1
|
Lüleci HB, Jones A, Çakır T. Multi-omics analyses highlight molecular differences between clinical and neuropathological diagnoses in Alzheimer's disease. Eur J Neurosci 2024; 60:4922-4936. [PMID: 39072881 DOI: 10.1111/ejn.16482] [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: 02/11/2024] [Revised: 05/14/2024] [Accepted: 07/13/2024] [Indexed: 07/30/2024]
Abstract
Both clinical diagnosis and neuropathological diagnosis are commonly used in literature to categorize individuals as Alzheimer's disease (AD) or non-AD in omics analyses. Whether these diagnostic strategies result in distinct profiles of molecular abnormalities is poorly understood. Here, we analysed one of the most commonly used AD omics datasets in the literature from the Religious Orders Study and Memory and Aging Project (ROSMAP) cohort and compared the two diagnosis strategies using brain transcriptome and metabolome by grouping individuals as non-AD and AD according to clinical or neuropathological diagnosis separately. Differentially expressed genes, associated pathways related with AD hallmarks and AD-related genes showed that the categorization based on neuropathological diagnosis more accurately reflects the disease state at the molecular level than the categorization based on clinical diagnosis. We further identified consensus biomarker candidates between the two diagnosis strategies such as 5-hydroxylysine, sphingomyelin and 1-myristoyl-2-palmitoyl-GPC as metabolite biomarkers and sphingolipid metabolism as a pathway biomarker, which could be robust AD biomarkers since they are independent of diagnosis strategies. We also used consensus AD and consensus non-AD individuals between the two diagnostic strategies to train a machine-learning based model, which we used to classify the individuals who were cognitively normal but diagnosed as AD based on neuropathological diagnosis (asymptomatic AD individuals). The majority of these individuals were classified as consensus AD patients for both omics data types. Our study provides a detailed characterization of both diagnostic strategies in terms of the association of the corresponding multi-omics profiles with AD.
Collapse
Affiliation(s)
| | - Attila Jones
- Clinical and Translational Neuroscience Section, Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Bethesda, Maryland, USA
| | - Tunahan Çakır
- Department of Bioengineering, Gebze Technical University, Kocaeli, Turkey
| |
Collapse
|
2
|
He K, Nie L, Yang C, Liu Z, Huang X, Li S, Yang X. Exhaustive exercise decreases tau phosphorylation and modifies biological processes associated with the protein translation and electron transport chain in P301L tau transgenic mice. Exp Gerontol 2024; 187:112375. [PMID: 38320733 DOI: 10.1016/j.exger.2024.112375] [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: 12/26/2023] [Revised: 01/30/2024] [Accepted: 02/02/2024] [Indexed: 02/13/2024]
Abstract
Stress response is a fundamental mechanism for cell survival, providing protection under unfavorable conditions. Mitochondrial stress, in particular, can trigger mitophagy, a process that restores cellular health. Exhaustive exercise (EE) is a form of acute mitochondrial stress. The objective of this current study is to investigate the impact of EE on tau pathology in pR5 mice, as well as the potential underlying mechanisms. To evaluate this, we examined the levels of total and phosphorylated tau in the hippocampus of pR5 mice, both with and without EE treatment. Furthermore, the application of weighted correlation network analysis (WGCNA) was employed to identify protein modules associated with the phenotype following the proteomic experiment. The findings of our study demonstrated a significant decrease in tau phosphorylation levels upon EE treatment, in comparison to the pR5 group. Moreover, the proteomic analysis provided additional insights, revealing that the mitigation of tau pathology was primarily attributed to the modulation of various pathways, such as translation factors and oxidative phosphorylation. Additionally, the analysis of heatmaps revealed a significant impact of EE treatment on the translation process and electron transport chain in pR5 mice. Furthermore, biochemical analysis provided further confirmation that EE treatment effectively modulated the ATP level in pR5 mice. In conclusion, our study suggests that the observed decrease in tau phosphorylation resulting from EE treatment may primarily be attributed to its regulation of the translation process and enhancement of mitochondrial function.
Collapse
Affiliation(s)
- Kaiwu He
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Medical Key Discipline of Health Toxicology (2020-2024), Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China; Department of Anesthesiology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), No. 1017, Dongmen North Road, Shenzhen 518020, China; Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, China
| | - Lulin Nie
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Medical Key Discipline of Health Toxicology (2020-2024), Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China; Institute of New Drug Research, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education, Jinan University College of Pharmacy, Jinan University, Guangzhou, China
| | - Chen Yang
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Medical Key Discipline of Health Toxicology (2020-2024), Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Zizhen Liu
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Xinfeng Huang
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Medical Key Discipline of Health Toxicology (2020-2024), Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Shupeng Li
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China.
| | - Xifei Yang
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Medical Key Discipline of Health Toxicology (2020-2024), Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China.
| |
Collapse
|
3
|
Kawade N, Yamanaka K. Novel insights into brain lipid metabolism in Alzheimer's disease: Oligodendrocytes and white matter abnormalities. FEBS Open Bio 2024; 14:194-216. [PMID: 37330425 PMCID: PMC10839347 DOI: 10.1002/2211-5463.13661] [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: 05/10/2023] [Revised: 06/07/2023] [Accepted: 06/14/2023] [Indexed: 06/19/2023] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia. A genome-wide association study has shown that several AD risk genes are involved in lipid metabolism. Additionally, epidemiological studies have indicated that the levels of several lipid species are altered in the AD brain. Therefore, lipid metabolism is likely changed in the AD brain, and these alterations might be associated with an exacerbation of AD pathology. Oligodendrocytes are glial cells that produce the myelin sheath, which is a lipid-rich insulator. Dysfunctions of the myelin sheath have been linked to white matter abnormalities observed in the AD brain. Here, we review the lipid composition and metabolism in the brain and myelin and the association between lipidic alterations and AD pathology. We also present the abnormalities in oligodendrocyte lineage cells and white matter observed in AD. Additionally, we discuss metabolic disorders, including obesity, as AD risk factors and the effects of obesity and dietary intake of lipids on the brain.
Collapse
Affiliation(s)
- Noe Kawade
- Department of Neuroscience and Pathobiology, Research Institute of Environmental MedicineNagoya UniversityJapan
- Department of Neuroscience and Pathobiology, Nagoya University Graduate School of MedicineNagoya UniversityJapan
| | - Koji Yamanaka
- Department of Neuroscience and Pathobiology, Research Institute of Environmental MedicineNagoya UniversityJapan
- Department of Neuroscience and Pathobiology, Nagoya University Graduate School of MedicineNagoya UniversityJapan
- Institute for Glyco‐core Research (iGCORE)Nagoya UniversityJapan
- Center for One Medicine Innovative Translational Research (COMIT)Nagoya UniversityJapan
| |
Collapse
|
4
|
Qiu F, Wang Y, Du Y, Zeng C, Liu Y, Pan H, Ke C. Current evidence for J147 as a potential therapeutic agent in nervous system disease: a narrative review. BMC Neurol 2023; 23:317. [PMID: 37674139 PMCID: PMC10481599 DOI: 10.1186/s12883-023-03358-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 08/03/2023] [Indexed: 09/08/2023] Open
Abstract
Curcumin has anti-inflammatory, antioxidant, and anticancer effects and is used to treat diseases such as dermatological diseases, infection, stress, depression, and anxiety. J147, an analogue of curcumin, is designed and synthesized with better stability and bioavailability. Accumulating evidence demonstrates the potential role of J147 in the prevention and treatment of Alzheimer's disease, diabetic neuropathy, ischemic stroke, depression, anxiety, and fatty liver disease. In this narrative review, we summarized the background and biochemical properties of J147 and discussed the role and mechanism of J147 in different diseases. Overall, the mechanical attributes of J147 connote it as a potential target for the prevention and treatment of neurological diseases.
Collapse
Affiliation(s)
- Fang Qiu
- Department of Burn and Plastic Surgery, Shenzhen Longhua District Central Hospital, Shenzhen, 518110, Guangdong, China
- Center for Human Tissues and Organs Degeneration, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, Guangdong, China
| | - Yanmei Wang
- Department of critical care medicine, Shenzhen Longhua District Central Hospital, Shenzhen, Guangdong, China
| | - Yunbo Du
- Department of critical care medicine, Shenzhen Longhua District Central Hospital, Shenzhen, Guangdong, China
| | - Changchun Zeng
- Department of Medical Laboratory, Shenzhen Longhua District Central Hospital, Affiliated Central Hospital of Shenzhen Longhua District, Guangdong Medical University, Shenzhen, Guangdong, China
| | - Yuqiang Liu
- Department of Anesthesiology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518025, Guangdong, China.
| | - Haobo Pan
- Center for Human Tissues and Organs Degeneration, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, Guangdong, China.
| | - Changneng Ke
- Department of Burn and Plastic Surgery, Shenzhen Longhua District Central Hospital, Shenzhen, 518110, Guangdong, China.
| |
Collapse
|
5
|
Cheng M, Ye C, Tian C, Zhao D, Li H, Sun Z, Miao Y, Zhang Q, Wang J, Dou Y. Engineered macrophage-biomimetic versatile nanoantidotes for inflammation-targeted therapy against Alzheimer's disease by neurotoxin neutralization and immune recognition suppression. Bioact Mater 2023; 26:337-352. [PMID: 36950153 PMCID: PMC10027514 DOI: 10.1016/j.bioactmat.2023.03.004] [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: 10/27/2022] [Revised: 02/21/2023] [Accepted: 03/06/2023] [Indexed: 03/17/2023] Open
Abstract
Immune recognition of excessive neurotoxins by microglia is a trigger for the onset of neuroinflammation in the brain, leading to neurodegeneration in Alzheimer's disease (AD). Blocking active recognition of microglia while removing neurotoxins holds promise for fundamentally alleviating neurotoxin-induced immune responses, but is very challenging. Herein, an engineered macrophage-biomimetic versatile nanoantidote (OT-Lipo@M) is developed for inflammation-targeted therapy against AD by neurotoxin neutralization and immune recognition suppression. Coating macrophage membranes can not only endow OT-Lipo@M with anti-phagocytic and inflammation-tropism capabilities to target inflammatory lesions in AD brain, but also efficiently reduce neurotoxin levels to prevent them from activating microglia. The loaded oxytocin (OT) can be slowly released to downregulate the expression of immune recognition site Toll-like receptor 4 (TLR4) on microglia, inhibiting TLR4-mediated pro-inflammatory signalling cascade. Benefiting from this two-pronged immunosuppressive strategy, OT-Lipo@M exhibits outstanding therapeutic effects on ameliorating cognitive deficits, inhibiting neuronal apoptosis, and enhancing synaptic plasticity in AD mice, accompanied by the delayed hippocampal atrophy and brain microstructural disruption by in vivo 9.4T MR imaging. This work provides new insights into potential AD therapeutics targeting microglia-mediated neuroinflammation at the source.
Collapse
Affiliation(s)
- Meng Cheng
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, PR China
| | - Caihua Ye
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, PR China
| | - Chunxiao Tian
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin, 300070, PR China
| | - Dongju Zhao
- School of Life Sciences, Tianjin University, Tianjin, 300072, PR China
| | - Haonan Li
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, PR China
| | - Zuhao Sun
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, PR China
| | - Yuyang Miao
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, 300052, PR China
| | - Qiang Zhang
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, 300052, PR China
- Corresponding author.
| | - Junping Wang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, PR China
- Corresponding author.
| | - Yan Dou
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, PR China
- Corresponding author.
| |
Collapse
|
6
|
Yin F. Lipid metabolism and Alzheimer's disease: clinical evidence, mechanistic link and therapeutic promise. FEBS J 2023; 290:1420-1453. [PMID: 34997690 PMCID: PMC9259766 DOI: 10.1111/febs.16344] [Citation(s) in RCA: 123] [Impact Index Per Article: 61.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 12/14/2021] [Accepted: 01/05/2022] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) is an age-associated neurodegenerative disorder with multifactorial etiology, intersecting genetic and environmental risk factors, and a lack of disease-modifying therapeutics. While the abnormal accumulation of lipids was described in the very first report of AD neuropathology, it was not until recent decades that lipid dyshomeostasis became a focus of AD research. Clinically, lipidomic and metabolomic studies have consistently shown alterations in the levels of various lipid classes emerging in early stages of AD brains. Mechanistically, decades of discovery research have revealed multifaceted interactions between lipid metabolism and key AD pathogenic mechanisms including amyloidogenesis, bioenergetic deficit, oxidative stress, neuroinflammation, and myelin degeneration. In the present review, converging evidence defining lipid dyshomeostasis in AD is summarized, followed by discussions on mechanisms by which lipid metabolism contributes to pathogenesis and modifies disease risk. Furthermore, lipid-targeting therapeutic strategies, and the modification of their efficacy by disease stage, ApoE status, and metabolic and vascular profiles, are reviewed.
Collapse
Affiliation(s)
- Fei Yin
- Center for Innovation in Brain Science, University of Arizona Health Sciences, Tucson, AZ, USA.,Department of Pharmacology, College of Medicine Tucson, University of Arizona, Tucson, AZ, USA.,Graduate Interdisciplinary Program in Neuroscience, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
7
|
Current Pharmacotherapy and Multi-Target Approaches for Alzheimer's Disease. Pharmaceuticals (Basel) 2022; 15:ph15121560. [PMID: 36559010 PMCID: PMC9781592 DOI: 10.3390/ph15121560] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/26/2022] [Accepted: 11/27/2022] [Indexed: 12/23/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by decreased synaptic transmission and cerebral atrophy with appearance of amyloid plaques and neurofibrillary tangles. Cognitive, functional, and behavioral alterations are commonly associated with the disease. Different pathophysiological pathways of AD have been proposed, some of which interact and influence one another. Current treatment for AD mainly involves the use of therapeutic agents to alleviate the symptoms in AD patients. The conventional single-target treatment approaches do not often cause the desired effect in the disease due to its multifactorial origin. Thus, multi-target strategies have since been undertaken, which aim to simultaneously target multiple targets involved in the development of AD. In this review, we provide an overview of the pathogenesis of AD and the current drug therapies for the disease. Additionally, rationales of the multi-target approaches and examples of multi-target drugs with pharmacological actions against AD are also discussed.
Collapse
|
8
|
Dewanjee S, Chakraborty P, Bhattacharya H, Chacko L, Singh B, Chaudhary A, Javvaji K, Pradhan SR, Vallamkondu J, Dey A, Kalra RS, Jha NK, Jha SK, Reddy PH, Kandimalla R. Altered glucose metabolism in Alzheimer's disease: Role of mitochondrial dysfunction and oxidative stress. Free Radic Biol Med 2022; 193:134-157. [PMID: 36206930 DOI: 10.1016/j.freeradbiomed.2022.09.032] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/16/2022] [Accepted: 09/29/2022] [Indexed: 12/06/2022]
Abstract
Increasing evidence suggests that abnormal cerebral glucose metabolism is largely present in Alzheimer's disease (AD). The brain utilizes glucose as its main energy source and a decline in its metabolism directly reflects on brain function. Weighing on recent evidence, here we systematically assessed the aberrant glucose metabolism associated with amyloid beta and phosphorylated tau accumulation in AD brain. Interlink between insulin signaling and AD highlighted the involvement of the IRS/PI3K/Akt/AMPK signaling, and GLUTs in the disease progression. While shedding light on the mitochondrial dysfunction in the defective glucose metabolism, we further assessed functional consequences of AGEs (advanced glycation end products) accumulation, polyol activation, and other contributing factors including terminal respiration, ROS (reactive oxygen species), mitochondrial permeability, PINK1/parkin defects, lysosome-mitochondrial crosstalk, and autophagy/mitophagy. Combined with the classic plaque and tangle pathologies, glucose hypometabolism with acquired insulin resistance and mitochondrial dysfunction potentiate these factors to exacerbate AD pathology. To this end, we further reviewed AD and DM (diabetes mellitus) crosstalk in disease progression. Taken together, the present work discusses the emerging role of altered glucose metabolism, contributing impact of insulin signaling, and mitochondrial dysfunction in the defective cerebral glucose utilization in AD.
Collapse
Affiliation(s)
- Saikat Dewanjee
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700 032, West Bengal, India
| | - Pratik Chakraborty
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700 032, West Bengal, India
| | - Hiranmoy Bhattacharya
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700 032, West Bengal, India
| | - Leena Chacko
- BioAnalytical Lab, Meso Scale Discovery, 1601 Research Blvd, Rockville, MD, USA
| | - Birbal Singh
- ICAR-Indian Veterinary Research Institute (IVRI), Regional Station, Palampur, 176061, Himachal Pradesh, India
| | - Anupama Chaudhary
- Orinin-BioSystems, LE-52, Lotus Road 4, CHD City, Karnal, 132001, Haryana, India
| | - Kalpana Javvaji
- CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, India
| | | | | | - Abhijit Dey
- Department of Life Sciences, Presidency University, Kolkata, 700073, India
| | - Rajkumar Singh Kalra
- Immune Signal Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, 9040495, Japan
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering & Technology (SET), Sharda University, UP, 201310, India; Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali, 140413, India; Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun, 248007, India
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering & Technology (SET), Sharda University, UP, 201310, India; Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali, 140413, India; Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun, 248007, India
| | - P Hemachandra Reddy
- Internal Medicine Department, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Neuroscience & Pharmacology, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Neurology Departments School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Public Health Department of Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Department of Speech, Language and Hearing Sciences, School Health Professions, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Ramesh Kandimalla
- CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, India; Department of Biochemistry, Kakatiya Medical College, Warangal, India.
| |
Collapse
|
9
|
Wang YW, Wang L, Yuan SJ, Zhang Y, Zhang X, Zhou LT. Postoperative Cognitive Dysfunction and Alzheimer’s Disease: A Transcriptome-Based Comparison of Animal Models. Front Aging Neurosci 2022; 14:900350. [PMID: 35837480 PMCID: PMC9273890 DOI: 10.3389/fnagi.2022.900350] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 06/08/2022] [Indexed: 12/02/2022] Open
Abstract
Background Postoperative cognitive dysfunction (POCD) is a common complication characterized by a significant cognitive decline. Increasing evidence suggests an association between the pathogenesis of POCD and Alzheimer’s disease (AD). However, a comprehensive understanding of their relationships is still lacking. Methods First, related databases were obtained from GEO, ArrayExpress, CNGB, and DDBJ repositories. De novo analysis was performed on the raw data using a uniform bioinformatics workflow. Then, macro- and micro-level comparisons were conducted between the transcriptomic changes associated with AD and POCD. Lastly, POCD was induced in male C57BL/6j mice and the hippocampal expression levels of mRNAs of interest were verified by PCR and compared to those in AD congenic models. Results There was a very weak correlation in the fold-changes in protein-coding transcripts between AD and POCD. Overall pathway-level comparison suggested that AD and POCD are two disease entities. Consistently, in the classical AD pathway, the mitochondrial complex and tubulin mRNAs were downregulated in both the POCD hippocampus and cortex. POCD and AD hippocampi might share the same pathways, such as tryptophan metabolism, but undergo different pathological changes in phagosome and transferrin endocytosis pathways. The core cluster in the hippocampal network was mainly enriched in mitosis-related pathways. The hippocampal expression levels of genes of interest detected by PCR showed good consistency with those generated by high throughput platforms. Conclusion POCD and AD are associated with different transcriptomic changes despite their similar clinical manifestations. This study provides a valuable resource for identifying biomarkers and therapeutic targets for POCD.
Collapse
Affiliation(s)
- Yi-Wei Wang
- Department of Anesthesiology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Liang Wang
- Department of Internal Medicine, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Sheng-Jie Yuan
- Department of Anesthesiology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Yuan Zhang
- Department of Internal Medicine, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Xin Zhang
- Department of Anesthesiology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
- Department of Anesthesiology, Center for Translational Pain Medicine, Duke University School of Medicine, Durham, NC, United States
- Xin Zhang,
| | - Le-Ting Zhou
- Department of Internal Medicine, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
- *Correspondence: Le-Ting Zhou,
| |
Collapse
|
10
|
Kepchia D, Huang L, Currais A, Liang Z, Fischer W, Maher P. The Alzheimer's disease drug candidate J147 decreases blood plasma fatty acid levels via modulation of AMPK/ACC1 signaling in the liver. Biomed Pharmacother 2022; 147:112648. [PMID: 35051863 PMCID: PMC8854339 DOI: 10.1016/j.biopha.2022.112648] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/07/2022] [Accepted: 01/12/2022] [Indexed: 12/30/2022] Open
Abstract
J147 is a novel drug candidate developed to treat neurological dysfunction. Numerous studies have demonstrated the beneficial effects of J147 in cellular and animal models of disease which has led to the transitioning of the compound into human clinical trials. However, no biomarkers for its target engagement have been identified. Here, we determined if specific metabolites in the plasma could be indicative of J147's activity in vivo. Plasma lipidomics data from three independent rodent studies were assessed along with liver lipidomics data from one of the studies. J147 consistently reduced plasma free fatty acid (FFA) levels across the independent studies. Decreased FFA levels were also found in the livers of J147-treated mice that correlated well with those in the plasma. These changes in the liver were associated with activation of the AMP-activated protein kinase/acetyl-CoA carboxylase 1 signaling pathway. A reduction in FFA levels by J147 was confirmed in HepG2 cells, where activation of the AMPK/ACC1 pathway was seen along with increases in acetyl-CoA and ATP levels which correlated with enhanced cellular bioenergetics. Our data show that J147 targets liver cells to activate the AMPK/ACC1 signaling pathway and preserve energy at the expense of inhibiting FFA synthesis.
Collapse
Affiliation(s)
- Devin Kepchia
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd., La Jolla, CA 92037, USA.
| | - Ling Huang
- The Razavi Newman Integrative Genomics and Bioinformatics
Core, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd. La Jolla,
CA 92037, USA
| | - Antonio Currais
- Cellular Neurobiology Laboratory, The Salk Institute for
Biological Studies, 10010 N. Torrey Pines Rd. La Jolla, CA 92037, USA
| | - Zhibin Liang
- Cellular Neurobiology Laboratory, The Salk Institute for
Biological Studies, 10010 N. Torrey Pines Rd. La Jolla, CA 92037, USA
| | - Wolfgang Fischer
- Cellular Neurobiology Laboratory, The Salk Institute for
Biological Studies, 10010 N. Torrey Pines Rd. La Jolla, CA 92037, USA
| | - Pamela Maher
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd., La Jolla, CA 92037, USA.
| |
Collapse
|
11
|
Alhowail A, Chigurupati S. Research advances on how metformin improves memory impairment in "chemobrain". Neural Regen Res 2022; 17:15-19. [PMID: 34100420 PMCID: PMC8451574 DOI: 10.4103/1673-5374.314284] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Cognitive impairment caused by chemotherapy, referred to as “chemobrain,” is observed in approximately 70% of cancer survivors. However, it is not completely understood how chemotherapy induces cognitive dysfunction, and clinical treatment strategies for this problem are lacking. Metformin, used as a first-line treatment for type 2 diabetes mellitus, is reported to reduce the effects of chemobrain. Recently, several studies have examined the effect of metformin in rescuing chemobrain. This review discusses recent clinical/preclinical studies that addressed some mechanisms of chemobrain and evaluates the effect of metformin in rescuing chemobrain and its potential mechanisms of action.
Collapse
Affiliation(s)
- Ahmad Alhowail
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraidah, Kingdom of Saudi Arabia
| | - Sridevi Chigurupati
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Buraydah, Kingdom of Saudi Arabia
| |
Collapse
|
12
|
Sharma NS, Karan A, Lee D, Yan Z, Xie J. Advances in Modeling Alzheimer's Disease In Vitro. ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202100097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Affiliation(s)
- Navatha Shree Sharma
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program University of Nebraska Medical Center Omaha NE 68198 USA
| | - Anik Karan
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program University of Nebraska Medical Center Omaha NE 68198 USA
| | - Donghee Lee
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program University of Nebraska Medical Center Omaha NE 68198 USA
| | - Zheng Yan
- Department of Mechanical & Aerospace Engineering and Department of Biomedical Biological and Chemical Engineering University of Missouri Columbia MO 65211 USA
| | - Jingwei Xie
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program University of Nebraska Medical Center Omaha NE 68198 USA
- Department of Mechanical and Materials Engineering College of Engineering University of Nebraska Lincoln Lincoln NE 68588 USA
| |
Collapse
|
13
|
Onyango IG, Bennett JP, Stokin GB. Regulation of neuronal bioenergetics as a therapeutic strategy in neurodegenerative diseases. Neural Regen Res 2021; 16:1467-1482. [PMID: 33433460 PMCID: PMC8323696 DOI: 10.4103/1673-5374.303007] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis are a heterogeneous group of debilitating disorders with multifactorial etiologies and pathogeneses that manifest distinct molecular mechanisms and clinical manifestations with abnormal protein dynamics and impaired bioenergetics. Mitochondrial dysfunction is emerging as an important feature in the etiopathogenesis of these age-related neurodegenerative diseases. The prevalence and incidence of these diseases is on the rise with the increasing global population and average lifespan. Although many therapeutic approaches have been tested, there are currently no effective treatment routes for the prevention or cure of these diseases. We present the current status of our knowledge and understanding of the involvement of mitochondrial dysfunction in these diseases and highlight recent advances in novel therapeutic strategies targeting neuronal bioenergetics as potential approach for treating these diseases.
Collapse
Affiliation(s)
- Isaac G Onyango
- Center for Translational Medicine, International Clinical Research Centre (ICRC), St. Anne's University Hospital, Brno, Czech Republic
| | - James P Bennett
- Neurodegeneration Therapeutics, 3050A Berkmar Drive, Charlottesville, VA, USA
| | - Gorazd B Stokin
- Center for Translational Medicine, International Clinical Research Centre (ICRC), St. Anne's University Hospital, Brno, Czech Republic
| |
Collapse
|
14
|
Xiao H, Li H, Song H, Kong L, Yan X, Li Y, Deng Y, Tai H, Wu Y, Ni Y, Li W, Chen J, Yang J. Shenzao jiannao oral liquid, an herbal formula, ameliorates cognitive impairments by rescuing neuronal death and triggering endogenous neurogenesis in AD-like mice induced by a combination of Aβ42 and scopolamine. JOURNAL OF ETHNOPHARMACOLOGY 2020; 259:112957. [PMID: 32416248 DOI: 10.1016/j.jep.2020.112957] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 04/28/2020] [Accepted: 05/07/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE According to the theory of traditional Chinese medicine (TCM), Alzheimer's disease (AD) is identified as "forgetfulness" or "dementia", and is mainly caused by "kidney essence deficiency" which ultimately induces "encephala reduction". Therefore, herbal formulas possessing the efficacy of nourishing kidney essence or replenishing brain marrow are commonly served as effective strategies for AD treatment. Shenzao jiannao oral liquid (SZJN), a traditional Chinese preparation approved by the China Food and Drug Administration (CFDA), is used for the treatment of insomnia and mind fatigue at present for its efficacy of nourishing kidneys. In present study, we found that SZJN could improve cognitive function of AD-like mice. AIMS OF STUDY This study aims to investigate the effects of SJZN on ameliorating cognitive deficits of AD-like mouse model, and to illuminate the underlying mechanisms from the perspective of neuroprotection and neurogenesis. MATERIALS AND METHODS Kunming mice (28 ± 2 g) were randomly allocated into seven groups: control, sham, model, donepezil and SZJN groups (low, middle and high). The AD mouse model was established by Aβ42 combined with scopolamine. SZJN were intragastrically administrated at doses of 0.3, 1.5 and 7.5 g/kg for 28 days. Morris water maze (MWM) test was applied to determine the cognitive function. Hematoxylin eosin (HE) and Nissl staining were carried out to evaluate pathological damages in the cortex and hippocampal tissues. To explore the protective effects of SZJN on multiple pathogenic factors of AD, protein levels of Aβ42, glial fibrillary acidic protein (GFAP), Bax, Bcl-2, Caspase-3, synaptophysin (SYP), brain-derived neurotrophic factor (BDNF), and neurogenesis related proteins were assessed using Immunofluorescence (IF) and western blot analysis. In vitro, the AD cell model was established by transduction of APP695swe genes into Neural stem cells (NSCs) isolated from the hippocampal tissues of neonatal C57BL/6 mice. Cell viability assay and neurosphere formation assay were carried out to verify the efficacy of SZJN on proliferation of NSCs. RESULTS Our results demonstrated that SZJN (1.5 g/kg and 7.5 g/kg) treatment significantly ameliorated cognitive deficits of AD-like mice. SZJN (7.5 g/kg) treatment significantly retarded the pathological damages including neuronal degeneration, neuronal apoptosis, Aβ peptides aggregation and reaction of astrocytes in AD-like mice. In addition, SZJN (7.5 g/kg) increased the expression of BDNF and SYP, and restored the abnormal level of MDA and SOD in the brain of AD-like mice. Furthermore, SZJN treatment for 28 days remarkably increased the proliferation of NSCs evidenced by more Nestin+ and BrdU+ cells in the hippocampal DG regions, and increased the amount of mature neurons marked by NeuN both in the cortex and hippocampal DG regions. In vitro, SZJN treatement (16, 32, 64 mg/ml) promoted the proliferation of NSCs evidenced by the increased amount and enlarged size of the neurospheres (p < 0.05). CONCLUSIONS Our findings indicated that SZJN could ameliorate cognitive deficits by protecting neurons from death and triggering endogenous neurogenesis. Therefore, SZJN may be considered as a promising agent to restore neuronal loss and deter the deterioration in AD patients.
Collapse
Affiliation(s)
- Honghe Xiao
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, 77 Life One Road, DD Port, Dalian, 116600, PR China.
| | - Hongyan Li
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, 77 Life One Road, DD Port, Dalian, 116600, PR China
| | - Huipeng Song
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, 77 Life One Road, DD Port, Dalian, 116600, PR China
| | - Liang Kong
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, 77 Life One Road, DD Port, Dalian, 116600, PR China
| | - Xin Yan
- Diaoyutai Pharmaceutical Group Jilin Tianqiang Pharmaceutical co. LTD, 309 Renmin Street, Tonghua, 135300, PR China
| | - Yan Li
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, 77 Life One Road, DD Port, Dalian, 116600, PR China
| | - Yan Deng
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, 77 Life One Road, DD Port, Dalian, 116600, PR China
| | - He Tai
- Key Laboratory of Ministry of Education for Traditional Chinese Medicine Visera-State Theory and Application, Liaoning University of Traditional Chinese Medicine, Huanggu District Chongshan Road No. 79, Shenyang, Liaoning, 110847, PR China
| | - Yutong Wu
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, 77 Life One Road, DD Port, Dalian, 116600, PR China
| | - Yingnan Ni
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, 77 Life One Road, DD Port, Dalian, 116600, PR China
| | - Wanyi Li
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, 77 Life One Road, DD Port, Dalian, 116600, PR China
| | - Jicong Chen
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, 77 Life One Road, DD Port, Dalian, 116600, PR China
| | - Jingxian Yang
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, 77 Life One Road, DD Port, Dalian, 116600, PR China.
| |
Collapse
|
15
|
Currais A, Huang L, Petrascheck M, Maher P, Schubert D. A chemical biology approach to identifying molecular pathways associated with aging. GeroScience 2020; 43:353-365. [PMID: 32705410 DOI: 10.1007/s11357-020-00238-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 07/19/2020] [Indexed: 12/26/2022] Open
Abstract
The understanding of how aging contributes to dementia remains obscure. To address this problem, a chemical biology approach was used employing CAD031, an Alzheimer's disease (AD) drug candidate identified using a discovery platform based upon phenotypic screens that mimic toxicities associated with the aging brain. Since CAD031 has therapeutic efficacy when fed to old symptomatic transgenic AD mice, the chemical biology hypothesis is that it can be used to determine the molecular pathways associated with age-related disease by identifying those that are modified by the compound. Here we show that when CAD031 was fed to rapidly aging SAMP8 mice starting in the last quadrant of their lifespan, it reduced many of the changes in gene, protein, and small molecule expression associated with mitochondrial aging, maintaining mitochondria at the younger molecular phenotype. Network analysis integrating the metabolomics and transcription data followed by mechanistic validation showed that CAD031 targets acetyl-CoA and fatty acid metabolism via the AMPK/ACC1 pathway. Importantly, CAD031 extended the median lifespan of SAMP8 mice by about 30%. These data show that specific alterations in mitochondrial composition and metabolism highly correlate with aging, supporting the use AD drug candidates that limit physiological aging in the brain.
Collapse
Affiliation(s)
- Antonio Currais
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd., La Jolla, CA, 92037, USA.
| | - Ling Huang
- The Razavi Newman Integrative Genomics and Bioinformatics Core, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd., La Jolla, CA, 92037, USA
| | - Michael Petrascheck
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Pamela Maher
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd., La Jolla, CA, 92037, USA
| | - David Schubert
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd., La Jolla, CA, 92037, USA.
| |
Collapse
|
16
|
Ates G, Goldberg J, Currais A, Maher P. CMS121, a fatty acid synthase inhibitor, protects against excess lipid peroxidation and inflammation and alleviates cognitive loss in a transgenic mouse model of Alzheimer's disease. Redox Biol 2020; 36:101648. [PMID: 32863221 PMCID: PMC7394765 DOI: 10.1016/j.redox.2020.101648] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/15/2020] [Accepted: 07/17/2020] [Indexed: 01/14/2023] Open
Abstract
The oxidative degradation of lipids has been shown to be implicated in the progression of several neurodegenerative diseases and modulating lipid peroxidation may be efficacious for treating Alzheimer’s disease (AD). This hypothesis is strengthened by recent findings suggesting that oxytosis/ferroptosis, a cell death process characterized by increased lipid peroxidation, plays an important role in AD-related toxicities. CMS121 is a small molecule developed against these aspects of neurodegeneration. Here we show that CMS121 alleviates cognitive loss, modulates lipid metabolism and reduces inflammation and lipid peroxidation in the brains of transgenic AD mice. We identify fatty acid synthase (FASN) as a molecular target of CMS121 and demonstrate that modulating lipid metabolism through the inhibition of FASN protects against several AD-related toxicities. These results support the involvement of lipid peroxidation and perturbed lipid metabolism in AD pathophysiology and propose FASN as a target in AD-associated toxicities. CMS121, a fisetin-derivative, alleviates memory decline in a double transgenic AD mouse model. CMS121 is able to reduce lipid peroxidation and neuroinflammation, both in vitro and in vivo. We identify fatty acid synthase (FASN), which shows increased protein levels in human AD patients, as a target of CMS121. Our results confirm the involvement of lipid peroxidation and perturbed lipid metabolism in AD pathophysiology. Decreasing lipid levels through FASN inhibition can be effective against excess lipid peroxidation.
Collapse
Affiliation(s)
- Gamze Ates
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Joshua Goldberg
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Antonio Currais
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Pamela Maher
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA.
| |
Collapse
|
17
|
Kepchia D, Huang L, Dargusch R, Rissman RA, Shokhirev MN, Fischer W, Schubert D. Diverse proteins aggregate in mild cognitive impairment and Alzheimer's disease brain. Alzheimers Res Ther 2020; 12:75. [PMID: 32560738 PMCID: PMC7305608 DOI: 10.1186/s13195-020-00641-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 06/04/2020] [Indexed: 01/04/2023]
Abstract
BACKGROUND All cells accumulate insoluble protein aggregates throughout their lifespan. While many studies have characterized the canonical disease-associated protein aggregates, such as those associated with amyloid plaques, additional, undefined proteins aggregate in the brain and may be directly associated with disease and lifespan. METHODS A proteomics approach was used to identify a large subset of insoluble proteins in the mild cognitively impaired (MCI) and Alzheimer's disease (AD) human brain. Cortical samples from control, MCI, and AD patients were separated into detergent-soluble and detergent-insoluble fractions, and high-resolution LC/MS/MS technology was used to determine which proteins became more insoluble in the disease state. Bioinformatics analyses were used to determine if the alteration of protein aggregation between AD and control patients was associated with any specific biological process. Western blots were used to validate the proteomics data and to assess the levels of secondary protein modifications in MCI and AD. RESULTS There was a stage-dependent increase in detergent-insoluble proteins, with more extreme changes occurring in the AD cohort. Glycolysis was the most significantly overrepresented gene ontology biological process associated with the alteration of protein aggregation between AD and control patients. It was further shown that many low molecular weight proteins that were enriched in the AD brain were also highly aggregated, migrating on SDS-PAGE far above their predicted molecular masses. Glucose-6-phosphate isomerase, ubiquitin carboxyl-terminal hydrolase isoenzyme L1 (UCHL1/PARK5), and the DNA damage repair enzyme KU70 were among the top insoluble proteins identified by proteomics and validated by Western blot to be increased in the insoluble fractions of both MCI and AD brain samples. CONCLUSIONS Diverse proteins became more detergent-insoluble in the brains of both MCI and AD patients compared to age-matched controls, suggesting that multiple proteins aggregate in these diseases, likely posing a direct toxic insult to neurons. Furthermore, detergent-insoluble proteins included those with important biological activities for critical cellular processes such as energetics, proteolysis, and DNA damage repair. Thus, reduced protein solubility likely promotes aggregation and limits functionality, reducing the efficiency of multiple aspects of cell physiology. Pharmaceutical interventions that increase autophagy may provide a useful therapeutic treatment to combat protein aggregation.
Collapse
Affiliation(s)
- Devin Kepchia
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, 92037, USA.
| | - Ling Huang
- The Razavi Newman Integrative Genomics and Bioinformatics Core, The Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Richard Dargusch
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Robert A Rissman
- Department of Neurosciences and Shiley-Marcos Alzheimer's Disease Research Center Neuropathology Core, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Maxim N Shokhirev
- The Razavi Newman Integrative Genomics and Bioinformatics Core, The Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Wolfgang Fischer
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - David Schubert
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| |
Collapse
|
18
|
Untangling human neurogenesis to understand and counteract brain disorders. Curr Opin Pharmacol 2019; 50:67-73. [PMID: 31901615 DOI: 10.1016/j.coph.2019.12.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 12/06/2019] [Accepted: 12/10/2019] [Indexed: 11/22/2022]
Abstract
Neurogenesis in the human postnatal brain occurs in two regions, the subventricular zone of the later ventricle and the dentate gyrus of the hippocampus. While it is well accepted that SVZ and hippocampal neurogenesis are active during juvenile stages in human, their contribution during adulthood and ageing as well as pathological states is recently animating the neural stem cell research field. In this review we will discuss recent evidence about the organization of SVZ and hippocampal neurogenic niches, and will report on how human adult neurogenesis may contribute to disease and appears to respond to neurodegeneration. In light of these novel findings, we will discuss how we can target human adult neurogenesis in order to influence brain disease trajectories.
Collapse
|
19
|
Currais A, Huang L, Goldberg J, Petrascheck M, Ates G, Pinto-Duarte A, Shokhirev MN, Schubert D, Maher P. Elevating acetyl-CoA levels reduces aspects of brain aging. eLife 2019; 8:47866. [PMID: 31742554 PMCID: PMC6882557 DOI: 10.7554/elife.47866] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 11/18/2019] [Indexed: 12/26/2022] Open
Abstract
Because old age is the greatest risk factor for dementia, a successful therapy will require an understanding of the physiological changes that occur in the brain with aging. Here, two structurally distinct Alzheimer's disease (AD) drug candidates, CMS121 and J147, were used to identify a unique molecular pathway that is shared between the aging brain and AD. CMS121 and J147 reduced cognitive decline as well as metabolic and transcriptional markers of aging in the brain when administered to rapidly aging SAMP8 mice. Both compounds preserved mitochondrial homeostasis by regulating acetyl-coenzyme A (acetyl-CoA) metabolism. CMS121 and J147 increased the levels of acetyl-CoA in cell culture and mice via the inhibition of acetyl-CoA carboxylase 1 (ACC1), resulting in neuroprotection and increased acetylation of histone H3K9 in SAMP8 mice, a site linked to memory enhancement. These data show that targeting specific metabolic aspects of the aging brain could result in treatments for dementia.
Collapse
Affiliation(s)
- Antonio Currais
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, United States
| | - Ling Huang
- The Razavi Newman Integrative Genomics and Bioinformatics Core, The Salk Institute for Biological Studies, La Jolla, United States
| | - Joshua Goldberg
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, United States
| | - Michael Petrascheck
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, United States
| | - Gamze Ates
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, United States
| | - António Pinto-Duarte
- Computational Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, United States
| | - Maxim N Shokhirev
- The Razavi Newman Integrative Genomics and Bioinformatics Core, The Salk Institute for Biological Studies, La Jolla, United States
| | - David Schubert
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, United States
| | - Pamela Maher
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, United States
| |
Collapse
|
20
|
Van der Auwera S, Ameling S, Wittfeld K, d'Harcourt Rowold E, Nauck M, Völzke H, Suhre K, Najafi-Shoushtari H, Methew J, Ramachandran V, Bülow R, Völker U, Grabe HJ. Association of childhood traumatization and neuropsychiatric outcomes with altered plasma micro RNA-levels. Neuropsychopharmacology 2019; 44:2030-2037. [PMID: 31284290 PMCID: PMC6898678 DOI: 10.1038/s41386-019-0460-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 06/27/2019] [Accepted: 06/28/2019] [Indexed: 12/21/2022]
Abstract
Childhood traumatization (CT) is associated with the development of several neuropsychiatric disorders in later life. Experimental data in animals and observational data in humans revealed evidence for biological alterations in response to CT that may contribute to its long-term consequences. This includes epigenetic changes in miRNA levels that contribute to complex alterations of gene expression. We investigated the association between CT and 121 miRNAs in a target sample of N = 150 subjects from the general population and patients from the Department of Psychiatry. We hypothesized that CT exhibits a long-term effect on miRNA plasma levels. We supported our findings using bioinformatics tools and databases. Among the 121 miRNAs 22 were nominally significantly associated with CT and four of them (let-7g-5p, miR-103a-3p, miR-107, and miR-142-3p) also after correction for multiple testing; most of them were previously associated with Alzheimer's disease (AD) or depression. Pathway analyses of target genes identified significant pathways involved in neurodevelopment, inflammation and intracellular transduction signaling. In an independent general population sample (N = 587) three of the four miRNAs were replicated. Extended analyses in the general population sample only (N = 687) showed associations of the four miRNAs with gender, memory, and brain volumes. We gained increasing evidence for a link between CT, depression and AD through miRNA alterations. We hypothesize that depression and AD not only share environmental factors like CT but also biological factors like altered miRNA levels. This miRNA pattern could serve as mediating factor on the biological path from CT to adult neuropsychiatric disorders.
Collapse
Affiliation(s)
- Sandra Van der Auwera
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany.
- German Centre for Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, Greifswald, Germany.
| | - Sabine Ameling
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany
| | - Katharina Wittfeld
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
- German Centre for Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, Greifswald, Germany
| | | | - Matthias Nauck
- German Centre for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Henry Völzke
- German Centre for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Karsten Suhre
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Ar-Rayyan, Qatar
| | - Hani Najafi-Shoushtari
- Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, 10021, NY, USA
- Division of Research, Weill Cornell Medicine-Qatar, Qatar Foundation, Education City, P.O. Box 24144, Doha, Qatar
| | - Jaicy Methew
- Division of Research, Weill Cornell Medicine-Qatar, Qatar Foundation, Education City, P.O. Box 24144, Doha, Qatar
| | - Vimal Ramachandran
- Division of Research, Weill Cornell Medicine-Qatar, Qatar Foundation, Education City, P.O. Box 24144, Doha, Qatar
| | - Robin Bülow
- Institute of Diagnostic Radiology and Neuroradiology, University Medicine Greifswald, Greifswald, Germany
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald, Germany
| | - Hans J Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany
- German Centre for Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, Greifswald, Germany
| |
Collapse
|
21
|
Wang N, Qiu P, Cui W, Yan X, Zhang B, He S. Recent Advances in Multi-target Anti-Alzheimer Disease Compounds (2013 Up to the Present). Curr Med Chem 2019; 26:5684-5710. [DOI: 10.2174/0929867326666181203124102] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 08/03/2018] [Accepted: 11/03/2018] [Indexed: 12/21/2022]
Abstract
:
Since the last century, when scientists proposed the lock-and-key model, the discovery of
drugs has focused on the development of drugs acting on single target. However, single-target drug
therapies are not effective to complex diseases with multi-factorial pathogenesis. Moreover, the
combination of single-target drugs readily causes drug resistance and side effects. In recent years,
multi-target drugs have increasingly been represented among FDA-approved drugs. Alzheimer’s
Disease (AD) is a complex and multi-factorial disease for which the precise molecular mechanisms
are still not fully understood. In recent years, rational multi-target drug design methods, which combine
the pharmacophores of multiple drugs, have been increasingly applied in the development of
anti-AD drugs. In this review, we give a brief description of the pathogenesis of AD and provide
detailed discussions about the recent development of chemical structures of anti-AD agents (2013 up
to present) that have multiple targets, such as amyloid-β peptide, Tau protein, cholinesterases,
monoamine oxidase, β-site amyloid-precursor protein-cleaving enzyme 1, free radicals, metal ions
(Fe2+, Cu2+, Zn2+) and so on. In this paper, we also added some novel targets or possible pathogenesis
which have been reported in recent years for AD therapy. We hope that these findings may provide
new perspectives for the pharmacological treatment of AD.
Collapse
Affiliation(s)
- Ning Wang
- Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, Ningbo University, Ningbo 315211, China
| | - Panpan Qiu
- Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, Ningbo University, Ningbo 315211, China
| | - Wei Cui
- Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, Ningbo University, Ningbo 315211, China
| | - Xiaojun Yan
- Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, Ningbo University, Ningbo 315211, China
| | - Bin Zhang
- Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, Ningbo University, Ningbo 315211, China
| | - Shan He
- Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, Ningbo University, Ningbo 315211, China
| |
Collapse
|
22
|
Hadad N, Masser DR, Blanco-Berdugo L, Stanford DR, Freeman WM. Early-life DNA methylation profiles are indicative of age-related transcriptome changes. Epigenetics Chromatin 2019; 12:58. [PMID: 31594536 PMCID: PMC6781367 DOI: 10.1186/s13072-019-0306-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 09/14/2019] [Indexed: 12/15/2022] Open
Abstract
Background Alterations to cellular and molecular programs with brain aging result in cognitive impairment and susceptibility to neurodegenerative disease. Changes in DNA methylation patterns, an epigenetic modification required for various CNS functions are observed with brain aging and can be prevented by anti-aging interventions, but the relationship of altered methylation to gene expression is poorly understood. Results Paired analysis of the hippocampal methylome and transcriptome with aging of male and female mice demonstrates that age-related differences in methylation and gene expression are anti-correlated within gene bodies and enhancers. Altered promoter methylation with aging was found to be generally un-related to altered gene expression. A more striking relationship was found between methylation levels at young age and differential gene expression with aging. Highly methylated gene bodies and promoters in early life were associated with age-related increases in gene expression even in the absence of significant methylation changes with aging. As well, low levels of methylation in early life were correlated to decreased expression with aging. This relationship was also observed in genes altered in two mouse Alzheimer’s models. Conclusion DNA methylation patterns established in youth, in combination with other epigenetic marks, were able to accurately predict changes in transcript trajectories with aging. These findings are consistent with the developmental origins of disease hypothesis and indicate that epigenetic variability in early life may explain differences in aging trajectories and age-related disease.
Collapse
Affiliation(s)
- Niran Hadad
- Oklahoma Center for Neuroscience, Oklahoma City, OK, USA.,Reynolds Oklahoma Center on Aging, SLY-BRC 1370, 975 NE 10th St, Oklahoma City, OK, 73104, USA.,The Jackson Laboratory, Bar Harbor, ME, 04609, USA
| | - Dustin R Masser
- Reynolds Oklahoma Center on Aging, SLY-BRC 1370, 975 NE 10th St, Oklahoma City, OK, 73104, USA.,Department of Physiology, Oklahoma City, OK, USA
| | | | - David R Stanford
- Reynolds Oklahoma Center on Aging, SLY-BRC 1370, 975 NE 10th St, Oklahoma City, OK, 73104, USA.,Department of Physiology, Oklahoma City, OK, USA.,Oklahoma Nathan Shock Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Willard M Freeman
- Oklahoma Center for Neuroscience, Oklahoma City, OK, USA. .,Reynolds Oklahoma Center on Aging, SLY-BRC 1370, 975 NE 10th St, Oklahoma City, OK, 73104, USA. .,Department of Physiology, Oklahoma City, OK, USA. .,Oklahoma Nathan Shock Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA. .,Oklahoma City VA Medical Center, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
23
|
Li C, Zhang J, Wu R, Liu Y, Hu X, Yan Y, Ling X. A novel strategy for rapidly and accurately screening biomarkers based on ultraperformance liquid chromatography-mass spectrometry metabolomics data. Anal Chim Acta 2019; 1063:47-56. [DOI: 10.1016/j.aca.2019.03.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 03/06/2019] [Accepted: 03/07/2019] [Indexed: 12/26/2022]
|
24
|
Dubey SK, Ram MS, Krishna KV, Saha RN, Singhvi G, Agrawal M, Ajazuddin, Saraf S, Saraf S, Alexander A. Recent Expansions on Cellular Models to Uncover the Scientific Barriers Towards Drug Development for Alzheimer's Disease. Cell Mol Neurobiol 2019; 39:181-209. [PMID: 30671696 DOI: 10.1007/s10571-019-00653-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 01/12/2019] [Indexed: 12/17/2022]
Abstract
Globally, the central nervous system (CNS) disorders appear as the most critical pathological threat with no proper cure. Alzheimer's disease (AD) is one such condition frequently observed with the aged population and sometimes in youth too. Most of the research utilizes different animal models for in vivo study of AD pathophysiology and to investigate the potency of the newly developed therapy. These in vivo models undoubtably provide a powerful investigation tool to study human brain. Although, it sometime fails to mimic the exact environment and responses as the human brain owing to the distinctive genetic and anatomical features of human and rodent brain. In such condition, the in vitro cell model derived from patient specific cell or human cell lines can recapitulate the human brain environment. In addition, the frequent use of animals in research increases the cost of study and creates various ethical issues. Instead, the use of in vitro cellular models along with animal models can enhance the translational values of in vivo models and represent a better and effective mean to investigate the potency of therapeutics. This strategy also limits the excessive use of laboratory animal during the drug development process. Generally, the in vitro cell lines are cultured from AD rat brain endothelial cells, the rodent models, human astrocytes, human brain capillary endothelial cells, patient derived iPSCs (induced pluripotent stem cells) and also from the non-neuronal cells. During the literature review process, we observed that there are very few reviews available which describe the significance and characteristics of in vitro cell lines, for AD investigation. Thus, in the present review article, we have compiled the various in vitro cell lines used in AD investigation including HBMEC, BCECs, SHSY-5Y, hCMEC/D3, PC-2 cell line, bEND3 cells, HEK293, hNPCs, RBE4 cells, SK-N-MC, BMVECs, CALU-3, 7W CHO, iPSCs and cerebral organoids cell lines and different types of culture media such as SCM, EMEM, DMEM/F12, RPMI, EBM and 3D-cell culture.
Collapse
Affiliation(s)
- Sunil Kumar Dubey
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS-PILANI), Pilani Campus, Rajasthan, India.
| | - Munnangi Siva Ram
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS-PILANI), Pilani Campus, Rajasthan, India
| | - Kowthavarapu Venkata Krishna
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS-PILANI), Pilani Campus, Rajasthan, India
| | - Ranendra Narayan Saha
- Department of Biotechnology, Birla Institute of Technology and Science, Pilani (BITS-PILANI), Dubai Campus, Dubai, United Arab Emirates
| | - Gautam Singhvi
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS-PILANI), Pilani Campus, Rajasthan, India
| | - Mukta Agrawal
- Department of Pharmaceutics, Rungta College of Pharmaceutical Sciences and Research, Kohka, Kurud Road, Bhilai, Chhattisgarh, 490024, India
| | - Ajazuddin
- Department of Pharmaceutics, Rungta College of Pharmaceutical Sciences and Research, Kohka, Kurud Road, Bhilai, Chhattisgarh, 490024, India
| | - Swarnlata Saraf
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur, 492 010, Chhattisgarh, India
| | - Shailendra Saraf
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur, 492 010, Chhattisgarh, India
- Hemchand Yadav University, Durg, Chhattisgarh, 491 001, India
| | - Amit Alexander
- Department of Pharmaceutics, Rungta College of Pharmaceutical Sciences and Research, Kohka, Kurud Road, Bhilai, Chhattisgarh, 490024, India.
| |
Collapse
|
25
|
Schubert D, Currais A, Goldberg J, Finley K, Petrascheck M, Maher P. Geroneuroprotectors: Effective Geroprotectors for the Brain. Trends Pharmacol Sci 2018; 39:1004-1007. [PMID: 30446211 DOI: 10.1016/j.tips.2018.09.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 09/19/2018] [Accepted: 09/25/2018] [Indexed: 01/30/2023]
Abstract
Geroprotectors are compounds that slow the rate of biological aging and therefore may reduce the incidence of age-associated diseases such as Alzheimer's disease (AD). However, few have therapeutic efficacy in mammalian AD models. Here we describe the identification of geroneuroprotectors (GNPs), novel AD drug candidates that meet the criteria for geroprotectors.
Collapse
Affiliation(s)
- David Schubert
- Cellular Neurobiology Laboratory CNB-S, Salk Institute, La Jolla, CA 92037-1002, USA
| | - Antonio Currais
- Cellular Neurobiology Laboratory CNB-S, Salk Institute, La Jolla, CA 92037-1002, USA
| | - Joshua Goldberg
- Cellular Neurobiology Laboratory CNB-S, Salk Institute, La Jolla, CA 92037-1002, USA
| | - Kim Finley
- BioScience Center (BSC), San Diego State University, San Diego, CA 92105, USA
| | - Michael Petrascheck
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, CA 92037-1000, USA
| | - Pamela Maher
- Cellular Neurobiology Laboratory CNB-S, Salk Institute, La Jolla, CA 92037-1002, USA.
| |
Collapse
|
26
|
Next-generation biomarker discovery in Alzheimer's disease using metabolomics - from animal to human studies. Bioanalysis 2018; 10:1525-1546. [PMID: 30198770 DOI: 10.4155/bio-2018-0135] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease (AD) is a complex disease driven mainly by neuronal loss due to accumulation of intracellular neurofibrillary tangles and amyloid β aggregates in the brain. The diagnosis of AD currently relies on clinical symptoms while the disease can only be confirmed at autopsy. The few available biomarkers allowing for diagnosis are typically detected many years after the onset of the disease. New diagnostic approaches, particularly in easily-accessible biofluids, are essential. By providing an exhaustive information of the phenotype, metabolomics is an ideal approach for identification of new biomarkers. This review investigates the current position of metabolomics in the field of AD research, focusing on animal and human studies, and discusses the improvements carried out over the past decade.
Collapse
|
27
|
Aguirre-Plans J, Piñero J, Menche J, Sanz F, Furlong LI, Schmidt HHHW, Oliva B, Guney E. Proximal Pathway Enrichment Analysis for Targeting Comorbid Diseases via Network Endopharmacology. Pharmaceuticals (Basel) 2018; 11:E61. [PMID: 29932108 PMCID: PMC6160959 DOI: 10.3390/ph11030061] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 06/15/2018] [Accepted: 06/19/2018] [Indexed: 01/13/2023] Open
Abstract
The past decades have witnessed a paradigm shift from the traditional drug discovery shaped around the idea of “one target, one disease” to polypharmacology (multiple targets, one disease). Given the lack of clear-cut boundaries across disease (endo)phenotypes and genetic heterogeneity across patients, a natural extension to the current polypharmacology paradigm is to target common biological pathways involved in diseases via endopharmacology (multiple targets, multiple diseases). In this study, we present proximal pathway enrichment analysis (PxEA) for pinpointing drugs that target common disease pathways towards network endopharmacology. PxEA uses the topology information of the network of interactions between disease genes, pathway genes, drug targets and other proteins to rank drugs by their interactome-based proximity to pathways shared across multiple diseases, providing unprecedented drug repurposing opportunities. Using PxEA, we show that many drugs indicated for autoimmune disorders are not necessarily specific to the condition of interest, but rather target the common biological pathways across these diseases. Finally, we provide high scoring drug repurposing candidates that can target common mechanisms involved in type 2 diabetes and Alzheimer’s disease, two conditions that have recently gained attention due to the increased comorbidity among patients.
Collapse
Affiliation(s)
- Joaquim Aguirre-Plans
- Research Programme on Biomedical Informatics, the Hospital del Mar Medical Research Institute and Pompeu Fabra University, Dr. Aiguader 88, 08003 Barcelona, Spain.
| | - Janet Piñero
- Research Programme on Biomedical Informatics, the Hospital del Mar Medical Research Institute and Pompeu Fabra University, Dr. Aiguader 88, 08003 Barcelona, Spain.
| | - Jörg Menche
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, AKH BT 25.3, A-1090 Vienna, Austria.
| | - Ferran Sanz
- Research Programme on Biomedical Informatics, the Hospital del Mar Medical Research Institute and Pompeu Fabra University, Dr. Aiguader 88, 08003 Barcelona, Spain.
| | - Laura I Furlong
- Research Programme on Biomedical Informatics, the Hospital del Mar Medical Research Institute and Pompeu Fabra University, Dr. Aiguader 88, 08003 Barcelona, Spain.
| | - Harald H H W Schmidt
- Department of Pharmacology and Personalised Medicine, CARIM, FHML, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands.
| | - Baldo Oliva
- Research Programme on Biomedical Informatics, the Hospital del Mar Medical Research Institute and Pompeu Fabra University, Dr. Aiguader 88, 08003 Barcelona, Spain.
| | - Emre Guney
- Research Programme on Biomedical Informatics, the Hospital del Mar Medical Research Institute and Pompeu Fabra University, Dr. Aiguader 88, 08003 Barcelona, Spain.
- Department of Pharmacology and Personalised Medicine, CARIM, FHML, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands.
| |
Collapse
|
28
|
Kim H, Han H. Computer-Aided Multi-Target Management of Emergent Alzheimer's Disease. Bioinformation 2018; 14:167-180. [PMID: 29983487 PMCID: PMC6016757 DOI: 10.6026/97320630014167] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 04/29/2018] [Accepted: 04/30/2018] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease (AD) represents an enormous global health burden in terms of human suffering and economic cost. AD management requires a shift from the prevailing paradigm targeting pathogenesis to design and develop effective drugs with adequate success in clinical trials. Therefore, it is of interest to report a review on amyloid beta (Aβ) effects and other multi-targets including cholinesterase, NFTs, tau protein and TNF associated with brain cell death to be neuro-protective from AD. It should be noted that these molecules have been generated either by target-based or phenotypic methods. Hence, the use of recent advancements in nanomedicine and other natural compounds screening tools as a feasible alternative for circumventing specific liabilities is realized. We review recent developments in the design and identification of neuro-degenerative compounds against AD generated using current advancements in computational multi-target modeling algorithms reflected by theragnosis (combination of diagnostic tests and therapy) concern.
Collapse
Affiliation(s)
- Hyunjo Kim
- Department of Medical Informatics, Ajou Medical University Hospital, Suwon, Kyeounggido province, South Korea
| | - Hyunwook Han
- Department of Informatics, School of Medicine, CHA University, Seongnam, South Korea
- Institute of Basic Medical Sciences, School of Medicine, CHA University, Seongnam, South Korea
| |
Collapse
|
29
|
Wilkins JM, Trushina E. Application of Metabolomics in Alzheimer's Disease. Front Neurol 2018; 8:719. [PMID: 29375465 PMCID: PMC5770363 DOI: 10.3389/fneur.2017.00719] [Citation(s) in RCA: 167] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 12/13/2017] [Indexed: 12/22/2022] Open
Abstract
Progress toward the development of efficacious therapies for Alzheimer’s disease (AD) is halted by a lack of understanding early underlying pathological mechanisms. Systems biology encompasses several techniques including genomics, epigenomics, transcriptomics, proteomics, and metabolomics. Metabolomics is the newest omics platform that offers great potential for the diagnosis and prognosis of neurodegenerative diseases as an individual’s metabolome reflects alterations in genetic, transcript, and protein profiles and influences from the environment. Advancements in the field of metabolomics have demonstrated the complexity of dynamic changes associated with AD progression underscoring challenges with the development of efficacious therapeutic interventions. Defining systems-level alterations in AD could provide insights into disease mechanisms, reveal sex-specific changes, advance the development of biomarker panels, and aid in monitoring therapeutic efficacy, which should advance individualized medicine. Since metabolic pathways are largely conserved between species, metabolomics could improve the translation of preclinical research conducted in animal models of AD into humans. A summary of recent developments in the application of metabolomics to advance the AD field is provided below.
Collapse
Affiliation(s)
- Jordan Maximillian Wilkins
- Mitochondrial Neurobiology and Therapeutics Laboratory, Department of Neurology, Mayo Clinic, Rochester, MN, United States
| | - Eugenia Trushina
- Mitochondrial Neurobiology and Therapeutics Laboratory, Department of Neurology, Mayo Clinic, Rochester, MN, United States.,Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
30
|
Onyango IG. Modulation of mitochondrial bioenergetics as a therapeutic strategy in Alzheimer's disease. Neural Regen Res 2018; 13:19-25. [PMID: 29451200 PMCID: PMC5840984 DOI: 10.4103/1673-5374.224362] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2018] [Indexed: 01/14/2023] Open
Abstract
Alzheimer's disease (AD) is an increasingly pressing worldwide public-health, social, political and economic concern. Despite significant investment in multiple traditional therapeutic strategies that have achieved success in preclinical models addressing the pathological hallmarks of the disease, these efforts have not translated into any effective disease-modifying therapies. This could be because interventions are being tested too late in the disease process. While existing therapies provide symptomatic and clinical benefit, they do not fully address the molecular abnormalities that occur in AD neurons. The pathophysiology of AD is complex; mitochondrial bioenergetic deficits and brain hypometabolism coupled with increased mitochondrial oxidative stress are antecedent and potentially play a causal role in the disease pathogenesis. Dysfunctional mitochondria accumulate from the combination of impaired mitophagy, which can also induce injurious inflammatory responses, and inadequate neuronal mitochondrial biogenesis. Altering the metabolic capacity of the brain by modulating/potentiating its mitochondrial bioenergetics may be a strategy for disease prevention and treatment. We present insights into the mechanisms of mitochondrial dysfunction in AD brain as well as an overview of emerging treatments with the potential to prevent, delay or reverse the neurodegenerative process by targeting mitochondria.
Collapse
|