1
|
Zhao M, Wang Y, Shen Y, Wei C, Zhang G, Sun L. A review of the roles of pathogens in Alzheimer's disease. Front Neurosci 2024; 18:1439055. [PMID: 39224577 PMCID: PMC11366636 DOI: 10.3389/fnins.2024.1439055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024] Open
Abstract
Alzheimer's disease (AD) is one of the leading causes of dementia and is characterized by memory loss, mental and behavioral abnormalities, and impaired ability to perform daily activities. Even as a global disease that threatens human health, effective treatments to slow the progression of AD have not been found, despite intensive research and significant investment. In recent years, the role of infections in the etiology of AD has sparked intense debate. Pathogens invade the central nervous system through a damaged blood-brain barrier or nerve trunk and disrupt the neuronal structure and function as well as homeostasis of the brain microenvironment through a series of molecular biological events. In this review, we summarize the various pathogens involved in AD pathology, discuss potential interactions between pathogens and AD, and provide an overview of the promising future of anti-pathogenic therapies for AD.
Collapse
Affiliation(s)
| | | | | | | | | | - Li Sun
- Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| |
Collapse
|
2
|
Rehman MU, Sehar N, Rasool I, Aldossari RM, Wani AB, Rashid SM, Wali AF, Ali A, Arafah A, Khan A. Glymphatic pathway: An emerging perspective in the pathophysiology of neurodegenerative diseases. Int J Geriatr Psychiatry 2024; 39:e6104. [PMID: 38877354 DOI: 10.1002/gps.6104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 05/23/2024] [Indexed: 06/16/2024]
Abstract
The central nervous system (CNS) is widely recognized as the only organ system without lymphatic capillaries to promote the removal of interstitial metabolic by-products. Thus, the newly identified glymphatic system which provides a pseudolymphatic activity in the nervous system has been focus of latest research in neurosciences. Also, findings reported that, sleep stimulates the elimination actions of glymphatic system and is linked to normal brain homeostatis. The CNS is cleared of potentially hazardous compounds via the glymphatic system, particularly during sleep. Any age-related alterations in brain functioning and pathophysiology of various neurodegenerative illnesses indicates the disturbance of the brain's glymphatic system. In this context, β-amyloid as well as tau leaves the CNS through the glymphatic system, it's functioning and CSF discharge markedly altered in elderly brains as per many findings. Thus, glymphatic failure may have a potential mechanism which may be therapeutically targetable in several neurodegenerative and age-associated cognitive diseases. Therefore, there is an urge to focus for more research into the connection among glymphatic system and several potential brain related diseases. Here, in our current review paper, we reviewed current research on the glymphatic system's involvement in a number of prevalent neurodegenerative and neuropsychiatric diseases and, we also discussed several therapeutic approaches, diet and life style modifications which might be used to acquire a more thorough performance and purpose of the glymphatic system to decipher novel prospects for clinical applicability for the management of these diseases.
Collapse
Affiliation(s)
- Muneeb U Rehman
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Nouroz Sehar
- Centre for Translational and Clinical Research, School of Chemical & Life Sciences, Jamia Hamdard, New Delhi, India
| | - Iyman Rasool
- Department of Pathology, Government Medical College (GMC-Srinagar), Srinagar, Jammu and Kashmir, India
| | - Rana M Aldossari
- Department of Pharmacology & Toxicology, College of Pharmacy, Prince Sattam Bin AbdulAziz University, Al Kharj, Saudi Arabia
| | - Amir Bashir Wani
- Division of Biotechnology, Sher-e-Kashmir University of Agricultural Sciences and Technology-Kashmir, Srinagar, Jammu and Kashmir, India
| | - Shahzada Mudasir Rashid
- Division of Veterinary Biochemistry, Faculty of Veterinary Science and Animal Husbandry, SKUAST-Kashmir, Alusteng, Shuhama, Srinagar, Jammu and Kashmir, India
| | - Adil Farooq Wali
- Department of Pharmaceutical Chemistry, RAK College of Pharmaceutical Sciences, RAK Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
| | - Aarif Ali
- Division of Veterinary Biochemistry, Faculty of Veterinary Science and Animal Husbandry, SKUAST-Kashmir, Alusteng, Shuhama, Srinagar, Jammu and Kashmir, India
| | - Azher Arafah
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Andleeb Khan
- Department of Biosciences, Faculty of Science, Integral University, Lucknow, Uttar Pradesh, India
| |
Collapse
|
3
|
Imtiaz A, Shimonaka S, Uddin MN, Elahi M, Ishiguro K, Hasegawa M, Hattori N, Motoi Y. Selection of lansoprazole from an FDA-approved drug library to inhibit the Alzheimer's disease seed-dependent formation of tau aggregates. Front Aging Neurosci 2024; 16:1368291. [PMID: 38633982 PMCID: PMC11022852 DOI: 10.3389/fnagi.2024.1368291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 03/15/2024] [Indexed: 04/19/2024] Open
Abstract
The efficacy of current treatments is still insufficient for Alzheimer's disease (AD), the most common cause of Dementia. Out of the two pathological hallmarks of AD amyloid-β plaques and neurofibrillary tangles, comprising of tau protein, tau pathology strongly correlates with the symptoms of AD. Previously, screening for inhibitors of tau aggregation that target recombinant tau aggregates have been attempted. Since a recent cryo-EM analysis revealed distinct differences in the folding patterns of heparin-induced recombinant tau filaments and AD tau filaments, this study focused on AD seed-dependent tau aggregation in drug repositioning for AD. We screened 763 compounds from an FDA-approved drug library using an AD seed-induced tau aggregation in SH-SY5Y cell-based assay. In the first screening, 180 compounds were selected, 72 of which were excluded based on the results of lactate dehydrogenase assay. In the third screening with evaluations of soluble and insoluble tau, 38 compounds were selected. In the fourth screening with 3 different AD seeds, 4 compounds, lansoprazole, calcipotriene, desogestrel, and pentamidine isethionate, were selected. After AD seed-induced real-time quaking-induced conversion, lansoprazole was selected as the most suitable drug for repositioning. The intranasal administration of lansoprazole for 4 months to AD seed-injected mice improved locomotor activity and reduced both the amount of insoluble tau and the extent of phosphorylated tau-positive areas. Alanine replacement of the predicted binding site to an AD filament indicated the involvement of Q351, H362, and K369 in lansoprazole and C-shaped tau filaments. These results suggest the potential of lansoprazole as a candidate for drug repositioning to an inhibitor of tau aggregate formation in AD.
Collapse
Affiliation(s)
- Ahmed Imtiaz
- Department of Diagnosis, Prevention and Treatment of Dementia, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan
- Department of Neurology, Juntendo University School of Medicine, Hongo, Bunkyo-ku, Tokyo, Japan
| | - Shotaro Shimonaka
- Research Institute for Diseases of Old Age, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Mohammad Nasir Uddin
- Department of Biochemistry & Molecular Biology, Faculty of Life Science, Mawlana Bhashani Science & Technology University, Tangail, Bangladesh
| | - Montasir Elahi
- Center for Birth Defect Research, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Koichi Ishiguro
- Department of Neurology, Juntendo University School of Medicine, Hongo, Bunkyo-ku, Tokyo, Japan
| | - Masato Hasegawa
- Department of Brain and Neuroscience, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University School of Medicine, Hongo, Bunkyo-ku, Tokyo, Japan
| | - Yumiko Motoi
- Medical Center for Dementia, Juntendo University Hospital, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
4
|
Ye Y, Tong HYK, Chong WH, Li Z, Tam PKH, Baptista-Hon DT, Monteiro O. A systematic review and meta-analysis of the effects of long-term antibiotic use on cognitive outcomes. Sci Rep 2024; 14:4026. [PMID: 38369574 PMCID: PMC10874946 DOI: 10.1038/s41598-024-54553-4] [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: 11/10/2023] [Accepted: 02/14/2024] [Indexed: 02/20/2024] Open
Abstract
Antibiotics are indispensable to infection management. However, use of antibiotics can cause gut microbiota dysbiosis, which has been linked to cognitive impairment by disrupting communication between the gut microbiota and the brain. We conducted a systematic review and meta-analysis on the effects of long-term antibiotic use on cognitive outcomes. We have searched PubMed, Web of Science, Embase, Cochrane Library and Scopus for English publications before March 2023 following the PRISMA guidelines. Screening, data extraction, and quality assessment were performed in duplicate. 960 articles were screened and 16 studies which evaluated the effect of any antibiotic compared to no antibiotics or placebo were included. Case-reports, in vitro and animal studies were excluded. We found that antibiotic use was associated with worse cognitive outcomes with a pooled effect estimate of - 0.11 (95% CI - 0.15, - 0.07, Z = 5.45; P < 0.00001). Subgroup analyses performed on adult vs pediatric patients showed a similar association of antibiotic on cognition in both subgroups. Antibiotic treatment was not associated with worse cognition on subjects with existing cognitive impairment. On the other hand, antibiotic treatment on subjects with no prior cognitive impairment was associated with worse cognitive performance later in life. This calls for future well-designed and well-powered studies to investigate the impact of antibiotics on cognitive performance.
Collapse
Affiliation(s)
- Yongqin Ye
- Faculty of Medicine, Medical Sciences Division, Macau University of Science and Technology, Avenida da Harmonia, Praia Park, Coloane, 999078, Macao SAR, China
| | | | - Wai Hong Chong
- Faculty of Medicine, Medical Sciences Division, Macau University of Science and Technology, Avenida da Harmonia, Praia Park, Coloane, 999078, Macao SAR, China
| | - Zhiqian Li
- Faculty of Medicine, Medical Sciences Division, Macau University of Science and Technology, Avenida da Harmonia, Praia Park, Coloane, 999078, Macao SAR, China
| | - Paul Kwong Hang Tam
- Faculty of Medicine, Medical Sciences Division, Macau University of Science and Technology, Avenida da Harmonia, Praia Park, Coloane, 999078, Macao SAR, China
| | - Daniel T Baptista-Hon
- Faculty of Medicine, Medical Sciences Division, Macau University of Science and Technology, Avenida da Harmonia, Praia Park, Coloane, 999078, Macao SAR, China
- Division of Systems Medicine, School of Medicine, University of Dundee, Dundee, UK
| | - Olivia Monteiro
- Faculty of Medicine, Medical Sciences Division, Macau University of Science and Technology, Avenida da Harmonia, Praia Park, Coloane, 999078, Macao SAR, China.
| |
Collapse
|
5
|
Liang C, Pereira R, Zhang Y, Rojas OL. Gut Microbiome in Alzheimer's Disease: from Mice to Humans. Curr Neuropharmacol 2024; 22:2314-2329. [PMID: 39403057 PMCID: PMC11451315 DOI: 10.2174/1570159x22666240308090741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 01/24/2024] [Accepted: 02/23/2024] [Indexed: 10/19/2024] Open
Abstract
Alzheimer's disease (AD) is the most prevalent type of dementia, but its etiopathogenesis is not yet fully understood. Recent preclinical studies and clinical evidence indicate that changes in the gut microbiome could potentially play a role in the accumulation of amyloid beta. However, the relationship between gut dysbiosis and AD is still elusive. In this review, the potential impact of the gut microbiome on AD development and progression is discussed. Pre-clinical and clinical literature exploring changes in gut microbiome composition is assessed, which can contribute to AD pathology including increased amyloid beta deposition and cognitive impairment. The gut-brain axis and the potential involvement of metabolites produced by the gut microbiome in AD are also highlighted. Furthermore, the potential of antibiotics, prebiotics, probiotics, fecal microbiota transplantation, and dietary interventions as complementary therapies for the management of AD is summarized. This review provides valuable insights into potential therapeutic strategies to modulate the gut microbiome in AD.
Collapse
Affiliation(s)
- Chang Liang
- Department of Gastroenterology, West China Hospital of Sichuan University, Chengdu, China
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Resel Pereira
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Yan Zhang
- Department of Gastroenterology, West China Hospital of Sichuan University, Chengdu, China
| | - Olga L. Rojas
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
| |
Collapse
|
6
|
Bhattacharjee A, Roy PK. Conjoint hepatobiliary-enterohepatic cycles for amyloid excretion and enhancing its drug-induced clearance: a systems biology approach to Alzheimer's disease. J Biomol Struct Dyn 2023; 41:10507-10524. [PMID: 36510663 DOI: 10.1080/07391102.2022.2154842] [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: 05/24/2022] [Accepted: 11/29/2022] [Indexed: 12/15/2022]
Abstract
The liver is the major organ responsible for metabolism of amyloid-beta, the primary toxic misfolded protein responsible for Alzheimer's disease (AD). The present study focuses on the crucial role of liver in AD. We have developed a framework that formulates and integrates two reciprocal transport processes of amyloid, via hepato-biliary and enterohepatic circulations (EHC). Our system analysis approach shows that activating the liver X-receptor (LXR) can reduce amyloid-beta formation by increasing expression of the genes: ATP-binding-cassette-transporter (ABCA1) and Stearoyl-CoA-desaturase (SCD). Besides, activating the pregnane-X-receptor (PXR) can enhance the clearance of amyloid-beta by increasing the expression of the genes: ATP-Binding-Cassette-Superfamily-G-member-2 (ABCG2) and multidrug-resistance protein-1 (MDR1). We also identified receptor-like apical sodium-dependent bile-acid transporter (ASBT) of intestinal enterocyte, showing affinity towards amyloid-beta, suggesting amyloid-beta's possible reuptake from intestinal contents to the systemic circulation through this receptor. Further, we have performed protein-protein interaction to evaluate the binding affinity of amyloid-beta to these receptors. Moreover, we undertook molecular docking and molecular dynamic simulation of some repurposed drugs (rifampicin, 24-hydroxycholesterols, resveratrol, cilostazol) which can target the aforesaid receptors to enhance amyloid-beta's fecal clearance, reduce amyloid-beta formation, and prevent the reuptake of amyloid-beta from intestinal feces. Additionally, network pharmacology and synergism analysis were utilized to validate our hypothesis and identify the drug combinations, respectively. Gene-ontology investigation, network pharmacology, and consolidated pathway analysis validate the alteration of the above-mentioned gene expression profiles. Furthermore, our neuropharmacological synergism study identifies the optimal combination of the repurposed drugs. Finally, our findings on candidate drugs are substantiated by clinical-trial outcomes.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Anindita Bhattacharjee
- School of Bio-Medical Engineering, Indian Institute of Technology (B.H.U.), Varanasi, India
| | - Prasun K Roy
- School of Bio-Medical Engineering, Indian Institute of Technology (B.H.U.), Varanasi, India
| |
Collapse
|
7
|
Cores Á, Carmona-Zafra N, Clerigué J, Villacampa M, Menéndez JC. Quinones as Neuroprotective Agents. Antioxidants (Basel) 2023; 12:1464. [PMID: 37508002 PMCID: PMC10376830 DOI: 10.3390/antiox12071464] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/12/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Quinones can in principle be viewed as a double-edged sword in the treatment of neurodegenerative diseases, since they are often cytoprotective but can also be cytotoxic due to covalent and redox modification of biomolecules. Nevertheless, low doses of moderately electrophilic quinones are generally cytoprotective, mainly due to their ability to activate the Keap1/Nrf2 pathway and thus induce the expression of detoxifying enzymes. Some natural quinones have relevant roles in important physiological processes. One of them is coenzyme Q10, which takes part in the oxidative phosphorylation processes involved in cell energy production, as a proton and electron carrier in the mitochondrial respiratory chain, and shows neuroprotective effects relevant to Alzheimer's and Parkinson's diseases. Additional neuroprotective quinones that can be regarded as coenzyme Q10 analogues are idobenone, mitoquinone and plastoquinone. Other endogenous quinones with neuroprotective activities include tocopherol-derived quinones, most notably vatiquinone, and vitamin K. A final group of non-endogenous quinones with neuroprotective activity is discussed, comprising embelin, APX-3330, cannabinoid-derived quinones, asterriquinones and other indolylquinones, pyrroloquinolinequinone and its analogues, geldanamycin and its analogues, rifampicin quinone, memoquin and a number of hybrid structures combining quinones with amino acids, cholinesterase inhibitors and non-steroidal anti-inflammatory drugs.
Collapse
Affiliation(s)
- Ángel Cores
- Unidad de Química Orgánica y Farmacéutica, Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense, Plaza de Ramón y Cajal sn, 28040 Madrid, Spain
| | - Noelia Carmona-Zafra
- Unidad de Química Orgánica y Farmacéutica, Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense, Plaza de Ramón y Cajal sn, 28040 Madrid, Spain
| | - José Clerigué
- Unidad de Química Orgánica y Farmacéutica, Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense, Plaza de Ramón y Cajal sn, 28040 Madrid, Spain
| | - Mercedes Villacampa
- Unidad de Química Orgánica y Farmacéutica, Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense, Plaza de Ramón y Cajal sn, 28040 Madrid, Spain
| | - J Carlos Menéndez
- Unidad de Química Orgánica y Farmacéutica, Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense, Plaza de Ramón y Cajal sn, 28040 Madrid, Spain
| |
Collapse
|
8
|
Zhang J, Li Y, Li H, Liu A, Cao J, Li X, Xia N, Zhang Z, Bai J, Zhang H. Aspirin ameliorates the neurotoxicity of benzo[a]pyrene in mice and HT22 cells: Possible role of miRNA-mRNA network. Food Chem Toxicol 2023:113919. [PMID: 37364758 DOI: 10.1016/j.fct.2023.113919] [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: 12/15/2022] [Revised: 06/20/2023] [Accepted: 06/23/2023] [Indexed: 06/28/2023]
Abstract
Benzo[a]pyrene (B[a]P) is neurotoxic, however, the mechanism and potential prevention are yet not clear. This study explored the miRNA-mRNA network in the B[a]P-induced neurotoxicity in mice and HT22 cells and the intervention of aspirin (ASP). HT22 cells were treated for 48 h with DMSO, B[a]P (20 μM), or both B[a]P (20 μM) and ASP (4 μM). Following B[a]P treatment, compared to the DMSO controls, HT22 cells showed injured cell morphology, reduced cell viability and neurotrophic factor concentrations, and increased LDH leakage, Aβ1-42, and inflammatory factor concentrations, which were improved by ASP. RNA sequencing and qPCR verified the significant differences of miRNA and mRNA profiles following B[a]P treatment, which were rescued by ASP. Bioinformatics analysis suggested the miRNA-mRNA network could be involved in the neurotoxicity of B[a]P and the intervention of ASP. The neurotoxicity and neuroinflammation were induced in mice's brains by B[a]P, and the target miRNA and mRNA were proved to be consistent with in vitro, which were ameliorated by ASP. The findings demonstrate a possible role of miRNA-mRNA network in the B[a]P-induced neurotoxicity. If this is confirmed by additional experiments, it will provide a promising pathway of intervention against B[a]P, using ASP or other agents with fewer toxic effects.
Collapse
Affiliation(s)
- Jinfeng Zhang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China; Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, China
| | - Yangyang Li
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China; Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, China
| | - Huan Li
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China; Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, China
| | - Aixiang Liu
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China; Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, China
| | - Jingjing Cao
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China; Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, China
| | - Xin Li
- Center of Disease Control and Prevention, Taiyuan Iron and Steel Company, Taiyuan, 030003, Shanxi, China
| | - Na Xia
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China; Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, China
| | - Zhihong Zhang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China; Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, China
| | - Jianying Bai
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China; Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, China
| | - Hongmei Zhang
- Department of Environmental Health, School of Public Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China; Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, China; Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Taiyuan, 030001, Shanxi, China; Key Laboratory of Cellular Physiology, Taiyuan, 030001, Shanxi, China; Department of Physiology, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
| |
Collapse
|
9
|
Balietti M, Casoli T, Giorgetti B, Colangeli R, Nicoletti C, Solazzi M, Pugliese A, Conti F. Generation and Characterization of the First Murine Model of Alzheimer's Disease with Mutated AβPP Inserted in a BALB/c Background (C.B6/J-APPswe). J Alzheimers Dis 2023:JAD230195. [PMID: 37182890 DOI: 10.3233/jad-230195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
BACKGROUND Numerous mouse models of Alzheimer's disease (AD) are available, but all suffer from certain limitations, thus prompting further attempts. To date, no one model exists with amyloidopathy in a BALB/c strain. OBJECTIVE To generate and characterize the C.B6/J-APPswe mouse, a model of AD with a mutated human gene for the amyloid-β protein precursor (AβPP) inserted in a BALB/c background. METHODS We analyzed five groups at different ages (3, 6, 9, 12, and 16-18 months) of C.B6/J-APPswe and wild-type mice (50% males and 50% females) for the main hallmarks of AD by western blotting, amyloid-β (Aβ) ELISA, immunocytochemistry, electrophysiology, and behavioral tests. RESULTS The C.B6/J-APPswe mouse displays early AβPP and Aβ production, late amyloid plaques formation, high level of tau phosphorylation, synaptic deficits (reduced density and functional impairment due to a reduced post-synaptic responsiveness), neurodegeneration caused by apoptosis and necroptosis/necrosis, microgliosis, astrocytic abnormalities, and sex-related differences in explorative behavior, anxiety-like behavior, and spatial long-term and working memories. Social housing is feasible despite the intra-cage aggressiveness of male animals. CONCLUSION C.B6/J-APPswe mice develop most of the distinctive features of AD and is a suitable model for the study of brain atrophy mechanisms and of the differences between males and females in the onset of cognitive/non-cognitive deficits.
Collapse
Affiliation(s)
- Marta Balietti
- Center for Neurobiology of Aging, IRCCS INRCA, Ancona, Italy
| | - Tiziana Casoli
- Center for Neurobiology of Aging, IRCCS INRCA, Ancona, Italy
| | | | - Roberto Colangeli
- Section of Neuroscience and Cell Biology, Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy
| | - Cristina Nicoletti
- Section of Neuroscience and Cell Biology, Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy
| | - Moreno Solazzi
- Center for Neurobiology of Aging, IRCCS INRCA, Ancona, Italy
| | - Arianna Pugliese
- Section of Neuroscience and Cell Biology, Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy
| | - Fiorenzo Conti
- Center for Neurobiology of Aging, IRCCS INRCA, Ancona, Italy
- Section of Neuroscience and Cell Biology, Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy
- Fondazione di Medicina Molecolare e Terapia Cellulare, Università Politecnica delle Marche, Ancona, Italy
| |
Collapse
|
10
|
Yuan M, Wang Y, Huang Z, Jing F, Qiao P, Zou Q, Li J, Cai Z. Impaired autophagy in amyloid-beta pathology: A traditional review of recent Alzheimer's research. J Biomed Res 2023; 37:30-46. [PMID: 36642915 PMCID: PMC9898044 DOI: 10.7555/jbr.36.20220145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder. The major pathological changes in AD progression are the generation and accumulation of amyloid-beta (Aβ) peptides as well as the presence of abnormally hyperphosphorylated tau proteins in the brain. Autophagy is a conserved degradation pathway that eliminates abnormal protein aggregates and damaged organelles. Previous studies have suggested that autophagy plays a key role in the production and clearance of Aβ peptides to maintain a steady-state of Aβ peptides levels. However, a growing body of evidence suggests that autophagy is significantly impaired in the pathogenesis of AD, especially in Aβ metabolism. Therefore, this article reviews the latest studies concerning the mechanisms of autophagy, the metabolism of Aβ peptides, and the defective autophagy in the production and clearance of Aβ peptides. Here, we also summarize the established and new strategies for targeting autophagy in vivo and through clinical AD trials to identify gaps in our knowledge and to generate further questions.
Collapse
Affiliation(s)
- Minghao Yuan
- Chongqing Medical University, Chongqing 400042, China,Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing 400013, China,Department of Neurology, Chongqing School, University of Chinese Academy of Sciences, Chongqing 400013, China,Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing 400013, China
| | - Yangyang Wang
- Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing 400013, China,Department of Neurology, Chongqing School, University of Chinese Academy of Sciences, Chongqing 400013, China
| | - Zhenting Huang
- Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing 400013, China,Department of Neurology, Chongqing School, University of Chinese Academy of Sciences, Chongqing 400013, China
| | - Feng Jing
- Chongqing Medical University, Chongqing 400042, China,Department of Neurology, Chongqing School, University of Chinese Academy of Sciences, Chongqing 400013, China,Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing 400013, China
| | - Peifeng Qiao
- Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing 400013, China,Department of Neurology, Chongqing School, University of Chinese Academy of Sciences, Chongqing 400013, China
| | - Qian Zou
- Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing 400013, China,Department of Neurology, Chongqing School, University of Chinese Academy of Sciences, Chongqing 400013, China
| | - Jing Li
- Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing 400013, China,Department of Neurology, Chongqing School, University of Chinese Academy of Sciences, Chongqing 400013, China
| | - Zhiyou Cai
- Chongqing Medical University, Chongqing 400042, China,Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing 400013, China,Department of Neurology, Chongqing School, University of Chinese Academy of Sciences, Chongqing 400013, China,Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing 400013, China,Zhiyou Cai, Department of Neurology, Chongqing General Hospital, University of Chinese Academy of Sciences, No.118, Xingguang Avenue, Liangjiang New Area, Chongqing 401147, China. Tel/Fax: +86-23-63515796/+86-23-63515796, E-mail:
| |
Collapse
|
11
|
Khan AN, Khan RH. Protein misfolding and related human diseases: A comprehensive review of toxicity, proteins involved, and current therapeutic strategies. Int J Biol Macromol 2022; 223:143-160. [PMID: 36356861 DOI: 10.1016/j.ijbiomac.2022.11.031] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022]
Abstract
Most of the cell's chemical reactions and structural components are facilitated by proteins. But proteins are highly dynamic molecules, where numerous modifications or changes in the cellular environment can affect their native conformational fold leading to protein aggregation. Various stress conditions, such as oxidative stress, mutations and metal toxicity may cause protein misfolding and aggregation by shifting the conformational equilibrium towards more aggregation-prone states. Most of the protein misfolding diseases (PMDs) involve aggregation of protein. We have discussed such proteins like Aβ peptide, α-synuclein, amylin and lysozyme involved in Alzheimer's, Parkinson's, type II diabetes and non-neuropathic systemic amyloidosis respectively. Till date, all advances in PMDs therapeutics help symptomatically but do not prevent the root cause of the disease, i.e., the aggregation of protein involved in the diseases. Current efforts focused on developing therapies for PMDs have employed diverse strategies; repositioning pre-existing drugs as it saves time and money; natural compounds that are touted as potential drug candidates have an advantage of being taken in diet normally and will induce lesser side effects. This review also covers recently developed therapeutic strategies like antisense drugs and disaggregases which has yielded therapeutic agents that have transitioned from preclinical studies into human clinical trials.
Collapse
Affiliation(s)
- Asra Nasir Khan
- Interdisciplinary Biotechnology Unit, AMU, Aligarh 202002, India
| | | |
Collapse
|
12
|
Zhou J, Tan Y, Hu L, Fu J, Li D, Chen J, Long Y. Inhibition of HSPA8 by rifampicin contributes to ferroptosis via enhancing autophagy. Liver Int 2022; 42:2889-2899. [PMID: 36254713 DOI: 10.1111/liv.15459] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 08/17/2022] [Accepted: 08/24/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIM Rifampicin is the most common pathogenic factor in anti-tuberculosis drug-induced liver injury (AT-DILI), the mechanisms that it promotes hepatocyte damage in AT-DILI are not yet to be thoroughly elucidated. In this study, we investigated the potential molecular mechanisms for ferroptosis involving rifampicin hepatotoxicity. METHODS Animal and cell injury models of rifampicin were constructed, and the toxicity of rifampicin was assessed by physicochemical staining and cell viability assay. Next, flow cytometry was employed to detect changes in ferroptosis-related markers, and Western blotting was used to detect protein expression. Then, the important role of autophagy and ferroptosis was verified with small molecule compound intervention. RESULTS We found that ferritinophagy-induced ferroptosis participates in the toxicity of rifampicin, and the mechanism is that rifampicin precisely activates high-throughput autophagy, which leads to the massive degradation of ferritin and the increase of free iron. Moreover, rifampicin exhibited conspicuous inhibition of Human 71 kDa heat shock cognate protein (HSPA8) that is intimately associated with Microtubule-associated protein light chain 3 isoform B (LC3B) expression, in turn, HSPA8 inducer attenuated intracellular autophagy flux. Of note, inducing HSPA8 or inhibition of autophagy and ferroptosis considerably relieved the hepatotoxicity of rifampicin in mouse model. CONCLUSIONS The present study highlights the crucial roles of the HSPA8 and autophagy in ferroptotic cell death driving by rifampicin, particularly illumines multiple promising regulatory nodes for therapeutic interventions in diseases involving AT-DILI.
Collapse
Affiliation(s)
- Juan Zhou
- Department of Infectious Diseases, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China
| | - Yingzheng Tan
- Department of Infectious Diseases, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China
| | - Lingli Hu
- Department of Infectious Diseases, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China
| | - Jingli Fu
- Department of Infectious Diseases, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China
| | - Dan Li
- Department of Infectious Diseases, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China
| | - Jun Chen
- Department of Liver Diseases, Third Hospital of Shenzhen, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Yunzhu Long
- Department of Infectious Diseases, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China
| |
Collapse
|
13
|
Zhou R, Zhu L, Zeng Z, Luo R, Zhang J, Guo R, Zhang L, Zhang Q, Bi W. Targeted brain delivery of RVG29-modified rifampicin-loaded nanoparticles for Alzheimer's disease treatment and diagnosis. Bioeng Transl Med 2022; 7:e10395. [PMID: 36176608 PMCID: PMC9472014 DOI: 10.1002/btm2.10395] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 05/19/2022] [Accepted: 06/07/2022] [Indexed: 11/16/2022] Open
Abstract
Alzheimer's disease (AD) is an aging-related neurodegenerative disease. The main pathological features of AD are β-amyloid protein (Aβ) deposition and tau protein hyperphosphorylation. Currently, there are no effective drugs for the etiological treatment of AD. Rifampicin (RIF) is a semi-synthetic broad-spectrum antibiotic with anti-β-amyloid deposition, anti-inflammatory, anti-apoptosis, and neuroprotective effects, but its application in AD treatment has been limited for its strong hydrophobicity, high toxicity, short half-life, low bioavailability, and blood-brain barrier hindrance. We designed a novel brain-targeted and MRI-characteristic nanomedicine via loading rabies virus protein 29 (RVG29), rifampicin, and Gd on poly (l-lactide) nanoparticles (RIF@PLA-PEG-Gd/Mal-RVG29). The cytotoxicity assay demonstrated that RIF@PLA-PEG-Gd/Mal-RVG29 had favorable biocompatibility and security. Fluorescence imaging in vivo showed that PLA-PEG-Gd/Mal-RVG29 could deliver rifampicin into the brain by enhancing cellular uptake and brain targeting performance, leading to improvement of the bioavailability of rifampicin. In in vivo study, RIF@PLA-PEG-Gd/Mal-RVG29 improved the spatial learning and memory capability of APP/PS1 mice in the Morris water maze, as compared to rifampicin. Immunofluorescence, TEM, immunoblotting, and H&E staining revealed that RIF@PLA-PEG-Gd/Mal-RVG29 reduced Aβ deposition in hippocampal and cortex of APP/PS1 mice, improved the damage of synaptic ultrastructure, increased the expression level of PSD95 and SYP, as well as reduced the necrosis of neurons. These findings suggest that RIF@PLA-PEG-Gd/Mal-RVG29 may be an effective strategy for the treatment of AD.
Collapse
Affiliation(s)
- Ruiyi Zhou
- Department of NeurologyThe First Affiliated Hospital, Jinan UniversityGuangzhouPeople's Republic of China
| | - Lihong Zhu
- Department of PathophysiologyKey Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan UniversityGuangzhouPeople's Republic of China
| | - Zhaohao Zeng
- Department of NeurologyThe First Affiliated Hospital, Jinan UniversityGuangzhouPeople's Republic of China
| | - Rixin Luo
- Department of NeurologyThe First Affiliated Hospital, Jinan UniversityGuangzhouPeople's Republic of China
| | - Jiawei Zhang
- Department of PathophysiologyKey Laboratory of State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Medicine, Jinan UniversityGuangzhouPeople's Republic of China
| | - Rui Guo
- Key Laboratory of Biomaterials of Guangdong Higher Education InstitutesGuangdong Provincial Engineering and Technological Research Center for Drug Carrier Development, Department of Biomedical Engineering, Jinan UniversityGuangzhouPeople's Republic of China
| | - Lei Zhang
- Department of Cerebrovascular DiseaseThe Fifth Affiliated Hospital, Sun Yat‐sen UniversityZhuhaiPeople's Republic of China
| | - Qunying Zhang
- Department of CardiologyThe Fifth Affiliated Hospital, Sun Yat‐sen UniversityZhuhaiPeople's Republic of China
| | - Wei Bi
- Department of NeurologyThe First Affiliated Hospital, Jinan UniversityGuangzhouPeople's Republic of China
| |
Collapse
|
14
|
Forloni G, La Vitola P, Balducci C. Oligomeropathies, inflammation and prion protein binding. Front Neurosci 2022; 16:822420. [PMID: 36081661 PMCID: PMC9445368 DOI: 10.3389/fnins.2022.822420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 07/28/2022] [Indexed: 11/13/2022] Open
Abstract
The central role of oligomers, small soluble aggregates of misfolded proteins, in the pathogenesis of neurodegenerative disorders is recognized in numerous experimental conditions and is compatible with clinical evidence. To underline this concept, some years ago we coined the term oligomeropathies to define the common mechanism of action of protein misfolding diseases like Alzheimer, Parkinson or prion diseases. Using simple experimental conditions, with direct application of synthetic β amyloid or α-synuclein oligomers intraventricularly at micromolar concentrations, we could detect differences and similarities in the biological consequences. The two oligomer species affected cognitive behavior, neuronal dysfunction and cerebral inflammatory reactions with distinct mechanisms. In these experimental conditions the proposed mediatory role of cellular prion protein in oligomer activities was not confirmed. Together with oligomers, inflammation at different levels can be important early in neurodegenerative disorders; both β amyloid and α-synuclein oligomers induce inflammation and its control strongly affects neuronal dysfunction. This review summarizes our studies with β-amyloid or α-synuclein oligomers, also considering the potential curative role of doxycycline, a well-known antibiotic with anti-amyloidogenic and anti-inflammatory activities. These actions are analyzed in terms of the therapeutic prospects.
Collapse
|
15
|
Tran VTA, Lee LP, Cho H. Neuroinflammation in neurodegeneration via microbial infections. Front Immunol 2022; 13:907804. [PMID: 36052093 PMCID: PMC9425114 DOI: 10.3389/fimmu.2022.907804] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 07/01/2022] [Indexed: 11/13/2022] Open
Abstract
Recent epidemiological studies show a noticeable correlation between chronic microbial infections and neurological disorders. However, the underlying mechanisms are still not clear due to the biological complexity of multicellular and multiorgan interactions upon microbial infections. In this review, we show the infection leading to neurodegeneration mediated by multiorgan interconnections and neuroinflammation. Firstly, we highlight three inter-organ communications as possible routes from infection sites to the brain: nose-brain axis, lung-brain axis, and gut-brain axis. Next, we described the biological crosstalk between microglia and astrocytes upon pathogenic infection. Finally, our study indicates how neuroinflammation is a critical player in pathogen-mediated neurodegeneration. Taken together, we envision that antibiotics targeting neuro-pathogens could be a potential therapeutic strategy for neurodegeneration.
Collapse
Affiliation(s)
- Van Thi Ai Tran
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, South Korea
| | - Luke P. Lee
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, South Korea
- Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Harvard Institute of Medicine, Harvard University, Boston, MA, United States
- *Correspondence: Hansang Cho, ; Luke P. Lee,
| | - Hansang Cho
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, South Korea
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, South Korea
- *Correspondence: Hansang Cho, ; Luke P. Lee,
| |
Collapse
|
16
|
Gupta C, Xu J, Jin T, Khullar S, Liu X, Alatkar S, Cheng F, Wang D. Single-cell network biology characterizes cell type gene regulation for drug repurposing and phenotype prediction in Alzheimer's disease. PLoS Comput Biol 2022; 18:e1010287. [PMID: 35849618 PMCID: PMC9333448 DOI: 10.1371/journal.pcbi.1010287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 07/28/2022] [Accepted: 06/07/2022] [Indexed: 12/03/2022] Open
Abstract
Dysregulation of gene expression in Alzheimer's disease (AD) remains elusive, especially at the cell type level. Gene regulatory network, a key molecular mechanism linking transcription factors (TFs) and regulatory elements to govern gene expression, can change across cell types in the human brain and thus serve as a model for studying gene dysregulation in AD. However, AD-induced regulatory changes across brain cell types remains uncharted. To address this, we integrated single-cell multi-omics datasets to predict the gene regulatory networks of four major cell types, excitatory and inhibitory neurons, microglia and oligodendrocytes, in control and AD brains. Importantly, we analyzed and compared the structural and topological features of networks across cell types and examined changes in AD. Our analysis shows that hub TFs are largely common across cell types and AD-related changes are relatively more prominent in some cell types (e.g., microglia). The regulatory logics of enriched network motifs (e.g., feed-forward loops) further uncover cell type-specific TF-TF cooperativities in gene regulation. The cell type networks are also highly modular and several network modules with cell-type-specific expression changes in AD pathology are enriched with AD-risk genes. The further disease-module-drug association analysis suggests cell-type candidate drugs and their potential target genes. Finally, our network-based machine learning analysis systematically prioritized cell type risk genes likely involved in AD. Our strategy is validated using an independent dataset which showed that top ranked genes can predict clinical phenotypes (e.g., cognitive impairment) of AD with reasonable accuracy. Overall, this single-cell network biology analysis provides a comprehensive map linking genes, regulatory networks, cell types and drug targets and reveals cell-type gene dysregulation in AD.
Collapse
Affiliation(s)
- Chirag Gupta
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Jielin Xu
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Ting Jin
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Saniya Khullar
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Xiaoyu Liu
- Department of Statistics, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Sayali Alatkar
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Computer Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Feixiong Cheng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
| | - Daifeng Wang
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Computer Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| |
Collapse
|
17
|
Quantum-dot-labeled synuclein seed assay identifies drugs modulating the experimental prion-like transmission. Commun Biol 2022; 5:636. [PMID: 35768587 PMCID: PMC9243017 DOI: 10.1038/s42003-022-03590-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 06/15/2022] [Indexed: 11/08/2022] Open
Abstract
Synucleinopathies are neurodegenerative disorders including Parkinson disease (PD), dementia with Lewy body (DLB), and multiple system atrophy (MSA) that involve deposits of the protein alpha-synuclein (α-syn) in the brain. The inoculation of α-syn aggregates derived from synucleinopathy or preformed fibrils (PFF) formed in vitro induces misfolding and deposition of endogenous α-syn. This is referred to as prion-like transmission, and the mechanism is still unknown. In this study, we label α-syn PFF with quantum dots and visualize their movement directly in acute slices of brain tissue inoculated with α-syn PFF seeds. Using this system, we find that the trafficking of α-syn seeds is dependent on fast axonal transport and the seed spreading is dependent on endocytosis and neuronal activity. We also observe pharmacological effects on α-syn seed spreading; clinically available drugs including riluzole are effective in reducing the spread of α-syn seeds and this effect is also observed in vivo. Our quantum-dot-labeled α-syn seed assay system combined with in vivo transmission experiment reveals an early phase of transmission, in which uptake and spreading of seeds occur depending on neuronal activity, and a later phase, in which seeds induce the propagation of endogenous misfolded α-syn.
Collapse
|
18
|
Gąsiorowski K, Brokos JB, Sochocka M, Ochnik M, Chojdak-Łukasiewicz J, Zajączkowska K, Fułek M, Leszek J. Current and Near-Future Treatment of Alzheimer's Disease. Curr Neuropharmacol 2022; 20:1144-1157. [PMID: 34856906 PMCID: PMC9886829 DOI: 10.2174/1570159x19666211202124239] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 10/19/2021] [Accepted: 11/26/2021] [Indexed: 11/22/2022] Open
Abstract
Recent findings have improved our understanding of the multifactorial nature of AD. While in early asymptomatic stages of AD, increased amyloid-β synthesis and tau hyperphosphorylation play a key role, while in the latter stages of the disease, numerous dysfunctions of homeostatic mechanisms in neurons, glial cells, and cerebrovascular endothelium determine the rate of progression of clinical symptoms. The main driving forces of advanced neurodegeneration include increased inflammatory reactions in neurons and glial cells, oxidative stress, deficiencies in neurotrophic growth and regenerative capacity of neurons, brain insulin resistance with disturbed metabolism in neurons, or reduction of the activity of the Wnt-β catenin pathway, which should integrate the homeostatic mechanisms of brain tissue. In order to more effectively inhibit the progress of neurodegeneration, combination therapies consisting of drugs that rectify several above-mentioned dysfunctions should be used. It should be noted that many widely-used drugs from various pharmacological groups, "in addition" to the main therapeutic indications, have a beneficial effect on neurodegeneration and may be introduced into clinical practice in combination therapy of AD. There is hope that complex treatment will effectively inhibit the progression of AD and turn it into a slowly progressing chronic disease. Moreover, as the mechanisms of bidirectional communication between the brain and microbiota are better understood, it is expected that these pathways will be harnessed to provide novel methods to enhance health and treat AD.
Collapse
Affiliation(s)
| | | | - Marta Sochocka
- Laboratory of Virology, Department of Immunology of Infectious Diseases, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Michał Ochnik
- Laboratory of Virology, Department of Immunology of Infectious Diseases, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | | | | | - Michał Fułek
- Department of Internal Medicine, Occupational Diseases, Hypertension and Clinical Oncology, Wrocław Medical University, Wrocław, Poland
| | - Jerzy Leszek
- Department of Psychiatry, Wrocław Medical University, Wrocław, Poland,Address correspondence to this author at the Department of Psychiatry, Wrocław Medical University, 10 Ludwika Pasteura Str., 50-367 Wrocław, Poland; Tel:+48603880572; E-mail:
| |
Collapse
|
19
|
Prebble DW, Er S, Hlushchuk I, Domanskyi A, Airavaara M, Ekins MG, Mellick GD, Carroll AR. α-Synuclein binding activity of the plant growth promoter asterubine. Bioorg Med Chem Lett 2022; 64:128677. [PMID: 35301136 DOI: 10.1016/j.bmcl.2022.128677] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 03/05/2022] [Accepted: 03/12/2022] [Indexed: 11/02/2022]
Abstract
Preventing the aggregation of certain amyloid proteins has the potential to slow down the progression of diseases like Alzheimer's, Parkinson's, and type 2 diabetes mellitus. During a high-throughput screen of 300 Australian marine invertebrate extracts, the extract of the marine sponge Thorectandra sp. 4408 displayed binding activity to the Parkinson's disease-associated protein, α-synuclein. Isolation of the active component led to its identification as the known plant growth promoter asterubine (1). This molecule shares distinct structural similarities with potent amyloid beta aggregation inhibitors tramiprosate (homotaurine) and ALZ-801. Herein we report the isolation, NMR data acquired in DMSO and α-synuclein binding activity of asterubine (1).
Collapse
Affiliation(s)
- Dale W Prebble
- School of Environment and Science, Griffith University (Gold Coast Campus), Parklands Drive, Southport, QLD 4222, Australia; Griffith Institute for Drug Discovery, Griffith University (Brisbane Innovation Park), Don Young Road, Nathan, QLD 4111, Australia
| | - Safak Er
- Faculty of Pharmacy, University of Helsinki, Helsinki 00014, Finland; Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki 00014, Finland
| | - Irena Hlushchuk
- Faculty of Pharmacy, University of Helsinki, Helsinki 00014, Finland
| | - Andrii Domanskyi
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki 00014, Finland
| | - Mikko Airavaara
- Faculty of Pharmacy, University of Helsinki, Helsinki 00014, Finland; Neuroscience Center, HiLIFE, University of Helsinki, Helsinki 00014, Finland
| | - Merrick G Ekins
- Griffith Institute for Drug Discovery, Griffith University (Brisbane Innovation Park), Don Young Road, Nathan, QLD 4111, Australia; Queensland Museum, South Brisbane BC, QLD 4101, Australia
| | - George D Mellick
- School of Environment and Science, Griffith University (Gold Coast Campus), Parklands Drive, Southport, QLD 4222, Australia; Griffith Institute for Drug Discovery, Griffith University (Brisbane Innovation Park), Don Young Road, Nathan, QLD 4111, Australia
| | - Anthony R Carroll
- School of Environment and Science, Griffith University (Gold Coast Campus), Parklands Drive, Southport, QLD 4222, Australia; Griffith Institute for Drug Discovery, Griffith University (Brisbane Innovation Park), Don Young Road, Nathan, QLD 4111, Australia.
| |
Collapse
|
20
|
C9orf72 Hexanucleotide Repeat Expansion-Related Neuropathology Is Attenuated by Nasal Rifampicin in Mice. Biomedicines 2022; 10:biomedicines10051080. [PMID: 35625816 PMCID: PMC9138602 DOI: 10.3390/biomedicines10051080] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/27/2022] [Accepted: 05/02/2022] [Indexed: 12/02/2022] Open
Abstract
The non-coding GGGGCC hexanucleotide repeat expansion (HRE) in C9orf72 gene is a dominant cause of frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS). This intronic mutation elicits the formation of nuclear and cytoplasmic inclusions containing RNA, RNA-binding proteins, and HRE-derived dipeptide repeat proteins (DPRs), leading to neurodegeneration via the gain-of-toxic function or loss-of-function of relevant proteins. Using C9-500 mice harboring ~500 repeats of the GGGGCC sequence in human C9orf72 gene, we investigated the effects of rifampicin against HRE-related pathological phenotypes. Rifampicin was administered intranasally to 4.5- to 5-month-old mice for 1 month, and their cognitive function and neuropathology were assessed by the Morris water maze test and immunohistochemical staining. Rifampicin treatment reduced the formation of RNA foci and cytoplasmic inclusions containing DPRs or phosphorylated TDP-43, and furthermore, the levels of phosphorylated double-strand RNA-dependent protein kinase (PKR) that regulates repeat-associated non-ATG (RAN) translation. Synapse loss in the hippocampus and neuronal loss and microglial activation in the prefrontal and motor cortices were also attenuated, and mouse memory was significantly improved. Our findings suggest a therapeutic potential of nasal rifampicin in the prevention of C9orf72-linked neurodegenerative disorders.
Collapse
|
21
|
Islam M, Shen F, Regmi D, Du D. Therapeutic strategies for tauopathies and drug repurposing as a potential approach. Biochem Pharmacol 2022; 198:114979. [PMID: 35219701 PMCID: PMC9159505 DOI: 10.1016/j.bcp.2022.114979] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 02/03/2022] [Accepted: 02/21/2022] [Indexed: 11/26/2022]
Abstract
Tauopathies are neurodegenerative diseases characterized by the deposition of abnormal tau in the brain. To date, there are no disease-modifying therapies approved by the U.S. Food and Drug Administration (US FDA) for the treatment of tauopathies. In the past decades, extensive efforts have been provided to develop disease-modifying therapies to treat tauopathies. Specifically, exploring existing drugs with the intent of repurposing for the treatment of tauopathies affords a reasonable alternative to discover potent drugs for treating these formidable diseases. Drug repurposing will not only reduce formulation and development stage effort and cost but will also take a key advantage of the established toxicological studies, which is one of the main causes of clinical trial failure of new molecules. In this review, we provide an overview of the current treatment strategies for tauopathies and the recent progress in drug repurposing as an alternative approach to treat tauopathies.
Collapse
Affiliation(s)
- Majedul Islam
- Department of Chemistry and Biochemistry, Florida Atlantic University, Boca Raton, FL 33431, United States.
| | - Fengyun Shen
- Department of Chemistry and Biochemistry, Florida Atlantic University, Boca Raton, FL 33431, United States
| | - Deepika Regmi
- Department of Chemistry and Biochemistry, Florida Atlantic University, Boca Raton, FL 33431, United States
| | - Deguo Du
- Department of Chemistry and Biochemistry, Florida Atlantic University, Boca Raton, FL 33431, United States.
| |
Collapse
|
22
|
Bartley SC, Proctor MT, Xia H, Ho E, Kang DS, Schuster K, Bicca MA, Seckler HS, Viola KL, Patrie SM, Kelleher NL, De Mello FG, Klein WL. An Essential Role for Alzheimer’s-Linked Amyloid Beta Oligomers in Neurodevelopment: Transient Expression of Multiple Proteoforms during Retina Histogenesis. Int J Mol Sci 2022; 23:ijms23042208. [PMID: 35216328 PMCID: PMC8875314 DOI: 10.3390/ijms23042208] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 02/11/2022] [Accepted: 02/14/2022] [Indexed: 12/04/2022] Open
Abstract
Human amyloid beta peptide (Aβ) is a brain catabolite that at nanomolar concentrations can form neurotoxic oligomers (AβOs), which are known to accumulate in Alzheimer’s disease. Because a predisposition to form neurotoxins seems surprising, we have investigated whether circumstances might exist where AβO accumulation may in fact be beneficial. Our investigation focused on the embryonic chick retina, which expresses the same Aβ as humans. Using conformation-selective antibodies, immunoblots, mass spectrometry, and fluorescence microscopy, we discovered that AβOs are indeed present in the developing retina, where multiple proteoforms are expressed in a highly regulated cell-specific manner. The expression of the AβO proteoforms was selectively associated with transiently expressed phosphorylated Tau (pTau) proteoforms that, like AβOs, are linked to Alzheimer’s disease (AD). To test whether the AβOs were functional in development, embryos were cultured ex ovo and then injected intravitreally with either a beta-site APP-cleaving enzyme 1 (BACE-1) inhibitor or an AβO-selective antibody to prematurely lower the levels of AβOs. The consequence was disrupted histogenesis resulting in dysplasia resembling that seen in various retina pathologies. We suggest the hypothesis that embryonic AβOs are a new type of short-lived peptidergic hormone with a role in neural development. Such a role could help explain why a peptide that manifests deleterious gain-of-function activity when it oligomerizes in the aging brain has been evolutionarily conserved.
Collapse
Affiliation(s)
- Samuel C. Bartley
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA; (S.C.B.); (M.T.P.); (H.X.); (E.H.); (D.S.K.); (K.S.); (M.A.B.); (K.L.V.)
| | - Madison T. Proctor
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA; (S.C.B.); (M.T.P.); (H.X.); (E.H.); (D.S.K.); (K.S.); (M.A.B.); (K.L.V.)
| | - Hongjie Xia
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA; (S.C.B.); (M.T.P.); (H.X.); (E.H.); (D.S.K.); (K.S.); (M.A.B.); (K.L.V.)
| | - Evelyn Ho
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA; (S.C.B.); (M.T.P.); (H.X.); (E.H.); (D.S.K.); (K.S.); (M.A.B.); (K.L.V.)
| | - Dong S. Kang
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA; (S.C.B.); (M.T.P.); (H.X.); (E.H.); (D.S.K.); (K.S.); (M.A.B.); (K.L.V.)
| | - Kristen Schuster
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA; (S.C.B.); (M.T.P.); (H.X.); (E.H.); (D.S.K.); (K.S.); (M.A.B.); (K.L.V.)
| | - Maíra A. Bicca
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA; (S.C.B.); (M.T.P.); (H.X.); (E.H.); (D.S.K.); (K.S.); (M.A.B.); (K.L.V.)
| | - Henrique S. Seckler
- Department of Chemistry, Northwestern University, Evanston, IL 60208, USA; (H.S.S.); (S.M.P.)
| | - Kirsten L. Viola
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA; (S.C.B.); (M.T.P.); (H.X.); (E.H.); (D.S.K.); (K.S.); (M.A.B.); (K.L.V.)
| | - Steven M. Patrie
- Department of Chemistry, Northwestern University, Evanston, IL 60208, USA; (H.S.S.); (S.M.P.)
| | - Neil L. Kelleher
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA;
| | - Fernando G. De Mello
- Instituto de Biofísica Carlos Chagas Filho (IBCCF), Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| | - William L. Klein
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA; (S.C.B.); (M.T.P.); (H.X.); (E.H.); (D.S.K.); (K.S.); (M.A.B.); (K.L.V.)
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University, Chicago, IL 60611, USA
- Correspondence: ; Tel.: +1-847-591-5510
| |
Collapse
|
23
|
Umeda T, Uekado R, Shigemori K, Eguchi H, Tomiyama T. Nasal Rifampicin Halts the Progression of Tauopathy by Inhibiting Tau Oligomer Propagation in Alzheimer Brain Extract-Injected Mice. Biomedicines 2022; 10:biomedicines10020297. [PMID: 35203506 PMCID: PMC8869211 DOI: 10.3390/biomedicines10020297] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/24/2022] [Accepted: 01/25/2022] [Indexed: 01/27/2023] Open
Abstract
The cell-to-cell transmission of tau aggregates is considered a mechanism underlying the intracerebral spreading of tau pathology in Alzheimer’s disease (AD) and other tauopathies. Recent studies suggest that tau oligomers, rather than fibrils, participate in this process. We previously showed that intranasal rifampicin inhibits tau oligomer accumulation and improves cognition in tauopathy mice. In the present study, we examined the effects of nasal rifampicin on tau propagation in a new mouse model of tauopathy. A tau oligomer-rich fraction prepared from the brain of an AD patient was injected into a unilateral hippocampus of tau264 mice that express both 3-repeat and 4-repeat wild-type human tau. Rifampicin administration was started one week after the injection and performed three times a week for 24 weeks. Cognitive function and tau pathology were assessed by the Morris water maze test and brain section staining. Rifampicin treatment inhibited the spreading of tau oligomers from the injection site to other brain regions and neurofibrillary tangle formation in the entorhinal cortex. Synapse and neuronal loss in the hippocampus were also prevented, and cognitive function remained normal. These results suggest that intranasal rifampicin could be a promising remedy that halts the progression of tauopathy by inhibiting tau oligomer propagation.
Collapse
Affiliation(s)
- Tomohiro Umeda
- Department of Translational Neuroscience, Graduate School of Medicine, Osaka City University, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan; (T.U.); (R.U.); (K.S.)
| | - Rumi Uekado
- Department of Translational Neuroscience, Graduate School of Medicine, Osaka City University, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan; (T.U.); (R.U.); (K.S.)
| | - Keiko Shigemori
- Department of Translational Neuroscience, Graduate School of Medicine, Osaka City University, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan; (T.U.); (R.U.); (K.S.)
| | - Hiroshi Eguchi
- Pharmaceutical Discovery Research Laboratories, Teijin Pharma Ltd., 4-3-2 Asahigaoka, Hino 191-8512, Japan;
| | - Takami Tomiyama
- Department of Translational Neuroscience, Graduate School of Medicine, Osaka City University, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan; (T.U.); (R.U.); (K.S.)
- Correspondence: ; Tel.: +81-6-6645-3921
| |
Collapse
|
24
|
Hideshima M, Kimura Y, Aguirre C, Kakuda K, Takeuchi T, Choong CJ, Doi J, Nabekura K, Yamaguchi K, Nakajima K, Baba K, Nagano S, Goto Y, Nagai Y, Mochizuki H, Ikenaka K. Two-step screening method to identify α-synuclein aggregation inhibitors for Parkinson's disease. Sci Rep 2022; 12:351. [PMID: 35013421 PMCID: PMC8748996 DOI: 10.1038/s41598-021-04131-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 12/13/2021] [Indexed: 12/14/2022] Open
Abstract
Parkinson's disease is a neurodegenerative disease characterized by the formation of neuronal inclusions of α-synuclein in patient brains. As the disease progresses, toxic α-synuclein aggregates transmit throughout the nervous system. No effective disease-modifying therapy has been established, and preventing α-synuclein aggregation is thought to be one of the most promising approaches to ameliorate the disease. In this study, we performed a two-step screening using the thioflavin T assay and a cell-based assay to identify α-synuclein aggregation inhibitors. The first screening, thioflavin T assay, allowed the identification of 30 molecules, among a total of 1262 FDA-approved small compounds, which showed inhibitory effects on α-synuclein fibrilization. In the second screening, a cell-based aggregation assay, seven out of these 30 candidates were found to prevent α-synuclein aggregation without causing substantial toxicity. Of the seven final candidates, tannic acid was the most promising compound. The robustness of our screening method was validated by a primary neuronal cell model and a Caenorhabditis elegans model, which demonstrated the effect of tannic acid against α-synuclein aggregation. In conclusion, our two-step screening system is a powerful method for the identification of α-synuclein aggregation inhibitors, and tannic acid is a promising candidate as a disease-modifying drug for Parkinson's disease.
Collapse
Affiliation(s)
- Makoto Hideshima
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yasuyoshi Kimura
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - César Aguirre
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Keita Kakuda
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Toshihide Takeuchi
- Department of Neurology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-sayama, Osaka, 589-8511, Japan
- Department of Neurotherapeutics, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Chi-Jing Choong
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Junko Doi
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Kei Nabekura
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Keiichi Yamaguchi
- Global Center for Medical Engineering and Informatics, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Kichitaro Nakajima
- Global Center for Medical Engineering and Informatics, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Kousuke Baba
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Seiichi Nagano
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
- Department of Neurotherapeutics, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yuji Goto
- Global Center for Medical Engineering and Informatics, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yoshitaka Nagai
- Department of Neurology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-sayama, Osaka, 589-8511, Japan
- Department of Neurotherapeutics, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hideki Mochizuki
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Kensuke Ikenaka
- Department of Neurology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
25
|
Umeda T, Sakai A, Shigemori K, Yokota A, Kumagai T, Tomiyama T. Oligomer-Targeting Prevention of Neurodegenerative Dementia by Intranasal Rifampicin and Resveratrol Combination - A Preclinical Study in Model Mice. Front Neurosci 2021; 15:763476. [PMID: 34966254 PMCID: PMC8710719 DOI: 10.3389/fnins.2021.763476] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 11/23/2021] [Indexed: 12/15/2022] Open
Abstract
Amyloidogenic protein oligomers are thought to play an important role in the pathogenesis of neurodegenerative dementia, including Alzheimer’s disease, frontotemporal dementia, and dementia with Lewy bodies. Previously we demonstrated that oral or intranasal rifampicin improved the cognition of APP-, tau-, and α-synuclein-transgenic mice by reducing the amount of Aβ, tau, and α-synuclein oligomers in the brain. In the present study, to explore more effective and safer medications for dementia, we tested the drug combination of rifampicin and resveratrol, which is a multifunctional natural polyphenol with the potential to antagonize the adverse effects of rifampicin. The mixture was intranasally administered to APP-, tau-, and α-synuclein-transgenic mice, and their memory and oligomer-related pathologies were evaluated. Compared with rifampicin and resveratrol alone, the combinatorial medicine significantly improved mouse cognition, reduced amyloid oligomer accumulation, and recovered synaptophysin levels in the hippocampus. The plasma levels of liver enzymes, which reflect hepatic injury and normally increase by rifampicin treatment, remained normal by the combination treatment. Notably, resveratrol alone and the combinatorial medicine, but not rifampicin alone, enhanced the levels of brain-derived neurotrophic factor (BDNF) and its precursor, pro-BDNF, in the hippocampus. Furthermore, the combination showed a synergistic effect in ameliorating mouse cognition. These results show the advantages of this combinatorial medicine with regards to safety and effectiveness over single-drug rifampicin. Our findings may provide a feasible means for the prevention of neurodegenerative dementia that targets toxic oligomers.
Collapse
Affiliation(s)
- Tomohiro Umeda
- Department of Translational Neuroscience, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Ayumi Sakai
- Department of Translational Neuroscience, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Keiko Shigemori
- Department of Translational Neuroscience, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Ayumi Yokota
- Department of Translational Neuroscience, Osaka City University Graduate School of Medicine, Osaka, Japan
| | | | - Takami Tomiyama
- Department of Translational Neuroscience, Osaka City University Graduate School of Medicine, Osaka, Japan.,Medilabo RFP, Inc., Kyoto, Japan
| |
Collapse
|
26
|
Minamisawa M, Sato Y, Ishiguro E, Taniai T, Sakamoto T, Kawai G, Saito T, Saido TC. Amelioration of Alzheimer's Disease by Gut-Pancreas-Liver-Brain Interaction in an App Knock-In Mouse Model. Life (Basel) 2021; 12:34. [PMID: 35054427 PMCID: PMC8778338 DOI: 10.3390/life12010034] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/19/2021] [Accepted: 12/20/2021] [Indexed: 02/07/2023] Open
Abstract
In this study, we observed disease progression, changes in the gut microbiota, and interactions among the brain, liver, pancreas, and intestine in a mouse model of Alzheimer's disease (AD), in addition to attempting to inhibit disease progression through the dietary supplementation of L-arginine and limonoids. Wild-type mice (WC) and AD mice were fed a normal diet (AC), a diet supplemented with L-arginine and limonoids (ALA), or a diet containing only limonoids (AL) for 12-64 weeks. The normal diet-fed WC and AC mice showed a decrease in the diversity of the gut microbiota, with an increase in the Firmicutes/Bacteroidetes ratio, and bacterial translocation. Considerable bacterial translocation to the pancreas and intense inflammation of the pancreas, liver, brain, and intestinal tissues were observed in the AC mice from alterations in the gut microbiota. The ALA diet or AL diet-fed mice showed increased diversity of the bacterial flora and suppressed oxidative stress and inflammatory responses in hepatocytes and pancreatic cells, bacterial translocation, and neurodegeneration of the brain. These findings suggest that L-arginine and limonoids help in maintaining the homeostasis of the gut microbiota, pancreas, liver, brain, and gut in AD mice.
Collapse
Affiliation(s)
- Mayumi Minamisawa
- Department of Life Science, Chiba Institute of Technology, Graduate School of Advanced Engineering, Chiba 275-0016, Japan; (Y.S.); (T.S.); (G.K.)
- Education Center, Faculty of Advanced Engineering, Chiba Institute of Technology, Chiba 275-0023, Japan;
| | - Yuma Sato
- Department of Life Science, Chiba Institute of Technology, Graduate School of Advanced Engineering, Chiba 275-0016, Japan; (Y.S.); (T.S.); (G.K.)
| | | | - Tetsuyuki Taniai
- Education Center, Faculty of Advanced Engineering, Chiba Institute of Technology, Chiba 275-0023, Japan;
| | - Taiichi Sakamoto
- Department of Life Science, Chiba Institute of Technology, Graduate School of Advanced Engineering, Chiba 275-0016, Japan; (Y.S.); (T.S.); (G.K.)
| | - Gota Kawai
- Department of Life Science, Chiba Institute of Technology, Graduate School of Advanced Engineering, Chiba 275-0016, Japan; (Y.S.); (T.S.); (G.K.)
| | - Takashi Saito
- RIKEN Center for Brain Science, Laboratory for Proteolytic Neuroscience, Saitama 351-0198, Japan; (T.S.); (T.C.S.)
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan
| | - Takaomi C. Saido
- RIKEN Center for Brain Science, Laboratory for Proteolytic Neuroscience, Saitama 351-0198, Japan; (T.S.); (T.C.S.)
| |
Collapse
|
27
|
Kaya-Tilki E, Dikmen M. Neuroprotective effects of some epigenetic modifying drugs' on Chlamydia pneumoniae-induced neuroinflammation: A novel model. PLoS One 2021; 16:e0260633. [PMID: 34847172 PMCID: PMC8631675 DOI: 10.1371/journal.pone.0260633] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 11/14/2021] [Indexed: 12/02/2022] Open
Abstract
Chlamydia pneumoniae (Cpn) is a gram-negative intracellular pathogen that causes a variety of pulmonary diseases, and there is growing evidence that it may play a role in Alzheimer's disease (AD) pathogenesis. Cpn can interact functionally with host histones, altering the host's epigenetic regulatory system by introducing bacterial products into the host tissue and inducing a persistent inflammatory response. Because Cpn is difficult to propagate, isolate, and detect, a modified LPS-like neuroinflammation model was established using lyophilized cell free supernatant (CFS) obtained from infected cell cultures, and the effects of CFS were compared to LPS. The neuroprotective effects of Trichostatin A (TSA), givinostat, and RG108, which are effective on epigenetic mechanisms, and the antibiotic rifampin, were studied in this newly introduced model and in the presence of amyloid beta (Aβ) 1-42. The neuroprotective effects of the drugs, as well as the effects of CFS and LPS, were evaluated in Aβ-induced neurotoxicity using a real-time cell analysis system, total ROS, and apoptotic impact. TSA, RG108, givinostat, and rifampin all demonstrated neuroprotective effects in both this novel model and Aβ-induced neurotoxicity. The findings are expected to provide early evidence on neuroprotective actions against Cpn-induced neuroinflammation and Aβ-induced neurotoxicity, which could represent a new treatment option for AD, for which there are currently few treatment options.
Collapse
Affiliation(s)
- Elif Kaya-Tilki
- Department of Pharmacology, Faculty of Pharmacy, Anadolu University, Eskisehir, Turkey
| | - Miriş Dikmen
- Department of Pharmacology, Faculty of Pharmacy, Anadolu University, Eskisehir, Turkey
| |
Collapse
|
28
|
Tau activates microglia via the PQBP1-cGAS-STING pathway to promote brain inflammation. Nat Commun 2021; 12:6565. [PMID: 34782623 PMCID: PMC8592984 DOI: 10.1038/s41467-021-26851-2] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 10/23/2021] [Indexed: 12/15/2022] Open
Abstract
Brain inflammation generally accompanies and accelerates neurodegeneration. Here we report a microglial mechanism in which polyglutamine binding protein 1 (PQBP1) senses extrinsic tau 3R/4R proteins by direct interaction and triggers an innate immune response by activating a cyclic GMP-AMP synthase (cGAS)-Stimulator of interferon genes (STING) pathway. Tamoxifen-inducible and microglia-specific depletion of PQBP1 in primary culture in vitro and mouse brain in vivo shows that PQBP1 is essential for sensing-tau to induce nuclear translocation of nuclear factor κB (NFκB), NFκB-dependent transcription of inflammation genes, brain inflammation in vivo, and eventually mouse cognitive impairment. Collectively, PQBP1 is an intracellular receptor in the cGAS-STING pathway not only for cDNA of human immunodeficiency virus (HIV) but also for the transmissible neurodegenerative disease protein tau. This study characterises a mechanism of brain inflammation that is common to virus infection and neurodegenerative disorders.
Collapse
|
29
|
Dai B, Zhong T, Chen ZX, Chen W, Zhang N, Liu XL, Wang LQ, Chen J, Liang Y. Myricetin slows liquid-liquid phase separation of Tau and activates ATG5-dependent autophagy to suppress Tau toxicity. J Biol Chem 2021; 297:101222. [PMID: 34560101 PMCID: PMC8551527 DOI: 10.1016/j.jbc.2021.101222] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 09/16/2021] [Accepted: 09/20/2021] [Indexed: 01/05/2023] Open
Abstract
Intraneuronal neurofibrillary tangles composed of Tau aggregates have been widely accepted as an important pathological hallmark of Alzheimer's disease. A current therapeutic avenue for treating Alzheimer's disease is aimed at inhibiting Tau accumulation with small molecules such as natural flavonoids. Liquid-liquid phase separation (LLPS) of Tau can lead to its aggregation, and Tau aggregates can then be degraded by autophagy. However, it is unclear whether natural flavonoids modulate the formation of phase-separated Tau droplets or promote autophagy and Tau clearance. Here, using confocal microscopy and fluorescence recovery after photobleaching assays, we report that a natural antioxidant flavonoid compound myricetin slows LLPS of full-length human Tau, shifting the equilibrium phase boundary to a higher protein concentration. This natural flavonoid also significantly inhibits pathological phosphorylation and abnormal aggregation of Tau in neuronal cells and blocks mitochondrial damage and apoptosis induced by Tau aggregation. Importantly, using coimmunoprecipitation and Western blotting, we show that treatment of cells with myricetin stabilizes the interaction between Tau and autophagy-related protein 5 (ATG5) to promote clearance of phosphorylated Tau to indirectly limit its aggregation. Consistently, this natural flavonoid inhibits mTOR pathway, activates ATG5-dependent Tau autophagy, and almost completely suppresses Tau toxicity in neuronal cells. Collectively, these results demonstrate how LLPS and abnormal aggregation of Tau are inhibited by natural flavonoids, bridging the gap between Tau LLPS and aggregation in neuronal cells, and also establish that myricetin could act as an ATG5-dependent autophagic activator to ameliorate the pathogenesis of Alzheimer's disease.
Collapse
Affiliation(s)
- Bin Dai
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China; Wuhan University Shenzhen Research Institute, Shenzhen, China
| | - Tao Zhong
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China; Wuhan University Shenzhen Research Institute, Shenzhen, China
| | - Zhi-Xian Chen
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China; Wuhan University Shenzhen Research Institute, Shenzhen, China
| | - Wang Chen
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China; Wuhan University Shenzhen Research Institute, Shenzhen, China
| | - Na Zhang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China; Wuhan University Shenzhen Research Institute, Shenzhen, China
| | - Xiao-Ling Liu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China; Wuhan University Shenzhen Research Institute, Shenzhen, China
| | - Li-Qiang Wang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China; Wuhan University Shenzhen Research Institute, Shenzhen, China
| | - Jie Chen
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China; Wuhan University Shenzhen Research Institute, Shenzhen, China
| | - Yi Liang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China; Wuhan University Shenzhen Research Institute, Shenzhen, China.
| |
Collapse
|
30
|
Ibrahim NF, Hamezah HS, Yanagisawa D, Tsuji M, Kiuchi Y, Ono K, Tooyama I. The effect of α-tocopherol, α- and γ-tocotrienols on amyloid-β aggregation and disaggregation in vitro. Biochem Biophys Rep 2021; 28:101131. [PMID: 34541343 PMCID: PMC8435913 DOI: 10.1016/j.bbrep.2021.101131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 09/07/2021] [Accepted: 09/07/2021] [Indexed: 11/27/2022] Open
Abstract
One of the neuropathological hallmarks of Alzheimer's disease (AD)-causing neurodegeneration and consequent memory deterioration, and eventually, cognitive decline-is amyloid-β (Aβ) aggregation forming amyloid plaques. Our previous study showed the potential of a tocotrienol-rich fraction-a mixture of naturally occurring of vitamin E analogs-to inhibit Aβ aggregation and restore cognitive function in an AD mouse model. The current study examined the effect of three vitamin E analogs-α-tocopherol (α-TOC), α-tocotrienol (α-T3), and γ-tocotrienol (γ-T3)-on Aβ aggregation, disaggregation, and oligomerization in vitro. Thioflavin T (ThT) assay showed α-T3 reduced Aβ aggregation at 10 μM concentration. Furthermore, both α-T3 and γ-T3 demonstrated Aβ disaggregation, as shown by the reduction of ThT fluorescence. However, α-TOC showed no significant effect. We confirmed the results for ThT assays with scanning electron microscopy imaging. Further investigation in photo-induced cross-linking of unmodified protein assay indicated a reduction in Aβ oligomerization by γ-T3. The present study thus revealed the individual effect of each tocotrienol analog in reducing Aβ aggregation and oligomerization as well as disaggregating preformed fibrils.
Collapse
Affiliation(s)
- Nor Faeizah Ibrahim
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Jalan Yaacob Latif, Cheras, Kuala Lumpur, 56000, Malaysia
| | - Hamizah Shahirah Hamezah
- Institute of Systems Biology (INBIOSIS), Universiti Kebangsaan Malaysia, 43600, UKM Bangi, Selangor, Malaysia
| | - Daijiro Yanagisawa
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, 520-2192, Japan
| | - Mayumi Tsuji
- Department of Pharmacology, Showa University School of Medicine, Shinagawa-ku, Tokyo, 142-8555, Japan
| | - Yuji Kiuchi
- Department of Pharmacology, Showa University School of Medicine, Shinagawa-ku, Tokyo, 142-8555, Japan
| | - Kenjiro Ono
- Department of Neurology, Showa University School of Medicine, Shinagawa-ku, Tokyo, 142-8666, Japan
| | - Ikuo Tooyama
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, 520-2192, Japan
| |
Collapse
|
31
|
Dow CT. Warm, Sweetened Milk at the Twilight of Immunity - Alzheimer's Disease - Inflammaging, Insulin Resistance, M. paratuberculosis and Immunosenescence. Front Immunol 2021; 12:714179. [PMID: 34421917 PMCID: PMC8375433 DOI: 10.3389/fimmu.2021.714179] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 07/09/2021] [Indexed: 01/22/2023] Open
Abstract
This article prosecutes a case against the zoonotic pathogen Mycobacterium avium ss. paratuberculosis (MAP) as a precipitant of Alzheimer’s disease (AD). Like the other major neurodegenerative diseases AD is, at its core, a proteinopathy. Aggregated extracellular amyloid protein plaques and intracellular tau protein tangles are the recognized protein pathologies of AD. Autophagy is the cellular housekeeping process that manages protein quality control and recycling, cellular metabolism, and pathogen elimination. Impaired autophagy and cerebral insulin resistance are invariant features of AD. With a backdrop of age-related low-grade inflammation (inflammaging) and heightened immune risk (immunosenescence), infection with MAP subverts glucose metabolism and further exhausts an already exhausted autophagic capacity. Increasingly, a variety of agents have been found to favorably impact AD; they are agents that promote autophagy and reduce insulin resistance. The potpourri of these therapeutic agents: mTOR inhibitors, SIRT1 activators and vaccines are seemingly random until one recognizes that all these agents also suppress intracellular mycobacterial infection. The zoonotic mycobacterial MAP causes a common fatal enteritis in ruminant animals. Humans are exposed to MAP from contaminated food products and from the environment. The enteritis in animals is called paratuberculosis or Johne’s disease; in humans, it is the putative cause of Crohn’s disease. Beyond Crohn’s, MAP is associated with an increasing number of inflammatory and autoimmune diseases: sarcoidosis, Blau syndrome, autoimmune diabetes, autoimmune thyroiditis, multiple sclerosis, and rheumatoid arthritis. Moreover, MAP has been associated with Parkinson’s disease. India is one county that has extensively studied the human bio-load of MAP; 30% of more than 28,000 tested individuals were found to harbor, or to have harbored, MAP. This article asserts an unfolding realization that MAP infection of humans 1) is widespread in its presence, 2) is wide-ranging in its zoonosis and 3) provides a plausible link connecting MAP to AD.
Collapse
Affiliation(s)
- Coad Thomas Dow
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
32
|
The Role of Autophagy in Anti-Cancer and Health Promoting Effects of Cordycepin. Molecules 2021; 26:molecules26164954. [PMID: 34443541 PMCID: PMC8400201 DOI: 10.3390/molecules26164954] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/10/2021] [Accepted: 08/12/2021] [Indexed: 12/18/2022] Open
Abstract
Cordycepin is an adenosine derivative isolated from Cordyceps sinensis, which has been used as an herbal complementary and alternative medicine with various biological activities. The general anti-cancer mechanisms of cordycepin are regulated by the adenosine A3 receptor, epidermal growth factor receptor (EGFR), mitogen-activated protein kinases (MAPKs), and glycogen synthase kinase (GSK)-3β, leading to cell cycle arrest or apoptosis. Notably, cordycepin also induces autophagy to trigger cell death, inhibits tumor metastasis, and modulates the immune system. Since the dysregulation of autophagy is associated with cancers and neuron, immune, and kidney diseases, cordycepin is considered an alternative treatment because of the involvement of cordycepin in autophagic signaling. However, the profound mechanism of autophagy induction by cordycepin has never been reviewed in detail. Therefore, in this article, we reviewed the anti-cancer and health-promoting effects of cordycepin in the neurons, kidneys, and the immune system through diverse mechanisms, including autophagy induction. We also suggest that formulation changes for cordycepin could enhance its bioactivity and bioavailability and lower its toxicity for future applications. A comprehensive understanding of the autophagy mechanism would provide novel mechanistic insight into the anti-cancer and health-promoting effects of cordycepin.
Collapse
|
33
|
Nasal Rifampicin Improves Cognition in a Mouse Model of Dementia with Lewy Bodies by Reducing α-Synuclein Oligomers. Int J Mol Sci 2021; 22:ijms22168453. [PMID: 34445158 PMCID: PMC8395129 DOI: 10.3390/ijms22168453] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 07/29/2021] [Accepted: 08/04/2021] [Indexed: 12/02/2022] Open
Abstract
α-Synuclein oligomers are thought to play an important role in the pathogenesis of dementia with Lewy bodies (DLB). There is no effective cure for DLB at present. Previously, we demonstrated that in APP- and tau-transgenic mice, oral or intranasal rifampicin reduced brain Aβ and tau oligomers and improved mouse cognition. In the present study, we expanded our research to DLB. Rifampicin was intranasally administered to 6-month-old A53T-mutant α-synuclein-transgenic mice at 0.1 mg/day for 1 month. The mice displayed memory impairment but no motor deficit at this age, indicating a suitable model of DLB. α-Synuclein pathologies were examined by the immunohistochemical/biochemical analyses of brain tissues. Cognitive function was evaluated by the Morris water maze test. Intranasal rifampicin significantly reduced the levels of [pSer129] α-synuclein in the hippocampus and α-synuclein oligomers in the visual cortex and hippocampus. The level of the presynaptic marker synaptophysin in the hippocampus was recovered to the level in non-transgenic littermates. In the Morris water maze, a significant improvement in spatial reference memory was observed in rifampicin-treated mice. Taken together with our previous findings, these results suggest that intranasal rifampicin is a promising remedy for the prevention of neurodegenerative dementia, including Alzheimer’s disease, frontotemporal dementia, and DLB.
Collapse
|
34
|
Autophagy and Tau Protein. Int J Mol Sci 2021; 22:ijms22147475. [PMID: 34299093 PMCID: PMC8303176 DOI: 10.3390/ijms22147475] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/22/2021] [Accepted: 07/01/2021] [Indexed: 12/19/2022] Open
Abstract
Neurofibrillary tangles, which consist of highly phosphorylated tau protein, and senile plaques (SPs) are pathological hallmarks of Alzheimer's disease (AD). In swollen axons, many autophagic vacuoles are observed around SP in the AD brain. This suggests that autophagy function is disturbed in AD. We used a neuronal cellular model of tauopathy (M1C cells), which harbors wild type tau (4R0N), to assess the effects of the lysosomotrophic agent NH4Cl, and autophagy inhibitors chloroquine and 3 methyladenine (3MA). It was found that chloroquine, NH4Cl and 3MA markedly increased tau accumulation. Thus, autophagy lysosomal system disturbances disturbed the degradation mechanisms of tau protein. Other studies also revealed that tau protein, including aggregated tau, is degraded via the autophagy lysosome system. Phosphorylated and C terminal truncated tau were also reported to disturb autophagy function. As a therapeutic strategy, autophagy upregulation was suggested. Thus far, as autophagy modulators, rapamycin, mTOCR1 inhibitor and its analogues, lithium, metformin, clonidine, curcumin, nicotinamide, bexaroten, and torehalose have been proposed. As a therapeutic strategy, autophagic modulation may be the next target of AD therapeutics.
Collapse
|
35
|
Rampa A, Gobbi S, Belluti F, Bisi A. Tackling Alzheimer's Disease with Existing Drugs: A Promising Strategy for Bypassing Obstacles. Curr Med Chem 2021; 28:2305-2327. [PMID: 32867634 DOI: 10.2174/0929867327666200831140745] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/22/2020] [Accepted: 08/08/2020] [Indexed: 11/22/2022]
Abstract
The unmet need for the development of effective drugs to treat Alzheimer 's disease has been steadily growing, representing a major challenge in drug discovery. In this context, drug repurposing, namely the identification of novel therapeutic indications for approved or investigational compounds, can be seen as an attractive attempt to obtain new medications reducing both the time and the economic burden usually required for research and development programs. In the last years, several classes of drugs have evidenced promising beneficial effects in neurodegenerative diseases, and for some of them, preliminary clinical trials have been started. This review aims to illustrate some of the most recent examples of drugs reprofiled for Alzheimer's disease, considering not only the finding of new uses for existing drugs but also the new hypotheses on disease pathogenesis that could promote previously unconsidered therapeutic regimens. Moreover, some examples of structural modifications performed on existing drugs in order to obtain multifunctional compounds will also be described.
Collapse
Affiliation(s)
- Angela Rampa
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, I-40126 Bologna, Italy
| | - Silvia Gobbi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, I-40126 Bologna, Italy
| | - Federica Belluti
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, I-40126 Bologna, Italy
| | - Alessandra Bisi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, I-40126 Bologna, Italy
| |
Collapse
|
36
|
Nakai T, Yamada K, Mizoguchi H. Alzheimer's Disease Animal Models: Elucidation of Biomarkers and Therapeutic Approaches for Cognitive Impairment. Int J Mol Sci 2021; 22:5549. [PMID: 34074018 PMCID: PMC8197360 DOI: 10.3390/ijms22115549] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 05/21/2021] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is an age-related and progressive neurodegenerative disorder. It is widely accepted that AD is mainly caused by the accumulation of extracellular amyloid β (Aβ) and intracellular neurofibrillary tau tangles. Aβ begins to accumulate years before the onset of cognitive impairment, suggesting that the benefit of currently available interventions would be greater if they were initiated in the early phases of AD. To understand the mechanisms of AD pathogenesis, various transgenic mouse models with an accelerated accumulation of Aβ and tau tangles have been developed. However, none of these models exhibit all pathologies present in human AD. To overcome these undesirable phenotypes, APP knock-in mice, which were presented with touchscreen-based tasks, were developed to better evaluate the efficacy of candidate therapeutics in mouse models of early-stage AD. This review assesses several AD mouse models from the aspect of biomarkers and cognitive impairment and discusses their potential as tools to provide novel AD therapeutic approaches.
Collapse
Affiliation(s)
- Tsuyoshi Nakai
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya 466-8560, Japan; (T.N.); (K.Y.)
| | - Kiyofumi Yamada
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya 466-8560, Japan; (T.N.); (K.Y.)
| | - Hiroyuki Mizoguchi
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya 466-8560, Japan; (T.N.); (K.Y.)
- Medical Interactive Research and Academia Industry Collaboration Center, Research Institute of Environmental Medicine, Nagoya University, Nagoya 464-8601, Japan
| |
Collapse
|
37
|
Anti-Alzheimer's Molecules Derived from Marine Life: Understanding Molecular Mechanisms and Therapeutic Potential. Mar Drugs 2021; 19:md19050251. [PMID: 33925063 PMCID: PMC8146595 DOI: 10.3390/md19050251] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 04/19/2021] [Accepted: 04/26/2021] [Indexed: 02/08/2023] Open
Abstract
Alzheimer’s disease (AD) is a devastating neurodegenerative disease and the most common cause of dementia. It has been confirmed that the pathological processes that intervene in AD development are linked with oxidative damage to neurons, neuroinflammation, tau phosphorylation, amyloid beta (Aβ) aggregation, glutamate excitotoxicity, and cholinergic deficit. Still, there is no available therapy that can cure AD. Available therapies only manage some of the AD symptoms at the early stages of AD. Various studies have revealed that bioactive compounds derived from marine organisms and plants can exert neuroprotective activities with fewer adverse events, as compared with synthetic drugs. Furthermore, marine organisms have been identified as a source of novel compounds with therapeutic potential. Thus, there is a growing interest regarding bioactive compounds derived from marine sources that have anti-AD potentials. Various marine drugs including bryostatin-1, homotaurine, anabaseine and its derivative, rifampicins, anhydroexfoliamycin, undecylprodigioisin, gracilins, 13-desmethyl spirolide-C, and dictyostatin displayed excellent bioavailability and efficacy against AD. Most of these marine drugs were found to be well-tolerated in AD patients, along with no significant drug-associated adverse events. In this review, we focus on the drugs derived from marine life that can be useful in AD treatment and also summarize the therapeutic agents that are currently used to treat AD.
Collapse
|
38
|
Medina L, González-Lizárraga F, Dominguez-Meijide A, Ploper D, Parrales V, Sequeira S, Cima-Omori MS, Zweckstetter M, Del Bel E, Michel PP, Outeiro TF, Raisman-Vozari R, Chehín R, Socias SB. Doxycycline Interferes With Tau Aggregation and Reduces Its Neuronal Toxicity. Front Aging Neurosci 2021; 13:635760. [PMID: 33828477 PMCID: PMC8020845 DOI: 10.3389/fnagi.2021.635760] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/22/2021] [Indexed: 01/15/2023] Open
Abstract
Tauopathies are neurodegenerative disorders with increasing incidence and still without cure. The extensive time required for development and approval of novel therapeutics highlights the need for testing and repurposing known safe molecules. Since doxycycline impacts α-synuclein aggregation and toxicity, herein we tested its effect on tau. We found that doxycycline reduces amyloid aggregation of the 2N4R and K18 isoforms of tau protein in a dose-dependent manner. Furthermore, in a cell free system doxycycline also prevents tau seeding and in cell culture reduces toxicity of tau aggregates. Overall, our results expand the spectrum of action of doxycycline against aggregation-prone proteins, opening novel perspectives for its repurposing as a disease-modifying drug for tauopathies.
Collapse
Affiliation(s)
- Luciana Medina
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (UNT-CONICET-SIPROSA), Tucumán, Argentina
| | - Florencia González-Lizárraga
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (UNT-CONICET-SIPROSA), Tucumán, Argentina
| | - Antonio Dominguez-Meijide
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Goettingen, Goettingen, Germany.,Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Diego Ploper
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (UNT-CONICET-SIPROSA), Tucumán, Argentina
| | - Valeria Parrales
- Sorbonne Université, Paris Brain Institute -ICM, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Assistance Publique - Hôpitaux de Paris (APHP), Hôpital de la Pitié Salpêtrière, Paris, France
| | - Sabrina Sequeira
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (UNT-CONICET-SIPROSA), Tucumán, Argentina
| | - Maria-Sol Cima-Omori
- German Center for Neurodegenerative Diseases Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Göttingen, Germany
| | - Markus Zweckstetter
- German Center for Neurodegenerative Diseases Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Göttingen, Germany.,Department for NMR-based Structural Biology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Elaine Del Bel
- Physiology- Dental School of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Patrick P Michel
- Sorbonne Université, Paris Brain Institute -ICM, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Assistance Publique - Hôpitaux de Paris (APHP), Hôpital de la Pitié Salpêtrière, Paris, France
| | - Tiago Fleming Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Goettingen, Goettingen, Germany.,Max Planck Institute for Experimental Medicine, Goettingen, Germany.,Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Rita Raisman-Vozari
- Sorbonne Université, Paris Brain Institute -ICM, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Assistance Publique - Hôpitaux de Paris (APHP), Hôpital de la Pitié Salpêtrière, Paris, France
| | - Rosana Chehín
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (UNT-CONICET-SIPROSA), Tucumán, Argentina
| | - Sergio B Socias
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (UNT-CONICET-SIPROSA), Tucumán, Argentina
| |
Collapse
|
39
|
Noori T, Dehpour AR, Sureda A, Sobarzo-Sanchez E, Shirooie S. Role of natural products for the treatment of Alzheimer's disease. Eur J Pharmacol 2021; 898:173974. [PMID: 33652057 DOI: 10.1016/j.ejphar.2021.173974] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 02/09/2021] [Accepted: 02/19/2021] [Indexed: 12/19/2022]
Abstract
Negative psychological and physiological consequences of neurodegenerative disorders represent a high social and health cost. Among the neurodegenerative disorders Alzheimer's disease (AD) is recognized as a leading neurodegenerative condition and a primary cause of dementia in the elderlys. AD is considered as neurodegenerative disorder that progressively impairs cognitive function and memory. According to current epidemiological data, about 50 milLion people worldwide are suffering from AD. The primary symptoms of AD are almost inappreciable and usually comprise forgetfulness of recent events. Numerous processes are involved in the development of AD, for example oxidative stress (OS) mainly due to mitochondrial dysfunction, intracellular the accumulation of hyperphosphorylated tau (τ) proteins in the form of neurofibrillary tangles, excessive the accumulation of extracellular plaques of beta-amyloid (Aβ), genetic and environmental factors. Running treatments only attenuate symptoms and temporarily reduce the rate of cognitive progression associated with AD. This means that most treatments focus only on controlLing symptoms, particularly in the initial stages of the disease. In the past, the first choice of treatment was based on natural ingredients. In this sense, diverse natural products (NPs) are capable to decrease the symptoms and alleviate the development of several diseases including AD attracting the attention of the scientific community and the pharmaceutical industry. Specifically, numerous NPs including flavonoids, gingerols, tannins, anthocyanins, triterpenes and alkaloids have been shown anti-inflammatory, antioxidant, anti-amyloidogenic, and anti-choLinesterase properties. This review provide a summary of the pathogenesis and the therapeutic goals of AD. It also discusses the available data on various plants and isolated natural compounds used to prevent and diminish the symptoms of AD.
Collapse
Affiliation(s)
- Tayebeh Noori
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ahmad Reza Dehpour
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Experimental Medicine Research Center, TUMS, Tehran, Iran
| | - Antoni Sureda
- Research Group on Community Nutrition and Oxidative Stress (NUCOX), University Research Institute of Health Sciences (IUNICS), and Health Research Institute of Balearic Islands (IdISBa), University of Balearic Islands, Palma de Mallorca E-07122, Balearic Islands, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029, Madrid, Spain
| | - Eduardo Sobarzo-Sanchez
- Instituto de Investigación y Postgrado, Facultad de Ciencias de la Salud, Universidad Central de Chile, Santiago, Chile; Department of Organic Chemistry, Faculty of Pharmacy, University of Santiago de Compostela, Spain
| | - Samira Shirooie
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
40
|
Takeda K, Uda A, Mitsubori M, Nagashima S, Iwasaki H, Ito N, Shiiba I, Ishido S, Matsuoka M, Inatome R, Yanagi S. Mitochondrial ubiquitin ligase alleviates Alzheimer's disease pathology via blocking the toxic amyloid-β oligomer generation. Commun Biol 2021; 4:192. [PMID: 33580194 PMCID: PMC7881000 DOI: 10.1038/s42003-021-01720-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 11/23/2020] [Indexed: 12/28/2022] Open
Abstract
Mitochondrial pathophysiology is implicated in the development of Alzheimer's disease (AD). An integrative database of gene dysregulation suggests that the mitochondrial ubiquitin ligase MITOL/MARCH5, a fine-tuner of mitochondrial dynamics and functions, is downregulated in patients with AD. Here, we report that the perturbation of mitochondrial dynamics by MITOL deletion triggers mitochondrial impairments and exacerbates cognitive decline in a mouse model with AD-related Aβ pathology. Notably, MITOL deletion in the brain enhanced the seeding effect of Aβ fibrils, but not the spontaneous formation of Aβ fibrils and plaques, leading to excessive secondary generation of toxic and dispersible Aβ oligomers. Consistent with this, MITOL-deficient mice with Aβ etiology exhibited worsening cognitive decline depending on Aβ oligomers rather than Aβ plaques themselves. Our findings suggest that alteration in mitochondrial morphology might be a key factor in AD due to directing the production of Aβ form, oligomers or plaques, responsible for disease development.
Collapse
Affiliation(s)
- Keisuke Takeda
- Laboratory of Molecular Biochemistry, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
- Department of Biology, University of Padova, Padova, Italy
- Venetian Institute of Molecular Medicine, Padova, Italy
| | - Aoi Uda
- Laboratory of Molecular Biochemistry, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| | - Mikihiro Mitsubori
- Laboratory of Molecular Biochemistry, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| | - Shun Nagashima
- Laboratory of Molecular Biochemistry, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| | - Hiroko Iwasaki
- Laboratory of Molecular Biochemistry, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| | - Naoki Ito
- Laboratory of Molecular Biochemistry, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
- Laboratory of Molecular Biochemistry, Department of Life Science, Faculty of Science, Gakushuin University, Toshima-ku, Tokyo, Japan
| | - Isshin Shiiba
- Laboratory of Molecular Biochemistry, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
- Laboratory of Molecular Biochemistry, Department of Life Science, Faculty of Science, Gakushuin University, Toshima-ku, Tokyo, Japan
| | - Satoshi Ishido
- Department of Microbiology, Hyogo College of Medicine, Nishinomiya, Japan
| | - Masaaki Matsuoka
- Department of Pharmacology, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| | - Ryoko Inatome
- Laboratory of Molecular Biochemistry, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
- Laboratory of Molecular Biochemistry, Department of Life Science, Faculty of Science, Gakushuin University, Toshima-ku, Tokyo, Japan
| | - Shigeru Yanagi
- Laboratory of Molecular Biochemistry, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan.
- Laboratory of Molecular Biochemistry, Department of Life Science, Faculty of Science, Gakushuin University, Toshima-ku, Tokyo, Japan.
| |
Collapse
|
41
|
Vigasova D, Nemergut M, Liskova B, Damborsky J. Multi-pathogen infections and Alzheimer's disease. Microb Cell Fact 2021; 20:25. [PMID: 33509204 PMCID: PMC7844946 DOI: 10.1186/s12934-021-01520-7] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 01/16/2021] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disease associated with the overproduction and accumulation of amyloid-β peptide and hyperphosphorylation of tau proteins in the brain. Despite extensive research on the amyloid-based mechanism of AD pathogenesis, the underlying cause of AD is not fully understood. No disease-modifying therapies currently exist, and numerous clinical trials have failed to demonstrate any benefits. The recent discovery that the amyloid-β peptide has antimicrobial activities supports the possibility of an infectious aetiology of AD and suggests that amyloid-β plaque formation might be induced by infection. AD patients have a weakened blood-brain barrier and immune system and are thus at elevated risk of microbial infections. Such infections can cause chronic neuroinflammation, production of the antimicrobial amyloid-β peptide, and neurodegeneration. Various pathogens, including viruses, bacteria, fungi, and parasites have been associated with AD. Most research in this area has focused on individual pathogens, with herpesviruses and periodontal bacteria being most frequently implicated. The purpose of this review is to highlight the potential role of multi-pathogen infections in AD. Recognition of the potential coexistence of multiple pathogens and biofilms in AD's aetiology may stimulate the development of novel approaches to its diagnosis and treatment. Multiple diagnostic tests could be applied simultaneously to detect major pathogens, followed by anti-microbial treatment using antiviral, antibacterial, antifungal, and anti-biofilm agents.
Collapse
Affiliation(s)
- Dana Vigasova
- International Clinical Research Center, St. Anne’s University Hospital Brno, Pekarska 53, 656 91 Brno, Czech Republic
- Department of Experimental Biology and RECETOX, Faculty of Science, Loschmidt Laboratories, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| | - Michal Nemergut
- Department of Experimental Biology and RECETOX, Faculty of Science, Loschmidt Laboratories, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| | - Barbora Liskova
- Department of Experimental Biology and RECETOX, Faculty of Science, Loschmidt Laboratories, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| | - Jiri Damborsky
- International Clinical Research Center, St. Anne’s University Hospital Brno, Pekarska 53, 656 91 Brno, Czech Republic
- Department of Experimental Biology and RECETOX, Faculty of Science, Loschmidt Laboratories, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| |
Collapse
|
42
|
The Microbiota-Gut-Brain Axis and Alzheimer's Disease: Neuroinflammation Is to Blame? Nutrients 2020; 13:nu13010037. [PMID: 33374235 PMCID: PMC7824474 DOI: 10.3390/nu13010037] [Citation(s) in RCA: 124] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 12/20/2020] [Accepted: 12/22/2020] [Indexed: 02/06/2023] Open
Abstract
For years, it has been reported that Alzheimer’s disease (AD) is the most common cause of dementia. Various external and internal factors may contribute to the early onset of AD. This review highlights a contribution of the disturbances in the microbiota–gut–brain (MGB) axis to the development of AD. Alteration in the gut microbiota composition is determined by increase in the permeability of the gut barrier and immune cell activation, leading to impairment in the blood–brain barrier function that promotes neuroinflammation, neuronal loss, neural injury, and ultimately AD. Numerous studies have shown that the gut microbiota plays a crucial role in brain function and changes in the behavior of individuals and the formation of bacterial amyloids. Lipopolysaccharides and bacterial amyloids synthesized by the gut microbiota can trigger the immune cells residing in the brain and can activate the immune response leading to neuroinflammation. Growing experimental and clinical data indicate the prominent role of gut dysbiosis and microbiota–host interactions in AD. Modulation of the gut microbiota with antibiotics or probiotic supplementation may create new preventive and therapeutic options in AD. Accumulating evidences affirm that research on MGB involvement in AD is necessary for new treatment targets and therapies for AD.
Collapse
|
43
|
Quinovic Acid Impedes Cholesterol Dyshomeostasis, Oxidative Stress, and Neurodegeneration in an Amyloid- β-Induced Mouse Model. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:9523758. [PMID: 33274012 PMCID: PMC7700034 DOI: 10.1155/2020/9523758] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 10/18/2020] [Accepted: 10/28/2020] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder typified by several neuropathological features including amyloid-beta (Aβ) plaque and neurofibrillary tangles (NFTs). Cholesterol retention and oxidative stress (OS) are the major contributors of elevated β- and γ-secretase activities, leading to excessive Aβ deposition, signifying the importance of altered cholesterol homeostasis and OS in the progression of Aβ-mediated neurodegeneration and cognitive deficit. However, the effect of Aβ on cholesterol metabolism is lesser-known. In this study, we evaluated the effect of quinovic acid (QA; 50 mg/kg body weight, i.p.) against the intracerebroventricular (i.c.v.) injection of Aβ (1-42)-induced cholesterol dyshomeostasis, oxidative stress, and neurodegeneration in the cortex and hippocampal brain regions of wild-type male C57BL/6J mice. Our results indicated that Aβ (1-42)-treated mice have increased Aβ oligomer formation along with increased β-secretase expression. The enhanced amyloidogenic pathway in Aβ (1-42)-treated mice intensified brain cholesterol accumulation due to increased expressions of p53 and 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR) enzyme. Importantly, we further confirmed the p53-mediated HMGCR axis activation by using pifithrin-α (PFT) in SH-SY5Y cells. Furthermore, the augmented brain cholesterol levels were also associated with increased OS. However, the QA administration to Aβ (1-42)-injected mice significantly ameliorated the Aβ burden, p53 expression, and cholesterol accumulation by deterring the oxidative stress through upregulating the Nrf2/HO-1 pathway. Moreover, the QA downregulated gliosis, neuroinflammatory mediators (p-NF-κB and IL-1β), and the expression of mitochondrial apoptotic markers (Bax, cleaved caspase-3, and cytochrome c). QA treatment also reversed the deregulated synaptic markers (PSD-95 and synaptophysin) and improved spatial learning and memory behaviors in the Aβ-treated mouse brains. These results suggest that Aβ (1-42) induces its acute detrimental effects on cognitive functions probably by increasing brain cholesterol levels through a possible activation of the p53/HMGCR axis. However, QA treatment reduces the cholesterol-induced oxidative stress, neuroinflammation, and neurodegeneration, leading to the restoration of cognitive deficit after Aβ (1-42) i.c.v. injection in mice.
Collapse
|
44
|
Antimicrobial therapy and the potential mechanisms in Alzheimer's disease. Neurosci Lett 2020; 741:135464. [PMID: 33166642 DOI: 10.1016/j.neulet.2020.135464] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/22/2020] [Accepted: 10/23/2020] [Indexed: 11/20/2022]
Abstract
Alzheimer's disease treatments have been a heavily investigated research area, however, new drugs have failed one after another. Some scientists have begun to reposition drugs, including antimicrobial agents. Here, the treatment effects of nine antimicrobial agents on Alzheimer's disease and their possible therapeutic mechanisms are described to clarify their efficacy. In vivo and in vitro studies are quite encouraging and tend to demonstrate that antimicrobial therapy is effective in Alzheimer's disease. Nevertheless, unsatisfactory clinical efficacy, side effects, and insufficient knowledge have yet to be overcome. Further laboratory and clinical studies are required to recommend antimicrobial treatment regimens.
Collapse
|
45
|
Terry C. Insights from nature: A review of natural compounds that target protein misfolding in vivo. CURRENT RESEARCH IN BIOTECHNOLOGY 2020. [DOI: 10.1016/j.crbiot.2020.10.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
46
|
Bulgart HR, Neczypor EW, Wold LE, Mackos AR. Microbial involvement in Alzheimer disease development and progression. Mol Neurodegener 2020; 15:42. [PMID: 32709243 PMCID: PMC7382139 DOI: 10.1186/s13024-020-00378-4] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 04/29/2020] [Indexed: 02/08/2023] Open
Abstract
Alzheimer disease (AD) is the most prominent form of dementia and the 5th leading cause of death in individuals over 65. AD is a complex disease stemming from genetic, environmental, and lifestyle factors. It is known that AD patients have increased levels of senile plaques, neurofibrillary tangles, and neuroinflammation; however, the mechanism(s) by which the plaques, tangles, and neuroinflammation manifest remain elusive. A recent hypothesis has emerged that resident bacterial populations contribute to the development and progression of AD by contributing to neuroinflammation, senile plaque formation, and potentially neurofibrillary tangle accumulation (Fig. 1). This review will highlight recent studies involved in elucidating microbial involvement in AD development and progression.
Collapse
Affiliation(s)
- Hannah R. Bulgart
- Biomedical Sciences Graduate Program, College of Medicine, The Ohio State University, Columbus, OH USA
| | - Evan W. Neczypor
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH USA
- College of Nursing, The Ohio State University, 1585 Neil Ave, Columbus, OH 43210 USA
| | - Loren E. Wold
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH USA
- College of Nursing, The Ohio State University, 1585 Neil Ave, Columbus, OH 43210 USA
- Department of Physiology and Cell Biology, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, OH USA
| | - Amy R. Mackos
- College of Nursing, The Ohio State University, 1585 Neil Ave, Columbus, OH 43210 USA
| |
Collapse
|
47
|
Iqbal UH, Zeng E, Pasinetti GM. The Use of Antimicrobial and Antiviral Drugs in Alzheimer's Disease. Int J Mol Sci 2020; 21:E4920. [PMID: 32664669 PMCID: PMC7404195 DOI: 10.3390/ijms21144920] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/09/2020] [Accepted: 07/10/2020] [Indexed: 12/15/2022] Open
Abstract
The aggregation and accumulation of amyloid-β plaques and tau proteins in the brain have been central characteristics in the pathophysiology of Alzheimer's disease (AD), making them the focus of most of the research exploring potential therapeutics for this neurodegenerative disease. With success in interventions aimed at depleting amyloid-β peptides being limited at best, a greater understanding of the physiological role of amyloid-β peptides is needed. The development of amyloid-β plaques has been determined to occur 10-20 years prior to AD symptom manifestation, hence earlier interventions might be necessary to address presymptomatic AD. Furthermore, recent studies have suggested that amyloid-β peptides may play a role in innate immunity as an antimicrobial peptide. These findings, coupled with the evidence of pathogens such as viruses and bacteria in AD brains, suggests that the buildup of amyloid-β plaques could be a response to the presence of viruses and bacteria. This has led to the foundation of the antimicrobial hypothesis for AD. The present review will highlight the current understanding of amyloid-β, and the role of bacteria and viruses in AD, and will also explore the therapeutic potential of antimicrobial and antiviral drugs in Alzheimer's disease.
Collapse
Affiliation(s)
| | | | - Giulio M. Pasinetti
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (U.H.I.); (E.Z.)
| |
Collapse
|
48
|
Panza F, Lozupone M, Solfrizzi V, Watling M, Imbimbo BP. Time to test antibacterial therapy in Alzheimer's disease. Brain 2020; 142:2905-2929. [PMID: 31532495 DOI: 10.1093/brain/awz244] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 05/24/2019] [Accepted: 06/14/2019] [Indexed: 12/19/2022] Open
Abstract
Alzheimer's disease is associated with cerebral accumulation of amyloid-β peptide and hyperphosphorylated tau. In the past 28 years, huge efforts have been made in attempting to treat the disease by reducing brain accumulation of amyloid-β in patients with Alzheimer's disease, with no success. While anti-amyloid-β therapies continue to be tested in prodromal patients with Alzheimer's disease and in subjects at risk of developing Alzheimer's disease, there is an urgent need to provide therapeutic support to patients with established Alzheimer's disease for whom current symptomatic treatment (acetylcholinesterase inhibitors and N-methyl d-aspartate antagonist) provide limited help. The possibility of an infectious aetiology for Alzheimer's disease has been repeatedly postulated over the past three decades. Infiltration of the brain by pathogens may act as a trigger or co-factor for Alzheimer's disease, with Herpes simplex virus type 1, Chlamydia pneumoniae, and Porphyromonas gingivalis being most frequently implicated. These pathogens may directly cross a weakened blood-brain barrier, reach the CNS and cause neurological damage by eliciting neuroinflammation. Alternatively, pathogens may cross a weakened intestinal barrier, reach vascular circulation and then cross blood-brain barrier or cause low grade chronic inflammation and subsequent neuroinflammation from the periphery. The gut microbiota comprises a complex community of microorganisms. Increased permeability of the gut and blood-brain barrier induced by microbiota dysbiosis may impact Alzheimer's disease pathogenesis. Inflammatory microorganisms in gut microbiota are associated with peripheral inflammation and brain amyloid-β deposition in subjects with cognitive impairment. Oral microbiota may also influence Alzheimer's disease risk through circulatory or neural access to the brain. At least two possibilities can be envisaged to explain the association of suspected pathogens and Alzheimer's disease. One is that patients with Alzheimer's disease are particularly prone to microbial infections. The other is that microbial infection is a contributing cause of Alzheimer's disease. Therapeutic trials with antivirals and/or antibacterials could resolve this dilemma. Indeed, antiviral agents are being tested in patients with Alzheimer's disease in double-blind placebo-controlled studies. Although combined antibiotic therapy was found to be effective in animal models of Alzheimer's disease, antibacterial drugs are not being widely investigated in patients with Alzheimer's disease. This is because it is not clear which bacterial populations in the gut of patients with Alzheimer's disease are overexpressed and if safe, selective antibacterials are available for them. On the other hand, a bacterial protease inhibitor targeting P. gingivalis toxins is now being tested in patients with Alzheimer's disease. Clinical studies are needed to test if countering bacterial infection may be beneficial in patients with established Alzheimer's disease.
Collapse
Affiliation(s)
- Francesco Panza
- Unit of Epidemiological Research on Aging, National Institute of Gastroenterology 'Saverio de Bellis', Research Hospital, Castellana Grotte, Bari, Italy
| | - Madia Lozupone
- Unit of Epidemiological Research on Aging, National Institute of Gastroenterology 'Saverio de Bellis', Research Hospital, Castellana Grotte, Bari, Italy
- Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, and Sense Organs, University of Bari Aldo Moro, Bari, Italy
| | - Vincenzo Solfrizzi
- 'C. Frugoni' Internal and Geriatric Medicine and Memory Unit, University of Bari Aldo Moro, Bari, Italy
| | - Mark Watling
- Department of Research and Development, Chiesi Farmaceutici, Parma, Italy
| | - Bruno P Imbimbo
- Department of Research and Development, Chiesi Farmaceutici, Parma, Italy
| |
Collapse
|
49
|
Anti-aggregation Effects of Phenolic Compounds on α-synuclein. Molecules 2020; 25:molecules25102444. [PMID: 32456274 PMCID: PMC7288075 DOI: 10.3390/molecules25102444] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 05/20/2020] [Accepted: 05/22/2020] [Indexed: 12/13/2022] Open
Abstract
The aggregation and deposition of α-synuclein (αS) are major pathologic features of Parkinson’s disease, dementia with Lewy bodies, and other α-synucleinopathies. The propagation of αS pathology in the brain plays a key role in the onset and progression of clinical phenotypes. Thus, there is increasing interest in developing strategies that attenuate αS aggregation and propagation. Based on cumulative evidence that αS oligomers are neurotoxic and critical species in the pathogenesis of α-synucleinopathies, we and other groups reported that phenolic compounds inhibit αS aggregation including oligomerization, thereby ameliorating αS oligomer-induced cellular and synaptic toxicities. Heterogeneity in gut microbiota may influence the efficacy of dietary polyphenol metabolism. Our recent studies on the brain-penetrating polyphenolic acids 3-hydroxybenzoic acid (3-HBA), 3,4-dihydroxybenzoic acid (3,4-diHBA), and 3-hydroxyphenylacetic acid (3-HPPA), which are derived from gut microbiota-based metabolism of dietary polyphenols, demonstrated an in vitro ability to inhibit αS oligomerization and mediate aggregated αS-induced neurotoxicity. Additionally, 3-HPPA, 3,4-diHBA, 3-HBA, and 4-hydroxybenzoic acid significantly attenuated intracellular αS seeding aggregation in a cell-based system. This review focuses on recent research developments regarding neuroprotective properties, especially anti-αS aggregation effects, of phenolic compounds and their metabolites by the gut microbiome, including our findings in the pathogenesis of α-synucleinopathies.
Collapse
|
50
|
Long-term exposure to constant light induces dementia, oxidative stress and promotes aggregation of sub-pathological Aβ42 in Wistar rats. Pharmacol Biochem Behav 2020; 192:172892. [DOI: 10.1016/j.pbb.2020.172892] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 03/02/2020] [Accepted: 03/02/2020] [Indexed: 12/17/2022]
|