1
|
Almutary AG, Begum MY, Kyada AK, Gupta S, Jyothi SR, Chaudhary K, Sharma S, Sinha A, Abomughaid MM, Imran M, Lakhanpal S, Babalghith AO, Abu-Seer EA, Avinash D, Alzahrani HA, Alhindi AA, Iqbal D, Kumar S, Jha NK, Alghamdi S. Inflammatory signaling pathways in Alzheimer's disease: Mechanistic insights and possible therapeutic interventions. Ageing Res Rev 2025; 104:102548. [PMID: 39419399 DOI: 10.1016/j.arr.2024.102548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 10/19/2024]
Abstract
The complex pathophysiology of Alzheimer's disease (AD) poses challenges for the development of therapies. Recently, neuroinflammation has been identified as a key pathogenic mechanism underlying AD, while inflammation has emerged as a possible target for the management and prevention of AD. Several prior studies have demonstrated that medications modulating neuroinflammation might lessen AD symptoms, mostly by controlling neuroinflammatory signaling pathways such as the NF-κB, MAPK, NLRP3, etc, and their respective signaling cascade. Moreover, targeting these inflammatory modalities with inhibitors, natural products, and metabolites has been the subject of intensive research because of their anti-inflammatory characteristics, with many studies demonstrating noteworthy pharmacological capabilities and potential clinical applications. Therefore, targeting inflammation is considered a promising strategy for treating AD. This review comprehensively elucidates the neuroinflammatory mechanisms underlying AD progression and the beneficial effects of inhibitors, natural products, and metabolites in AD treatment.
Collapse
Affiliation(s)
- Abdulmajeed G Almutary
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, P.O. Box 59911, Abu Dhabi, United Arab Emirates
| | - M Yasmin Begum
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Ashish Kumar Kyada
- Marwadi University Research Center, Department of Pharmaceutical Sciences, Faculty of Health Sciences, Marwadi University, Rajkot, Gujarat 360003, India
| | - Saurabh Gupta
- Department of Biotechnology, GLA University, Mathura, Uttar Pradesh, India
| | - S Renuka Jyothi
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Kamlesh Chaudhary
- Department of Neurology, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| | - Swati Sharma
- Chandigarh Pharmacy College, Chandigarh Group of Colleges, Jhanjeri, Mohali, Punjab 140307, India
| | - Aashna Sinha
- School of Applied and Life Sciences, Division of Research and Innovation, Uttaranchal University, Dehradun, Uttarakhand
| | - Mosleh Mohammad Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha 61922, Saudi Arabia
| | - Mohd Imran
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia; Center for Health Research, Northern Border University, Arar, Saudi Arabia
| | - Sorabh Lakhanpal
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India
| | - Ahmad O Babalghith
- Medical Genetics Department, College of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Eman Adnan Abu-Seer
- Department of Epidemiology and Medical Statistic, Faculty of Public Health and Health Informatics, Umm Al-Qura University, Makkah, Saudi Arabia
| | - D Avinash
- Center for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, India
| | - Hassan A Alzahrani
- Department of Respiratory Care, Medical Cities at the Minister of Interior, MCMOl, Riyadh, Saudi Arabia
| | | | - Danish Iqbal
- Department of Health Information Management, College of Applied Medical Sciences, Buraydah Private Colleges, Buraydah 51418, Saudi Arabia
| | - Sandeep Kumar
- School of Pharmacy, Sharda University, Greater Noida, India; DST-FIST Laboratory, Sharda University, Greater Noida, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Biosciences and Technology (SBT), Galgotias University, Greater Noida, India; Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura 140401, Punjab, India.
| | - Saad Alghamdi
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| |
Collapse
|
2
|
Beckers L, Rashid M, Lee AJ, Chatila ZK, Tamucci KA, Talcoff RC, Hall JL, Bennett DA, Vardarajan BN, Bradshaw EM. CD33 and SHP-1/ PTPN6 Interaction in Alzheimer's Disease. Genes (Basel) 2024; 15:1204. [PMID: 39336795 PMCID: PMC11431297 DOI: 10.3390/genes15091204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 09/04/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
Large-scale genetic studies have identified numerous genetic risk factors that suggest a central role for innate immune cells in susceptibility to Alzheimer's disease (AD). CD33, an immunomodulatory transmembrane sialic acid binding protein expressed on myeloid cells, was identified as one such genetic risk factor associated with Alzheimer's disease. Several studies explored the molecular outcomes of genetic variation at the CD33 locus. It has been determined that the risk variant associated with AD increases the expression of the large isoform of CD33 (CD33M) in innate immune cells and alters its biological functions. CD33 is thought to signal via the interaction of its ITIM motif and the protein tyrosine phosphatase, SHP-1. Here, we utilize different molecular and computational approaches to investigate how AD-associated genetic variation in CD33 affects its interaction with SHP-1 in human microglia and microglia-like cells. Our findings demonstrate a genotype-dependent interaction between CD33 and SHP-1, which may functionally contribute to the AD risk associated with this CD33 variant. We also found that CD33-PTPN6 (SHP-1) gene-gene interactions impact AD-related traits, while CD33-PTPN11 (SHP-2) interactions do not.
Collapse
Affiliation(s)
- Lien Beckers
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA; (L.B.); (M.R.); (A.J.L.); (Z.K.C.); (K.A.T.); (R.C.T.); (J.L.H.); (B.N.V.)
| | - Mamunur Rashid
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA; (L.B.); (M.R.); (A.J.L.); (Z.K.C.); (K.A.T.); (R.C.T.); (J.L.H.); (B.N.V.)
- Division of Translational Neurobiology, Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
- The Carol and Gene Ludwig Center for Research on Neurodegeneration, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Annie J. Lee
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA; (L.B.); (M.R.); (A.J.L.); (Z.K.C.); (K.A.T.); (R.C.T.); (J.L.H.); (B.N.V.)
- Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
- The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, New York, NY 10032, USA
| | - Zena K. Chatila
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA; (L.B.); (M.R.); (A.J.L.); (Z.K.C.); (K.A.T.); (R.C.T.); (J.L.H.); (B.N.V.)
- Division of Translational Neurobiology, Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
- The Carol and Gene Ludwig Center for Research on Neurodegeneration, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Kirstin A. Tamucci
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA; (L.B.); (M.R.); (A.J.L.); (Z.K.C.); (K.A.T.); (R.C.T.); (J.L.H.); (B.N.V.)
- Division of Translational Neurobiology, Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
- The Carol and Gene Ludwig Center for Research on Neurodegeneration, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Ryan C. Talcoff
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA; (L.B.); (M.R.); (A.J.L.); (Z.K.C.); (K.A.T.); (R.C.T.); (J.L.H.); (B.N.V.)
- Division of Translational Neurobiology, Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
- The Carol and Gene Ludwig Center for Research on Neurodegeneration, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Jennifer L. Hall
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA; (L.B.); (M.R.); (A.J.L.); (Z.K.C.); (K.A.T.); (R.C.T.); (J.L.H.); (B.N.V.)
- Division of Translational Neurobiology, Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
- The Carol and Gene Ludwig Center for Research on Neurodegeneration, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - David A. Bennett
- Rush Alzheimer Disease Center, Rush University Medical Center, Chicago, IL 60612, USA;
| | - Badri N. Vardarajan
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA; (L.B.); (M.R.); (A.J.L.); (Z.K.C.); (K.A.T.); (R.C.T.); (J.L.H.); (B.N.V.)
- Gertrude H. Sergievsky Center, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
- The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, New York, NY 10032, USA
| | - Elizabeth M. Bradshaw
- Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA; (L.B.); (M.R.); (A.J.L.); (Z.K.C.); (K.A.T.); (R.C.T.); (J.L.H.); (B.N.V.)
- Division of Translational Neurobiology, Department of Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
- The Carol and Gene Ludwig Center for Research on Neurodegeneration, Columbia University Irving Medical Center, New York, NY 10032, USA
- The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, New York, NY 10032, USA
| |
Collapse
|
3
|
Cheng M, Yuan C, Ju Y, Liu Y, Shi B, Yang Y, Jin S, He X, Zhang L, Min D. Quercetin Attenuates Oxidative Stress and Apoptosis in Brain Tissue of APP/PS1 Double Transgenic AD Mice by Regulating Keap1/Nrf2/HO-1 Pathway to Improve Cognitive Impairment. Behav Neurol 2024; 2024:5698119. [PMID: 39233848 PMCID: PMC11374423 DOI: 10.1155/2024/5698119] [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: 12/18/2023] [Revised: 06/02/2024] [Accepted: 06/20/2024] [Indexed: 09/06/2024] Open
Abstract
Objective: The objective of the study is to investigate whether quercetin ameliorates Alzheimer's disease (AD)-like pathology in APP/PS1 double transgenic mice and its hypothesized mechanism, contributing to the comprehension of AD pathogenesis. Methods: A total of 30 APP/PS1 transgenic mice were randomized into model group (APP/PS1), quercetin group (APP/PS1+Q), and donepezil hydrochloride group (APP/PS1+DON). Simultaneously, there were 10 C57 mice of the same age served as a control group. Three months posttreatment, the effects of quercetin on AD mice were evaluated using the Morris water maze (MWM) test, Y maze experiment, immunohistochemistry, immunofluorescence, and western blotting. Results: Results from the water maze and Y maze indicated that quercetin significantly improved cognitive impairment in APP/PS1 transgenic AD mice. Additionally, serum enzyme-linked immunosorbent assay (ELISA) results demonstrated that quercetin elevated MDA, superoxide dismutase (SOD), CAT, GSH, acetylcholine (ACh), and acetylcholinesterase (AChE) levels in AD mice. Hematoxylin-eosin (HE) staining, Nissl staining, and hippocampal tissue thioflavine staining revealed that quercetin reduced neuronal damage and Aβ protein accumulation in AD mice. Western blot validated protein expression in the Kelch-like ECH-associated protein 1 (Keap1)/nuclear factor erythroid 2-related factor 2 (Nrf2)/HO-1 pathway associated with oxidative stress and apoptosis, confirming quercetin's potential molecular mechanism of enhancing AD mouse cognition. Furthermore, western blot findings indicate that quercetin significantly alters protein expression in the Keap1/Nrf2/HO-1 pathway. Moreover, molecular docking analysis suggests that Keap1, NQO1, HO-1, caspase-3, Bcl-2, and Bax proteins in the Keap1/Nrf2/HO-1 pathway may be potential regulatory targets of quercetin. These findings will provide a molecular basis for quercetin's clinical application in AD treatment. Conclusion: Quercetin can improve cognitive impairment and AD-like pathology in APP/PS1 double transgenic mice, potentially related to quercetin's activation of the Keap1/Nrf2/HO-1 pathway and reduction of cell apoptosis.
Collapse
Affiliation(s)
- Meijia Cheng
- Affiliated Hospital of Liaoning University of Traditional Chinese MedicineExperimental Center of Traditional Chinese Medicine, Shenyang 110032, China
| | - Changbin Yuan
- Liaoning University of Traditional Chinese Medicine, Shenyang 110847, China
| | - Yetao Ju
- Affiliated Hospital of Liaoning University of Traditional Chinese MedicineExperimental Center of Traditional Chinese Medicine, Shenyang 110032, China
| | - Yongming Liu
- Affiliated Hospital of Liaoning University of Traditional Chinese MedicineExperimental Center of Traditional Chinese Medicine, Shenyang 110032, China
| | - Baorui Shi
- Liaoning University of Traditional Chinese Medicine, Shenyang 110847, China
| | - Yali Yang
- Liaoning University of Traditional Chinese Medicine, Shenyang 110847, China
| | - Sian Jin
- Liaoning University of Traditional Chinese Medicine, Shenyang 110847, China
| | - Xiaoming He
- Liaoning University of Traditional Chinese Medicine, Shenyang 110847, China
| | - Li Zhang
- Liaoning University of Traditional Chinese Medicine, Shenyang 110847, China
| | - Dongyu Min
- Key Laboratory of Ministry of Education for TCM Viscera-State Theory and ApplicationsLiaoning University of Traditional Chinese Medicine, Shenyang 110847, China
| |
Collapse
|
4
|
Kepp KP, Robakis NK, Høilund-Carlsen PF, Sensi SL, Vissel B. The amyloid cascade hypothesis: an updated critical review. Brain 2023; 146:3969-3990. [PMID: 37183523 DOI: 10.1093/brain/awad159] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 04/20/2023] [Accepted: 04/23/2023] [Indexed: 05/16/2023] Open
Abstract
Results from recent clinical trials of antibodies that target amyloid-β (Aβ) for Alzheimer's disease have created excitement and have been heralded as corroboration of the amyloid cascade hypothesis. However, while Aβ may contribute to disease, genetic, clinical, imaging and biochemical data suggest a more complex aetiology. Here we review the history and weaknesses of the amyloid cascade hypothesis in view of the new evidence obtained from clinical trials of anti-amyloid antibodies. These trials indicate that the treatments have either no or uncertain clinical effect on cognition. Despite the importance of amyloid in the definition of Alzheimer's disease, we argue that the data point to Aβ playing a minor aetiological role. We also discuss data suggesting that the concerted activity of many pathogenic factors contribute to Alzheimer's disease and propose that evolving multi-factor disease models will better underpin the search for more effective strategies to treat the disease.
Collapse
Affiliation(s)
- Kasper P Kepp
- Section of Biophysical and Biomedicinal chemistry, DTU Chemistry, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Nikolaos K Robakis
- Icahn School of Medicine at Mount Sinai Medical Center, New York, NY 10029, USA
| | - Poul F Høilund-Carlsen
- Department of Nuclear Medicine, Odense University Hospital, 5000 Odense C, Denmark
- Department of Clinical Research, University of Southern Denmark, 5000 Odense C, Denmark
| | - Stefano L Sensi
- Center for Advanced Studies and Technology-CAST, and Institute for Advanced Biotechnology (ITAB), University G. d'Annunzio of Chieti-Pescara, Chieti, 66013, Italy
- Department of Neuroscience, Imaging, and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Chieti, 66013, Italy
| | - Bryce Vissel
- St Vincent's Hospital Centre for Applied Medical Research, St Vincent's Hospital, Sydney, 2010, Australia
- School of Clinical Medicine, UNSW Medicine and Health, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, Sydney, NSW 2052, Australia
| |
Collapse
|
5
|
Rarinca V, Nicoara MN, Ureche D, Ciobica A. Exploitation of Quercetin's Antioxidative Properties in Potential Alternative Therapeutic Options for Neurodegenerative Diseases. Antioxidants (Basel) 2023; 12:1418. [PMID: 37507955 PMCID: PMC10376113 DOI: 10.3390/antiox12071418] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 07/05/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
Oxidative stress (OS) is a condition in which there is an excess of reactive oxygen species (ROS) in the body, which can lead to cell and tissue damage. This occurs when there is an overproduction of ROS or when the body's antioxidant defense systems are overwhelmed. Quercetin (Que) is part of a group of compounds called flavonoids. It is found in high concentrations in vegetables, fruits, and other foods. Over the past decade, a growing number of studies have highlighted the therapeutic potential of flavonoids to modulate neuronal function and prevent age-related neurodegeneration. Therefore, Que has been shown to have antioxidant, anticancer, and anti-inflammatory properties, both in vitro and in vivo. Due to its antioxidant character, Que alleviates oxidative stress, thus improving cognitive function, reducing the risk of neurodegenerative diseases. On the other hand, Que can also help support the body's natural antioxidant defense systems, thus being a potentially practical supplement for managing OS. This review focuses on experimental studies supporting the neuroprotective effects of Que in Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), and epilepsy.
Collapse
Affiliation(s)
- Viorica Rarinca
- Doctoral School of Geosciences, Faculty of Geography and Geology, Alexandru Ioan Cuza University of Iasi, No 20A, Carol I Avenue, 700505 Iasi, Romania
- Doctoral School of Biology, Faculty of Biology, Alexandru Ioan Cuza University of Iasi, No 20A, Carol I Avenue, 700506 Iasi, Romania
| | - Mircea Nicusor Nicoara
- Doctoral School of Geosciences, Faculty of Geography and Geology, Alexandru Ioan Cuza University of Iasi, No 20A, Carol I Avenue, 700505 Iasi, Romania
- Department of Biology, Faculty of Biology, Alexandru Ioan Cuza University of Iasi, No 20A, Carol I Avenue, 700505 Iasi, Romania
| | - Dorel Ureche
- Department of Biology, Ecology and Environmental Protection, Faculty of Sciences, University Vasile Alecsandri of Bacau, Calea Marasesti Street, No 157, 600115 Bacau, Romania
| | - Alin Ciobica
- Department of Biology, Faculty of Biology, Alexandru Ioan Cuza University of Iasi, No 20A, Carol I Avenue, 700505 Iasi, Romania
- Center of Biomedical Research, Romanian Academy, No 8, Carol I Avenue, 700506 Iasi, Romania
- Academy of Romanian Scientists, No 54, Independence Street, Sector 5, 050094 Bucharest, Romania
| |
Collapse
|
6
|
Abstract
Alzheimer's disease (AD) is a debilitating age-related neurodegenerative condition. Unbiased genetic studies have implicated a central role for microglia, the resident innate immune cells of the central nervous system, in AD pathogenesis. On-going efforts are clarifying the biology underlying these associations and the microglial pathways that are dysfunctional in AD. Several genetic risk factors converge to decrease the function of activating microglial receptors and increase the function of inhibitory receptors, resulting in a seemingly dampened microglial phenotype in AD. Moreover, many of these microglial proteins that are genetically associated with AD appear to interact and share pathways or regulatory mechanisms, presenting several points of convergence that may be strategic targets for therapeutic intervention. Here, we review some of these studies and their implications for microglial participation in AD pathogenesis.
Collapse
|
7
|
Kepp KP, Sensi SL, Johnsen KB, Barrio JR, Høilund-Carlsen PF, Neve RL, Alavi A, Herrup K, Perry G, Robakis NK, Vissel B, Espay AJ. The Anti-Amyloid Monoclonal Antibody Lecanemab: 16 Cautionary Notes. J Alzheimers Dis 2023; 94:497-507. [PMID: 37334596 DOI: 10.3233/jad-230099] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
After the CLARITY-AD clinical trial results of lecanemab were interpreted as positive, and supporting the amyloid hypothesis, the drug received accelerated Food and Drug Administration approval. However, we argue that benefits of lecanemab treatment are uncertain and may yield net harm for some patients, and that the data do not support the amyloid hypothesis. We note potential biases from inclusion, unblinding, dropouts, and other issues. Given substantial adverse effects and subgroup heterogeneity, we conclude that lecanemab's efficacy is not clinically meaningful, consistent with numerous analyses suggesting that amyloid-β and its derivatives are not the main causative agents of Alzheimer's disease dementia.
Collapse
Affiliation(s)
- Kasper P Kepp
- Department of Chemistry, Section of Biophysical and Biomedicinal Chemistry, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Stefano L Sensi
- Center for Advanced Studies and Technology - CAST, and Institute for Advanced Biotechnology (ITAB), University G. d'Annunzio of Chieti-Pescara, Italy
- Department of Neuroscience, Imaging, and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Italy
| | - Kasper B Johnsen
- Department of Health Science and Technology, Neurobiology Research and Drug Delivery Group, Aalborg University, Aalborg, Denmark
| | - Jorge R Barrio
- Department of Molecular and Medical Pharmacology, UCLA School of Medicine, Los Angeles, CA, USA
| | - Poul F Høilund-Carlsen
- Department of Nuclear Medicine, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Rachael L Neve
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Abass Alavi
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA USA
| | - Karl Herrup
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - George Perry
- Department of Neuroscience, Developmental and Regenerative Biology, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Nikolaos K Robakis
- Icahn School of Medicine at Mount Sinai Medical Center, New York, NY, USA
| | - Bryce Vissel
- St Vincent's Hospital Centre for Applied Medical Research, St Vincent's Hospital, Darlinghurst, NSW, Australia
- School of Clinical Medicine, UNSW Medicine & Health, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Alberto J Espay
- Department of Neurology, James J. and Joan A. Gardner Family Center for Parkinson's Disease and Movement Disorders, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
8
|
Abubakar MB, Sanusi KO, Ugusman A, Mohamed W, Kamal H, Ibrahim NH, Khoo CS, Kumar J. Alzheimer’s Disease: An Update and Insights Into Pathophysiology. Front Aging Neurosci 2022; 14:742408. [PMID: 35431894 PMCID: PMC9006951 DOI: 10.3389/fnagi.2022.742408] [Citation(s) in RCA: 88] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 02/25/2022] [Indexed: 12/17/2022] Open
Abstract
Alzheimer’s disease (AD) is an irreversible brain disorder associated with slow, progressive loss of brain functions mostly in older people. The disease processes start years before the symptoms are manifested at which point most therapies may not be as effective. In the hippocampus, the key proteins involved in the JAK2/STAT3 signaling pathway, such as p-JAK2-Tyr1007 and p-STAT3-Tyr705 were found to be elevated in various models of AD. In addition to neurons, glial cells such as astrocytes also play a crucial role in the progression of AD. Without having a significant effect on tau and amyloid pathologies, the JAK2/STAT3 pathway in reactive astrocytes exhibits a behavioral impact in the experimental models of AD. Cholinergic atrophy in AD has been traced to a trophic failure in the NGF metabolic pathway, which is essential for the survival and maintenance of basal forebrain cholinergic neurons (BFCN). In AD, there is an alteration in the conversion of the proNGF to mature NGF (mNGF), in addition to an increase in degradation of the biologically active mNGF. Thus, the application of exogenous mNGF in experimental studies was shown to improve the recovery of atrophic BFCN. Furthermore, it is now coming to light that the FGF7/FGFR2/PI3K/Akt signaling pathway mediated by microRNA-107 is also involved in AD pathogenesis. Vascular dysfunction has long been associated with cognitive decline and increased risk of AD. Vascular risk factors are associated with higher tau and cerebral beta-amyloid (Aβ) burden, while synergistically acting with Aβ to induce cognitive decline. The apolipoprotein E4 polymorphism is not just one of the vascular risk factors, but also the most prevalent genetic risk factor of AD. More recently, the research focus on AD shifted toward metabolisms of various neurotransmitters, major and minor nutrients, thus giving rise to metabolomics, the most important “omics” tool for the diagnosis and prognosis of neurodegenerative diseases based on an individual’s metabolome. This review will therefore proffer a better understanding of novel signaling pathways associated with neural and glial mechanisms involved in AD, elaborate potential links between vascular dysfunction and AD, and recent developments in “omics”-based biomarkers in AD.
Collapse
Affiliation(s)
- Murtala Bello Abubakar
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, Usmanu Danfodiyo University, Sokoto, Nigeria
- Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University, Sokoto, Nigeria
| | - Kamaldeen Olalekan Sanusi
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, Usmanu Danfodiyo University, Sokoto, Nigeria
- Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University, Sokoto, Nigeria
| | - Azizah Ugusman
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Wael Mohamed
- Department of Basic Medical Science, Kulliyyah of Medicine, International Islamic University Malaysia, Kuantan, Malaysia
- Department of Clinical Pharmacology, Faculty of Medicine, Menoufia University, Shebin El-Kom, Egypt
| | - Haziq Kamal
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Nurul Husna Ibrahim
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Ching Soong Khoo
- Neurology Unit, Department of Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Jaya Kumar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
- *Correspondence: Jaya Kumar,
| |
Collapse
|
9
|
Hossain MF, Wang N, Chen R, Li S, Roy J, Uddin MG, Li Z, Lim LW, Song YQ. Exploring the multifunctional role of melatonin in regulating autophagy and sleep to mitigate Alzheimer's disease neuropathology. Ageing Res Rev 2021; 67:101304. [PMID: 33610813 DOI: 10.1016/j.arr.2021.101304] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 02/12/2021] [Accepted: 02/15/2021] [Indexed: 12/12/2022]
Abstract
Melatonin (MLT) is a neurohormone that is regulated by the circadian clock and plays multifunctional roles in numerous neurodegenerative disorders, such as Alzheimer's disease (AD). AD is the most common form of dementia and is associated with the degradation of axons and synapses resulting in memory loss and cognitive impairment. Despite extensive research, there is still no effective cure or specific treatment to prevent the progression of AD. The pathogenesis of AD involves atrophic alterations in the brain that also result in circadian alterations, sleep disruption, and autophagic dysfunction. In this scenario, MLT and autophagy play a central role in removing the misfolded protein aggregations. MLT also promotes autophagy through inhibiting methamphetamine toxicity to protect against neuronal cell death in AD brain. Besides, MLT plays critical roles as either a pro-autophagic indicator or anti-autophagic regulator depending on the phase of autophagy. MLT also has antioxidant properties that can counteract mitochondrial damage, oxidative stress, and apoptosis. Aging, a major risk factor for AD, can change sleep patterns and sleep quality, and MLT can improve sleep quality through regulating sleep cycles. The primary purpose of this review is to explore the putative mechanisms of the beneficial effects of MLT in AD patients. Furthermore, we also summarize the findings from preclinical and clinical studies on the multifunctional roles of MLT on autophagic regulation, the control of the circadian clock-associated genes, and sleep regulation.
Collapse
|
10
|
Uddin MS, Hasana S, Ahmad J, Hossain MF, Rahman MM, Behl T, Rauf A, Ahmad A, Hafeez A, Perveen A, Ashraf GM. Anti-Neuroinflammatory Potential of Polyphenols by Inhibiting NF-κB to Halt Alzheimer's Disease. Curr Pharm Des 2021; 27:402-414. [PMID: 33213314 DOI: 10.2174/1381612826666201118092422] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 10/01/2020] [Indexed: 11/22/2022]
Abstract
Alzheimer's disease (AD) is an irrevocable chronic brain disorder featured by neuronal loss, microglial accumulation, and progressive cognitive impairment. The proper pathophysiology of this life-threatening disorder is not completely understood and no exact remedies have been found yet. Over the last few decades, research on AD has mainly highlighted pathomechanisms linked to a couple of the major pathological hallmarks, including extracellular senile plaques made of amyloid-β (Aβ) peptides, and intracellular neurofibrillary tangles (NFTs) made of tau proteins. Aβ can induce apoptosis, trigger an inflammatory response, and inhibit the synaptic plasticity of the hippocampus, which ultimately contributes to reducing cognitive functions and memory impairment. Recently, a third disease hallmark, the neuroinflammatory reaction that is mediated by cerebral innate immune cells, has become a spotlight in the current research area, assured by pre-clinical, clinical, and genetic investigations. Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), a cytokine producer, is significantly associated with physiological inflammatory proceedings and thus shows a promising candidate for inflammation- based AD therapy. Recent data reveal that phytochemicals, mainly polyphenol compounds, exhibit potential neuroprotective functions and these may be considered as a vital resource for discovering several drug candidates against AD. Interestingly, phytochemicals can easily interfere with the signaling pathway of NF-κB. This review represents the anti-neuroinflammatory potential of polyphenols as inhibitors of NF-κB to combat AD pathogenesis.
Collapse
Affiliation(s)
- Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh
| | - Sharifa Hasana
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh
| | - Jamil Ahmad
- Department of Human Nutrition, The University of Agriculture Peshawar, Khyber Pakhtunkhwa, Pakistan
| | | | | | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Abdur Rauf
- Department of Chemistry, University of Swabi, Swabi, Anbar, Khyber Pakhtunkhwa, Pakistan
| | - Ausaf Ahmad
- Amity Institute of Biotechnology, Amity University Uttar Pradesh Lucknow Campus, Uttar Pradesh, India
| | - Abdul Hafeez
- Glocal School of Pharmacy, Glocal University, Saharanpur, India
| | - Asma Perveen
- Glocal School of Life Sciences, Glocal University, Saharanpur, India
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
11
|
He C, Huang ZS, Yu CC, Wang HH, Zhou H, Kong LH. Epigenetic Regulation of Amyloid-beta Metabolism in Alzheimer's Disease. Curr Med Sci 2021; 40:1022-1030. [PMID: 33428129 DOI: 10.1007/s11596-020-2283-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 10/05/2020] [Indexed: 11/28/2022]
Abstract
Senile plaques (SPs) are one of the pathological features of Alzheimer's disease (AD) and they are formed by the overproduction and aggregation of amyloid-beta (Aβ) peptides derived from the abnormal cleavage of amyloid precursor protein (APP). Thus, understanding the regulatory mechanisms during Aβ metabolism is of great importance to elucidate AD pathogenesis. Recent studies have shown that epigenetic modulation-including DNA methylation, non-coding RNA alterations, and histone modifications-is of great significance in regulating Aβ metabolism. In this article, we review the aberrant epigenetic regulation of Aβ metabolism.
Collapse
Affiliation(s)
- Chuan He
- Hubei University of Chinese Medicine, Wuhan, 430060, China
| | | | - Chao-Chao Yu
- Department of Tuina, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518000, China.,The 4th Clinical College of Guangzhou University of Chinese Medicine, Shenzhen, 518000, China
| | - Hai-Hua Wang
- Hospital of Traditional Chinese Medicine of Fengrun District, Tangshan, 064000, China
| | - Hua Zhou
- Hubei University of Chinese Medicine, Wuhan, 430060, China.
| | - Li-Hong Kong
- Hubei University of Chinese Medicine, Wuhan, 430060, China.
| |
Collapse
|
12
|
Mehra R, Kepp KP. Identification of Structural Calcium Binding Sites in Membrane-Bound Presenilin 1 and 2. J Phys Chem B 2020; 124:4697-4711. [PMID: 32420742 DOI: 10.1021/acs.jpcb.0c01712] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Variants of presenilin (PS1 and PS2) are the main genetic risk factors of familial Alzheimer's disease and thus central to the disease etiology. Although mostly studied as catalytic units of γ-secretase controlling Aβ production, presenilins also affect calcium levels, which are disturbed in Alzheimer's disease. We investigated the interaction of calcium with both PS1 and PS2 using all-atom molecular dynamics (MD) simulations in realistic membrane models, with the specific aim to identify any Ca2+ sites. We did not observe any complete Ca2+ leak event, but we identified four persistent Ca2+ sites in membrane-bound PS1 and PS2: One in HL2 near the C-terminal of TM6, one in HL2 toward the N-terminal of TM7, a site at the catalytic aspartate on TM7, and a site at the PALP motif on TM9. The sites feature negatively charged glutamates and aspartates typical of calcium binding. Structural homology to diaspartate calcium transport proteins and mutation studies of calcium efflux support our identified calcium sites. Calcium consistently dampens HL2 motions in all comparisons (PS1, protonated PS1, PS2, protonated PS2). Due to their location in HL2 and the active site, we propose that the calcium sites control autoproteolytic maturation of presenilin by a pH-dependent conformational restriction of the HL2 recognition loop, which also regulates calcium transport proteins such as inositol 1,4,5-triphosphate receptor and ryanodine receptor. Our structural dynamics could provide a possible molecular basis for the need of both calcium and presenilin for lysosome proteolytic function, perhaps relevant also to other protein misfolding diseases.
Collapse
Affiliation(s)
- Rukmankesh Mehra
- Technical University of Denmark, DTU Chemistry, DK-2800 Kongens Lyngby, Denmark
| | - Kasper P Kepp
- Technical University of Denmark, DTU Chemistry, DK-2800 Kongens Lyngby, Denmark
| |
Collapse
|
13
|
Khan H, Ullah H, Aschner M, Cheang WS, Akkol EK. Neuroprotective Effects of Quercetin in Alzheimer's Disease. Biomolecules 2019; 10:59. [PMID: 31905923 PMCID: PMC7023116 DOI: 10.3390/biom10010059] [Citation(s) in RCA: 250] [Impact Index Per Article: 41.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 12/21/2019] [Accepted: 12/22/2019] [Indexed: 02/05/2023] Open
Abstract
Quercetin is a flavonoid with notable pharmacological effects and promising therapeutic potential. It is widely distributed among plants and found commonly in daily diets predominantly in fruits and vegetables. Neuroprotection by quercetin has been reported in several in vitro studies. It has been shown to protect neurons from oxidative damage while reducing lipid peroxidation. In addition to its antioxidant properties, it inhibits the fibril formation of amyloid-β proteins, counteracting cell lyses and inflammatory cascade pathways. In this review, we provide a synopsis of the recent literature exploring the relationship between quercetin and cognitive performance in Alzheimer's disease and its potential as a lead compound in clinical applications.
Collapse
Affiliation(s)
- Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan 23200, Pakistan;
| | - Hammad Ullah
- Department of Pharmacy, Abdul Wali Khan University, Mardan 23200, Pakistan;
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Forchheimer 209, 1300 Morris Park Avenue, Bronx, NY 10461, USA;
| | - Wai San Cheang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macau, China;
| | - Esra Küpeli Akkol
- Department of Pharmacognosy, Faculty of Pharmacy Gazi University, 06330 Etiler/Ankara, Turkey;
| |
Collapse
|
14
|
Kepp KP, Squitti R. Copper imbalance in Alzheimer’s disease: Convergence of the chemistry and the clinic. Coord Chem Rev 2019. [DOI: 10.1016/j.ccr.2019.06.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
|
15
|
Rezazadeh M, Hosseinzadeh H, Moradi M, Salek Esfahani B, Talebian S, Parvin S, Gharesouran J. Genetic discoveries and advances in late-onset Alzheimer's disease. J Cell Physiol 2019; 234:16873-16884. [PMID: 30790294 DOI: 10.1002/jcp.28372] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 01/20/2019] [Accepted: 01/24/2019] [Indexed: 01/05/2025]
Abstract
Alzheimer's disease (AD) is a heterogeneous disorder with multiple patterns of clinical manifestations. Recently, due to the advance of linkage studies, next-generation sequencing and genome-wide association studies, a large number of putative risk genes for AD have been identified using acquired genome mega data. The genetic association between three causal genes, including amyloid precursor protein, presenilin1, and presenilin2 in early-onset AD (EOAD), was discovered over the past few decades. These discoveries showed that there should be additional genetic risk factors for both EOAD and late-onset AD (LOAD) to help fully explain the leading molecular mechanisms in a single pathophysiological entity. This study reviews the clinical features and genetic etiology of LOAD and discusses a variety of AD-mediated genes that are involved in cholesterol and lipid metabolism, endocytosis, and immune response according to their mutations for more efficient selection of functional candidate genes for LOAD. New mechanisms and pathways have been identified as a result.
Collapse
Affiliation(s)
- Maryam Rezazadeh
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Division of Medical Genetics, Tabriz Children's Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mohsen Moradi
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behnaz Salek Esfahani
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shahrzad Talebian
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shaho Parvin
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jalal Gharesouran
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Division of Medical Genetics, Tabriz Children's Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
16
|
Fišar Z, Jirák R, Zvěřová M, Setnička V, Habartová L, Hroudová J, Vaníčková Z, Raboch J. Plasma amyloid beta levels and platelet mitochondrial respiration in patients with Alzheimer's disease. Clin Biochem 2019; 72:71-80. [PMID: 30954436 DOI: 10.1016/j.clinbiochem.2019.04.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 04/03/2019] [Indexed: 12/12/2022]
Abstract
OBJECTIVES Altered amyloid metabolism and mitochondrial dysfunction play key roles in the development of Alzheimer's disease (AD). We asked whether an association exists between disturbed platelet mitochondrial respiration and the plasma concentrations of Aβ40 and Aβ42 in patients with AD. DESIGN AND METHODS Plasma Aβ40 and Aβ42 concentrations and mitochondrial respiration in intact and permeabilized platelets were measured in 50 patients with AD, 15 patients with vascular dementia and 25 control subjects. A pilot longitudinal study was performed to monitor the progression of AD in a subgroup 11 patients with AD. RESULTS The mean Aβ40, Aβ42 and Aβ42/Aβ40 levels were not significantly altered in patients with AD compared with controls. The mitochondrial respiratory rate in intact platelets was significantly reduced in patients with AD compared to controls, particularly the basal respiratory rate, maximum respiratory capacity, and respiratory reserve; however, the flux control ratio for basal respiration was increased. A correlation between the plasma Aβ42 concentration and mitochondrial respiration in both intact and permeabilized platelets differs in controls and patients with AD. CONCLUSIONS Based on our data, (1) mitochondrial respiration in intact platelets, but not the Aβ level itself, may be included in a panel of biomarkers for AD; (2) dysfunctional mitochondrial respiration in platelets is not explained by changes in plasma Aβ concentrations; and (3) the association between mitochondrial respiration in platelets and plasma Aβ levels differs in patients with AD and controls. The results supported the hypothesis that mitochondrial dysfunction is the primary factor contributing to the development of AD.
Collapse
Affiliation(s)
- Zdeněk Fišar
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic.
| | - Roman Jirák
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic.
| | - Martina Zvěřová
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic.
| | - Vladimír Setnička
- Department of Analytical Chemistry, University of Chemistry and Technology, Prague, Czech Republic.
| | - Lucie Habartová
- Department of Analytical Chemistry, University of Chemistry and Technology, Prague, Czech Republic.
| | - Jana Hroudová
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic.
| | - Zdislava Vaníčková
- Institute of Medical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic.
| | - Jiří Raboch
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic.
| |
Collapse
|
17
|
Banning LCP, Ramakers IHGB, Deckers K, Verhey FRJ, Aalten P. Apolipoprotein E and affective symptoms in mild cognitive impairment and Alzheimer's disease dementia: A systematic review and meta-analysis. Neurosci Biobehav Rev 2018; 96:302-315. [PMID: 30513312 DOI: 10.1016/j.neubiorev.2018.11.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 10/24/2018] [Accepted: 11/30/2018] [Indexed: 01/17/2023]
Abstract
OBJECTIVE APOE status has been associated to affective symptoms in cognitively impaired subjects, with conflicting results. METHODS Databases CINAHL, Embase, PsychINFO and PubMed were searched for studies evaluating APOE genotype with affective symptoms in MCI and AD dementia. Symptoms were meta-analyzed separately and possible sources of heterogeneity were examined. RESULTS Fifty-three abstracts fulfilled the eligibility criteria. No association was found between the individual symptoms and APOE ε4 carriership or zygosity. For depression and anxiety, only pooled unadjusted estimates showed positive associations with between-study heterogeneity, which could be explained by variation in study design, setting and way of symptom assessment. CONCLUSIONS There is no evidence that APOE ε4 carriership or zygosity is associated with the presence of depression, anxiety, apathy, agitation, irritability or sleep disturbances in cognitively impaired subjects. Future research should shift its focus from this single polymorphism to a more integrated view of other biological factors.
Collapse
Affiliation(s)
- Leonie C P Banning
- Alzheimer Center Limburg, Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands.
| | - Inez H G B Ramakers
- Alzheimer Center Limburg, Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands.
| | - Kay Deckers
- Alzheimer Center Limburg, Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands.
| | - Frans R J Verhey
- Alzheimer Center Limburg, Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands.
| | - Pauline Aalten
- Alzheimer Center Limburg, Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands.
| |
Collapse
|
18
|
Halaris A. Neuroinflammation and neurotoxicity contribute to neuroprogression in neurological and psychiatric disorders. FUTURE NEUROLOGY 2018. [DOI: 10.2217/fnl-2017-0039] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The immune system and inflammatory processes contribute to brain-related pathologies in most, if not all, neurological and psychiatric disorders. Stress is a key factor in inducing immune system dysregulation in conjunction with genetic, epigenetic and environmental factors. Activation of the immune response can alter neurotransmission leading, among others, to serotonin deficiency, and increased production of neurotoxic substances contributing to disease progression. The concept of neuroprogression is gaining acceptance among clinicians and researches as it seeks to explain the mechanism(s) responsible for disease chronicity, recurrence and treatment resistance. Therefore, measurement of neuroinflammatory biomarkers along with assessment of neurotoxic metabolites, oxidative stress and neuroplasticity impairment, will ultimately be useful tools to predict and possibly prevent the development and progression of neuropsychiatric disorders as well as to identify the most efficacious treatments.
Collapse
Affiliation(s)
- Angelos Halaris
- Department of Psychiatry & Behavioral Neuroscience, Loyola University Stritch School of Medicine, Loyola University Medical Center, Maywood, IL 60153, USA
| |
Collapse
|
19
|
Cui D, Xu X. DNA Methyltransferases, DNA Methylation, and Age-Associated Cognitive Function. Int J Mol Sci 2018; 19:E1315. [PMID: 29710796 PMCID: PMC5983821 DOI: 10.3390/ijms19051315] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 04/20/2018] [Accepted: 04/20/2018] [Indexed: 12/16/2022] Open
Abstract
Ageing, a leading cause of the decline/deficits in human learning, memory, and cognitive abilities, is a major risk factor for age-associated neurodegenerative disorders such as Alzheimer’s disease. Emerging evidence suggests that epigenetics, an inheritable but reversible biochemical process, plays a crucial role in the pathogenesis of age-related neurological disorders. DNA methylation, the best-known epigenetic mark, has attracted most attention in this regard. DNA methyltransferases (DNMTs) are key enzymes in mediating the DNA methylation process, by which a methyl group is transferred, faithfully or anew, to genomic DNA sequences. Biologically, DNMTs are important for gene imprinting. Accumulating evidence suggests that DNMTs not only play critical roles, including gene imprinting and transcription regulation, in early development stages of the central nervous system (CNS), but also are indispensable in adult learning, memory, and cognition. Therefore, the impact of DNMTs and DNA methylation on age-associated cognitive functions and neurodegenerative diseases has emerged as a pivotal topic in the field. In this review, the effects of each DNMT on CNS development and healthy and pathological ageing are discussed.
Collapse
Affiliation(s)
- Di Cui
- Max Planck Institute for Biology of Ageing, 50931 Cologne, Germany.
| | - Xiangru Xu
- Max Planck Institute for Biology of Ageing, 50931 Cologne, Germany.
- Department of Anesthesiology, Yale University School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
20
|
|
21
|
Scholz CJ, Weber H, Jungwirth S, Danielczyk W, Reif A, Tragl KH, Fischer P, Riederer P, Deckert J, Grünblatt E. Explorative results from multistep screening for potential genetic risk loci of Alzheimer's disease in the longitudinal VITA study cohort. J Neural Transm (Vienna) 2017; 125:77-87. [PMID: 29027019 DOI: 10.1007/s00702-017-1796-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 10/04/2017] [Indexed: 01/21/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that preferentially affects individuals of advanced age. Heritability estimates for AD range between 60 and 80%, but only few genetic risk factors have been identified so far. In the present explorative study, we aimed at characterizing the genetic contribution to late-onset AD in participants of the Vienna Transdanube Aging (VITA) longitudinal birth cohort study in a two-step approach. First, we performed a genome-wide screen of pooled DNA samples (n = 588) to identify allele frequency differences between AD patients and non-AD individuals using life-time diagnoses made at the age of 80 (t = 60 months). This analysis suggested a high proportion of brain-expressed genes required for cell adhesion, cell signaling and cell morphogenesis, and also scored in known AD risk genes. In a second step, we confirmed associations using individual genotypes of top-ranked markers examining AD diagnoses as well as the dimensional scores: FULD and MMSE determined up to the age of 82.5 (t = 90 months). Taken together, our study proposes genes ANKS1B, ENST00000414107, LOC100505811, SLC22A14, QRFPR, ZDHHC8P1, ADAMTS3 and PPFIA1 as possible new candidates involved in the etiology of late-onset AD, with further research being needed to clarify their exact roles.
Collapse
Affiliation(s)
- Claus-Jürgen Scholz
- Core Unit Systems Medicine, University Hospital of Würzburg, Würzburg, Germany
| | - Heike Weber
- Department of Psychiatry, Psychosomatics, Psychotherapy, University Hospital Frankfurt/Main, Frankfurt/Main, Germany
- Center of Mental Health, Clinic and Policlinic of Psychiatry, Psychosomatics and Psychotherapy, University Hospital of Würzburg, Würzburg, Germany
| | - Susanne Jungwirth
- Ludwig Boltzmann Society, L. Boltzmann Institute of Aging Research, Vienna, Austria
- Department of Psychiatry, Social Medicine Center East- Donauspital, Vienna, Austria
| | - Walter Danielczyk
- Ludwig Boltzmann Society, L. Boltzmann Institute of Aging Research, Vienna, Austria
| | - Andreas Reif
- Department of Psychiatry, Psychosomatics, Psychotherapy, University Hospital Frankfurt/Main, Frankfurt/Main, Germany
| | - Karl-Heinz Tragl
- Ludwig Boltzmann Society, L. Boltzmann Institute of Aging Research, Vienna, Austria
| | - Peter Fischer
- Ludwig Boltzmann Society, L. Boltzmann Institute of Aging Research, Vienna, Austria
- Department of Psychiatry, Social Medicine Center East- Donauspital, Vienna, Austria
| | - Peter Riederer
- Center of Mental Health, Clinic and Policlinic of Psychiatry, Psychosomatics and Psychotherapy, University Hospital of Würzburg, Würzburg, Germany
| | - Jürgen Deckert
- Center of Mental Health, Clinic and Policlinic of Psychiatry, Psychosomatics and Psychotherapy, University Hospital of Würzburg, Würzburg, Germany
| | - Edna Grünblatt
- Center of Mental Health, Clinic and Policlinic of Psychiatry, Psychosomatics and Psychotherapy, University Hospital of Würzburg, Würzburg, Germany.
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital of Psychiatry Zurich, University of Zurich, Wagistrasse 12, Schlieren, 8952, Zurich, Switzerland.
| |
Collapse
|
22
|
Bakhtiari M, Panahi Y, Ameli J, Darvishi B. Protective effects of flavonoids against Alzheimer's disease-related neural dysfunctions. Biomed Pharmacother 2017. [PMID: 28641164 DOI: 10.1016/j.biopha.2017.06.010] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Senile ages of human life is mostly associated with developmental of several neurological complicated conditions including decreased cognition and reasoning, increased memory loss and impaired language performance. Alzheimer's disease (AD) is the most prevalent neural disorder associated with dementia, consisting of about 70% of dementia reported cases. Failure of currently approved chemical anti-AD therapeutic agents has once again brought up the idea of administering naturally occurring compounds as effective alternative and/or complementary regimens in AD treatment. Polyphenol structured neuroprotecting agents are group of biologically active compounds abundantly found in plants with significant protecting effects against neural injuries and degeneration. As a subclass of this family, Flavonoids are potent anti-oxidant, anti-inflammatory and signalling pathways modulatory agents. Phosphatidylinositol 3-kinase (PI3K)/AKT and mitogen activated protein kinase (MAPK) pathways are both affected by Flavonoids. Regulation of pro-survival transcription factors and induction of specific genes expression in hippocampus are other important anti AD therapeutic activities of Flavonoids. These agents are also capable of inhibiting specific enzymes involved in phosphorylation of tau proteins including β-secretases, cyclin dependent kinase 5 and glycogen synthase. Other significant anti AD effects of Flavonoids include neural rehabilitation and lost cognitive performance recovery. In this review, first we briefly describe the pathophysiology and important pathways involved in pathology of AD and then describe the most important mechanisms through which Flavonoids demonstrate their significant neuroprotective effects in AD therapy.
Collapse
Affiliation(s)
- Mahsa Bakhtiari
- Department of Clinical Pharmacy, Faculty of Pharmacy, Pharmaceutical Science Branch, Islamic Azad University, IAUPS, Tehran, Iran
| | - Yunes Panahi
- Chemical Injuries Research Center, Baghiatallah University of Medical Science, Tehran, Iran
| | - Javad Ameli
- Department of Neurology, Baghiatallah University of Medical Science, Tehran, Iran
| | - Behrad Darvishi
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran.
| |
Collapse
|
23
|
Fan Z, Brooks DJ, Okello A, Edison P. An early and late peak in microglial activation in Alzheimer's disease trajectory. Brain 2017; 140:792-803. [PMID: 28122877 DOI: 10.1093/brain/aww349] [Citation(s) in RCA: 190] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 11/18/2016] [Indexed: 12/11/2022] Open
Abstract
Amyloid-β deposition, neuroinflammation and tau tangle formation all play a significant role in Alzheimer's disease. We hypothesized that there is microglial activation early on in Alzheimer's disease trajectory, where in the initial phase, microglia may be trying to repair the damage, while later on in the disease these microglia could be ineffective and produce proinflammatory cytokines leading to progressive neuronal damage. In this longitudinal study, we have evaluated the temporal profile of microglial activation and its relationship between fibrillar amyloid load at baseline and follow-up in subjects with mild cognitive impairment, and this was compared with subjects with Alzheimer's disease. Thirty subjects (eight mild cognitive impairment, eight Alzheimer's disease and 14 controls) aged between 54 and 77 years underwent 11C-(R)PK11195, 11C-PIB positron emission tomography and magnetic resonance imaging scans. Patients were followed-up after 14 ± 4 months. Region of interest and Statistical Parametric Mapping analysis were used to determine longitudinal alterations. Single subject analysis was performed to evaluate the individualized pathological changes over time. Correlations between levels of microglial activation and amyloid deposition at a voxel level were assessed using Biological Parametric Mapping. We demonstrated that both baseline and follow-up microglial activation in the mild cognitive impairment cohort compared to controls were increased by 41% and 21%, respectively. There was a longitudinal reduction of 18% in microglial activation in mild cognitive impairment cohort over 14 months, which was associated with a mild elevation in fibrillar amyloid load. Cortical clusters of microglial activation and amyloid deposition spatially overlapped in the subjects with mild cognitive impairment. Baseline microglial activation was increased by 36% in Alzheimer's disease subjects compared with controls. Longitudinally, Alzheimer's disease subjects showed an increase in microglial activation. In conclusion, this is one of the first longitudinal positron emission tomography studies evaluating longitudinal changes in microglial activation in mild cognitive impairment and Alzheimer's disease subjects. We found there is an initial longitudinal reduction in microglial activation in subjects with mild cognitive impairment, while subjects with Alzheimer's disease showed an increase in microglial activation. This could reflect that activated microglia in mild cognitive impairment initially may adopt a protective activation phenotype, which later change to a cidal pro-inflammatory phenotype as disease progresses and amyloid clearance fails. Thus, we speculate that there might be two peaks of microglial activation in the Alzheimer's disease trajectory; an early protective peak and a later pro-inflammatory peak. If so, anti-microglial agents targeting the pro-inflammatory phenotype would be most beneficial in the later stages of the disease.
Collapse
Affiliation(s)
- Zhen Fan
- Neurology Imaging Unit, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| | - David J Brooks
- Neurology Imaging Unit, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK.,Department of Nuclear Medicine, Institute of Medicine, Aarhus University, Denmark
| | - Aren Okello
- Neurology Imaging Unit, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| | - Paul Edison
- Neurology Imaging Unit, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| |
Collapse
|
24
|
Dos Santos LR, Pimassoni LHS, Sena GGS, Camporez D, Belcavello L, Trancozo M, Morelato RL, Errera FIV, Bueno MRP, de Paula F. Validating GWAS Variants from Microglial Genes Implicated in Alzheimer's Disease. J Mol Neurosci 2017; 62:215-221. [PMID: 28477215 DOI: 10.1007/s12031-017-0928-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 04/26/2017] [Indexed: 11/25/2022]
Abstract
Late-onset Alzheimer's disease (LOAD) is a multifactorial neurodegenerative disorder that corresponds to most Alzheimer's disease (AD) cases. Inflammation is frequently related to AD, whereas microglial cells are the major phagocytes in the brain and mediate the removal of Aβ peptides. Microglial cell dsyregulation might contribute to the formation of amyloid plaques, a hallmark of AD. Genome-wide association studies have reported genetic loci associated with the inflammatory pathway involved in AD. Among them, rs3865444 CD33, rs3764650 ABCA7, rs6656401 CR1, and rs610932 MS4A6A variants in microglial genes are associated with LOAD. These variants are proposed to participate in the clearance of Aβ peptides. However, their association with LOAD was not validated in all case-control studies. Thus, the present work aimed to assess the involvement of CD33 (rs3865444), ABCA7 (rs3764650), CR1 (rs6656401), and MS4A6A (rs610932) with LOAD in a sample from southeastern Brazil. The genotype frequencies were assessed in 79 AD patients and 145 healthy elders matched for sex and age. We found that rs3865444 CD33 acts as a protective factor against LOAD. These results support a role for the inflammatory pathway in LOAD.
Collapse
Affiliation(s)
- Lígia Ramos Dos Santos
- Laboratório de Genética Humana e Molecular, Departamento de Ciências Biológicas, Centro de CiênciasHumanas e Naturais, Universidade Federal do Espírito Santo, Av. Fernando Ferrari, 514, Prédio BárbaraWeinberg, Sala 102, Vitória, ES, 29075-910, Brazil. .,Programa de Pós-Graduação em Biotecnologia, Universidade Federal do Espírito Santo, Vitória, ES, Brazil.
| | | | - Geralda Gillian Silva Sena
- Laboratório de Genética Humana e Molecular, Departamento de Ciências Biológicas, Centro de CiênciasHumanas e Naturais, Universidade Federal do Espírito Santo, Av. Fernando Ferrari, 514, Prédio BárbaraWeinberg, Sala 102, Vitória, ES, 29075-910, Brazil.,Departamento de Educação Integrada em Saúde, Centro de Ciências da Saúde, Universidade Federal do Espírito Santo, Vitória, ES, Brazil
| | - Daniela Camporez
- Laboratório de Genética Humana e Molecular, Departamento de Ciências Biológicas, Centro de CiênciasHumanas e Naturais, Universidade Federal do Espírito Santo, Av. Fernando Ferrari, 514, Prédio BárbaraWeinberg, Sala 102, Vitória, ES, 29075-910, Brazil.,Programa de Pós-Graduação em Biotecnologia, Universidade Federal do Espírito Santo, Vitória, ES, Brazil
| | - Luciano Belcavello
- Laboratório de Genética Humana e Molecular, Departamento de Ciências Biológicas, Centro de CiênciasHumanas e Naturais, Universidade Federal do Espírito Santo, Av. Fernando Ferrari, 514, Prédio BárbaraWeinberg, Sala 102, Vitória, ES, 29075-910, Brazil
| | - Maíra Trancozo
- Laboratório de Genética Humana e Molecular, Departamento de Ciências Biológicas, Centro de CiênciasHumanas e Naturais, Universidade Federal do Espírito Santo, Av. Fernando Ferrari, 514, Prédio BárbaraWeinberg, Sala 102, Vitória, ES, 29075-910, Brazil.,Programa de Pós-Graduação em Biotecnologia, Universidade Federal do Espírito Santo, Vitória, ES, Brazil
| | - Renato Lírio Morelato
- Escola Superior de Ciências da Santa Casa de Misericórdia de Vitória, Vitória, ES, Brazil.,Hospital da Santa Casa de Misericórdia de Vitória, Escola Superior de Ciências da Santa Casa de Misericórdia de Vitória, Vitória, ES, Brazil
| | - Flavia Imbroisi Valle Errera
- Programa de Pós-Graduação em Biotecnologia, Universidade Federal do Espírito Santo, Vitória, ES, Brazil.,Escola Superior de Ciências da Santa Casa de Misericórdia de Vitória, Vitória, ES, Brazil
| | | | - Flavia de Paula
- Laboratório de Genética Humana e Molecular, Departamento de Ciências Biológicas, Centro de CiênciasHumanas e Naturais, Universidade Federal do Espírito Santo, Av. Fernando Ferrari, 514, Prédio BárbaraWeinberg, Sala 102, Vitória, ES, 29075-910, Brazil.,Programa de Pós-Graduação em Biotecnologia, Universidade Federal do Espírito Santo, Vitória, ES, Brazil
| |
Collapse
|
25
|
Epigenetics in Alzheimer's Disease: Perspective of DNA Methylation. Mol Neurobiol 2017; 55:1026-1044. [PMID: 28092081 DOI: 10.1007/s12035-016-0357-6] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2016] [Accepted: 12/28/2016] [Indexed: 12/12/2022]
Abstract
Research over the years has shown that causes of Alzheimer's disease are not well understood, but over the past years, the involvement of epigenetic mechanisms in the developing memory formation either under pathological or physiological conditions has become clear. The term epigenetics represents the heredity of changes in phenotype that are independent of altered DNA sequences. Different studies validated that cytosine methylation of genomic DNA decreases with age in different tissues of mammals, and therefore, the role of epigenetic factors in developing neurological disorders in aging has been under focus. In this review, we summarized and reviewed the involvement of different epigenetic mechanisms especially the DNA methylation in Alzheimer's disease (AD), late-onset Alzheimer's disease (LOAD), familial Alzheimer's disease (FAD), and autosomal dominant Alzheimer's disease (ADAD). Down to the minutest of details, we tried to discuss the methylation patterns like mitochondrial DNA methylation and ribosomal DNA (rDNA) methylation. Additionally, we mentioned some therapeutic approaches related to epigenetics, which could provide a potential cure for AD. Moreover, we reviewed some recent studies that validate DNA methylation as a potential biomarker and its role in AD. We hope that this review will provide new insights into the understanding of AD pathogenesis from the epigenetic perspective especially from the perspective of DNA methylation.
Collapse
|
26
|
Kepp KP. Ten Challenges of the Amyloid Hypothesis of Alzheimer’s Disease. J Alzheimers Dis 2016; 55:447-457. [DOI: 10.3233/jad-160550] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
27
|
Burke SL, Maramaldi P, Cadet T, Kukull W. Associations between depression, sleep disturbance, and apolipoprotein E in the development of Alzheimer's disease: dementia. Int Psychogeriatr 2016; 28:1409-24. [PMID: 27020605 PMCID: PMC4963299 DOI: 10.1017/s1041610216000405] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is a neurodegenerative brain disease that causes cognitive impairment and dementia. Within the US, AD is the most common form of dementia in the elderly, affecting 1 in 10 people over the age of 65. Sleep disturbance has been called a "public health epidemic" and, like depression, is a prodromal symptom of AD but may also contribute to the risk of developing AD. It was hypothesized that sleep disturbance, depression, and the apolipoprotein E (APOE) genotype increase the likelihood of AD. METHODS Utilizing data from the National Alzheimer's Coordinating Center, information from evaluations of 11,453 cognitively asymptomatic participants was analyzed. Survival analysis was used to explore the independent relationships between depression, sleep disturbance, and APOE genotypes with eventual AD diagnosis. Cox proportional hazard models were utilized to explore the main effects and synergistic effects of psychosocial factors as moderated by APOE genotypes. RESULTS This study reinforced the association between APOE and AD. The hazard of developing AD was eight times higher for those with recent depression and the Ɛ4 homozygote (HR = 8.15 [3.70-17.95]). Among Ɛ4 carriers with clinician-verified depression, the hazard was ten times that of the reference group (HR = 10.11 [4.43-23.09]). The hazard for Ɛ4 carriers reporting sleep disturbance was almost 7 times greater than the reference group (HR = 6.79 [2.38-19.37]). CONCLUSION Findings suggest that sleep disturbance, depression, and APOE Ɛ4 genotype are associated with AD during follow-up evaluations among a group of initially cognitively asymptomatic participants. This study contributes to the literature base exploring an increased hazard or risk of AD due to potential modifiable risk factors as well as genetic biomarkers, such as APOE.
Collapse
Affiliation(s)
- Shanna L Burke
- Florida International University,Robert Stempel College of Public Health and Social Work,School of Social Work,Miami,FL,USA
| | | | - Tamara Cadet
- Simmons College School of Social Work,Boston,MA,USA
| | - Walter Kukull
- National Alzheimer's Coordinating Center (NACC),University of Washington School of Public Health,Department of Epidemiology,Seattle,WA,USA
| |
Collapse
|
28
|
Genetic Association of CHAT rs3810950 and rs2177369 Polymorphisms with the Risk of Alzheimer's Disease: A Meta-Analysis. BIOMED RESEARCH INTERNATIONAL 2016; 2016:9418163. [PMID: 27597977 PMCID: PMC5002460 DOI: 10.1155/2016/9418163] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 06/03/2016] [Accepted: 07/03/2016] [Indexed: 12/28/2022]
Abstract
Choline acetyltransferase (CHAT) rs3810950 and rs2177369 polymorphisms have been implicated in susceptibility to Alzheimer's disease (AD). Due to the inconsistent results from previous studies, a meta-analysis was performed to estimate the association between these polymorphisms and AD risk more precisely. Pooled results of our meta-analysis indicated CHAT rs2177369 polymorphism was correlated with decreasing AD risk in one of five genetic models (dominant: OR = 0.77, 95% CI: 0.62–0.96), while rs3810950 mutant was associated with AD development in three models (allelic: OR = 1.18, 95% CI: 1.01–1.37, homozygous: OR = 1.63, 95% CI: 1.09–2.42, and recessive: OR = 1.65, 95% CI: 1.20–2.26). In subgroup analysis by ethnicity, the association between CHAT rs3810950 polymorphism and AD risk was just found in the recessive model (OR = 1.47, 95% CI: 1.05–2.07) among Caucasians, while four genetic models (allelic: OR = 1.23, 95% CI: 1.01–1.48; homozygous: OR = 2.24, 95% CI: 1.48–3.39; dominant: OR = 1.21, 95% CI: 1.06–1.40; and recessive: OR = 2.18, 95% CI: 1.45–3.29) assumed this association in Asians. In conclusion, our meta-analysis indicated CHAT rs2177369 polymorphism might play a protective role in AD, while rs3810950 variant was a risk factor for AD but its single heterozygous mutations might not influence susceptibility to AD.
Collapse
|
29
|
Kepp KP. Alzheimer's disease due to loss of function: A new synthesis of the available data. Prog Neurobiol 2016; 143:36-60. [PMID: 27327400 DOI: 10.1016/j.pneurobio.2016.06.004] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 06/10/2016] [Accepted: 06/11/2016] [Indexed: 12/11/2022]
Abstract
Alzheimer's Disease (AD) is a highly complex disease involving a broad range of clinical, cellular, and biochemical manifestations that are currently not understood in combination. This has led to many views of AD, e.g. the amyloid, tau, presenilin, oxidative stress, and metal hypotheses. The amyloid hypothesis has dominated the field with its assumption that buildup of pathogenic β-amyloid (Aβ) peptide causes disease. This paradigm has been criticized, yet most data suggest that Aβ plays a key role in the disease. Here, a new loss-of-function hypothesis is synthesized that accounts for the anomalies of the amyloid hypothesis, e.g. the curious pathogenicity of the Aβ42/Aβ40 ratio, the loss of Aβ caused by presenilin mutation, the mixed phenotypes of APP mutations, the poor clinical-biochemical correlations for genetic variant carriers, and the failure of Aβ reducing drugs. The amyloid-loss view accounts for recent findings on the structure and chemical features of Aβ variants and their coupling to human patient data. The lost normal function of APP/Aβ is argued to be metal transport across neuronal membranes, a view with no apparent anomalies and substantially more explanatory power than the gain-of-function amyloid hypothesis. In the loss-of-function scenario, the central event of Aβ aggregation is interpreted as a loss of soluble, functional monomer Aβ rather than toxic overload of oligomers. Accordingly, new research models and treatment strategies should focus on remediation of the functional amyloid balance, rather than strict containment of Aβ, which, for reasons rationalized in this review, has failed clinically.
Collapse
Affiliation(s)
- Kasper P Kepp
- Technical University of Denmark, DTU Chemistry, DK-2800 Kongens Lyngby, Denmark.
| |
Collapse
|
30
|
Laury-Kleintop LD, Mulgrew JR, Heletz I, Nedelcoviciu RA, Chang MY, Harris DM, Koch WJ, Schneider MD, Muller AJ, Prendergast GC. Cardiac-specific disruption of Bin1 in mice enables a model of stress- and age-associated dilated cardiomyopathy. J Cell Biochem 2016; 116:2541-51. [PMID: 25939245 DOI: 10.1002/jcb.25198] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 04/14/2015] [Indexed: 12/21/2022]
Abstract
Non-compensated dilated cardiomyopathy (DCM) leading to death from heart failure is rising rapidly in developed countries due to aging demographics, and there is a need for informative preclinical models to guide the development of effective therapeutic strategies to prevent or delay disease onset. In this study, we describe a novel model of heart failure based on cardiac-specific deletion of the prototypical mammalian BAR adapter-encoding gene Bin1, a modifier of age-associated disease. Bin1 deletion during embryonic development causes hypertrophic cardiomyopathy and neonatal lethality, but there is little information on how Bin1 affects cardiac function in adult animals. Here we report that cardiomyocyte-specific loss of Bin1 causes age-associated dilated cardiomyopathy (DCM) beginning by 8-10 months of age. Echocardiographic analysis showed that Bin1 loss caused a 45% reduction in ejection fraction during aging. Younger animals rapidly developed DCM if cardiac pressure overload was created by transverse aortic constriction. Heterozygotes exhibited an intermediate phenotype indicating Bin1 is haplo-insufficient to sustain normal heart function. Bin1 loss increased left ventricle (LV) volume and diameter during aging, but it did not alter LV volume or diameter in hearts from heterozygous mice nor did it affect LV mass. Bin1 loss increased interstitial fibrosis and mislocalization of the voltage-dependent calcium channel Cav 1.2, and the lipid raft scaffold protein caveolin-3, which normally complexes with Bin1 and Cav 1.2 in cardiomyocyte membranes. Our findings show how cardiac deficiency in Bin1 function causes age- and stress-associated heart failure, and they establish a new preclinical model of this terminal cardiac disease.
Collapse
Affiliation(s)
| | | | - Ido Heletz
- Lankenau Medical Center, Wynnewood, Pennsylvania
| | | | - Mee Young Chang
- Lankenau Institute for Medical Research, Wynnewood, Pennsylvania
| | - David M Harris
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | - Walter J Koch
- Center for Translational Medicine, Temple University Medical School, Philadelphia, Pennsylvania
| | - Michael D Schneider
- National Heart and Lung Institute, British Heart Foundation Centre of Research Excellence, Faculty of Medicine, Imperial College London, London, UK
| | | | - George C Prendergast
- Lankenau Institute for Medical Research, Wynnewood, Pennsylvania.,Department of Pathology, Anatomy and Cell Biology, Sidney Kimmel Medical School and Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| |
Collapse
|
31
|
Burke SL, Maramaldi P, Cadet T, Kukull W. Neuropsychiatric symptoms and Apolipoprotein E: Associations with eventual Alzheimer's disease development. Arch Gerontol Geriatr 2016; 65:231-8. [PMID: 27111252 DOI: 10.1016/j.archger.2016.04.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 04/10/2016] [Accepted: 04/11/2016] [Indexed: 11/17/2022]
Abstract
OBJECTIVE Alzheimer's disease (AD) is the result of neurodegeneration, which manifests clinically as deficits in memory, thinking, and behavior. It was hypothesized that neuropsychiatric symptoms and the apolipoprotein E genotype increase the likelihood of Alzheimer's disease development. METHODS Utilizing data from the National Alzheimer's Coordinating Center, information from evaluations of 11,453 cognitively intact participants was analyzed. Survival analysis was used to explore relationships between individual neuropsychiatric symptoms as determined by the Neuropsychiatric Inventory Questionnaire, apolipoprotein E, and eventual AD diagnosis. Cox proportional hazard models were utilized to explore the main effects and synergistic (additive and multiplicative) interactions. RESULTS This study provided evidence for an increased hazard of developing AD among participants with any of the symptoms assessed by the NPI-Q. The hazard of developing AD was almost thirteen times higher for ε4 carriers with delusions and eleven times greater for those with apathy and disinhibition. Statistically significant hazards (p>0.001) were also realized by ε4 carriers with hallucinations; agitation; depression; anxiety; elation; apathy; irritability; and motor, sleep, and appetite disturbances. CONCLUSIONS Findings suggest that neuropsychiatric symptoms are associated with eventual AD diagnosis among a group of cognitively asymptomatic participants at baseline. Many studies begin with a group of participants already impacted by AD diagnosis. The longitudinal analysis of a group of participants who, at baseline, demonstrated no observable signs of AD was a strength of this study. This investigation contributes to the literature exploring an increased hazard of AD due to potential modifiable risk factors and genetic biomarkers such as apolipoprotein E.
Collapse
Affiliation(s)
- Shanna L Burke
- Florida International University, Robert Stempel College of Public Health and Social Work, School of Social Work, 11200 S.W. 8th Street, AHC5 564, Miami, FL 33199, USA.
| | - Peter Maramaldi
- Simmons College School of Social Work, Boston, MA 02115-5820, USA; Oral Health Policy and Epidemiology, Harvard School of Dental Medicine, USA; Department of Social and Behavioral Sciences, Harvard T.H. Chan School of Public Health, USA.
| | - Tamara Cadet
- Simmons College School of Social Work, Boston, MA 02115-5820, USA; Oral Health Policy and Epidemiology, Harvard School of Dental Medicine, USA.
| | - Walter Kukull
- National Alzheimer's Coordinating Center (NACC) University of Washington, Department of Epidemiology Box 357236, Seattle, WA 98195-7236, USA.
| |
Collapse
|
32
|
Somavarapu AK, Kepp KP. Loss of stability and hydrophobicity of presenilin 1 mutations causing Alzheimer's disease. J Neurochem 2016; 137:101-11. [PMID: 26756738 DOI: 10.1111/jnc.13535] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 12/23/2015] [Accepted: 12/29/2015] [Indexed: 12/15/2022]
Abstract
Nearly 200 mutations in the gene coding for presenilin 1 (PSEN1) cause early-onset Alzheimer's disease, yet the molecular mechanism remains obscure. As a meta-analysis, we compiled available clinical and biochemical data for PSEN1 variants and correlated these to chemical properties of the mutants. We found statistically significant relationships between relative Aβ42 levels and clinical age of onset. We then computed chemical properties of the mutants from a variety of computational chemistry tools. Relative Aβ42 levels correlated significantly (95% confidence or more from p-values of linear regression) with loss of hydrophobicity for four different regression analyses (squared correlation coefficient of linear regression R(2) of 0.41-0.53) and with increased polarity (R(2) = 0.47, 0.59) and loss of protein stability (R(2) = 0.39, 0.63) for two independent data sets. Age of onset of patients carrying PSEN1 variants correlated with increased polarity (R(2) = 0.49, 0.40) and loss of stability (R(2) = 0.75, 0.44) of the protein for both data sets. These relations suggest that mutants impair the membrane-associated structural integrity of presenilin by reducing hydrophobic membrane association and overall protein stability. This explains why the many mutations that spread out across the protein and far from the catalytic aspartates can cause disease. The identified molecular determinants of clinical age of symptom onset may be relevant to future presenilin-modulating therapies specifically directed towards increasing the structural integrity and packing of the protein. Close to 200 mutations in presenilin 1 (PSEN1) cause Alzheimer's disease, but the biochemical relating these to disease remains debated. The chemical properties of PSEN1 variants were computed and correlated against clinical age of symptom onset. Loss of stability and hydrophobicity and gain of polarity relate to disease onset, suggesting that mutants impair the membrane structure of PSEN1 and that therapies should increase PSEN1 structural integrity.
Collapse
Affiliation(s)
| | - Kasper P Kepp
- Technical University of Denmark, DTU Chemistry, Kongens Lyngby, Denmark
| |
Collapse
|
33
|
Liu W, Li G, Hölscher C, Li L. Neuroprotective effects of geniposide on Alzheimer's disease pathology. Rev Neurosci 2015; 26:371-83. [PMID: 25879319 DOI: 10.1515/revneuro-2015-0005] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Accepted: 02/25/2015] [Indexed: 12/20/2022]
Abstract
A growing body of evidence has linked two of the most common aged-related diseases: type 2 diabetes mellitus (T2DM) and Alzheimer's disease (AD). It has led to the notion that drugs developed for the treatment of T2DM may be beneficial in modifying the pathophysiology of AD. As a receptor agonist of glucagon-like peptide-1 (GLP-1R), which is a newer drug class to treat T2DM, geniposide shows clear effects in inhibiting pathological processes underlying AD, such as promoting neurite outgrowth. In the present article, we review the possible molecular mechanisms of geniposide to protect the brain from pathologic damages underlying AD: reducing amyloid plaques, inhibiting τ phosphorylation, preventing memory impairment and loss of synapses, reducing oxidative stress and the chronic inflammatory response, and promoting neurite outgrowth via the GLP-1R signaling pathway. In summary, the Chinese herb geniposide shows great promise as a novel treatment for AD.
Collapse
|
34
|
Pistollato F, Cavanaugh SE, Chandrasekera PC. A Human-Based Integrated Framework forAlzheimer’s Disease Research. J Alzheimers Dis 2015; 47:857-68. [DOI: 10.3233/jad-150281] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
35
|
Halaris A. Neurological disorders, depression and inflammation: is there a common link? FUTURE NEUROLOGY 2015. [DOI: 10.2217/fnl.15.18] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
To understand the origin of co-morbidity between neurological disorders and depressive illness, a multifactorial model is in order. Diverse approaches have been undertaken to elucidate the co-morbidity. Of these, the concept that inflammatory processes contribute to brain-related pathologies has been gaining traction. Inflammatory processes have been identified in most, if not all, neurological conditions. Similarly, major depressive disorder has been associated with a chronic proinflammatory status. Activation of the immune response can alter neurotransmission leading, among others, to serotonin deficiency, and increased production of neurotoxic substances contributing to primary disease progression. Therefore, inflammatory factors might serve as biomarkers to predict and ultimately prevent the development and progression of neuropsychiatric disorders as well as to identify the most efficacious treatments.
Collapse
Affiliation(s)
- Angelos Halaris
- Professor of Psychiatry, Department of Psychiatry & Behavioral Sciences, Loyola University Chicago Stritch School of Medicine, Loyola University Medical Center, 2160 South First Avenue, Maywood, IL 60153, USA
| |
Collapse
|
36
|
Luo J, Li S, Qin X, Peng Q, Liu Y, Yang S, Qin X, Xiong Y, Zeng Z. Association of the NQO1 C609T polymorphism with Alzheimer's disease in Chinese populations: a meta-analysis. Int J Neurosci 2015; 126:199-204. [DOI: 10.3109/00207454.2015.1004573] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
37
|
Sulfhydryl-mediated redox signaling in inflammation: role in neurodegenerative diseases. Arch Toxicol 2015; 89:1439-67. [DOI: 10.1007/s00204-015-1496-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 02/25/2015] [Indexed: 01/05/2023]
|
38
|
Jiang T, Tan MS, Tan L, Yu JT. Application of next-generation sequencing technologies in Neurology. ANNALS OF TRANSLATIONAL MEDICINE 2015; 2:125. [PMID: 25568878 DOI: 10.3978/j.issn.2305-5839.2014.11.11] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/20/2013] [Accepted: 03/01/2013] [Indexed: 12/11/2022]
Abstract
Genetic risk factors that underlie many rare and common neurological diseases remain poorly understood because of the multi-factorial and heterogeneous nature of these disorders. Although genome-wide association studies (GWAS) have successfully uncovered numerous susceptibility genes for these diseases, odds ratios associated with risk alleles are generally low and account for only a small proportion of estimated heritability. These results implicated that there are rare (present in <5% of the population) but not causative variants exist in the pathogenesis of these diseases, which usually have large effect size and cannot be captured by GWAS. With the decreasing cost of next-generation sequencing (NGS) technologies, whole-genome sequencing (WGS) and whole-exome sequencing (WES) have enabled the rapid identification of rare variants with large effect size, which made huge progress in understanding the basis of many Mendelian neurological conditions as well as complex neurological diseases. In this article, recent NGS-based studies that aimed to investigate genetic causes for neurological diseases, including Alzheimer's disease, Parkinson's disease, epilepsy, multiple sclerosis, stroke, amyotrophic lateral sclerosis and spinocerebellar ataxias, have been reviewed. In addition, we also discuss the future directions of NGS applications in this article.
Collapse
Affiliation(s)
- Teng Jiang
- 1 Department of Neurology, Qingdao Municipal Hospital, Nanjing Medical University, Qingdao 266071, China ; 2 College of Medicine and Pharmaceutics, Ocean University of China, Qingdao 266071, China ; 3 Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao 266071, China
| | - Meng-Shan Tan
- 1 Department of Neurology, Qingdao Municipal Hospital, Nanjing Medical University, Qingdao 266071, China ; 2 College of Medicine and Pharmaceutics, Ocean University of China, Qingdao 266071, China ; 3 Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao 266071, China
| | - Lan Tan
- 1 Department of Neurology, Qingdao Municipal Hospital, Nanjing Medical University, Qingdao 266071, China ; 2 College of Medicine and Pharmaceutics, Ocean University of China, Qingdao 266071, China ; 3 Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao 266071, China
| | - Jin-Tai Yu
- 1 Department of Neurology, Qingdao Municipal Hospital, Nanjing Medical University, Qingdao 266071, China ; 2 College of Medicine and Pharmaceutics, Ocean University of China, Qingdao 266071, China ; 3 Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao 266071, China
| |
Collapse
|
39
|
Wang J, Tang J, Lai M, Zhang H. 5-Hydroxymethylcytosine and disease. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2014; 762:167-75. [PMID: 25475423 DOI: 10.1016/j.mrrev.2014.09.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 08/27/2014] [Accepted: 09/29/2014] [Indexed: 11/27/2022]
Abstract
Epigenetics is the study of inherited changes in phenotype or gene expression that do not alter DNA sequence. Recently, scientists have focused their attention on 5-hydroxymethylcytosine (5hmC), a newly discovered epigenetic marker, also known as sixth DNA base of the genome. In mammals, this novel epigenetic marker is derived from 5-methylcytosine (5mC) in a process catalyzed by ten-eleven translocation (TET) enzymes. Although 5hmC has only been subjected to study for a short while, a great deal of data has been accumulated regarding its generation, distribution, demethylation, function, and disease implications. All this information suggested that 5hmC acts not only as an intermediate in the DNA demethylation process but also as an independent epigenetic marker, playing an important role in the regulation of gene expression. This review focuses on recent progress in the study of the relationship between 5hmC and human diseases, such as cancer and Rett syndrome (RTT).
Collapse
Affiliation(s)
- Jingyu Wang
- Department of Pathology, School of Medicine, Zhejiang University, Zhejiang, PR China; Department of Pathology, The First Hospital of Jiaxing, Zhejiang, PR China
| | - Jinlong Tang
- Department of Pathology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, PR China
| | - Maode Lai
- Department of Pathology, School of Medicine, Zhejiang University, Zhejiang, PR China; Key Laboratory of Disease Proteomics of Zhejiang Province, PR China.
| | - Honghe Zhang
- Department of Pathology, School of Medicine, Zhejiang University, Zhejiang, PR China; Key Laboratory of Disease Proteomics of Zhejiang Province, PR China.
| |
Collapse
|
40
|
Cissé M, Checler F. Eph receptors: new players in Alzheimer's disease pathogenesis. Neurobiol Dis 2014; 73:137-49. [PMID: 25193466 DOI: 10.1016/j.nbd.2014.08.028] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 08/01/2014] [Accepted: 08/22/2014] [Indexed: 12/23/2022] Open
Abstract
Alzheimer's disease (AD) is devastating and leads to permanent losses of memory and other cognitive functions. Although recent genetic evidences strongly argue for a causative role of Aβ in AD onset and progression (Jonsson et al., 2012), its role in AD etiology remains a matter of debate. However, even if not the sole culprit or pathological trigger, genetic and anatomical evidences in conjunction with numerous pharmacological studies, suggest that Aβ peptides, at least contribute to the disease. How Aβ contributes to memory loss remains largely unknown. Soluble Aβ species referred to as Aβ oligomers have been shown to be neurotoxic and induce network failure and cognitive deficits in animal models of the disease. In recent years, several proteins were described as potential Aβ oligomers receptors, amongst which are the receptor tyrosine kinases of Eph family. These receptors together with their natural ligands referred to as ephrins have been involved in a plethora of physiological and pathological processes, including embryonic neurogenesis, learning and memory, diabetes, cancers and anxiety. Here we review recent discoveries on Eph receptors-mediated protection against Aβ oligomers neurotoxicity as well as their potential as therapeutic targets in AD pathogenesis.
Collapse
Affiliation(s)
- Moustapha Cissé
- Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275 CNRS/UNS, "Labex Distalz", 660 route des Lucioles, 06560, Sophia-Antipolis, Valbonne, France..
| | - Frédéric Checler
- Institut de Pharmacologie Moléculaire et Cellulaire, UMR7275 CNRS/UNS, "Labex Distalz", 660 route des Lucioles, 06560, Sophia-Antipolis, Valbonne, France..
| |
Collapse
|
41
|
Abstract
Positive predictions were made in the aftermath of the development of induced pluripotent stem cell technology for the use of patient-specific iPSCs to model neurological diseases, including dementia. Here, we review the current state of the field and explore how close we are to the goal of in vitro models that capture all aspects of the cell and molecular biology of dementia.
Collapse
Affiliation(s)
- Frederick J Livesey
- Gurdon Institute, Cambridge Stem Cell Institute & Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge CB2 1QN, UK
| |
Collapse
|
42
|
Liu L, Chan C. The role of inflammasome in Alzheimer's disease. Ageing Res Rev 2014; 15:6-15. [PMID: 24561250 PMCID: PMC4029867 DOI: 10.1016/j.arr.2013.12.007] [Citation(s) in RCA: 138] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Revised: 12/12/2013] [Accepted: 12/23/2013] [Indexed: 12/18/2022]
Abstract
Alzheimer's disease (AD) is a chronic, progressive and irreversible neurodegenerative disease with clinical characteristics of memory loss, dementia and cognitive impairment. Although the pathophysiologic mechanism is not fully understood, inflammation has been shown to play a critical role in the pathogenesis of AD. Inflammation in the central nervous system (CNS) is characterized by the activation of glial cells and release of proinflammatory cytokines and chemokines. Accumulating evidence demonstrates that inflammasomes, which cleave precursors of interleukin-1β (IL-1β) and IL-18 to generate their active forms, play an important role in the inflammatory response in the CNS and in AD pathogenesis. Therefore, modulating inflammasome complex assembly and activation could be a potential strategy for suppressing inflammation in the CNS. This review aims to provide insight into the role of inflammasomes in the CNS, with respect to the pathogenesis of AD, and may provide possible clues for devising novel therapeutic strategies.
Collapse
Affiliation(s)
- Li Liu
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI 48824, United States; Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Christina Chan
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824, United States; Department of Chemical Engineering and Materials Science, Michigan State University, East Lansing, MI 48824, United States.
| |
Collapse
|
43
|
Richens JL, Morgan K, O'Shea P. Reverse engineering of Alzheimer's disease based on biomarker pathways analysis. Neurobiol Aging 2014; 35:2029-38. [PMID: 24684789 DOI: 10.1016/j.neurobiolaging.2014.02.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Revised: 02/18/2014] [Accepted: 02/26/2014] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) poses an increasingly profound problem to society, yet progress toward a genuine understanding of the disease remains worryingly slow. Perhaps, the most outstanding problem with the biology of AD is the question of its mechanistic origins, that is, it remains unclear wherein the molecular failures occur that underlie the disease. We demonstrate how molecular biomarkers could help define the nature of AD in terms of the early biochemical events that correlate with disease progression. We use a novel panel of biomolecules that appears in cerebrospinal fluid of AD patients. As changes in the relative abundance of these molecular markers are associated with progression to AD from mild cognitive impairment, we make the assumption that by tracking their origins we can identify the biochemical conditions that predispose their presence and consequently cause the onset of AD. We couple these protein markers with an analysis of a series of genetic factors and together this hypothesis essentially allows us to redefine AD in terms of the molecular pathways that underlie the disease.
Collapse
Affiliation(s)
- Joanna L Richens
- Cell Biophysics Group, School of Life Sciences, Faculty of Medicine & Health Sciences, University Park, University of Nottingham, Nottingham, UK
| | - Kevin Morgan
- Humans Genetics Research Group, School of Life Sciences, Faculty of Medicine & Health Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, UK
| | - Paul O'Shea
- Cell Biophysics Group, School of Life Sciences, Faculty of Medicine & Health Sciences, University Park, University of Nottingham, Nottingham, UK.
| |
Collapse
|
44
|
Is encephalopathic brain genetically more prone to dementia? Open Med (Wars) 2013. [DOI: 10.2478/s11536-013-0229-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
45
|
Schutte DL. Genetic testing and Alzheimer's disease: implications for psychiatric-mental health nursing. J Psychosoc Nurs Ment Health Serv 2013; 51:14-8. [PMID: 24124693 DOI: 10.3928/02793695-20131010-01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Alzheimer's disease (AD), the most common cause of irreversible dementia, continues to grow in prevalence as well as public health impact. Extensive research into the genetic etiology of AD has yielded knowledge of some genetic factors that are causative and other genetic factors that increase risk for disease. Consequently, the possibility of genetic testing in individuals with or at risk for AD is a question that nurses may be asked. Psychiatric-mental health (PMH) professionals are in key positions to influence the care of individuals who are considering the effect of genetic information on their health care decisions. Whether by working within interdisciplinary genetic counseling teams to provide direct specialty services or by developing skills to identify and refer individuals at risk for or concerned about their risk for AD, PMH nurses can play an important role in the health care of individuals and families experiencing AD.
Collapse
|
46
|
Abstract
The initial preclinical phase of Alzheimer disease (AD), which has no symptoms, is followed by a phase whereby cognitive impairment, but no functional impairment is present (mild cognitive impairment), after which comes the third phase of dementia. Diagnosis of AD has primarily been one of exclusion of all other causes of reversible and irreversible dementia. Overlapping clinical presentations of diseases causing neurodegeneration, however, create challenges for accurate diagnosis. Algorithms are provided for the most current guidelines. Use of clinical magnetic resonance and PET imaging modalities increase the specificity of diagnosis, and several new promising experimental approaches are being developed.
Collapse
Affiliation(s)
- Robert M Cohen
- Neuroscience Program, Graduate Division of Biological and Medical Sciences, Department of Psychiatry and Behavioral Neuroscience, Emory University, 101 Woodruff Circle, Suite WMB4003, Atlanta, GA 30322, USA.
| |
Collapse
|
47
|
Fung WLA, Naylor MG, Bennett DA, Lange C, Blacker D. Principal components methods for narrow-sense heritability in the analysis of multidimensional longitudinal cognitive phenotypes. Am J Med Genet B Neuropsychiatr Genet 2013; 162B:770-8. [PMID: 23650207 PMCID: PMC3758806 DOI: 10.1002/ajmg.b.32151] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Accepted: 02/27/2013] [Indexed: 11/06/2022]
Abstract
BACKGROUND Genetic association studies of longitudinal cognitive phenotypes are an alternate approach to discovering genetic risk factors for Alzheimer's disease (AD). However, the standard linear mixed model approach is limited in the face of multidimensional longitudinal data and multiple genotypes. In this setting, the principal components of heritability (PCH) approach may increase efficiency by deriving a linear combination of phenotypes to maximize the heritability attributable to a particular genetic locus. The current study investigated the performance of two PCH methods, the Principal Components of Heritability Association Test (PCHAT) and C2BAT, in detecting association of the known AD susceptibility allele APOE-ϵ4 with cognitive function at baseline and decline in cognition over time. METHODS PCHAT, C2BAT, and standard linear mixed models were used to test for association between APOE-ϵ4 allele and performance on 19 neuropsychological tests using subjects without dementia at baseline from the Religious Orders Study (ROS) (n = 693) and Memory and Aging Project (MAP) (n = 778). Analyses were conducted across the three methods for three nested phenotype definitions (all 19 measures, executive function and episodic memory measures, and episodic memory only), and for baseline data only versus longitudinal change. RESULTS In all cases, APOE-ϵ4 was significantly associated with baseline level of and change over time in cognitive function, and PCHAT and C2BAT yielded evidence of association comparable to or stronger than conventional methods. CONCLUSION PCHAT, C2BAT, and other PCH methods may have utility for genetic association studies of multidimensional cognitive and other phenotypes by maximizing genetic information while limiting multiple comparisons.
Collapse
Affiliation(s)
- Wai Lun Alan Fung
- Department of Epidemiology, Harvard School of Public Health, Boston, MA, USA
,Department of Psychiatry, University of Toronto Faculty of Medicine, Toronto, ON, Canada
| | - Melissa G. Naylor
- Department of Biostatistics, Harvard School of Public Health, Boston, MA, USA
,Pritzker School of Medicine, University of Chicago, Chicago, IL, USA
| | - David A. Bennett
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Christoph Lange
- Department of Biostatistics, Harvard School of Public Health, Boston, MA, USA
,Channing Laboratory, Brigham and Women’s Hospital, Boston, MA, USA
| | - Deborah Blacker
- Department of Epidemiology, Harvard School of Public Health, Boston, MA, USA
,Department of Psychiatry, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| |
Collapse
|
48
|
Wang KS, Xu N, Wang L, Aragon L, Ciubuc R, Arana TB, Mao C, Petty L, Briones D, Su BB, Luo X, Camarillo C, Escamilla MA, Xu C. NRG3 gene is associated with the risk and age at onset of Alzheimer disease. J Neural Transm (Vienna) 2013; 121:183-92. [PMID: 24061483 DOI: 10.1007/s00702-013-1091-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Accepted: 09/03/2013] [Indexed: 10/26/2022]
Abstract
The Neuregulin 3 (NRG3) gene at 10q22-q24 has been implicated in multiple psychiatric traits such as cognitive impairment. We therefore hypothesized that NRG3 gene polymorphisms may play a role in Alzheimer disease (AD). This present study explored the association of NRG3 with the age at onset (AAO) of AD and the risk of developing AD. Secondary data analysis of 257 single-nucleotide polymorphisms (SNPs) in NRG3 gene was performed in 806 Alzheimer's disease patients and 782 controls using logistic regression and linear regression analyses. Eight SNPs were associated with the risk of AD (p < 0.05), while linear regression analysis showed 33 SNPs associated with the AAO of AD (p < 0.05). Two-SNP haplotype analyses based on UNPHASED revealed that the G-C haplotype from rs17685233 and rs17101017 was significantly associated with AD (p = 0.0031) and the A-G haplotype from rs504522 and rs474018 as well as the A-G haplotype from rs504522 and rs2483295 were more significantly associated with the AAO of AD (p = 6.72 × 10(-5)). Using an independent family-based sample, we found one SNP rs11192423 associated with AAO both in the case-control sample (p = 0.0155) and in the family sample (p = 0.0166). In addition, we observed nominally significant associations with AD and AAO for several flanking SNPs (p < 0.05). This is the first study demonstrating that genetic variants in the NRG3 gene play a role in AD. Our results also revealed that SNPs in the NRG3 genes were more strongly associated with AAO of AD.
Collapse
Affiliation(s)
- Ke-Sheng Wang
- Department of Biostatistics and Epidemiology, College of Public Health, East Tennessee State University, PO Box 70259, Lamb Hall, Johnson City, TN, 37614-1700, USA,
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Rongve A, Årsland D, Graff C. [Alzheimer's disease and genetics]. TIDSSKRIFT FOR DEN NORSKE LEGEFORENING 2013; 133:1449-52. [PMID: 23929292 DOI: 10.4045/tidsskr.12.0873] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
BACKGROUND Alzheimer's disease is the most frequent cause of dementia. Recent knowledge reveals several new risk genes. We wish to summarise the knowledge of genetic factors related to Alzheimer's disease. METHOD This article is based on findings in Alzgene, a database that summarises genetic association studies in Alzheimer's disease, a literature search in PubMed and the authors' own experience in dementia research. RESULTS Several mutations of the genes APP, PSEN1 and PSEN2 are described. These cause around half of all cases of the rare early onset autosomal dominant form of Alzheimer's disease. Heritability, or how much of the development of the disease in an individual that is explained by genetics, is between 60 and 80% in the most common late onset form of Alzheimer's disease. APOE ε4 is the most robust risk gene for the development of this form of the disease, but recently ten new genes that increase the risk of developing Alzheimer's disease were identified by applying genome-wide association studies. These genes code for proteins that are central in the metabolism of cholesterol, activation of the immune system and synaptic cell membrane processes. INTERPRETATION New hypotheses on the disease mechanisms for Alzheimer's disease are suggested based on the identification of new risk genes. These hypotheses partly replace and partly supplement the previously dominant amyloid pathway hypothesis. The new risk genes point to the potential for new biomarkers for specific disease processes and to possible new targets for future disease modifying therapies.
Collapse
Affiliation(s)
- Arvid Rongve
- Seksjon for alderspsykiatri, Klinikk for psykisk helsevern, Helse-Fonna, Haugesund, Norway.
| | | | | |
Collapse
|
50
|
Binding and repressive activities of apolipoprotein E3 and E4 isoforms on the human ApoD promoter. Mol Neurobiol 2013; 48:669-80. [PMID: 23715769 PMCID: PMC7090986 DOI: 10.1007/s12035-013-8456-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Accepted: 04/09/2013] [Indexed: 11/04/2022]
Abstract
Apolipoprotein D (ApoD) gene expression is increased in several neurological disorders such as Alzheimer’s disease (AD) and multiple sclerosis. We previously showed that transgenic mice that overexpress human ApoD show a better resistance against paraquat or OC43 coronavirus-induced neurodegeneration. Here, we identified several nuclear factors from the cortex of control and OC43-infected mice which bind a fragment of the proximal ApoD promoter in vitro. Of interest, we detected apolipoprotein E (ApoE). Human ApoE consists of three isoforms (E2, E3, and E4) with the E4 and E2 alleles representing a greater and a lower risk for developping AD, respectively. Our results show that ApoE is located in the nucleus and on the ApoD promoter in human hepatic and glioblastoma cells lines. Furthermore, overexpression of ApoE3 and ApoE4 isoforms but not ApoE2 significantly inhibited the ApoD promoter activity in U87 cells (E3/E3 genotype) cultured under normal or different stress conditions while ApoE knock-down by siRNA had a converse effect. Consistent with these results, we also demonstrated by ChIP assay that E3 and E4 isoforms, but not E2, bind the ApoD promoter. Moreover, using the Allen Brain Atlas in situ hybridization database, we observed an inverse correlation between ApoD and ApoE mRNA expression during development and in several regions of the mouse brain, notably in the cortex, hippocampus, plexus choroid, and cerebellum. This negative correlation was also observed for cortex layers IV–VI based on a new Transcriptomic Atlas of the Mouse Neocortical Layers. These findings reveal a new function for ApoE by regulating ApoD gene expression.
Collapse
|