023; 91:102068. [PMID: 37704050 DOI: 10.1016/j.arr.2023.102068] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/05/2023] [Accepted: 09/07/2023] [Indexed: 09/15/2023]
Abstract
Alzheimer's disease (AD) represents the most frequent type of dementia in elderly people. Two major forms of the disease exist: sporadic - the causes of which have not yet been fully understood - and familial - inherited within families from generation to generation, with a clear autosomal dominant transmission of mutations in Presenilin 1 (PSEN1), 2 (PSEN2) or Amyloid Precursors Protein (APP) genes. The main hallmark of AD consists of extracellular deposits of amyloid-beta (Aβ) peptide and intracellular deposits of the hyperphosphorylated form of the tau protein. An ever-growing body of research supports the viral infectious hypothesis of sporadic forms of AD. In particular, it has been shown that several herpes viruses (i.e., HHV-1, HHV-2, HHV-3 or varicella zoster virus, HHV-4 or Epstein Barr virus, HHV-5 or cytomegalovirus, HHV-6A and B, HHV-7), flaviviruses (i.e., Zika virus, Dengue fever virus, Japanese encephalitis virus) as well as Human Immunodeficiency Virus (HIV), hepatitis viruses (HAV, HBV, HCV, HDV, HEV), SARS-CoV2, Ljungan virus (LV), Influenza A virus and Borna disease virus, could increase the risk of AD. Here, we summarized and discussed these results. Based on these findings, significant issues for future studies are also put forward.
Collapse
Affiliation(s)
- Francesco Bruno
- Regional Neurogenetic Centre (CRN), Department of Primary Care, Azienda Sanitaria Provinciale Di Catanzaro, Viale A. Perugini, 88046 Lamezia Terme, CZ, Italy; Association for Neurogenetic Research (ARN), Lamezia Terme, CZ, Italy
| | - Paolo Abondio
- Laboratory of Ancient DNA, Department of Cultural Heritage, University of Bologna, Via degli Ariani 1, 48121 Ravenna, Italy.
| | - Rossella Bruno
- Sudent at the Department of Medical and Surgical Sciences, Magna Graecia University of Catanzaro, 88050 Catanzaro, Italy
| | - Leognano Ceraudo
- Sudent at the Department of Medical and Surgical Sciences, University of Parma, 43121 Parma, Italy
| | - Ersilia Paparazzo
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende 87036, Italy
| | - Luigi Citrigno
- National Research Council (CNR) - Institute for Biomedical Research and Innovation - (IRIB), 87050 Mangone, Cosenza, Italy
| | - Donata Luiselli
- Laboratory of Ancient DNA, Department of Cultural Heritage, University of Bologna, Via degli Ariani 1, 48121 Ravenna, Italy
| | - Amalia C Bruni
- Regional Neurogenetic Centre (CRN), Department of Primary Care, Azienda Sanitaria Provinciale Di Catanzaro, Viale A. Perugini, 88046 Lamezia Terme, CZ, Italy; Association for Neurogenetic Research (ARN), Lamezia Terme, CZ, Italy
| | - Giuseppe Passarino
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende 87036, Italy
| | - Rosanna Colao
- Regional Neurogenetic Centre (CRN), Department of Primary Care, Azienda Sanitaria Provinciale Di Catanzaro, Viale A. Perugini, 88046 Lamezia Terme, CZ, Italy
| | - Raffaele Maletta
- Regional Neurogenetic Centre (CRN), Department of Primary Care, Azienda Sanitaria Provinciale Di Catanzaro, Viale A. Perugini, 88046 Lamezia Terme, CZ, Italy; Association for Neurogenetic Research (ARN), Lamezia Terme, CZ, Italy
| | - Alberto Montesanto
- Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende 87036, Italy.
| |
Collapse
|
6
|
The Nerve Growth Factor Receptor (NGFR/p75 NTR): A Major Player in Alzheimer's Disease. Int J Mol Sci 2023; 24:ijms24043200. [PMID: 36834612 PMCID: PMC9965628 DOI: 10.3390/ijms24043200] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 02/01/2023] [Accepted: 02/03/2023] [Indexed: 02/09/2023] Open
Abstract
Alzheimer's disease (AD) represents the most prevalent type of dementia in elderly people, primarily characterized by brain accumulation of beta-amyloid (Aβ) peptides, derived from Amyloid Precursor Protein (APP), in the extracellular space (amyloid plaques) and intracellular deposits of the hyperphosphorylated form of the protein tau (p-tau; tangles or neurofibrillary aggregates). The Nerve growth factor receptor (NGFR/p75NTR) represents a low-affinity receptor for all known mammalians neurotrophins (i.e., proNGF, NGF, BDNF, NT-3 e NT-4/5) and it is involved in pathways that determine both survival and death of neurons. Interestingly, also Aβ peptides can blind to NGFR/p75NTR making it the "ideal" candidate in mediating Aβ-induced neuropathology. In addition to pathogenesis and neuropathology, several data indicated that NGFR/p75NTR could play a key role in AD also from a genetic perspective. Other studies suggested that NGFR/p75NTR could represent a good diagnostic tool, as well as a promising therapeutic target for AD. Here, we comprehensively summarize and review the current experimental evidence on this topic.
Collapse
|
7
|
Differential dysregulation of CREB and synaptic genes in transgenic Drosophila melanogaster expressing shaggy (GSK3), Tau WT, or Amyloid-beta. Mol Biol Rep 2023; 50:1101-1108. [PMID: 36399243 DOI: 10.1007/s11033-022-08059-9] [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: 08/26/2022] [Accepted: 10/25/2022] [Indexed: 11/19/2022]
Abstract
BACKGROUND Tau, Amyloid-beta (Aβ42), and Glycogen synthase kinase 3 (GSK3) contribute to synaptic dysfunction observed in Alzheimer's disease (AD), the most common form of dementia. In the current study, the effect of pan-neuronal expression of TauWT, Aβ42, or shaggy (orthologue of GSK3) in Drosophila melanogaster was assessed on the locomotor function, ethanol sensitivity, synaptic genes and CREB expression. The effect of TauWT and Aβ42 on the expression of shaggy was also determined. METHODS AND RESULTS Gene expression analysis was performed using quantitative real-time RT-PCR method. While syt1, SNAP25 and CREB (upstream transcription factor of syt1 and SNAP25) were upregulated in flies expressing TauWT or Aβ42, a prominent decline was observed in those genes in shaggy expressing flies. Although all transgenic flies showed climbing disability and higher sensitivity to ethanol, abnormality in these features was significantly more prominent in transgenic flies expressing shaggy compared to TauWT or Aβ42. Despite a significant upregulation of shaggy transcription in TauWT expressing flies, Aβ42 transgenic flies witnessed no significant changes. CONCLUSIONS TauWT, Aβ42, and shaggy may affect synaptic plasticity through dysregulation of synaptic genes and CREB, independently. However shaggy has more detrimental effect on synaptic genes expression, locomotor ability and sensitivity to ethanol. It is important when it comes to drug discovery. It appears that CREB is a direct effector of changes in synaptic genes expression as they showed similar pattern of alteration and it is likely to be a part of compensatory mechanisms independent of the GSK3/CREB pathway in TauWT or Aβ42 expressing flies.
Collapse
|
8
|
Guin PS, Roy S. Recently Reported Ru-Metal Organic Coordination Complexes and Their Application (A Review). RUSS J GEN CHEM+ 2022. [DOI: 10.1134/s1070363222080242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
9
|
Bruno F, Malvaso A, Canterini S, Bruni AC. Antimicrobial Peptides (AMPs) in the Pathogenesis of Alzheimer's Disease: Implications for Diagnosis and Treatment. Antibiotics (Basel) 2022; 11:726. [PMID: 35740133 PMCID: PMC9220182 DOI: 10.3390/antibiotics11060726] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 05/25/2022] [Accepted: 05/26/2022] [Indexed: 02/04/2023] Open
Abstract
Alzheimer's disease (AD) represents the most frequent type of dementia in elderly people. There are two major forms of the disease: sporadic (SAD)-whose causes are not completely understood-and familial (FAD)-with clear autosomal dominant inheritance. The two main hallmarks of AD are extracellular deposits of amyloid-beta (Aβ) peptide and intracellular deposits of the hyperphosphorylated form of the tau protein (P-tau). An ever-growing body of research supports the infectious hypothesis of sporadic forms of AD. Indeed, it has been documented that some pathogens, such as herpesviruses and certain bacterial species, are commonly present in AD patients, prompting recent clinical research to focus on the characterization of antimicrobial peptides (AMPs) in this pathology. The literature also demonstrates that Aβ can be considered itself as an AMP; thus, representing a type of innate immune defense peptide that protects the host against a variety of pathogens. Beyond Aβ, other proteins with antimicrobial activity, such as lactoferrin, defensins, cystatins, thymosin β4, LL37, histatin 1, and statherin have been shown to be involved in AD. Here, we summarized and discussed these findings and explored the diagnostic and therapeutic potential of AMPs in AD.
Collapse
Affiliation(s)
- Francesco Bruno
- Regional Neurogenetic Centre (CRN), Department of Primary Care, ASP Catanzaro, 88046 Lamezia Terme, Italy
- Association for Neurogenetic Research (ARN), 88046 Lamezia Terme, Italy;
| | - Antonio Malvaso
- Neurology Unit, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy;
| | - Sonia Canterini
- Division of Neuroscience, Department of Psychology, University La Sapienza, 00158 Rome, Italy;
| | | |
Collapse
|
10
|
Biyong EF, Tremblay C, Leclerc M, Caron V, Alfos S, Helbling JC, Rodriguez L, Pernet V, Bennett DA, Pallet V, Calon F. Role of Retinoid X Receptors (RXRs) and dietary vitamin A in Alzheimer's disease: Evidence from clinicopathological and preclinical studies. Neurobiol Dis 2021; 161:105542. [PMID: 34737043 DOI: 10.1016/j.nbd.2021.105542] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 10/27/2021] [Accepted: 10/31/2021] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Vitamin A (VitA), via its active metabolite retinoic acid (RA), is critical for the maintenance of memory function with advancing age. Although its role in Alzheimer's disease (AD) is not well understood, data suggest that impaired brain VitA signaling is associated with the accumulation of β-amyloid peptides (Aβ), and could thus contribute to the onset of AD. METHODS We evaluated the protective action of a six-month-long dietary VitA-supplementation (20 IU/g), starting at 8 months of age, on the memory and the neuropathology of the 3xTg-AD mouse model of AD (n = 11-14/group; including 4-6 females and 7-8 males). We also measured protein levels of Retinoic Acid Receptor β (RARβ) and Retinoid X Receptor γ (RXRγ) in homogenates from the inferior parietal cortex of 60 participants of the Religious Orders study (ROS) divided in three groups: no cognitive impairment (NCI) (n = 20), mild cognitive impairment (MCI) (n = 20) and AD (n = 20). RESULTS The VitA-enriched diet preserved spatial memory of 3xTg-AD mice in the Y maze. VitA-supplementation affected hippocampal RXR expression in an opposite way according to sex by tending to increase in males and decrease in females their mRNA expression. VitA-enriched diet also reduced the amount of hippocampal Aβ40 and Aβ42, as well as the phosphorylation of tau protein at sites Ser396/Ser404 (PHF-1) in males. VitA-supplementation had no effect on tau phosphorylation in females but worsened their hippocampal Aβ load. However, the expression of Rxr-β in the hippocampus was negatively correlated with the amount of both soluble and insoluble Aβ in both males and females. Western immunoblotting in the human cortical samples of the ROS study did not reveal differences in RARβ levels. However, it evidenced a switch from a 60-kDa-RXRγ to a 55-kDa-RXRγ in AD, correlating with ante mortem cognitive decline and the accumulation of neuritic plaques in the brain cortex. CONCLUSION Our data suggest that (i) an altered expression of RXRs receptors is a contributor to β-amyloid pathology in both humans and 3xTg-AD mice, (ii) a chronic exposure of 3xTg-AD mice to a VitA-enriched diet may be protective in males, but not in females.
Collapse
Affiliation(s)
- Essi F Biyong
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France; Faculté de pharmacie, Université Laval, Québec, Québec, Canada; Centre de recherche du CHU de Québec-Université Laval (CHUL), Axe Neurosciences, 2705 Boulevard Laurier, Québec, Québec, Canada; Institut sur la Nutrition et les Aliments Fonctionnels (INAF), Québec, Québec, Canada; LIA OptiNutriBrain - Laboratoire International Associé (NutriNeuro France-INAF Canada), Canada
| | - Cyntia Tremblay
- Faculté de pharmacie, Université Laval, Québec, Québec, Canada; Centre de recherche du CHU de Québec-Université Laval (CHUL), Axe Neurosciences, 2705 Boulevard Laurier, Québec, Québec, Canada
| | - Manon Leclerc
- Faculté de pharmacie, Université Laval, Québec, Québec, Canada; Centre de recherche du CHU de Québec-Université Laval (CHUL), Axe Neurosciences, 2705 Boulevard Laurier, Québec, Québec, Canada
| | - Vicky Caron
- Faculté de pharmacie, Université Laval, Québec, Québec, Canada; Centre de recherche du CHU de Québec-Université Laval (CHUL), Axe Neurosciences, 2705 Boulevard Laurier, Québec, Québec, Canada
| | - Serge Alfos
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | | | - Léa Rodriguez
- CUO-Recherche, Centre de Recherche du CHU de Québec, Québec, QC, Canada; Département d'ophtalmologie, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - Vincent Pernet
- CUO-Recherche, Centre de Recherche du CHU de Québec, Québec, QC, Canada; Département d'ophtalmologie, Faculté de Médecine, Université Laval, Québec, QC, Canada
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Véronique Pallet
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France; LIA OptiNutriBrain - Laboratoire International Associé (NutriNeuro France-INAF Canada), Canada
| | - Frédéric Calon
- Faculté de pharmacie, Université Laval, Québec, Québec, Canada; Centre de recherche du CHU de Québec-Université Laval (CHUL), Axe Neurosciences, 2705 Boulevard Laurier, Québec, Québec, Canada; Institut sur la Nutrition et les Aliments Fonctionnels (INAF), Québec, Québec, Canada; LIA OptiNutriBrain - Laboratoire International Associé (NutriNeuro France-INAF Canada), Canada.
| |
Collapse
|
11
|
Shinjyo N, Kagaya W, Pekna M. Interaction Between the Complement System and Infectious Agents - A Potential Mechanistic Link to Neurodegeneration and Dementia. Front Cell Neurosci 2021; 15:710390. [PMID: 34408631 PMCID: PMC8365172 DOI: 10.3389/fncel.2021.710390] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Accepted: 07/09/2021] [Indexed: 12/24/2022] Open
Abstract
As part of the innate immune system, complement plays a critical role in the elimination of pathogens and mobilization of cellular immune responses. In the central nervous system (CNS), many complement proteins are locally produced and regulate nervous system development and physiological processes such as neural plasticity. However, aberrant complement activation has been implicated in neurodegeneration, including Alzheimer's disease. There is a growing list of pathogens that have been shown to interact with the complement system in the brain but the short- and long-term consequences of infection-induced complement activation for neuronal functioning are largely elusive. Available evidence suggests that the infection-induced complement activation could be protective or harmful, depending on the context. Here we summarize how various infectious agents, including bacteria (e.g., Streptococcus spp.), viruses (e.g., HIV and measles virus), fungi (e.g., Candida spp.), parasites (e.g., Toxoplasma gondii and Plasmodium spp.), and prion proteins activate and manipulate the complement system in the CNS. We also discuss the potential mechanisms by which the interaction between the infectious agents and the complement system can play a role in neurodegeneration and dementia.
Collapse
Affiliation(s)
- Noriko Shinjyo
- Laboratory of Immune Homeostasis, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
- School of Tropical Medicine and Global Health, Nagasaki University, Nagasaki, Japan
| | - Wataru Kagaya
- Department of Parasitology and Research Center for Infectious Disease Sciences, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Marcela Pekna
- Laboratory of Regenerative Neuroimmunology, Center for Brain Repair, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| |
Collapse
|
12
|
Raj A. Graph Models of Pathology Spread in Alzheimer's Disease: An Alternative to Conventional Graph Theoretic Analysis. Brain Connect 2021; 11:799-814. [PMID: 33858198 DOI: 10.1089/brain.2020.0905] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Background: Graph theory and connectomics are new techniques for uncovering disease-induced changes in the brain's structural network. Most prior studied have focused on network statistics as biomarkers of disease. However, an emerging body of work involves exploring how the network serves as a conduit for the propagation of disease factors in the brain and has successfully mapped the functional and pathological consequences of disease propagation. In Alzheimer's disease (AD), progressive deposition of misfolded proteins amyloid and tau is well-known to follow fiber projections, under a "prion-like" trans-neuronal transmission mechanism, through which misfolded proteins cascade along neuronal pathways, giving rise to network spread. Methods: In this review, we survey the state of the art in mathematical modeling of connectome-mediated pathology spread in AD. Then we address several open questions that are amenable to mathematically precise parsimonious modeling of pathophysiological processes, extrapolated to the whole brain. We specifically identify current formal models of how misfolded proteins are produced, aggregate, and disseminate in brain circuits, and attempt to understand how this process leads to stereotyped progression in Alzheimer's and other related diseases. Conclusion: This review serves to unify current efforts in modeling of AD progression that together have the potential to explain observed phenomena and serve as a test-bed for future hypothesis generation and testing in silico. Impact statement Graph theory is a powerful new approach that is transforming the study of brain processes. There do not exist many focused reviews of the subfield of graph modeling of how Alzheimer's and other dementias propagate within the brain network, and how these processes can be mapped mathematically. By providing timely and topical review of this subfield, we fill a critical gap in the community and present a unified view that can serve as an in silico test-bed for future hypothesis generation and testing. We also point to several open and unaddressed questions and controversies that future practitioners can tackle.
Collapse
Affiliation(s)
- Ashish Raj
- Department of Radiology and Biomedical Imaging, University of California at San Francisco, San Francisco, California, USA
| |
Collapse
|
13
|
Sánchez-Sarasúa S, Fernández-Pérez I, Espinosa-Fernández V, Sánchez-Pérez AM, Ledesma JC. Can We Treat Neuroinflammation in Alzheimer's Disease? Int J Mol Sci 2020; 21:E8751. [PMID: 33228179 PMCID: PMC7699542 DOI: 10.3390/ijms21228751] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 11/13/2020] [Accepted: 11/16/2020] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease (AD), considered the most common type of dementia, is characterized by a progressive loss of memory, visuospatial, language and complex cognitive abilities. In addition, patients often show comorbid depression and aggressiveness. Aging is the major factor contributing to AD; however, the initial cause that triggers the disease is yet unknown. Scientific evidence demonstrates that AD, especially the late onset of AD, is not the result of a single event, but rather it appears because of a combination of risk elements with the lack of protective ones. A major risk factor underlying the disease is neuroinflammation, which can be activated by different situations, including chronic pathogenic infections, prolonged stress and metabolic syndrome. Consequently, many therapeutic strategies against AD have been designed to reduce neuro-inflammation, with very promising results improving cognitive function in preclinical models of the disease. The literature is massive; thus, in this review we will revise the translational evidence of these early strategies focusing in anti-diabetic and anti-inflammatory molecules and discuss their therapeutic application in humans. Furthermore, we review the preclinical and clinical data of nutraceutical application against AD symptoms. Finally, we introduce new players underlying neuroinflammation in AD: the activity of the endocannabinoid system and the intestinal microbiota as neuroprotectors. This review highlights the importance of a broad multimodal approach to treat successfully the neuroinflammation underlying AD.
Collapse
Affiliation(s)
| | | | | | - Ana María Sánchez-Pérez
- Neurobiotechnology Group, Department of Medicine, Health Science Faculty, Universitat Jaume I, 12071 Castellón, Spain; (S.S.-S.); (I.F.-P.); (V.E.-F.)
| | - Juan Carlos Ledesma
- Neurobiotechnology Group, Department of Medicine, Health Science Faculty, Universitat Jaume I, 12071 Castellón, Spain; (S.S.-S.); (I.F.-P.); (V.E.-F.)
| |
Collapse
|
14
|
Angeli S, Kousiappa I, Stavrou M, Sargiannidou I, Georgiou E, Papacostas SS, Kleopa KA. Altered Expression of Glial Gap Junction Proteins Cx43, Cx30, and Cx47 in the 5XFAD Model of Alzheimer's Disease. Front Neurosci 2020; 14:582934. [PMID: 33117125 PMCID: PMC7575794 DOI: 10.3389/fnins.2020.582934] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 09/14/2020] [Indexed: 11/13/2022] Open
Abstract
Glial gap junction proteins, called connexins (Cxs), form gap junctions in the central nervous system (CNS) to allow the bidirectional cytosolic exchange of molecules between adjacent cells. Their involvement in inheritable diseases and the use of experimental animal models that closely mimic such diseases revealed the critical role of glial GJs in myelination and homeostasis. Cxs are also implicated in acquired demyelinating disorders, such as Multiple Sclerosis (MS) and Alzheimer's disease (AD). Animal and human studies have revealed a role of the astrocytic Cx43 in the progression of AD but the role of Cx47, which is the main partner of Cx43 in the astrocyte-oligodendrocyte GJs is still unknown. The aim of this study was to investigate the astrocytic connexins, Cx43 and Cx30 in relation to oligodendrocytic Cx47 in the cortex and thalamus of the 5XFAD mouse model of AD. The model was characterized by increased Aβ deposition, gliosis, neuronal loss, and memory impairment. Compared to wild-type mice, Cx43 and Cx30 showed increased immunoreactivity in older 5XFAD mice, reflecting astrogliosis, while Cx47 immunoreactivity was reduced. Moreover, Cx47 GJ plaques showed reduced colocalization with Cx43 plaques. Oligodendrocyte precursor cells (OPCs) and mature oligodendrocyte populations were also depleted, and myelin deficits were observed. Our findings indicate reduced astrocyte-oligodendrocyte gap junction connectivity and possibly a shift in Cx43 expression toward astrocyte-astrocyte gap junctions and/or hemichannels, that could impair oligodendrocyte homeostasis and myelination. However, other factors, such as Aβ toxicity, could directly affect oligodendrocyte survival in AD. Our study provides evidence that Cxs might have implications in the progression of AD, although the role of oligodendrocyte Cxs in AD requires further investigation.
Collapse
Affiliation(s)
- Stella Angeli
- Neurobiology Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
- Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | - Ioanna Kousiappa
- Neurobiology Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
- Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | - Marios Stavrou
- Department of Electrical and Computer Engineering, Faculty of Engineering, University of Cyprus, Nicosia, Cyprus
| | - Irene Sargiannidou
- Cyprus School of Molecular Medicine, Nicosia, Cyprus
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Elena Georgiou
- Cyprus School of Molecular Medicine, Nicosia, Cyprus
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Savvas S. Papacostas
- Neurobiology Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
- Cyprus School of Molecular Medicine, Nicosia, Cyprus
- Dementia and Cognitive Disorders Center, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
- Medical School, University of Nicosia, Nicosia, Cyprus
| | - Kleopas A. Kleopa
- Cyprus School of Molecular Medicine, Nicosia, Cyprus
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
- Center for Neuromuscular disorders, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
- Center for Multiple Sclerosis and Related Disorders, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| |
Collapse
|
15
|
Poon CH, Wang Y, Fung ML, Zhang C, Lim LW. Rodent Models of Amyloid-Beta Feature of Alzheimer's Disease: Development and Potential Treatment Implications. Aging Dis 2020; 11:1235-1259. [PMID: 33014535 PMCID: PMC7505263 DOI: 10.14336/ad.2019.1026] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 10/26/2019] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder worldwide and causes severe financial and social burdens. Despite much research on the pathogenesis of AD, the neuropathological mechanisms remain obscure and current treatments have proven ineffective. In the past decades, transgenic rodent models have been used to try to unravel this disease, which is crucial for early diagnosis and the assessment of disease-modifying compounds. In this review, we focus on transgenic rodent models used to study amyloid-beta pathology in AD. We also discuss their possible use as promising tools for AD research. There is still no effective treatment for AD and the development of potent therapeutics are urgently needed. Many molecular pathways are susceptible to AD, ranging from neuroinflammation, immune response, and neuroplasticity to neurotrophic factors. Studying these pathways may shed light on AD pathophysiology as well as provide potential targets for the development of more effective treatments. This review discusses the advantages and limitations of these models and their potential therapeutic implications for AD.
Collapse
Affiliation(s)
- Chi Him Poon
- 1School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yingyi Wang
- 1School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Man-Lung Fung
- 1School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Chengfei Zhang
- 2Endodontology, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Lee Wei Lim
- 1School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
16
|
Li J, Wang G, Qin Y, Zhang X, Wang HF, Liu HW, Zhu LJ, Yao XS. Neuroprotective constituents from the aerial parts of Cannabis sativa L. subsp. sativa. RSC Adv 2020; 10:32043-32049. [PMID: 35518127 PMCID: PMC9056577 DOI: 10.1039/d0ra04565a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 08/12/2020] [Indexed: 11/21/2022] Open
Abstract
Five new compounds including three new cannabinoids, cannabisativas A-C (1-3), two new phenolic acids, (7Z,9Z)-cannabiphenolic acid A (4) and (8S,9Z)-cannabiphenolic acid B (5), together with twelve known compounds (6-17), were isolated from the aerial parts of Cannabis sativa L. subsp. sativa. The structures of 1-5 were established on the basis of extensive 1D, 2D NMR and HRESIMS analysis. The absolute configurations were determined by comparison between their experimental and calculated spectra of electronic circular dichroism (ECD) or the modified Mosher's method. The neuroprotective effects of the compounds 1-17 were evaluated on PC 12 cells. Compounds 12, 13 and 15 showed potential protective effects against H2O2-induced damage.
Collapse
Affiliation(s)
- Jia Li
- College of Traditional Chinese Materia Medica, Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University Shenyang 110016 China
| | - Guan Wang
- College of Traditional Chinese Materia Medica, Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University Shenyang 110016 China
- State Key Laboratory of Biotherapy, Cancer Center, Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy Chengdu 610041 China
| | - Yu Qin
- College of Traditional Chinese Materia Medica, Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University Shenyang 110016 China
| | - Xue Zhang
- College of Traditional Chinese Materia Medica, Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University Shenyang 110016 China
| | - Hai-Feng Wang
- College of Traditional Chinese Materia Medica, Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University Shenyang 110016 China
| | - Hong-Wei Liu
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences Beijing 100101 China
| | - Ling-Juan Zhu
- College of Traditional Chinese Materia Medica, Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University Shenyang 110016 China
- State Key Laboratory of Biotherapy, Cancer Center, Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy Chengdu 610041 China
| | - Xin-Sheng Yao
- College of Traditional Chinese Materia Medica, Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University Shenyang 110016 China
| |
Collapse
|
17
|
Cavalcante SFDA, Simas ABC, Barcellos MC, de Oliveira VGM, Sousa RB, Cabral PADM, Kuča K, França TCC. Acetylcholinesterase: The "Hub" for Neurodegenerative Diseases and Chemical Weapons Convention. Biomolecules 2020; 10:E414. [PMID: 32155996 PMCID: PMC7175162 DOI: 10.3390/biom10030414] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 03/02/2020] [Accepted: 03/03/2020] [Indexed: 12/12/2022] Open
Abstract
This article describes acetylcholinesterase (AChE), an enzyme involved in parasympathetic neurotransmission, its activity, and how its inhibition can be pharmacologically useful for treating dementia, caused by Alzheimer's disease, or as a warfare method due to the action of nerve agents. The chemical concepts related to the irreversible inhibition of AChE, its reactivation, and aging are discussed, along with a relationship to the current international legislation on chemical weapons.
Collapse
Affiliation(s)
- Samir F. de A. Cavalcante
- Institute of Chemical, Biological, Radiological and Nuclear Defense (IDQBRN), Brazilian Army Technological Center (CTEx), Avenida das Américas 28705, Rio de Janeiro 23020-470, Brazil; (M.C.B.); (V.G.M.d.O.); (R.B.S.); (P.A.d.M.C.)
- Walter Mors Institute of Research on Natural Products (IPPN), Federal University of Rio de Janeiro (UFRJ), CCS, Bloco H, Rio de Janeiro 21941-902, Brazil
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Rokitanskeho 62, 50003 Hradec Králové, Czech Republic
| | - Alessandro B. C. Simas
- Walter Mors Institute of Research on Natural Products (IPPN), Federal University of Rio de Janeiro (UFRJ), CCS, Bloco H, Rio de Janeiro 21941-902, Brazil
| | - Marcos C. Barcellos
- Institute of Chemical, Biological, Radiological and Nuclear Defense (IDQBRN), Brazilian Army Technological Center (CTEx), Avenida das Américas 28705, Rio de Janeiro 23020-470, Brazil; (M.C.B.); (V.G.M.d.O.); (R.B.S.); (P.A.d.M.C.)
| | - Victor G. M. de Oliveira
- Institute of Chemical, Biological, Radiological and Nuclear Defense (IDQBRN), Brazilian Army Technological Center (CTEx), Avenida das Américas 28705, Rio de Janeiro 23020-470, Brazil; (M.C.B.); (V.G.M.d.O.); (R.B.S.); (P.A.d.M.C.)
| | - Roberto B. Sousa
- Institute of Chemical, Biological, Radiological and Nuclear Defense (IDQBRN), Brazilian Army Technological Center (CTEx), Avenida das Américas 28705, Rio de Janeiro 23020-470, Brazil; (M.C.B.); (V.G.M.d.O.); (R.B.S.); (P.A.d.M.C.)
| | - Paulo A. de M. Cabral
- Institute of Chemical, Biological, Radiological and Nuclear Defense (IDQBRN), Brazilian Army Technological Center (CTEx), Avenida das Américas 28705, Rio de Janeiro 23020-470, Brazil; (M.C.B.); (V.G.M.d.O.); (R.B.S.); (P.A.d.M.C.)
| | - Kamil Kuča
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Rokitanskeho 62, 50003 Hradec Králové, Czech Republic
| | - Tanos C. C. França
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Rokitanskeho 62, 50003 Hradec Králové, Czech Republic
- Laboratory of Molecular Modelling Applied to Chemical and Biological Defense (LMACBD), Military Institute of Engineering (IME), Praça General Tibúrcio 80, Rio de Janeiro 22290-270, Brazil
| |
Collapse
|
18
|
Gao Y, Yan Y, Fang Q, Zhang N, Kumar G, Zhang J, Song LJ, Yu J, Zhao L, Zhang HT, Ma CG. The Rho kinase inhibitor fasudil attenuates Aβ 1-42-induced apoptosis via the ASK1/JNK signal pathway in primary cultures of hippocampal neurons. Metab Brain Dis 2019; 34:1787-1801. [PMID: 31482248 DOI: 10.1007/s11011-019-00487-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 08/27/2019] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD), a chronic, progressive, neurodegenerative disorder, is the most common type of dementia. Beta amyloid (Aβ) peptide aggregation and phosphorylated tau protein accumulation are considered as one of the causes for AD. Our previous studies have demonstrated the neuroprotective effect of the Rho kinase inhibitor fasudil, but the mechanism remains elucidated. In the present study, we examined the effects of fasudil on Aβ1-42 aggregation and apoptosis and identified the intracellular signaling pathways involved in these actions in primary cultures of mouse hippocampal neurons. The results showed that fasudil increased neurite outgrowth (52.84%), decreased Aβ burden (46.65%), Tau phosphorylation (96.84%), and ROCK-II expression. In addition, fasudil reversed Aβ1-42-induced decreased expression of Bcl-2 and increases in caspase-3, cleaved-PARP, phospho-JNK(Thr183/Tyr185), and phospho-ASK1(Ser966). Further, fasudil decreased mitochondrial membrane potential and intracellular calcium overload in the neurons treated with Aβ1-42. These results suggest that inhibition of Rho kinase by fasudil reverses Aβ1-42-induced neuronal apoptosis via the ASK1/JNK signal pathway, calcium ions, and mitochondrial membrane potential. Fasudil could be a drug of choice for treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Ye Gao
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, China
| | - Yuqing Yan
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, China.
| | - Qingli Fang
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, China
| | - Nianping Zhang
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, China
| | - Gajendra Kumar
- Department of Biomedical Sciences, City University of Hong Kong, Tat Chee Avenue, Hong Kong
- Bio-Signal technologies (HK) Limited, 9th Floor, Amtel Building,148 Des Voeux Road Central, Central, Hong Kong
| | - Jihong Zhang
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, China
| | - Li-Juan Song
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Jiezhong Yu
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, China
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Linhu Zhao
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, China
| | - Han-Ting Zhang
- Departments of Neuroscience and Behavioral Medicine & Psychiatry, the Rockefeller Neurosciences Institute, West Virginia University Health Sciences Center, Morgantown, WV, 26506, USA.
| | - Cun-Gen Ma
- Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, China.
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Taiyuan, China.
| |
Collapse
|
19
|
New barrigenol-type triterpenoids with anti-Alzheimer’s disease activity from Koelreuteria paniculata Laxm. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.103459] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
20
|
Zhang X, Zhang S, Yang Y, Wang D, Gao H. Natural barrigenol-like triterpenoids: A comprehensive review of their contributions to medicinal chemistry. PHYTOCHEMISTRY 2019; 161:41-74. [PMID: 30818173 DOI: 10.1016/j.phytochem.2019.01.017] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 01/03/2019] [Accepted: 01/30/2019] [Indexed: 05/07/2023]
Abstract
Barrigenol-like triterpenoids (BATs), which contain an unusual oleanane substituted by many hydroxyl groups as the skeleton, are subdivided into five subtypes: barrigenol A1, barrigenol A2, barrigenol R1, barringtogenol C, and 16-deoxybarringtogenol C. The variations in acyl derivatives, hydroxyl groups, and carbohydrate chains in their structures have enhanced the diversity of BATs. Moreover, the stable polyhydroxy-replaced pentacyclic skeleton provides an ideal platform for structural modifications. To date, more than 500 BAT derivatives have been isolated from plants. Synchronously, BATs possess anti-tumour, anti-Alzheimer's disease, anti-inflammatory, anti-microbial, anti-obesity and anti-allergic activities by regulating numerous cellular molecules. Some BAT derivatives, such as escin obtained from Aesculus hippocastanum L. and xanthoceraside isolated from Xanthoceras sorbifolia Bunge, have been used to treat encephaloedema or inflammatory diseases. This review aims to provide comprehensive information about the chemistry, sources, bioavailability, and anti-tumour effects of BATs, with a particular emphasis on the molecular mechanisms of action. The pharmacokinetics and clinical progress are also concerned. More than 300 structures identified over past 25 years are summarized here (249 compounds) and in the supplementary information (114 compounds). Accordingly, the pharmaceutical activity of barrigenol triterpenoids suggests that some compounds should be developed as promising anti-tumour or anti- Alzheimer's disease agents in future.
Collapse
Affiliation(s)
- Xinxin Zhang
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Song Zhang
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Yiren Yang
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Da Wang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China.
| | - Huiyuan Gao
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China.
| |
Collapse
|
21
|
Nanoemulsions in CNS drug delivery: recent developments, impacts and challenges. Drug Discov Today 2019; 24:1104-1115. [PMID: 30914298 DOI: 10.1016/j.drudis.2019.03.021] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 03/03/2019] [Accepted: 03/20/2019] [Indexed: 02/06/2023]
Abstract
Despite enormous efforts, treatment of CNS diseases remains challenging. One of the main issues causing this situation is limited CNS access for the majority of drugs used as part of the therapeutic regimens against life-threatening CNS diseases. Regarding the inarguable position of the nanocarrier systems in neuropharmacokinetic enhancement of the CNS drugs, this review discusses the latest findings on nanoemulsions (NEs) as one of the most promising candidates of this type, to overcome the challenges of CNS drug delivery. Future development of NE-based CNS drug delivery needs to consider so many aspects not only from a physicochemical point of view but also related to the biointerface of these very small droplets before achieving clinical value.
Collapse
|
22
|
Mechanisms Associated with Type 2 Diabetes as a Risk Factor for Alzheimer-Related Pathology. Mol Neurobiol 2019; 56:5815-5834. [PMID: 30684218 DOI: 10.1007/s12035-019-1475-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 01/10/2019] [Indexed: 12/19/2022]
Abstract
Current evidence suggests dementia and pathology in Alzheimer's Disease (AD) are both dependent and independent of amyloid processing and can be induced by multiple 'hits' on vital neuronal functions. Type 2 diabetes (T2D) poses the most important risk factor for developing AD after ageing and dysfunctional IR/PI3K/Akt signalling is a major contributor in both diseases. We developed a model of T2D, coupling subdiabetogenic doses of streptozotocin (STZ) with a human junk food (HJF) diet to more closely mimic the human condition. Over 35 weeks, this induced classic signs of T2D (hyperglycemia and insulin dysfunction) and a modest, but stable deficit in spatial recognition memory, with very little long-term modification of proteins in or associated with IR/PI3K/Akt signalling in CA1 of the hippocampus. Intracerebroventricular infusion of soluble amyloid beta 42 (Aβ42) to mimic the early preclinical rise in Aβ alone induced a more severe, but short-lasting deficits in memory and deregulation of proteins. Infusion of Aβ on the T2D phenotype exacerbated and prolonged the memory deficits over approximately 4 months, and induced more severe aberrant regulation of proteins associated with autophagy, inflammation and glucose uptake from the periphery. A mild form of environmental enrichment transiently rescued memory deficits and could reverse the regulation of some, but not all protein changes. Together, these data identify mechanisms by which T2D could create a modest dysfunctional neuronal milieu via multiple and parallel inputs that permits the development of pathological events identified in AD and memory deficits when Aβ levels are transiently effective in the brain.
Collapse
|
23
|
Chornenkyy Y, Wang W, Wei A, Nelson PT. Alzheimer's disease and type 2 diabetes mellitus are distinct diseases with potential overlapping metabolic dysfunction upstream of observed cognitive decline. Brain Pathol 2019; 29:3-17. [PMID: 30106209 PMCID: PMC6427919 DOI: 10.1111/bpa.12655] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 08/09/2018] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease (AD) and type 2 diabetes mellitus (T2DM) are highly prevalent aging-related diseases associated with significant morbidity and mortality. Some findings in human and animal models have linked T2DM to AD-type dementia. Despite epidemiological associations between the T2DM and cognitive impairment, the interrelational mechanisms are unclear. The preponderance of evidence in longitudinal studies with autopsy confirmation have indicated that vascular mechanisms, rather than classic AD-type pathologies, underlie the cognitive decline often seen in self-reported T2DM. T2DM is associated with cardiovascular and cerebrovascular disease (CVD), and is associated with increased risk of infarcts and small vessel disease in the brain and other organs. Neuropathological examinations of post-mortem brains demonstrated evidence of cerebrovascular disease and little to no correlation between T2DM and β-amyloid deposits or neurofibrillary tangles. Nevertheless, the mechanisms upstream of early AD-specific pathology remain obscure. In this regard, there may indeed be overlap between the pathologic mechanisms of T2DM/"metabolic syndrome," and AD. More specifically, cerebral insulin processing, glucose metabolism, mitochondrial function, and/or lipid metabolism could be altered in patients in early AD and directly influence symptomatology and/or neuropathology.
Collapse
Affiliation(s)
| | - Wang‐Xia Wang
- University of Kentucky College of MedicineLexingtonKY
- Sanders‐Brown Center on Aging, Department of PathologyUniversity of KentuckyLexingtonKY
| | - Angela Wei
- Department of BiologyUniversity of KentuckyLexingtonKY
| | - Peter T. Nelson
- University of Kentucky College of MedicineLexingtonKY
- Sanders‐Brown Center on Aging, Department of PathologyUniversity of KentuckyLexingtonKY
| |
Collapse
|
24
|
Morris G, Berk M, Maes M, Puri BK. Could Alzheimer's Disease Originate in the Periphery and If So How So? Mol Neurobiol 2019; 56:406-434. [PMID: 29705945 PMCID: PMC6372984 DOI: 10.1007/s12035-018-1092-y] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Accepted: 04/17/2018] [Indexed: 12/11/2022]
Abstract
The classical amyloid cascade model for Alzheimer's disease (AD) has been challenged by several findings. Here, an alternative molecular neurobiological model is proposed. It is shown that the presence of the APOE ε4 allele, altered miRNA expression and epigenetic dysregulation in the promoter region and exon 1 of TREM2, as well as ANK1 hypermethylation and altered levels of histone post-translational methylation leading to increased transcription of TNFA, could variously explain increased levels of peripheral and central inflammation found in AD. In particular, as a result of increased activity of triggering receptor expressed on myeloid cells 2 (TREM-2), the presence of the apolipoprotein E4 (ApoE4) isoform, and changes in ANK1 expression, with subsequent changes in miR-486 leading to altered levels of protein kinase B (Akt), mechanistic (previously mammalian) target of rapamycin (mTOR) and signal transducer and activator of transcription 3 (STAT3), all of which play major roles in microglial activation, proliferation and survival, there is activation of microglia, leading to the subsequent (further) production of cytokines, chemokines, nitric oxide, prostaglandins, reactive oxygen species, inducible nitric oxide synthase and cyclooxygenase-2, and other mediators of inflammation and neurotoxicity. These changes are associated with the development of amyloid and tau pathology, mitochondrial dysfunction (including impaired activity of the electron transport chain, depleted basal mitochondrial potential and oxidative damage to key tricarboxylic acid enzymes), synaptic dysfunction, altered glycogen synthase kinase-3 (GSK-3) activity, mTOR activation, impairment of autophagy, compromised ubiquitin-proteasome system, iron dyshomeostasis, changes in APP translation, amyloid plaque formation, tau hyperphosphorylation and neurofibrillary tangle formation.
Collapse
Affiliation(s)
- Gerwyn Morris
- IMPACT Strategic Research Centre, School of Medicine, Barwon Health, Deakin University, P.O. Box 291, Geelong, Victoria, Australia
| | - Michael Berk
- IMPACT Strategic Research Centre, School of Medicine, Barwon Health, Deakin University, P.O. Box 291, Geelong, Victoria, Australia
- Department of Psychiatry, Level 1 North, Main Block, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia
- Florey Institute for Neuroscience and Mental Health, Kenneth Myer Building, University of Melbourne, 30 Royal Parade, Parkville, Victoria, Australia
- Orygen, The National Centre of Excellence in Youth Mental Health, 35 Poplar Rd, Parkville, Victoria, Australia
| | - Michael Maes
- IMPACT Strategic Research Centre, School of Medicine, Barwon Health, Deakin University, P.O. Box 291, Geelong, Victoria, Australia
- Department of Psychiatry, Chulalongkorn University, Bangkok, Thailand
| | - Basant K Puri
- Department of Medicine, Hammersmith Hospital, Imperial College London, London, UK.
| |
Collapse
|
25
|
De Loof A, Schoofs L. Intraluminal Farnesol and Farnesal in the Mealworm's Alimentary Canal: An Unusual Storage Site Uncovering Hidden Eukaryote Ca 2+-Homeostasis-Dependent "Golgicrine" Activities. Front Endocrinol (Lausanne) 2019; 10:885. [PMID: 31920991 PMCID: PMC6930878 DOI: 10.3389/fendo.2019.00885] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Accepted: 12/03/2019] [Indexed: 12/15/2022] Open
Abstract
Farnesol, the sesquiterpenoid precursor of the six presently known insect juvenile hormones (JHs) was for the first time chemically identified in 1961, not in JH synthesizing glands or whole body extracts, but in excrements of the mealworm Tenebrio molitor. This finding was thought to be irrelevant and remained unexplored. In 1970, it was reported that the fall to zero of the JH titer in both prediapausing adults and in last instar larvae of the Colorado potato beetle causes severe malfunctioning of the Golgi system in the fat body, among various other effects. This endomembrane system in the cytoplasm resides at the intersection of the secretory, lysosomal, and endocytic pathways and is required for the processing of secretory proteins. Why the Golgi needs farnesol-like endogenous sesquiterpenoids (FLS) for its proper functioning has also never been further investigated. In 1999, farnesol was found to be a natural endogenous ligand for particular types of voltage-gated Ca2+ channels in mammalian cells, a finding that also remained undervalued. Only since 2014 more attention has been paid to the functional research of the "noble unknown" farnesol, in particular to its Ca2+-homeostasis-related juvenilizing and anti-apoptotic activities. Here, we introduce the term "Golgicrine activity" that addresses the secretory activity of the RER-Golgi system from its role in Ca2+-homeostasis rather than from its conventional role in mere protein secretion. Golgicrine activity attributes the so far forgotten role of farnesol-like sesquiterpenoids in proper Golgi functioning, and unites the endocrine, exocrine and enterocrine functions of these sesquiterpenoids. This out of the box view may open novel perspectives for the better understanding of particular inflammatory bowel diseases and of neurodegenerative diseases as well, because the early initiation of Alzheimer's disease may possibly result from malfunctioning of the mevalonate-farnesol-cholesterol biosynthetic pathway and thus might be a farnesol- and Ca2+-homeostasis-dependent Golgicrine issue.
Collapse
|
26
|
Deak T, Savage LM. Preface: Setting the stage for understanding alcohol effects in late aging: A special issue including both human and rodent studies. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2019; 148:xiii-xxv. [PMID: 31733669 PMCID: PMC6998208 DOI: 10.1016/s0074-7742(19)30116-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
It is widely recognized that people worldwide are living longer than in previous decades, with formidable projections regarding the expansion of elderly age groups in the decades to come. Older individuals are also sustaining higher levels of alcohol consumption later in life, and binge drinking remains a prevalent pastime in a significant proportion of aged individuals. Older people are more sensitive to neurobehavioral effects of alcohol, and as individuals age, the cumulative impact of lifetime alcohol intake begins to emerge. This brief review provides a perspective on the emerging field of how alcohol interacts with the aging brain and sets the stage for understanding the relationship between alcohol and overall brain health. In doing so, we introduce a set of articles collected in this book series (all chapters available on PubMed) which spans human epidemiology and clinical outcomes, along with a series of neurobehavioral studies in preclinical (rodent) models. Because both natural aging as well as alcohol use and abuse include tell-tale signs of neuroinflammation (heightened expression of neuroimmune genes, activation of inflammatory signaling pathways, and signs of glial activation), particular emphasis is placed on the role of neuroinflammation in both aging- and alcohol-related alterations in neurobehavioral function, with special emphasis on the spectrum of cognitive dysfunction ranging from mild cognitive impairment to Alzheimer's associated brain pathology.
Collapse
Affiliation(s)
- Terrence Deak
- Behavioral Neuroscience Program, Department of Psychology, Developmental Exposure Alcohol Research Center, Binghamton University-SUNY, Binghamton, NY, United States.
| | - Lisa M Savage
- Behavioral Neuroscience Program, Department of Psychology, Developmental Exposure Alcohol Research Center, Binghamton University-SUNY, Binghamton, NY, United States
| |
Collapse
|
27
|
Abstract
This study was performed to identify the association between smoking and Alzheimer's disease (AD). To perform this meta-analysis based on case-control and cohort studies, PubMed, Google Scholar, and the CNKI electronic databases were searched through April 30, 2017. Our meta-analysis included 27 studies, including 16 that reported odds ratios (ORs) and 11 that reported hazard ratios (HRs) or ratio risks. No significant association was found between smoking and AD among the studies that reported ORs (1.020, 95% confidence interval [CI] = 0.812-1.281, I = 67.9%, random model, p < 0.001). A subgroup analysis revealed no significant difference between different smoking statuses. The pooled HRs revealed a significant association between smoking and AD (HR = 1.520, 95% CI = 1.194-1.934, I = 83.6%, random model, p < 0.001). Cumulative meta-analysis of the HRs revealed that the effect of smoking on AD tended to be stable over time. Smoking may confer an increased risk of AD, and this effect has tended to be stable over time.
Collapse
|
28
|
Pfeffer A, Munder T, Schreyer S, Klein C, Rasińska J, Winter Y, Steiner B. Behavioral and psychological symptoms of dementia (BPSD) and impaired cognition reflect unsuccessful neuronal compensation in the pre-plaque stage and serve as early markers for Alzheimer's disease in the APP23 mouse model. Behav Brain Res 2018; 347:300-313. [PMID: 29572105 DOI: 10.1016/j.bbr.2018.03.030] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 02/28/2018] [Accepted: 03/19/2018] [Indexed: 12/15/2022]
Abstract
Recent research on Alzheimer's disease (AD) focuses on processes prior to amyloid-beta plaque deposition accounting for the progress of the disease. However, early mechanisms of AD are still poorly understood and predictors of the disease in the pre-plaque stage essential for initiating an early therapy are lacking. Behavioral and psychological symptoms of dementia (BPSD) and potentially impaired cognition may serve as predictors and early clinical diagnostic markers for AD. To investigate potential BPSD and cognitive impairments in association with neuronal cell development as such markers for AD in the pre-plaque stage, female APP23 mice at eight, 19 and 31 weeks of age and corresponding control animals were tested for BPSD (elevated zero maze; sucrose preference test), motor coordination (rotarod), spatial memory and reversal learning (Morris water maze) and hippocampal neurogenesis as a neuronal correlate for hippocampus-dependent behavior. To evaluate a potential therapeutic effect of physical, cognitive and social stimulation, animals were exposed to environmental enrichment (EE) for one, twelve or 24 weeks from five weeks of age. In APP23, decreased anxiety accompanied increased agitation from eight weeks of age. Impairment of spatial memory and learning flexibility prior to plaque deposition involved an insufficient use of spatial search strategies associated with an unsuccessful compensatory increase of neurogenesis. EE had an overall beneficial effect on behavior and neurogenesis and thus constitutes a therapeutic tool to slow disease progression. BPSD, cognition and associated impaired neurogenesis complement clinical diagnostic markers for pre-plaque AD and contribute to an early detection essential to halt disease progression.
Collapse
Affiliation(s)
- Anna Pfeffer
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Neurology, Charitéplatz 1, 10117, Berlin, Germany
| | - Tonia Munder
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Neurology, Charitéplatz 1, 10117, Berlin, Germany
| | - Stefanie Schreyer
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Neurology, Charitéplatz 1, 10117, Berlin, Germany
| | - Charlotte Klein
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Neurology, Charitéplatz 1, 10117, Berlin, Germany
| | - Justyna Rasińska
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Neurology, Charitéplatz 1, 10117, Berlin, Germany
| | - York Winter
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, NeuroCure Cluster of Excellence, Charitéplatz 1, 10117, Berlin, Germany
| | - Barbara Steiner
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Neurology, Charitéplatz 1, 10117, Berlin, Germany.
| |
Collapse
|
29
|
Pretorius E, Bester J, Kell DB. A Bacterial Component to Alzheimer's-Type Dementia Seen via a Systems Biology Approach that Links Iron Dysregulation and Inflammagen Shedding to Disease. J Alzheimers Dis 2018; 53:1237-56. [PMID: 27340854 PMCID: PMC5325058 DOI: 10.3233/jad-160318] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The progression of Alzheimer's disease (AD) is accompanied by a great many observable changes, both molecular and physiological. These include oxidative stress, neuroinflammation, and (more proximal to cognitive decline) the death of neuronal and other cells. A systems biology approach seeks to organize these observed variables into pathways that discriminate those that are highly involved (i.e., causative) from those that are more usefully recognized as bystander effects. We review the evidence that iron dysregulation is one of the central causative pathway elements here, as this can cause each of the above effects. In addition, we review the evidence that dormant, non-growing bacteria are a crucial feature of AD, that their growth in vivo is normally limited by a lack of free iron, and that it is this iron dysregulation that is an important factor in their resuscitation. Indeed, bacterial cells can be observed by ultrastructural microscopy in the blood of AD patients. A consequence of this is that the growing cells can shed highly inflammatory components such as lipopolysaccharides (LPS). These too are known to be able to induce (apoptotic and pyroptotic) neuronal cell death. There is also evidence that these systems interact with elements of vitamin D metabolism. This integrative systems approach has strong predictive power, indicating (as has indeed been shown) that both natural and pharmaceutical iron chelators might have useful protective roles in arresting cognitive decline, and that a further assessment of the role of microbes in AD development is more than highly warranted.
Collapse
Affiliation(s)
- Etheresia Pretorius
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Arcadia, South Africa
| | - Janette Bester
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Arcadia, South Africa
| | - Douglas B Kell
- School of Chemistry, The University of Manchester, Manchester, Lancs, UK.,The Manchester Institute of Biotechnology, The University of Manchester, Manchester, Lancs, UK.,Centre for Synthetic Biology of Fine and Speciality Chemicals, The University of Manchester, Manchester, Lancs, UK
| |
Collapse
|
30
|
Seddighi S, Varma VR, An Y, Varma S, Beason-Held LL, Tanaka T, Kitner-Triolo MH, Kraut MA, Davatzikos C, Thambisetty M. SPARCL1 Accelerates Symptom Onset in Alzheimer's Disease and Influences Brain Structure and Function During Aging. J Alzheimers Dis 2018; 61:401-414. [PMID: 29154276 PMCID: PMC5934753 DOI: 10.3233/jad-170557] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
We recently reported that alpha-2 macroglobulin (A2M) is a biomarker of neuronal injury in Alzheimer's disease (AD) and identified a network of nine genes co-expressed with A2M in the brain. This network includes the gene encoding SPARCL1, a protein implicated in synaptic maintenance. Here, we examine whether SPARCL1 is associated with longitudinal changes in brain structure and function in older individuals at risk for AD in the Baltimore Longitudinal Study of Aging. Using data from the Gene-Tissue Expression Project, we first identified two single nucleotide polymorphisms (SNPs), rs9998212 and rs7695558, associated with lower brain SPARCL1 gene expression. We then analyzed longitudinal trajectories of cognitive performance in 591 participants who remained cognitively normal (average follow-up interval: 11.8 years) and 129 subjects who eventually developed MCI or AD (average follow-up interval: 9.4 years). Cognitively normal minor allele carriers of rs7695558 who developed incident AD showed accelerated memory loss prior to disease onset. Next, we compared longitudinal changes in brain volumes (MRI; n = 120 participants; follow-up = 6.4 years; 826 scans) and resting-state cerebral blood flow (rCBF; 15O-water PET; n = 81 participants; follow-up = 7.7 years; 664 scans) in cognitively normal participants. Cognitively normal minor allele carriers of rs9998212 showed accelerated atrophy in several global, lobar, and regional brain volumes. Minor allele carriers of both SNPs showed longitudinal changes in rCBF in several brain regions, including those vulnerable to AD pathology. Our findings suggest that SPARCL1 accelerates AD pathogenesis and thus link neuroinflammation with widespread changes in brain structure and function during aging.
Collapse
Affiliation(s)
- Sahba Seddighi
- Clinical and Translational Neuroscience Unit, Laboratory of Behavioral Neuroscience, National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, MD, USA
| | - Vijay R. Varma
- Clinical and Translational Neuroscience Unit, Laboratory of Behavioral Neuroscience, National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, MD, USA
| | - Yang An
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD, USA
| | | | - Lori L. Beason-Held
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Baltimore, MD, USA
| | - Toshiko Tanaka
- Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| | | | - Michael A. Kraut
- Department of Radiology and Radiological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Christos Davatzikos
- Department of Radiology, Center for Biomedical Image Computing and Analytics, University of Pennsylvania, Philadelphia, PA, USA
| | - Madhav Thambisetty
- Clinical and Translational Neuroscience Unit, Laboratory of Behavioral Neuroscience, National Institute on Aging (NIA), National Institutes of Health (NIH), Baltimore, MD, USA
| |
Collapse
|
31
|
Sahadevan P, Allen BG. MK5: A novel regulator of cardiac fibroblast function? IUBMB Life 2017; 69:785-794. [PMID: 28941148 DOI: 10.1002/iub.1677] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 08/21/2017] [Indexed: 12/28/2022]
Abstract
MAP kinase-activated protein kinases (MKs), protein serine/threonine kinases downstream of the MAPKs, regulate a number of biological functions. MK5 was initially identified as a substrate for p38 MAPK but subsequent studies revealed that MK5 activity is regulated by atypical MAPKs ERK3 and ERK4. However, the roles of these MAPKs in activating MK5 remain controversial. The interactome and physiological function of MK5 are just beginning to be understood. Here, we provide an overview of the structure-function of MK5 including recent progress in determining its role in cardiac structure and function. The cardiac phenotype of MK5 haplodeficient mice, and the effect of reduced MK5 expression on cardiac remodeling, is also discussed. © 2017 IUBMB Life, 69(10):785-794, 2017.
Collapse
Affiliation(s)
- Pramod Sahadevan
- Department of Biochemistry and Molecular Medicine, Université de Montréal and Montreal Heart Institute, Montréal, Québec, Canada
| | - Bruce G Allen
- Department of Biochemistry and Molecular Medicine, Université de Montréal and Montreal Heart Institute, Montréal, Québec, Canada.,Department of Pharmacology and Physiology, Université de Montréal, Montréal, Québec, Canada.,Department of Medicine, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
32
|
Fei YL, Lv HJ, Li YB, Liu J, Qian YH, Yang WN, Ma KG, Li HB, Qu QM. Tongxinluo improves cognition by decreasing β-amyloid in spontaneous hypertensive rats. Brain Res 2017; 1663:151-160. [DOI: 10.1016/j.brainres.2017.03.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 03/02/2017] [Accepted: 03/03/2017] [Indexed: 12/31/2022]
|
33
|
T Lymphocytes and Inflammatory Mediators in the Interplay between Brain and Blood in Alzheimer's Disease: Potential Pools of New Biomarkers. J Immunol Res 2017; 2017:4626540. [PMID: 28293644 PMCID: PMC5331319 DOI: 10.1155/2017/4626540] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 12/22/2016] [Accepted: 01/23/2017] [Indexed: 02/07/2023] Open
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disorder and the main cause of dementia. The disease is among the leading medical concerns of the modern world, because only symptomatic therapies are available, and no reliable, easily accessible biomarkers exist for AD detection and monitoring. Therefore extensive research is conducted to elucidate the mechanisms of AD pathogenesis, which seems to be heterogeneous and multifactorial. Recently much attention has been given to the neuroinflammation and activation of glial cells in the AD brain. Reports also highlighted the proinflammatory role of T lymphocytes infiltrating the AD brain. However, in AD molecular and cellular alterations involving T cells and immune mediators occur not only in the brain, but also in the blood and the cerebrospinal fluid (CSF). Here we review alterations concerning T lymphocytes and related immune mediators in the AD brain, CSF, and blood and the mechanisms by which peripheral T cells cross the blood brain barrier and the blood-CSF barrier. This knowledge is relevant for better AD therapies and for identification of novel biomarkers for improved AD diagnostics in the blood and the CSF. The data will be reviewed with the special emphasis on possibilities for development of AD biomarkers.
Collapse
|
34
|
Mitochondrial-Targeted Catalase: Extended Longevity and the Roles in Various Disease Models. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 146:203-241. [PMID: 28253986 DOI: 10.1016/bs.pmbts.2016.12.015] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The free-radical theory of aging was proposed more than 50 years ago. As one of the most popular mechanisms explaining the aging process, it has been extensively studied in several model organisms. However, the results remain controversial. The mitochondrial version of free-radical theory of aging proposes that mitochondria are both the primary sources of reactive oxygen species (ROS) and the primary targets of ROS-induced damage. One critical ROS is hydrogen peroxide, which is naturally degraded by catalase in peroxisomes or glutathione peroxidase within mitochondria. Our laboratory developed mice-overexpressing catalase targeted to mitochondria (mCAT), peroxisomes (pCAT), or the nucleus (nCAT) in order to investigate the role of hydrogen peroxide in different subcellular compartments in aging and age-related diseases. The mCAT mice have demonstrated the largest effects on life span and healthspan extension. This chapter will discuss the mCAT phenotype and review studies using mCAT to investigate the roles of mitochondrial oxidative stresses in various disease models, including metabolic syndrome and atherosclerosis, cardiac aging, heart failure, skeletal muscle pathology, sensory defect, neurodegenerative diseases, and cancer. As ROS has been increasingly recognized as essential signaling molecules that may be beneficial in hormesis, stress response and immunity, the potential pleiotropic, or adverse effects of mCAT are also discussed. Finally, the development of small-molecule mitochondrial-targeted therapeutic approaches is reviewed.
Collapse
|
35
|
Bester J, Soma P, Kell DB, Pretorius E. Viscoelastic and ultrastructural characteristics of whole blood and plasma in Alzheimer-type dementia, and the possible role of bacterial lipopolysaccharides (LPS). Oncotarget 2016; 6:35284-303. [PMID: 26462180 PMCID: PMC4742105 DOI: 10.18632/oncotarget.6074] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 09/28/2015] [Indexed: 12/22/2022] Open
Abstract
Alzheimer-type dementia (AD) is a neurodegenerative disorder and the most common form of dementia. Patients typically present with neuro- and systemic inflammation and iron dysregulation, associated with oxidative damage that reflects in hypercoagulability. Hypercoagulability is closely associated with increased fibrin(ogen) and in AD patients fibrin(ogen) has been implicated in the development of neuroinflammation and memory deficits. There is still no clear reason precisely why (a) this hypercoagulable state, (b) iron dysregulation and (c) increased fibrin(ogen) could together lead to the loss of neuronal structure and cognitive function. Here we suggest an alternative hypothesis based on previous ultrastructural evidence of the presence of a (dormant) blood microbiome in AD. Furthermore, we argue that bacterial cell wall components, such as the endotoxin lipopolysaccharide (LPS) of Gram-negative strains, might be the cause of the continuing and low-grade inflammation, characteristic of AD. Here, we follow an integrated approach, by studying the viscoelastic and ultrastructural properties of AD plasma and whole blood by using scanning electron microscopy, Thromboelastography (TEG®) and the Global Thrombosis Test (GTT®). Ultrastructural analysis confirmed the presence and close proximity of microbes to erythrocytes. TEG® analysis showed a hypercoagulable state in AD. TEG® results where LPS was added to naive blood showed the same trends as were found with the AD patients, while the GTT® results (where only platelet activity is measured), were not affected by the added LPS, suggesting that LPS does not directly impact platelet function. Our findings reinforce the importance of further investigating the role of LPS in AD.
Collapse
Affiliation(s)
- Janette Bester
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Arcadia, South Africa
| | - Prashilla Soma
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Arcadia, South Africa
| | - Douglas B Kell
- School of Chemistry and The Manchester Institute of Biotechnology, The University of Manchester, Manchester, UK
| | - Etheresia Pretorius
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Arcadia, South Africa
| |
Collapse
|
36
|
Upregulation of Aβ42 in the Brain and Bodily Fluids of Rhesus Monkeys with Aging. J Mol Neurosci 2016; 61:79-87. [PMID: 27647310 DOI: 10.1007/s12031-016-0840-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 09/06/2016] [Indexed: 10/21/2022]
Abstract
The cerebral accumulation of amyloid beta (Aβ) is one of the key pathological hallmarks of Alzheimer's disease (AD). Aβ is also found in bodily fluids such as the cerebrospinal fluid (CSF) and plasma. However, the significance of Aβ accumulation in the brain and different bodily pools, as well as its correlation with aging and cerebral amyloid pathology, is not completely understood. To better understand this question, we selected the rhesus monkey, which is phylogenetically and physiologically highly similar to the human, as a model to study. We quantified the levels of the two main Aβ isoforms (Aβ42 and Aβ40) in different sections of the brain (frontal cortex, temporal cortex, and hippocampus) and bodily fluids (CSF and plasma) of rhesus monkeys at different developmental phases (young, 5-9 years of age; mature, 10-19 years of age; and old, 21-24 years of age). We found that the levels of neuronal and insoluble Aβ42 increased significantly in the brain with aging, suggesting that this specific isoform might be directly involved in aging and AD-like pathophysiology. There was no significant change in the Aβ40 level in the brain with aging. In addition, the Aβ42 level, but not the Aβ40 level, in both the CSF and plasma increased with aging. We also identified a positive correlation between Aβ42 in the CSF and plasma and Aβ42 in the brain. Taken collectively, our results indicate that there is an association between Aβ accumulation and age. These results support the increased incidence of AD with aging.
Collapse
|
37
|
Vyas NA, Ramteke SN, Kumbhar AS, Kulkarni PP, Jani V, Sonawane UB, Joshi RR, Joshi B, Erxleben A. Ruthenium(II) polypyridyl complexes with hydrophobic ancillary ligand as Aβ aggregation inhibitors. Eur J Med Chem 2016; 121:793-802. [PMID: 27406812 DOI: 10.1016/j.ejmech.2016.06.038] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Revised: 06/15/2016] [Accepted: 06/20/2016] [Indexed: 11/28/2022]
Abstract
The synthesis, spectral and electrochemical characterization of the complexes of the type [Ru(NN)2(txbg)](2+) where NN is 2,2'-bipyridine (bpy) (1), 1,10-phenanthroline (phen) (2), dipyrido [3,2-d:2',3f] quinoxaline (dpq) (3), and dipyrido[3,2-a:2',3'-c]phenazine (dppz) (4) which incorporate the tetra-xylene bipyridine glycoluril (txbg) as the ancillary ligand are described in detail. Crystal structures of ligand txbg and complex 2 were solved by single crystal X-ray diffraction. Thioflavin T (ThT) fluorescence and Transmission Electron Microscopy (TEM) results indicated that at micromolar concentration all complexes exhibit significant potential of Aβ aggregation inhibition, while the ligand txbg displayed weak activity towards Aβ aggregation. Complex 1 showed relatively low inhibition (70%) while complexes 2-4 inhibited nearly 100% Aβ aggregation after 240 h of incubation. The similar potential of complexes 2-4 and absence of any trend in their activity with the planarity of polypyridyl ligands suggests there is no marked effect of planarity of coligands on their inhibitory potential. Further studies on acetylcholinesterase (AChE) inhibition indicated very weak activity of these complexes against AChE. Detailed interactions of Aβ with both ligand and complex 2 have been studied by molecular modeling. Complex 2 showed interactions involving all three polypyridyl ligands with hydrophobic region of Aβ. Furthermore, the toxicity of these complexes towards human neuroblastoma cells was evaluated by MTT assay and except complex 4, the complexes displayed very low toxicity.
Collapse
Affiliation(s)
- Nilima A Vyas
- Department of Chemistry, Savitribai Phule Pune University, Pune, 411007, India
| | | | - Avinash S Kumbhar
- Department of Chemistry, Savitribai Phule Pune University, Pune, 411007, India.
| | | | - Vinod Jani
- Bioinformatics Group, Centre for Development of Advanced Computing (C-DAC), Savitribai Phule Pune University, Pune, 411007, India
| | - Uddhavesh B Sonawane
- Bioinformatics Group, Centre for Development of Advanced Computing (C-DAC), Savitribai Phule Pune University, Pune, 411007, India
| | - Rajendra R Joshi
- Bioinformatics Group, Centre for Development of Advanced Computing (C-DAC), Savitribai Phule Pune University, Pune, 411007, India
| | - Bimba Joshi
- Bioprospecting, Agharkar Research Institute, Pune, 411004, India
| | - Andrea Erxleben
- School of Chemistry, National University of Ireland, Galway, Ireland
| |
Collapse
|
38
|
Amyloid- and FDG-PET imaging in amyotrophic lateral sclerosis. Eur J Nucl Med Mol Imaging 2016; 43:2050-60. [DOI: 10.1007/s00259-016-3434-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 05/30/2016] [Indexed: 12/31/2022]
|
39
|
Yan R, Fan Q, Zhou J, Vassar R. Inhibiting BACE1 to reverse synaptic dysfunctions in Alzheimer's disease. Neurosci Biobehav Rev 2016; 65:326-40. [PMID: 27044452 PMCID: PMC4856578 DOI: 10.1016/j.neubiorev.2016.03.025] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 03/25/2016] [Accepted: 03/29/2016] [Indexed: 12/21/2022]
Abstract
Over the past two decades, many studies have identified significant contributions of toxic β-amyloid peptides (Aβ) to the etiology of Alzheimer's disease (AD), which is the most common age-dependent neurodegenerative disease. AD is also recognized as a disease of synaptic failure. Aβ, generated by sequential proteolytic cleavages of amyloid precursor protein (APP) by BACE1 and γ-secretase, is one of major culprits that cause this failure. In this review, we summarize current findings on how BACE1-cleaved APP products impact learning and memory through proteins localized on glutamatergic, GABAergic, and dopaminergic synapses. Considering the broad effects of Aβ on all three types of synapses, BACE1 inhibition emerges as a practical approach for ameliorating Aβ-mediated synaptic dysfunctions. Since BACE1 inhibitory drugs are currently in clinical trials, this review also discusses potential complications arising from BACE1 inhibition. We emphasize that the benefits of BACE1 inhibitory drugs will outweigh the concerns.
Collapse
Affiliation(s)
- Riqiang Yan
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
| | - Qingyuan Fan
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - John Zhou
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Robert Vassar
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
| |
Collapse
|
40
|
Grieb P. Intracerebroventricular Streptozotocin Injections as a Model of Alzheimer's Disease: in Search of a Relevant Mechanism. Mol Neurobiol 2016; 53:1741-1752. [PMID: 25744568 PMCID: PMC4789228 DOI: 10.1007/s12035-015-9132-3] [Citation(s) in RCA: 249] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 02/22/2015] [Indexed: 01/26/2023]
Abstract
Streptozotocin (STZ), a glucosamine-nitrosourea compound derived from soil bacteria and originally developed as an anticancer agent, in 1963 has been found to induce diabetes in experimental animals. Since then, systemic application of STZ became the most frequently studied experimental model of insulin-dependent (type 1) diabetes. The compound is selectively toxic toward insulin-producing pancreatic beta cells, which is explained as the result of its cellular uptake by the low-affinity glucose transporter 2 (GLUT2) protein located in their cell membranes. STZ cytotoxicity is mainly due to DNA alkylation which results in cellular necrosis. Besides pancreatic beta cells, STZ applied systemically damages also other organs expressing GLUT2, such as kidney and liver, whereas brain is not affected directly because blood-brain barrier lacks this transporter protein. However, single or double intracerebroventricular (icv) STZ injection(s) chronically decrease cerebral glucose uptake and produce multiple other effects that resemble molecular, pathological, and behavioral features of Alzheimer's disease (AD). Taking into consideration that glucose hypometabolism is an early and persistent sign of AD and that Alzheimer's brains present features of impaired insulin signaling, icv STZ injections are exploited by some investigators as a non-transgenic model of this disease and used for preclinical testing of pharmacological therapies for AD. While it has been assumed that icv STZ produces cerebral glucose hypometabolism and other effects directly through desensitizing brain insulin receptors, the evidence for such mechanism is poor. On the other hand, early data on insulin immunoreactivity showed intense insulin expression in the rodent brain, and the possibility of local production of insulin in the mammalian brain has never been conclusively excluded. Also, there are GLUT2-expressing cells in the brain, in particular in the circumventricular organs and hypothalamus; some of these cells may be involved in glucose sensing. Thus, icv STZ may damage brain glucose insulin producing cells and/or brain glucose sensors. Mechanistic explanation of the mode of action of icv STZ, which is currently lacking, would provide a valuable contribution to the field of animal models of Alzheimer's disease.
Collapse
Affiliation(s)
- Paweł Grieb
- Department of Experimental Pharmacology, Mossakowski Medical Research Centre, Polish Academy of Sciences, Str. Pawinskiego 5, 02-106, Warsaw, Poland.
| |
Collapse
|
41
|
Ultra rapid in vivo screening for anti-Alzheimer anti-amyloid drugs. Sci Rep 2016; 6:23349. [PMID: 27000658 PMCID: PMC4802339 DOI: 10.1038/srep23349] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 03/02/2016] [Indexed: 12/20/2022] Open
Abstract
More than 46 million people worldwide suffer from Alzheimer’s disease. A
large number of potential treatments have been proposed; among these, the inhibition
of the aggregation of amyloid β-peptide (Aβ), considered one
of the main culprits in Alzheimer’s disease. Limitations in monitoring
the aggregation of Aβ in cells and tissues restrict the screening of
anti-amyloid drugs to in vitro studies in most cases. We have developed a
simple but powerful method to track Aβ aggregation in vivo in
real-time, using bacteria as in vivo amyloid reservoir. We use the specific
amyloid dye Thioflavin-S (Th-S) to stain bacterial inclusion bodies (IBs), in this
case mainly formed of Aβ in amyloid conformation. Th-S binding to
amyloids leads to an increment of fluorescence that can be monitored. The
quantification of the Th-S fluorescence along the time allows tracking
Aβ aggregation and the effect of potential anti-aggregating agents.
Collapse
|
42
|
Kim Y, Kim C, Son SM, Song H, Hong HS, Han SH, Mook-Jung I. The novel RAGE interactor PRAK is associated with autophagy signaling in Alzheimer's disease pathogenesis. Mol Neurodegener 2016; 11:4. [PMID: 26758977 PMCID: PMC4709948 DOI: 10.1186/s13024-016-0068-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 01/04/2016] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND The receptor for advanced glycation end products (RAGE) has been found to interact with amyloid β (Aβ). Although RAGE does not have any kinase motifs in its cytosolic domain, the interaction between RAGE and Aβ triggers multiple cellular signaling involved in Alzheimer's disease (AD). However, the mechanism of signal transduction by RAGE remains still unknown. Therefore, identifying binding proteins of RAGE may provide novel therapeutic targets for AD. RESULTS In this study, we identified p38-regulated/activated protein kinase (PRAK) as a novel RAGE interacting molecule. To investigate the effect of Aβ on PRAK mediated RAGE signaling pathway, we treated SH-SY5Y cells with monomeric form of Aβ. We demonstrated that Aβ significantly increased the phosphorylation of PRAK as well as the interaction between PRAK and RAGE. We showed that knockdown of PRAK rescued mTORC1 inactivation induced by Aβ treatment and decreased the formation of Aβ-induced autophagosome. CONCLUSIONS We provide evidence that PRAK plays a critical role in AD pathology as a key interactor of RAGE. Thus, our data suggest that PRAK might be a potential therapeutic target of AD involved in RAGE-mediated cell signaling induced by Aβ.
Collapse
Affiliation(s)
- Yoonhee Kim
- Department of Biochemistry and Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongro-gu, Seoul, 110-799, Korea.
| | - Chaeyoung Kim
- Department of Biochemistry and Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongro-gu, Seoul, 110-799, Korea.
| | - Sung Min Son
- Department of Biochemistry and Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongro-gu, Seoul, 110-799, Korea.
| | - Hyundong Song
- Department of Biochemistry and Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongro-gu, Seoul, 110-799, Korea.
| | - Hyun Seok Hong
- Department of Biochemistry and Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongro-gu, Seoul, 110-799, Korea.
| | - Sun-ho Han
- Department of Biochemistry and Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongro-gu, Seoul, 110-799, Korea.
| | - Inhee Mook-Jung
- Department of Biochemistry and Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongro-gu, Seoul, 110-799, Korea.
| |
Collapse
|
43
|
Müller-Schiffmann A, Herring A, Abdel-Hafiz L, Chepkova AN, Schäble S, Wedel D, Horn AHC, Sticht H, de Souza Silva MA, Gottmann K, Sergeeva OA, Huston JP, Keyvani K, Korth C. Amyloid-β dimers in the absence of plaque pathology impair learning and synaptic plasticity. Brain 2015; 139:509-25. [DOI: 10.1093/brain/awv355] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 10/17/2015] [Indexed: 11/12/2022] Open
Abstract
Abstract
Despite amyloid plaques, consisting of insoluble, aggregated amyloid-β peptides, being a defining feature of Alzheimer’s disease, their significance has been challenged due to controversial findings regarding the correlation of cognitive impairment in Alzheimer’s disease with plaque load. The amyloid cascade hypothesis defines soluble amyloid-β oligomers, consisting of multiple amyloid-β monomers, as precursors of insoluble amyloid-β plaques. Dissecting the biological effects of single amyloid-β oligomers, for example of amyloid-β dimers, an abundant amyloid-β oligomer associated with clinical progression of Alzheimer’s disease, has been difficult due to the inability to control the kinetics of amyloid-β multimerization. For investigating the biological effects of amyloid-β dimers, we stabilized amyloid-β dimers by an intermolecular disulphide bridge via a cysteine mutation in the amyloid-β peptide (Aβ-S8C) of the amyloid precursor protein. This construct was expressed as a recombinant protein in cells and in a novel transgenic mouse, termed tgDimer mouse. This mouse formed constant levels of highly synaptotoxic soluble amyloid-β dimers, but not monomers, amyloid-β plaques or insoluble amyloid-β during its lifespan. Accordingly, neither signs of neuroinflammation, tau hyperphosphorylation or cell death were observed. Nevertheless, these tgDimer mice did exhibit deficits in hippocampal long-term potentiation and age-related impairments in learning and memory, similar to what was observed in classical Alzheimer’s disease mouse models. Although the amyloid-β dimers were unable to initiate the formation of insoluble amyloid-β aggregates in tgDimer mice, after crossbreeding tgDimer mice with the CRND8 mouse, an amyloid-β plaque generating mouse model, Aβ-S8C dimers were sequestered into amyloid-β plaques, suggesting that amyloid-β plaques incorporate neurotoxic amyloid-β dimers that by themselves are unable to self-assemble. Our results suggest that within the fine interplay between different amyloid-β species, amyloid-β dimer neurotoxic signalling, in the absence of amyloid-β plaque pathology, may be involved in causing early deficits in synaptic plasticity, learning and memory that accompany Alzheimer’s disease.
10.1093/brain/awv355_video_abstract awv355_video_abstract
Collapse
Affiliation(s)
| | - Arne Herring
- 2 Institute of Neuropathology, University of Duisburg-Essen, Germany
| | - Laila Abdel-Hafiz
- 3 Centre for Behavioural Neuroscience, Heinrich Heine University, Düsseldorf, Germany
| | - Aisa N. Chepkova
- 4 Institute for Neuro- and Sensory Physiology, Heinrich Heine University, Düsseldorf, Germany
| | - Sandra Schäble
- 3 Centre for Behavioural Neuroscience, Heinrich Heine University, Düsseldorf, Germany
- *Present address: Comparative Psychology, Institute of Experimental Psychology, Heinrich Heine University, Düsseldorf, Germany
| | - Diana Wedel
- 1 Department Neuropathology, Heinrich Heine University, Düsseldorf, Germany
| | - Anselm H. C. Horn
- 5 Institute for Biochemistry, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Heinrich Sticht
- 5 Institute for Biochemistry, Friedrich Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | | | - Kurt Gottmann
- 4 Institute for Neuro- and Sensory Physiology, Heinrich Heine University, Düsseldorf, Germany
| | - Olga A. Sergeeva
- 4 Institute for Neuro- and Sensory Physiology, Heinrich Heine University, Düsseldorf, Germany
| | - Joseph P. Huston
- 3 Centre for Behavioural Neuroscience, Heinrich Heine University, Düsseldorf, Germany
| | - Kathy Keyvani
- 2 Institute of Neuropathology, University of Duisburg-Essen, Germany
| | - Carsten Korth
- 1 Department Neuropathology, Heinrich Heine University, Düsseldorf, Germany
| |
Collapse
|
44
|
Agresta AM, De Palma A, Bardoni A, Salvini R, Iadarola P, Mauri PL. Proteomics as an innovative tool to investigate frontotemporal disorders. Proteomics Clin Appl 2015; 10:457-69. [DOI: 10.1002/prca.201500090] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 10/14/2015] [Accepted: 10/28/2015] [Indexed: 12/12/2022]
Affiliation(s)
- Anna Maria Agresta
- Proteomics and Metabolomics Unit; Institute for Biomedical Technologies (ITB-CNR); Segrate (MI) Italy
- Department of Biology and Biotechnologies; Biochemistry Unit; University of Pavia; Pavia Italy
- Doctorate School of Molecular and Translational Medicine; University of Milan; Segrate (MI) Italy
| | - Antonella De Palma
- Proteomics and Metabolomics Unit; Institute for Biomedical Technologies (ITB-CNR); Segrate (MI) Italy
| | - Anna Bardoni
- Biochemistry Unit; Department of Molecular Medicine; University of Pavia; Pavia Italy
| | - Roberta Salvini
- Biochemistry Unit; Department of Molecular Medicine; University of Pavia; Pavia Italy
| | - Paolo Iadarola
- Department of Biology and Biotechnologies; Biochemistry Unit; University of Pavia; Pavia Italy
| | - Pier Luigi Mauri
- Proteomics and Metabolomics Unit; Institute for Biomedical Technologies (ITB-CNR); Segrate (MI) Italy
| |
Collapse
|
45
|
Foley AM, Ammar ZM, Lee RH, Mitchell CS. Systematic review of the relationship between amyloid-β levels and measures of transgenic mouse cognitive deficit in Alzheimer's disease. J Alzheimers Dis 2015; 44:787-95. [PMID: 25362040 PMCID: PMC4346318 DOI: 10.3233/jad-142208] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Amyloid-β (Aβ) is believed to directly affect memory and learning in Alzheimer’s disease (AD). It is widely suggested that there is a relationship between Aβ40 and Aβ42 levels and cognitive performance. In order to explore the validity of this relationship, we performed a meta-analysis of 40 peer-reviewed, published AD transgenic mouse studies that quantitatively measured Aβ levels in brain tissue after assessing cognitive performance. We examined the relationship between Aβ levels (Aβ40, Aβ42, or the ratio of Aβ42 to Aβ40) and cognitive function as measured by escape latency times in the Morris water maze or exploratory preference percentage in the novel object recognition test. Our systematic review examined five mouse models (Tg2576, APP, PS1, 3xTg, APP(OSK)-Tg), gender, and age. The overall result revealed no statistically significant correlation between quantified Aβ levels and experimental measures of cognitive function. However, enough of the trends were of the same sign to suggest that there probably is a very weak qualitative trend visible only across many orders of magnitude. In summary, the results of the systematic review revealed that mice bred to show elevated levels of Aβ do not perform significantly worse in cognitive tests than mice that do not have elevated Aβ levels. Our results suggest two lines of inquiry: 1) Aβ is a biochemical “side effect” of the AD pathology; or 2) learning and memory deficits in AD are tied to the presence of qualitatively “high” levels of Aβ but are not quantitatively sensitive to the levels themselves.
Collapse
Affiliation(s)
- Avery M Foley
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Zeena M Ammar
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Robert H Lee
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Cassie S Mitchell
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| |
Collapse
|
46
|
Talwar P, Sinha J, Grover S, Rawat C, Kushwaha S, Agarwal R, Taneja V, Kukreti R. Dissecting Complex and Multifactorial Nature of Alzheimer's Disease Pathogenesis: a Clinical, Genomic, and Systems Biology Perspective. Mol Neurobiol 2015; 53:4833-64. [PMID: 26351077 DOI: 10.1007/s12035-015-9390-0] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 08/11/2015] [Indexed: 01/14/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by loss of memory and other cognitive functions. AD can be classified into familial AD (FAD) and sporadic AD (SAD) based on heritability and into early onset AD (EOAD) and late onset AD (LOAD) based on age of onset. LOAD cases are more prevalent with genetically complex architecture. In spite of significant research focused on understanding the etiological mechanisms, search for diagnostic biomarker(s) and disease-modifying therapy is still on. In this article, we aim to comprehensively review AD literature on established etiological mechanisms including role of beta-amyloid and apolipoprotein E (APOE) along with promising newer etiological factors such as epigenetic modifications that have been associated with AD suggesting its multifactorial nature. As genomic studies have recently played a significant role in elucidating AD pathophysiology, a systematic review of findings from genome-wide linkage (GWL), genome-wide association (GWA), genome-wide expression (GWE), and epigenome-wide association studies (EWAS) was conducted. The availability of multi-dimensional genomic data has further coincided with the advent of computational and network biology approaches in recent years. Our review highlights the importance of integrative approaches involving genomics and systems biology perspective in elucidating AD pathophysiology. The promising newer approaches may provide reliable means of early and more specific diagnosis and help identify therapeutic interventions for LOAD.
Collapse
Affiliation(s)
- Puneet Talwar
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB) Campus, New Delhi, India.,Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Delhi, 110 007, India
| | - Juhi Sinha
- Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Delhi, 110 007, India
| | - Sandeep Grover
- Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Delhi, 110 007, India.,Department of Paediatrics, Division of Pneumonology-Immunology, Charité University Medical Centre, Berlin, Germany
| | - Chitra Rawat
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB) Campus, New Delhi, India.,Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Delhi, 110 007, India
| | - Suman Kushwaha
- Institute of Human Behaviour and Allied Sciences (IHBAS), Delhi, India
| | - Rachna Agarwal
- Institute of Human Behaviour and Allied Sciences (IHBAS), Delhi, India
| | - Vibha Taneja
- Department of Research, Sir Ganga Ram Hospital, New Delhi, India
| | - Ritushree Kukreti
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB) Campus, New Delhi, India. .,Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Delhi, 110 007, India.
| |
Collapse
|
47
|
Anastasio TJ. Computational identification of potential multi-drug combinations for reduction of microglial inflammation in Alzheimer disease. Front Pharmacol 2015; 6:116. [PMID: 26097457 PMCID: PMC4456568 DOI: 10.3389/fphar.2015.00116] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 05/19/2015] [Indexed: 12/31/2022] Open
Abstract
Like other neurodegenerative diseases, Alzheimer Disease (AD) has a prominent inflammatory component mediated by brain microglia. Reducing microglial inflammation could potentially halt or at least slow the neurodegenerative process. A major challenge in the development of treatments targeting brain inflammation is the sheer complexity of the molecular mechanisms that determine whether microglia become inflammatory or take on a more neuroprotective phenotype. The process is highly multifactorial, raising the possibility that a multi-target/multi-drug strategy could be more effective than conventional monotherapy. This study takes a computational approach in finding combinations of approved drugs that are potentially more effective than single drugs in reducing microglial inflammation in AD. This novel approach exploits the distinct advantages of two different computer programming languages, one imperative and the other declarative. Existing programs written in both languages implement the same model of microglial behavior, and the input/output relationships of both programs agree with each other and with data on microglia over an extensive test battery. Here the imperative program is used efficiently to screen the model for the most efficacious combinations of 10 drugs, while the declarative program is used to analyze in detail the mechanisms of action of the most efficacious combinations. Of the 1024 possible drug combinations, the simulated screen identifies only 7 that are able to move simulated microglia at least 50% of the way from a neurotoxic to a neuroprotective phenotype. Subsequent analysis shows that of the 7 most efficacious combinations, 2 stand out as superior both in strength and reliability. The model offers many experimentally testable and therapeutically relevant predictions concerning effective drug combinations and their mechanisms of action.
Collapse
Affiliation(s)
- Thomas J Anastasio
- Computational Neurobiology Laboratory, Department of Molecular and Integrative Physiology, Beckman Institute, University of Illinois at Urbana-Champaign Urbana, IL, USA
| |
Collapse
|
48
|
The Ubiquitin-Proteasome System and Molecular Chaperone Deregulation in Alzheimer's Disease. Mol Neurobiol 2015; 53:905-931. [PMID: 25561438 DOI: 10.1007/s12035-014-9063-4] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 12/09/2014] [Indexed: 12/18/2022]
Abstract
One of the shared hallmarks of neurodegenerative diseases is the accumulation of misfolded proteins. Therefore, it is suspected that normal proteostasis is crucial for neuronal survival in the brain and that the malfunction of this mechanism may be the underlying cause of neurodegenerative diseases. The accumulation of amyloid plaques (APs) composed of amyloid-beta peptide (Aβ) aggregates and neurofibrillary tangles (NFTs) composed of misfolded Tau proteins are the defining pathological markers of Alzheimer's disease (AD). The accumulation of these proteins indicates a faulty protein quality control in the AD brain. An impaired ubiquitin-proteasome system (UPS) could lead to negative consequences for protein regulation, including loss of function. Another pivotal mechanism for the prevention of misfolded protein accumulation is the utilization of molecular chaperones. Molecular chaperones, such as heat shock proteins (HSPs) and FK506-binding proteins (FKBPs), are highly involved in protein regulation to ensure proper folding and normal function. In this review, we elaborate on the molecular basis of AD pathophysiology using recent data, with a particular focus on the role of the UPS and molecular chaperones as the defensive mechanism against misfolded proteins that have prion-like properties. In addition, we propose a rational therapy approach based on this mechanism.
Collapse
|
49
|
Sarvaiya J, Agrawal Y. Chitosan as a suitable nanocarrier material for anti-Alzheimer drug delivery. Int J Biol Macromol 2015; 72:454-65. [DOI: 10.1016/j.ijbiomac.2014.08.052] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 08/24/2014] [Accepted: 08/28/2014] [Indexed: 11/25/2022]
|
50
|
Intranasal deferoxamine engages multiple pathways to decrease memory loss in the APP/PS1 model of amyloid accumulation. Neurosci Lett 2015; 584:362-7. [DOI: 10.1016/j.neulet.2014.11.013] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 11/03/2014] [Accepted: 11/07/2014] [Indexed: 11/18/2022]
|