51
|
Kaeberlein M, Galvan V. Rapamycin and Alzheimer's disease: Time for a clinical trial? Sci Transl Med 2019; 11:eaar4289. [PMID: 30674654 PMCID: PMC6762017 DOI: 10.1126/scitranslmed.aar4289] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 06/08/2018] [Indexed: 12/16/2022]
Abstract
The drug rapamycin has beneficial effects in a number of animal models of neurodegeneration and aging including mouse models of Alzheimer's disease. Despite its compelling preclinical record, no clinical trials have tested rapamycin or other mTOR inhibitors in patients with Alzheimer's disease. We argue that such clinical trials should be undertaken.
Collapse
Affiliation(s)
- Matt Kaeberlein
- Department of Pathology, University of Washington, Seattle, WA 98045, USA.
| | - Veronica Galvan
- Department of Cellular and Integrative Physiology, Barshop Institute for Longevity and Aging Studies and Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health San Antonio, San Antonio, TX 78229, USA.
- Department of Veterans Affairs, South Texas Veterans Health Care System and Geriatric Research Education and Clinical Center, San Antonio, TX 78229, USA
| |
Collapse
|
52
|
Morris G, Berk M, Maes M, Puri BK. Could Alzheimer's Disease Originate in the Periphery and If So How So? Mol Neurobiol 2019; 56:406-434. [PMID: 29705945 PMCID: PMC6372984 DOI: 10.1007/s12035-018-1092-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Accepted: 04/17/2018] [Indexed: 12/11/2022]
Abstract
The classical amyloid cascade model for Alzheimer's disease (AD) has been challenged by several findings. Here, an alternative molecular neurobiological model is proposed. It is shown that the presence of the APOE ε4 allele, altered miRNA expression and epigenetic dysregulation in the promoter region and exon 1 of TREM2, as well as ANK1 hypermethylation and altered levels of histone post-translational methylation leading to increased transcription of TNFA, could variously explain increased levels of peripheral and central inflammation found in AD. In particular, as a result of increased activity of triggering receptor expressed on myeloid cells 2 (TREM-2), the presence of the apolipoprotein E4 (ApoE4) isoform, and changes in ANK1 expression, with subsequent changes in miR-486 leading to altered levels of protein kinase B (Akt), mechanistic (previously mammalian) target of rapamycin (mTOR) and signal transducer and activator of transcription 3 (STAT3), all of which play major roles in microglial activation, proliferation and survival, there is activation of microglia, leading to the subsequent (further) production of cytokines, chemokines, nitric oxide, prostaglandins, reactive oxygen species, inducible nitric oxide synthase and cyclooxygenase-2, and other mediators of inflammation and neurotoxicity. These changes are associated with the development of amyloid and tau pathology, mitochondrial dysfunction (including impaired activity of the electron transport chain, depleted basal mitochondrial potential and oxidative damage to key tricarboxylic acid enzymes), synaptic dysfunction, altered glycogen synthase kinase-3 (GSK-3) activity, mTOR activation, impairment of autophagy, compromised ubiquitin-proteasome system, iron dyshomeostasis, changes in APP translation, amyloid plaque formation, tau hyperphosphorylation and neurofibrillary tangle formation.
Collapse
Affiliation(s)
- Gerwyn Morris
- IMPACT Strategic Research Centre, School of Medicine, Barwon Health, Deakin University, P.O. Box 291, Geelong, Victoria, Australia
| | - Michael Berk
- IMPACT Strategic Research Centre, School of Medicine, Barwon Health, Deakin University, P.O. Box 291, Geelong, Victoria, Australia
- Department of Psychiatry, Level 1 North, Main Block, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia
- Florey Institute for Neuroscience and Mental Health, Kenneth Myer Building, University of Melbourne, 30 Royal Parade, Parkville, Victoria, Australia
- Orygen, The National Centre of Excellence in Youth Mental Health, 35 Poplar Rd, Parkville, Victoria, Australia
| | - Michael Maes
- IMPACT Strategic Research Centre, School of Medicine, Barwon Health, Deakin University, P.O. Box 291, Geelong, Victoria, Australia
- Department of Psychiatry, Chulalongkorn University, Bangkok, Thailand
| | - Basant K Puri
- Department of Medicine, Hammersmith Hospital, Imperial College London, London, UK.
| |
Collapse
|
53
|
Hernández F, Llorens-Martín M, Bolós M, Pérez M, Cuadros R, Pallas-Bazarra N, Zabala JC, Avila J. New Beginnings in Alzheimer’s Disease: The Most Prevalent Tauopathy. J Alzheimers Dis 2018; 64:S529-S534. [DOI: 10.3233/jad-179916] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Félix Hernández
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
- CIBERNED, Madrid, Spain
| | - María Llorens-Martín
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
- CIBERNED, Madrid, Spain
- Department of Molecular Biology, Faculty of Science, Universidad Autonoma de Madrid, Spain
| | - Marta Bolós
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
- CIBERNED, Madrid, Spain
| | - Mar Pérez
- Departamento de Anatomía Histología y Neurociencia, Facultad de Medicina UAM, Madrid, Spain
| | - Raquel Cuadros
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
- CIBERNED, Madrid, Spain
| | - Noemí Pallas-Bazarra
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
- CIBERNED, Madrid, Spain
| | - Juan C. Zabala
- Departamento de Biología Molecular, Facultad de Medicina, Universidad de Cantabria, Santander, Spain
| | - Jesús Avila
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain
- CIBERNED, Madrid, Spain
| |
Collapse
|
54
|
Braidy N, Essa MM, Poljak A, Selvaraju S, Al-Adawi S, Manivasagm T, Thenmozhi AJ, Ooi L, Sachdev P, Guillemin GJ. Consumption of pomegranates improves synaptic function in a transgenic mice model of Alzheimer's disease. Oncotarget 2018; 7:64589-64604. [PMID: 27486879 PMCID: PMC5323101 DOI: 10.18632/oncotarget.10905] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 06/17/2016] [Indexed: 01/18/2023] Open
Abstract
Alzheimer's Disease (AD) is a progressive neurodegenerative disorder characterized by extracellular plaques containing abnormal Amyloid Beta (Aβ) aggregates, intracellular neurofibrillary tangles containing hyperphosphorylated tau protein, microglia-dominated neuroinflammation, and impairments in synaptic plasticity underlying cognitive deficits. Therapeutic strategies for the treatment of AD are currently limited. In this study, we investigated the effects of dietary supplementation of 4% pomegranate extract to a standard chow diet on neuroinflammation, and synaptic plasticity in APPsw/Tg2576 mice brain. Treatment with a custom mixed diet (pellets) containing 4% pomegranate for 15 months ameliorated the loss of synaptic structure proteins, namely PSD-95, Munc18-1, and SNAP25, synaptophysin, phosphorylation of Calcium/Calmodulin Dependent Protein Kinase IIα (p-CaMKIIα/ CaMKIIα), and phosphorylation of Cyclic AMP-Response Element Binding Protein (pCREB/CREB), inhibited neuroinflammatory activity, and enhanced autophagy, and activation of the phophoinositide-3-kinase-Akt-mammalian target of rapamycin signaling pathway. These neuroprotective effects were associated with reduced β-site cleavage of Amyloid Precursor Protein in APPsw/Tg2576 mice. Therefore, long-term supplementation with pomegranates can attenuate AD pathology by reducing inflammation, and altering APP-dependent processes.
Collapse
Affiliation(s)
- Nady Braidy
- Centre for Healthy Brain Ageing, School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Musthafa Mohamed Essa
- Department of Food Science and Nutrition, College of Agricultural and Marine Sciences, Sultan Qaboos University, Al Khoudh, Oman.,Ageing and Dementia Research Group, Sultan Qaboos University, Al Khoudh, Oman
| | - Anne Poljak
- Centre for Healthy Brain Ageing, School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, Australia.,College of Medicine and Health Sciences, Sultan Qaboos University, Al Khoudh, Oman
| | - Subash Selvaraju
- Department of Food Science and Nutrition, College of Agricultural and Marine Sciences, Sultan Qaboos University, Al Khoudh, Oman.,Ageing and Dementia Research Group, Sultan Qaboos University, Al Khoudh, Oman
| | - Samir Al-Adawi
- Department of Food Science and Nutrition, College of Agricultural and Marine Sciences, Sultan Qaboos University, Al Khoudh, Oman.,College of Medicine and Health Sciences, Sultan Qaboos University, Al Khoudh, Oman
| | | | | | - Lezanne Ooi
- Illawarra Health and Medical Research Institute, University of Wollongong, NSW, Australia
| | - Perminder Sachdev
- Centre for Healthy Brain Ageing, School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, Australia.,Neuropsychiatric Institute, The Prince of Wales Hospital, Sydney, Australia
| | - Gilles J Guillemin
- Neuroinflammation Group, MND and Neurodegenerative Diseases Research Centre, Macquarie University, NSW, Australia
| |
Collapse
|
55
|
Lee HK, Kwon B, Lemere CA, de la Monte S, Itamura K, Ha AY, Querfurth HW. mTORC2 (Rictor) in Alzheimer's Disease and Reversal of Amyloid-β Expression-Induced Insulin Resistance and Toxicity in Rat Primary Cortical Neurons. J Alzheimers Dis 2018; 56:1015-1036. [PMID: 28035937 DOI: 10.3233/jad-161029] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Mammalian target of rapamycin complex 1 (mTORC1), a nutrient sensor and central controller of cell growth and proliferation, is altered in various models of Alzheimer's disease (AD). Even less studied or understood in AD is mammalian target of rapamycin complex 2 (mTORC2) that influences cellular metabolism, in part through the regulations of Akt/PKB and SGK. Dysregulation of insulin/PI3K/Akt signaling is another important feature of AD pathogenesis. We found that both total mTORC1 and C2 protein levels and individual C1 and C2 enzymatic activities were decreased in human AD brain samples. In two rodent AD models, mTORC1 and C2 activities were also decreased. In a neuronal culture model of AD characterized by accumulation of cellular amyloid-β (Aβ)42, mTORC1 activity was reduced. Autophagic vesicles and markers were correspondingly increased and new protein synthesis was inhibited, consistent with mTORC1 hypofunction. Interestingly, mTORC2 activity in neural culture seemed resistant to the effects of intracellular amyloid. In various cell lines, Aβ expression provoked insulin resistance, characterized by inhibition of stimulated Akt phosphorylation, and an increase in negative mTORC1 regular, p-AMPK, itself a nutrient sensor. Rapamycin decreased phospho-mTOR and to lesser degree p-Rictor. This further suppression of mTORC1 activity protected cells from Aβ-induced toxicity and insulin resistance. More striking, Rictor over-expression fully reversed the Aβ-effects on primary neuronal cultures. Finally, using in vitro assay, Rictor protein addition completely overcame oligomeric Aβ-induced inhibition of the PDK-Akt activation step. We conclude that striking a new balance by restoring mTORC2 abundance and/or inhibition of mTORC1 has therapeutic potential in AD.
Collapse
Affiliation(s)
- Han-Kyu Lee
- Department of Neurology, Rhode Island Hospital and Brown University Warren Alpert Medical School, Providence, RI, USA
| | - Bumsup Kwon
- Department of Neurology, Rhode Island Hospital and Brown University Warren Alpert Medical School, Providence, RI, USA
| | - Cynthia A Lemere
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Suzanne de la Monte
- Department of Pathology, Rhode Island Hospital and Brown University Warren Alpert Medical School, Providence, RI, USA
| | - Kyohei Itamura
- Department of Neurology, Rhode Island Hospital and Brown University Warren Alpert Medical School, Providence, RI, USA
| | - Austin Y Ha
- Department of Neurology, Rhode Island Hospital and Brown University Warren Alpert Medical School, Providence, RI, USA
| | - Henry W Querfurth
- Department of Neurology, Rhode Island Hospital and Brown University Warren Alpert Medical School, Providence, RI, USA
| |
Collapse
|
56
|
Increased Levels of Rictor Prevent Mutant Huntingtin-Induced Neuronal Degeneration. Mol Neurobiol 2018; 55:7728-7742. [DOI: 10.1007/s12035-018-0956-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 02/06/2018] [Indexed: 01/21/2023]
|
57
|
Uddin MS, Stachowiak A, Mamun AA, Tzvetkov NT, Takeda S, Atanasov AG, Bergantin LB, Abdel-Daim MM, Stankiewicz AM. Autophagy and Alzheimer's Disease: From Molecular Mechanisms to Therapeutic Implications. Front Aging Neurosci 2018; 10:04. [PMID: 29441009 PMCID: PMC5797541 DOI: 10.3389/fnagi.2018.00004] [Citation(s) in RCA: 264] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 01/08/2018] [Indexed: 01/07/2023] Open
Abstract
Alzheimer’s disease (AD) is the most common cause of progressive dementia in the elderly. It is characterized by a progressive and irreversible loss of cognitive abilities and formation of senile plaques, composed mainly of amyloid β (Aβ), and neurofibrillary tangles (NFTs), composed of tau protein, in the hippocampus and cortex of afflicted humans. In brains of AD patients the metabolism of Aβ is dysregulated, which leads to the accumulation and aggregation of Aβ. Metabolism of Aβ and tau proteins is crucially influenced by autophagy. Autophagy is a lysosome-dependent, homeostatic process, in which organelles and proteins are degraded and recycled into energy. Thus, dysfunction of autophagy is suggested to lead to the accretion of noxious proteins in the AD brain. In the present review, we describe the process of autophagy and its importance in AD. Additionally, we discuss mechanisms and genes linking autophagy and AD, i.e., the mTOR pathway, neuroinflammation, endocannabinoid system, ATG7, BCL2, BECN1, CDK5, CLU, CTSD, FOXO1, GFAP, ITPR1, MAPT, PSEN1, SNCA, UBQLN1, and UCHL1. We also present pharmacological agents acting via modulation of autophagy that may show promise in AD therapy. This review updates our knowledge on autophagy mechanisms proposing novel therapeutic targets for the treatment of AD.
Collapse
Affiliation(s)
- Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh
| | - Anna Stachowiak
- Department of Experimental Embryology, Institute of Genetics and Animal Breeding, Polish Academy of Sciences, Magdalenka, Poland
| | | | - Nikolay T Tzvetkov
- Department of Molecular Biology and Biochemical Pharmacology, Institute of Molecular Biology "Roumen Tsanev", Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Shinya Takeda
- Department of Clinical Psychology, Tottori University Graduate School of Medical Sciences, Tottori, Japan
| | - Atanas G Atanasov
- Department of Molecular Biology, Institute of Genetics and Animal Breeding, Polish Academy of Sciences, Magdalenka, Poland.,Department of Pharmacognosy, University of Vienna, Vienna, Austria
| | - Leandro B Bergantin
- Department of Pharmacology, Federal University of São Paulo, São Paulo, Brazil
| | - Mohamed M Abdel-Daim
- Department of Pharmacology, Suez Canal University, Ismailia, Egypt.,Department of Ophthalmology and Micro-technology, Yokohama City University, Yokohama, Japan
| | - Adrian M Stankiewicz
- Department of Molecular Biology, Institute of Genetics and Animal Breeding, Polish Academy of Sciences, Magdalenka, Poland
| |
Collapse
|
58
|
Ntsapi C, Lumkwana D, Swart C, du Toit A, Loos B. New Insights Into Autophagy Dysfunction Related to Amyloid Beta Toxicity and Neuropathology in Alzheimer's Disease. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 336:321-361. [DOI: 10.1016/bs.ircmb.2017.07.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
59
|
Morgan AH, Rees DJ, Andrews ZB, Davies JS. Ghrelin mediated neuroprotection - A possible therapy for Parkinson's disease? Neuropharmacology 2017; 136:317-326. [PMID: 29277488 DOI: 10.1016/j.neuropharm.2017.12.027] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 12/15/2017] [Accepted: 12/17/2017] [Indexed: 12/31/2022]
Abstract
Parkinson's disease is a common age-related neurodegenerative disorder affecting 10 million people worldwide, but the mechanisms underlying its pathogenesis are still unclear. The disease is characterised by dopamine nerve cell loss in the mid-brain and intra-cellular accumulation of α-synuclein that results in motor and non-motor dysfunction. In this review, we discuss the neuroprotective effects of the stomach hormone, ghrelin, in models of Parkinson's disease. Recent findings suggest that it may modulate mitochondrial function and autophagic clearance of impaired organelle in response to changes in cellular energy balance. We consider the putative cellular mechanisms underlying ghrelin-action and the possible role of ghrelin mimetics in slowing or preventing Parkinson's disease progression. This article is part of the Special Issue entitled 'Metabolic Impairment as Risk Factors for Neurodegenerative Disorders.'
Collapse
Affiliation(s)
- Alwena H Morgan
- Molecular Neurobiology, Institute of Life Science, Medical School, Swansea University, UK
| | - Daniel J Rees
- Molecular Neurobiology, Institute of Life Science, Medical School, Swansea University, UK
| | - Zane B Andrews
- Biomedicine Discovery Institute & Department of Physiology, Monash University, Melbourne, Victoria, Australia
| | - Jeffrey S Davies
- Molecular Neurobiology, Institute of Life Science, Medical School, Swansea University, UK.
| |
Collapse
|
60
|
Morgan AH, Andrews ZB, Davies JS. Less is more: Caloric regulation of neurogenesis and adult brain function. J Neuroendocrinol 2017; 29. [PMID: 28771924 DOI: 10.1111/jne.12512] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 07/20/2017] [Accepted: 07/26/2017] [Indexed: 12/11/2022]
Abstract
Calorie intake is essential for regulating normal physiological processes and is fundamental to maintaining life. Indeed, both extremes of calorie intake result in increased morbidity and mortality. In this review, we discuss the effect of calorie intake on adult brain function, with an emphasis on the beneficial effects of mild calorie restriction. Recent findings relating to the regenerative and protective effects of the gastrointestinal hormone, ghrelin, suggest that it may underlie the beneficial effects of calorie restriction. We discuss the putative cellular mechanisms underlying the action of ghrelin and their possible role in supporting healthy brain ageing.
Collapse
Affiliation(s)
- A H Morgan
- Molecular Neurobiology, Institute of Life Science, School of Medicine, Swansea University, Swansea, UK
| | - Z B Andrews
- Department of Physiology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - J S Davies
- Molecular Neurobiology, Institute of Life Science, School of Medicine, Swansea University, Swansea, UK
| |
Collapse
|
61
|
Lee KH, Lee SJ, Lee HJ, Choi GE, Jung YH, Kim DI, Gabr AA, Ryu JM, Han HJ. Amyloid β1-42 (Aβ1-42) Induces the CDK2-Mediated Phosphorylation of Tau through the Activation of the mTORC1 Signaling Pathway While Promoting Neuronal Cell Death. Front Mol Neurosci 2017; 10:229. [PMID: 28790888 PMCID: PMC5522873 DOI: 10.3389/fnmol.2017.00229] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 07/05/2017] [Indexed: 01/08/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder, characterized by cognitive impairment and memory loss. Amyloid β1-42 (Aβ) and hyper-phosphorylation of microtubule-associated protein tau have been considered as major histological features in AD. However, the mechanism of how Aβ induces the hyper-phosphorylation of tau remains to be clarified. In the present study, we investigated the underlying cellular mechanisms of Aβ with regard to the cell cycle regulatory protein-mediated phosphorylation of tau in promoting neuronal cell death. The oligomer Aβ (5 μM) significantly increased the level of caspase 3 cleavage and has the ability to induce cytotoxicity in human neuroblastoma SK-N-MC cells. Aβ induced the degree of extracellular calcium influx via the L-type channel to facilitate the production of reactive oxygen species (ROS). Aβ signaling through ROS production is uniquely mediated by the activation of PI3K/Akt, which is in turn required for mammalian target of rapamycin complex 1 (mTORC1) phosphorylation. mTORC1 activated by Aβ further increased the phosphorylation of eukaryotic translation initiation factor 4E (eIF4E), a binding protein (4E-BP1) and p70S6K1 to stimulate the HIF1α synthesis responsible for the induction of cyclinD1/cyclin-dependent kinase 4 (CDK4) and cyclinE/CDK2, whereas it significantly attenuated the activation of autophagy. Aβ distinctively induced the CDK2-mediated phosphorylation of tau, which is responsible for microtubule destabilization in promoting neuronal apoptosis. In mouse hippocampal primary neurons, the apoptotic cell death induced by Aβ is highly susceptible to the mTORC1 signaling pathway. These results demonstrate that Aβ efficiently stimulates the mTORC1 signaling pathway to facilitate HIF1α synthesis and autophagy inhibition to promote the expression of cell cycle regulatory proteins, during which CDK2 uniquely stimulates tau phosphorylation for microtubule destabilization-mediated neuronal apoptosis.
Collapse
Affiliation(s)
- Ki Hoon Lee
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 PLUS Program for Creative Veterinary Science Research Center, Seoul National UniversitySeoul, South Korea
| | - Sei-Jung Lee
- Department of Pharmaceutical Engineering, Daegu Haany UniversityGyeongsan, South Korea
| | - Hyun Jik Lee
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 PLUS Program for Creative Veterinary Science Research Center, Seoul National UniversitySeoul, South Korea
| | - Gee Euhn Choi
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 PLUS Program for Creative Veterinary Science Research Center, Seoul National UniversitySeoul, South Korea
| | - Young Hyun Jung
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 PLUS Program for Creative Veterinary Science Research Center, Seoul National UniversitySeoul, South Korea
| | - Dah Ihm Kim
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 PLUS Program for Creative Veterinary Science Research Center, Seoul National UniversitySeoul, South Korea
| | - Amr Ahmed Gabr
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 PLUS Program for Creative Veterinary Science Research Center, Seoul National UniversitySeoul, South Korea.,Department of Physiology, Faculty of Veterinary Medicine, Cairo UniversityGiza, Egypt
| | - Jung Min Ryu
- Department of Veterinary Physiology, College of Veterinary Medicine, Chonnam National UniversityGwangju, South Korea
| | - Ho Jae Han
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 PLUS Program for Creative Veterinary Science Research Center, Seoul National UniversitySeoul, South Korea
| |
Collapse
|
62
|
García-Escudero V, Gargini R, Martín-Maestro P, García E, García-Escudero R, Avila J. Tau mRNA 3'UTR-to-CDS ratio is increased in Alzheimer disease. Neurosci Lett 2017; 655:101-108. [PMID: 28689927 DOI: 10.1016/j.neulet.2017.07.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 07/01/2017] [Accepted: 07/05/2017] [Indexed: 11/30/2022]
Abstract
Neurons frequently show an imbalance in expression of the 3' untranslated region (3'UTR) relative to the coding DNA sequence (CDS) region of mature messenger RNAs (mRNA). The ratio varies among different cells or parts of the brain. The Map2 protein levels per cell depend on the 3'UTR-to-CDS ratio rather than the total mRNA amount, which suggests powerful regulation of protein expression by 3'UTR sequences. Here we found that MAPT (the microtubule-associated protein tau gene) 3'UTR levels are particularly high with respect to other genes; indeed, the 3'UTR-to-CDS ratio of MAPT is balanced in healthy brain in mouse and human. The tau protein accumulates in Alzheimer diseased brain. We nonetheless observed that the levels of RNA encoding MAPT/tau were diminished in these patients' brains. To explain this apparently contradictory result, we studied MAPT mRNA stoichiometry in coding and non-coding regions, and found that the 3'UTR-to-CDS ratio was higher in the hippocampus of Alzheimer disease patients, with higher tau protein but lower total mRNA levels. Our data indicate that changes in the 3'UTR-to-CDS ratio have a regulatory role in the disease. Future research should thus consider not only mRNA levels, but also the ratios between coding and non-coding regions.
Collapse
Affiliation(s)
- Vega García-Escudero
- Centro de Biología Molecular Severo Ochoa (UAM-CSIC), Nicolás Cabrera 1, Cantoblanco, 28049 Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Valderrebollo 5, 28031 Madrid, Spain
| | - Ricardo Gargini
- Centro de Biología Molecular Severo Ochoa (UAM-CSIC), Nicolás Cabrera 1, Cantoblanco, 28049 Madrid, Spain
| | - Patricia Martín-Maestro
- Centro de Biología Molecular Severo Ochoa (UAM-CSIC), Nicolás Cabrera 1, Cantoblanco, 28049 Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Valderrebollo 5, 28031 Madrid, Spain
| | - Esther García
- Centro de Biología Molecular Severo Ochoa (UAM-CSIC), Nicolás Cabrera 1, Cantoblanco, 28049 Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Valderrebollo 5, 28031 Madrid, Spain
| | - Ramón García-Escudero
- Molecular Oncology Unit, CIEMAT, Complutense 40, 28040 Madrid, Spain; Biomedical Research Institute I+12, University Hospital 12 de Octubre, Avenida de Córdoba s/n, 28041 Madrid, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Monforte de Lemos 3-5, 28029 Madrid, Spain
| | - Jesús Avila
- Centro de Biología Molecular Severo Ochoa (UAM-CSIC), Nicolás Cabrera 1, Cantoblanco, 28049 Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Valderrebollo 5, 28031 Madrid, Spain.
| |
Collapse
|
63
|
Shafei MA, Harris M, Conway ME. Divergent Metabolic Regulation of Autophagy and mTORC1-Early Events in Alzheimer's Disease? Front Aging Neurosci 2017. [PMID: 28626421 PMCID: PMC5454035 DOI: 10.3389/fnagi.2017.00173] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive disease associated with the production and deposition of amyloid β-peptide (Aβ) aggregates and neurofibrillary tangles, which lead to synaptic and neuronal damage. Reduced autophagic flux has been widely associated with the accumulation of autophagic vacuoles (AV), which has been proposed to contribute to aggregate build-up observed in AD. As such, targeting autophagy regulation has received wide review, where an understanding as to how this mechanism can be controlled will be important to neuronal health. The mammalian target of rapamycin complex 1 (mTORC1), which was found to be hyperactive in AD brain, regulates autophagy and is considered to be mechanistically important to aberrant autophagy in AD. Hormones and nutrients such as insulin and leucine, respectively, positively regulate mTORC1 activation and are largely considered to inhibit autophagy. However, in AD brain there is a dysregulation of nutrient metabolism, linked to insulin resistance, where a role for insulin treatment to improve cognition has been proposed. Recent studies have highlighted that mitochondrial proteins such as glutamate dehydrogenase and the human branched chain aminotransferase protein, through metabolism of leucine and glutamate, differentially regulate mTORC1 and autophagy. As the levels of the hBCAT proteins are significantly increased in AD brain relative to aged-matched controls, we discuss how these metabolic pathways offer new potential therapeutic targets. In this review article, we highlight the core regulation of autophagy through mTORC1, focusing on how insulin and leucine will be important to consider in particular with respect to our understanding of nutrient load and AD pathogenesis.
Collapse
Affiliation(s)
- Mai A Shafei
- Department of Applied Science, The University of the West of EnglandBristol, United Kingdom
| | - Matthew Harris
- Department of Applied Science, The University of the West of EnglandBristol, United Kingdom
| | - Myra E Conway
- Department of Applied Science, The University of the West of EnglandBristol, United Kingdom
| |
Collapse
|
64
|
Melnik BC, Schmitz G. Milk's Role as an Epigenetic Regulator in Health and Disease. Diseases 2017; 5:diseases5010012. [PMID: 28933365 PMCID: PMC5456335 DOI: 10.3390/diseases5010012] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Revised: 03/02/2017] [Accepted: 03/07/2017] [Indexed: 12/16/2022] Open
Abstract
It is the intention of this review to characterize milk's role as an epigenetic regulator in health and disease. Based on translational research, we identify milk as a major epigenetic modulator of gene expression of the milk recipient. Milk is presented as an epigenetic "doping system" of mammalian development. Milk exosome-derived micro-ribonucleic acids (miRNAs) that target DNA methyltransferases are implicated to play the key role in the upregulation of developmental genes such as FTO, INS, and IGF1. In contrast to miRNA-deficient infant formula, breastfeeding via physiological miRNA transfer provides the appropriate signals for adequate epigenetic programming of the newborn infant. Whereas breastfeeding is restricted to the lactation period, continued consumption of cow's milk results in persistent epigenetic upregulation of genes critically involved in the development of diseases of civilization such as diabesity, neurodegeneration, and cancer. We hypothesize that the same miRNAs that epigenetically increase lactation, upregulate gene expression of the milk recipient via milk-derived miRNAs. It is of critical concern that persistent consumption of pasteurized cow's milk contaminates the human food chain with bovine miRNAs, that are identical to their human analogs. Commercial interest to enhance dairy lactation performance may further increase the epigenetic miRNA burden for the milk consumer.
Collapse
Affiliation(s)
- Bodo C Melnik
- Department of Dermatology, Environmental Medicine and Health Theory, Faculty of Human Sciences, University of Osnabrück, Am Finkenhügel 7a, D-49076 Osnabrück, Germany.
| | - Gerd Schmitz
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, University of Regensburg, Franz-Josef-Strauß-Allee 11, D-93053 Regensburg, Germany.
| |
Collapse
|
65
|
Di Domenico F, Barone E, Perluigi M, Butterfield DA. The Triangle of Death in Alzheimer's Disease Brain: The Aberrant Cross-Talk Among Energy Metabolism, Mammalian Target of Rapamycin Signaling, and Protein Homeostasis Revealed by Redox Proteomics. Antioxid Redox Signal 2017; 26:364-387. [PMID: 27626216 DOI: 10.1089/ars.2016.6759] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
SIGNIFICANCE Alzheimer's disease (AD) is a multifactorial neurodegenerative disorder and represents one of the most disabling conditions. AD shares many features in common with systemic insulin resistance diseases, suggesting that it can be considered as a metabolic disease, characterized by reduced insulin-stimulated growth and survival signaling, increased oxidative stress (OS), proinflammatory cytokine activation, mitochondrial dysfunction, impaired energy metabolism, and altered protein homeostasis. Recent Advances: Reduced glucose utilization and energy metabolism in AD have been associated with the buildup of amyloid-β peptide and hyperphosphorylated tau, increased OS, and the accumulation of unfolded/misfolded proteins. Mammalian target of rapamycin (mTOR), which is aberrantly activated in AD since early stages, plays a key role during AD neurodegeneration by, on one side, inhibiting insulin signaling as a negative feedback mechanism and, on the other side, regulating protein homeostasis (synthesis/clearance). CRITICAL ISSUES It is likely that the concomitant and mutual alterations of energy metabolism-mTOR signaling-protein homeostasis might represent a self-sustaining triangle of harmful events that trigger the degeneration and death of neurons and the development and progression of AD. Intriguingly, the altered cross-talk between the components of such a triangle of death, beyond altering the redox homeostasis of the neuron, is further exacerbated by increased levels of OS that target and impair key components of the pathways involved. Redox proteomic studies in human samples and animal models of AD-like dementia led to identification of oxidatively modified components of the pathways composing the triangle of death, therefore revealing the crucial role of OS in fueling this aberrant vicious cycle. FUTURE DIRECTIONS The identification of compounds able to restore the function of the pathways targeted by oxidative damage might represent a valuable therapeutic approach to slow or delay AD. Antioxid. Redox Signal. 26, 364-387.
Collapse
Affiliation(s)
- Fabio Di Domenico
- 1 Department of Biochemical Sciences, Sapienza University of Rome , Rome, Italy
| | - Eugenio Barone
- 1 Department of Biochemical Sciences, Sapienza University of Rome , Rome, Italy .,2 Facultad de Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile , Santiago, Chile
| | - Marzia Perluigi
- 1 Department of Biochemical Sciences, Sapienza University of Rome , Rome, Italy
| | - D Allan Butterfield
- 3 Department of Chemistry, Sanders-Brown Center of Aging, University of Kentucky , Lexington, Kentucky
| |
Collapse
|
66
|
Franco R, Martínez-Pinilla E, Navarro G, Zamarbide M. Potential of GPCRs to modulate MAPK and mTOR pathways in Alzheimer's disease. Prog Neurobiol 2017; 149-150:21-38. [PMID: 28189739 DOI: 10.1016/j.pneurobio.2017.01.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 01/20/2017] [Accepted: 01/22/2017] [Indexed: 02/08/2023]
Abstract
Despite efforts to understand the mechanism of neuronal cell death, finding effective therapies for neurodegenerative diseases is still a challenge. Cognitive deficits are often associated with neurodegenerative diseases. Remarkably, in the absence of consensus biomarkers, diagnosis of diseases such as Alzheimer's still relies on cognitive tests. Unfortunately, all efforts to translate findings in animal models to the patients have been unsuccessful. Alzheimer's disease may be addressed from two different points of view, neuroprotection or cognitive enhancement. Based on recent data, the mammalian target of rapamycin (mTOR) pathway arises as a versatile player whose modulation may impact on mechanisms of both neuroprotection and cognition. Whereas direct targeting of mTOR does not seem to constitute a convenient approach in drug discovery, its indirect modulation by other signaling pathways seems promising. In fact, G-protein-coupled receptors (GPCRs) remain the most common 'druggable' targets and as such pharmacological manipulation of GPCRs with selective ligands may modulate phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K), mitogen-activated protein (MAP) kinase and mTOR signaling pathways. Thus, GPCRs become important targets for potential drug treatments in different neurodegenerative disorders including, but not limited to, Alzheimer's disease. GPCR-mediated modulation of mTOR may take advantage of different GPCRs coupled to different G-dependent and G-independent signal transduction routes, of functional selectivity and/or of biased agonism. Signals mediated by GPCRs may act as coincidence detectors to achieve different benefits in different stages of the neurodegenerative disease.
Collapse
Affiliation(s)
- Rafael Franco
- Department of Biochemistry and Molecular Biomedicine and IBUB (Institute of Biomedicine of the University of Barcelona), University of Barcelona, Barcelona, Spain; Centro de investigación en Red: Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Eva Martínez-Pinilla
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), Departamento de Morfología y Biología Celular, Facultad de Medicina, Universidad de Oviedo, Asturias, Spain
| | - Gemma Navarro
- Department of Biochemistry and Molecular Biomedicine and IBUB (Institute of Biomedicine of the University of Barcelona), University of Barcelona, Barcelona, Spain; Centro de investigación en Red: Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | | |
Collapse
|
67
|
Bordi M, Berg MJ, Mohan PS, Peterhoff CM, Alldred MJ, Che S, Ginsberg SD, Nixon RA. Autophagy flux in CA1 neurons of Alzheimer hippocampus: Increased induction overburdens failing lysosomes to propel neuritic dystrophy. Autophagy 2016; 12:2467-2483. [PMID: 27813694 PMCID: PMC5173282 DOI: 10.1080/15548627.2016.1239003] [Citation(s) in RCA: 238] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Defective autophagy contributes to Alzheimer disease (AD) pathogenesis although evidence is conflicting on whether multiple stages are impaired. Here, for the first time, we have comprehensively evaluated the entire autophagic process specifically in CA1 pyramidal neurons of hippocampus from early and late-stage AD subjects and nondemented controls. CA1 neurons aspirated by laser capture microdissection were analyzed using a custom-designed microarray comprising 578 neuropathology- and neuroscience-associated genes. Striking upregulation of autophagy-related genes, exceeding that of other gene ontology groups, reflected increases in autophagosome formation and lysosomal biogenesis beginning at early AD stages. Upregulated autophagosome formation was further indicated by elevated gene and protein expression levels for autophagosome components and increased LC3-positive puncta. Increased lysosomal biogenesis was evidenced by activation of MiTF/TFE family transcriptional regulators, particularly TFE3 (transcription factor binding to IGHM enhancer 3) and by elevated expression of their target genes and encoded proteins. Notably, TFEB (transcription factor EB) activation was associated more strongly with glia than neurons. These findings establish that autophagic sequestration is both competent and upregulated in AD. Autophagosome-lysosome fusion is not evidently altered. Despite this early disease response, however, autophagy flux is progressively impeded due to deficient substrate clearance, as reflected by autolysosomal accumulation of LC3-II and SQSTM1/p62 and expansion of autolysosomal size and total area. We propose that sustained induction of autophagy in the face of progressively declining lysosomal clearance of substrates explains the uncommonly robust autophagic pathology and neuritic dystrophy implicated in AD pathogenesis.
Collapse
Affiliation(s)
- Matteo Bordi
- a Center for Dementia Research, Nathan Kline Institute , Orangeburg , NY , USA.,b Department of Psychiatry , New York University Langone Medical Center , New York , NY , USA
| | - Martin J Berg
- a Center for Dementia Research, Nathan Kline Institute , Orangeburg , NY , USA
| | - Panaiyur S Mohan
- a Center for Dementia Research, Nathan Kline Institute , Orangeburg , NY , USA.,b Department of Psychiatry , New York University Langone Medical Center , New York , NY , USA
| | | | - Melissa J Alldred
- a Center for Dementia Research, Nathan Kline Institute , Orangeburg , NY , USA.,b Department of Psychiatry , New York University Langone Medical Center , New York , NY , USA
| | - Shaoli Che
- a Center for Dementia Research, Nathan Kline Institute , Orangeburg , NY , USA.,b Department of Psychiatry , New York University Langone Medical Center , New York , NY , USA
| | - Stephen D Ginsberg
- a Center for Dementia Research, Nathan Kline Institute , Orangeburg , NY , USA.,b Department of Psychiatry , New York University Langone Medical Center , New York , NY , USA.,d Department of Neuroscience and Physiology , New York University Langone Medical Center , New York , NY , USA
| | - Ralph A Nixon
- a Center for Dementia Research, Nathan Kline Institute , Orangeburg , NY , USA.,b Department of Psychiatry , New York University Langone Medical Center , New York , NY , USA.,c Department of Cell Biology , New York University Langone Medical Center , New York , NY , USA
| |
Collapse
|
68
|
Wahl D, Cogger VC, Solon-Biet SM, Waern RVR, Gokarn R, Pulpitel T, Cabo RD, Mattson MP, Raubenheimer D, Simpson SJ, Le Couteur DG. Nutritional strategies to optimise cognitive function in the aging brain. Ageing Res Rev 2016; 31:80-92. [PMID: 27355990 PMCID: PMC5035589 DOI: 10.1016/j.arr.2016.06.006] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 06/17/2016] [Accepted: 06/23/2016] [Indexed: 12/15/2022]
Abstract
Old age is the greatest risk factor for most neurodegenerative diseases. During recent decades there have been major advances in understanding the biology of aging, and the development of nutritional interventions that delay aging including calorie restriction (CR) and intermittent fasting (IF), and chemicals that influence pathways linking nutrition and aging processes. CR influences brain aging in many animal models and recent findings suggest that dietary interventions can influence brain health and dementia in older humans. The role of individual macronutrients in brain aging also has been studied, with conflicting results about the effects of dietary protein and carbohydrates. A new approach known as the Geometric Framework (GF) has been used to unravel the complex interactions between macronutrients (protein, fat, and carbohydrate) and total energy on outcomes such as aging. These studies have shown that low-protein, high-carbohydrate (LPHC) diets are optimal for lifespan in ad libitum fed animals, while total calories have minimal effect once macronutrients are taken into account. One of the primary purposes of this review is to explore the notion that macronutrients may have a more translational potential than CR and IF in humans, and therefore there is a pressing need to use GF to study the impact of diet on brain aging. Furthermore, given the growing recognition of the role of aging biology in dementia, such studies might provide a new approach for dietary interventions for optimizing brain health and preventing dementia in older people.
Collapse
Affiliation(s)
- Devin Wahl
- Charles Perkins Centre, University of Sydney, Sydney 2006 Australia; Aging and Alzheimers Institute, ANZAC Research Institute, Concord Clinical School/Sydney Medical School, Concord, 2139 Australia
| | - Victoria C Cogger
- Charles Perkins Centre, University of Sydney, Sydney 2006 Australia; Aging and Alzheimers Institute, ANZAC Research Institute, Concord Clinical School/Sydney Medical School, Concord, 2139 Australia
| | - Samantha M Solon-Biet
- Charles Perkins Centre, University of Sydney, Sydney 2006 Australia; Aging and Alzheimers Institute, ANZAC Research Institute, Concord Clinical School/Sydney Medical School, Concord, 2139 Australia
| | - Rosilene V R Waern
- Charles Perkins Centre, University of Sydney, Sydney 2006 Australia; School of Life and Environmental Sciences, University of Sydney, Sydney 2006, Australia
| | - Rahul Gokarn
- Charles Perkins Centre, University of Sydney, Sydney 2006 Australia; Aging and Alzheimers Institute, ANZAC Research Institute, Concord Clinical School/Sydney Medical School, Concord, 2139 Australia
| | - Tamara Pulpitel
- Charles Perkins Centre, University of Sydney, Sydney 2006 Australia
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Mark P Mattson
- Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - David Raubenheimer
- Charles Perkins Centre, University of Sydney, Sydney 2006 Australia; Faculty of Veterinary Science, University of Sydney, Sydney 2006, Australia; School of Life and Environmental Sciences, University of Sydney, Sydney 2006, Australia
| | - Stephen J Simpson
- Charles Perkins Centre, University of Sydney, Sydney 2006 Australia; School of Life and Environmental Sciences, University of Sydney, Sydney 2006, Australia
| | - David G Le Couteur
- Charles Perkins Centre, University of Sydney, Sydney 2006 Australia; Aging and Alzheimers Institute, ANZAC Research Institute, Concord Clinical School/Sydney Medical School, Concord, 2139 Australia.
| |
Collapse
|
69
|
Tramutola A, Lanzillotta C, Di Domenico F. Targeting mTOR to reduce Alzheimer-related cognitive decline: from current hits to future therapies. Expert Rev Neurother 2016; 17:33-45. [PMID: 27690737 DOI: 10.1080/14737175.2017.1244482] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION The mTOR pathway is involved in the regulation of a wide repertoire of cellular functions in the brain and its dysregulation is emerging as a leitmotif in a large number of neurological disorders. In AD, altered mTOR signaling contributes to the inhibition of autophagy deposition of Aβ and tau aggregates and to the alteration of several neuronal metabolic pathways. Areas covered: In this review, we report all the current findings on the use of mTOR inhibitors (rapamycin, rapalogues) in the treatment of AD. These results support the role of mTOR inhibitors as potential therapeutic agents able to reduce AD hallmarks and recover cognitive performances. Expert commentary: Despite mTOR inhibitors appearing to be ideal compounds to counteract AD, further studies are needed in order to gain knowledge on the involvement of aberrant mTOR in AD, and to standardize a valuable therapeutic approach that can be translated to humans.
Collapse
Affiliation(s)
- Antonella Tramutola
- a Department of Biochemical Sciences , Sapienza University of Rome , Rome , Italy
| | - Chiara Lanzillotta
- a Department of Biochemical Sciences , Sapienza University of Rome , Rome , Italy
| | - Fabio Di Domenico
- a Department of Biochemical Sciences , Sapienza University of Rome , Rome , Italy
| |
Collapse
|
70
|
Dulla CG, Coulter DA, Ziburkus J. From Molecular Circuit Dysfunction to Disease: Case Studies in Epilepsy, Traumatic Brain Injury, and Alzheimer's Disease. Neuroscientist 2016; 22:295-312. [PMID: 25948650 PMCID: PMC4641826 DOI: 10.1177/1073858415585108] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Complex circuitry with feed-forward and feed-back systems regulate neuronal activity throughout the brain. Cell biological, electrical, and neurotransmitter systems enable neural networks to process and drive the entire spectrum of cognitive, behavioral, and motor functions. Simultaneous orchestration of distinct cells and interconnected neural circuits relies on hundreds, if not thousands, of unique molecular interactions. Even single molecule dysfunctions can be disrupting to neural circuit activity, leading to neurological pathology. Here, we sample our current understanding of how molecular aberrations lead to disruptions in networks using three neurological pathologies as exemplars: epilepsy, traumatic brain injury (TBI), and Alzheimer's disease (AD). Epilepsy provides a window into how total destabilization of network balance can occur. TBI is an abrupt physical disruption that manifests in both acute and chronic neurological deficits. Last, in AD progressive cell loss leads to devastating cognitive consequences. Interestingly, all three of these neurological diseases are interrelated. The goal of this review, therefore, is to identify molecular changes that may lead to network dysfunction, elaborate on how altered network activity and circuit structure can contribute to neurological disease, and suggest common threads that may lie at the heart of molecular circuit dysfunction.
Collapse
Affiliation(s)
- Chris G Dulla
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| | - Douglas A Coulter
- Department of Pediatrics and Neuroscience, University of Pennsylvania Perleman School of Medicine, Philadelphia, PA, USA Division of Neurology and the Research Institute of Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jokubas Ziburkus
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| |
Collapse
|
71
|
Nacarelli T, Azar A, Sell C. Mitochondrial stress induces cellular senescence in an mTORC1-dependent manner. Free Radic Biol Med 2016; 95:133-54. [PMID: 27016071 DOI: 10.1016/j.freeradbiomed.2016.03.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 03/07/2016] [Accepted: 03/12/2016] [Indexed: 11/25/2022]
Abstract
Although mitochondrial stress is a key determinant of cellular homeostasis, the intracellular mechanisms by which this stress is communicated to the nucleus and its impact on cell fate decisions are not well defined. In this study, we report that activation of mTORC1 signaling triggered by mitochondrial-generated reactive oxygen species (ROS) results in activation of the senescence program. We show that exposure of human fibroblasts to nucleoside analogs commonly used in antiretroviral therapies, and known to induce mitochondrial dysfunction, increases mitochondrial ROS and leads to a rise in intracellular ROS concomitant with activation of mTORC1. In this setting, it appears that mTORC1 activates senescence through HDM2 phosphorylation, facilitating a p53-mediated response. Inhibition of mTORC1 by rapamycin decreases HDM2 phosphorylation and blocks activation of the senescence program in human cells. In addition, decreasing mitochondrial ROS directly blocks mTORC1 signaling and prevents the onset of senescence. Consistent with these results, both total and mitochondrial-specific ROS increased in cells undergoing replicative senescence along with ribosomal p70 phosphorylation. The results reveal a novel link between mitochondrial dysfunction, mTORC1 signaling, and the senescence program.
Collapse
Affiliation(s)
- Timothy Nacarelli
- Drexel University College of Medicine, 245 N 15th Street, Philadelphia, PA 19102, United States
| | - Ashley Azar
- Drexel University College of Medicine, 245 N 15th Street, Philadelphia, PA 19102, United States
| | - Christian Sell
- Drexel University College of Medicine, 245 N 15th Street, Philadelphia, PA 19102, United States.
| |
Collapse
|
72
|
Biever A, Valjent E, Puighermanal E. Ribosomal Protein S6 Phosphorylation in the Nervous System: From Regulation to Function. Front Mol Neurosci 2015; 8:75. [PMID: 26733799 PMCID: PMC4679984 DOI: 10.3389/fnmol.2015.00075] [Citation(s) in RCA: 152] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 11/23/2015] [Indexed: 01/31/2023] Open
Abstract
Since the discovery of the phosphorylation of the 40S ribosomal protein S6 (rpS6) about four decades ago, much effort has been made to uncover the molecular mechanisms underlying the regulation of this post-translational modification. In the field of neuroscience, rpS6 phosphorylation is commonly used as a readout of the mammalian target of rapamycin complex 1 signaling activation or as a marker for neuronal activity. Nevertheless, its biological role in neurons still remains puzzling. Here we review the pharmacological and physiological stimuli regulating this modification in the nervous system as well as the pathways that transduce these signals into rpS6 phosphorylation. Altered rpS6 phosphorylation observed in various genetic and pathophysiological mouse models is also discussed. Finally, we examine the current state of knowledge on the physiological role of this post-translational modification and highlight the questions that remain to be addressed.
Collapse
Affiliation(s)
- Anne Biever
- Centre National de la Recherche Scientifique, UMR5203, Institut de Génomique FonctionnelleMontpellier, France; Institut National de la Santé et de la Recherche Médicale, U1191Montpellier, France; Université de Montpellier, UMR-5203Montpellier, France
| | - Emmanuel Valjent
- Centre National de la Recherche Scientifique, UMR5203, Institut de Génomique FonctionnelleMontpellier, France; Institut National de la Santé et de la Recherche Médicale, U1191Montpellier, France; Université de Montpellier, UMR-5203Montpellier, France
| | - Emma Puighermanal
- Centre National de la Recherche Scientifique, UMR5203, Institut de Génomique FonctionnelleMontpellier, France; Institut National de la Santé et de la Recherche Médicale, U1191Montpellier, France; Université de Montpellier, UMR-5203Montpellier, France
| |
Collapse
|
73
|
Galvan V, Hart MJ. Vascular mTOR-dependent mechanisms linking the control of aging to Alzheimer's disease. Biochim Biophys Acta Mol Basis Dis 2015; 1862:992-1007. [PMID: 26639036 DOI: 10.1016/j.bbadis.2015.11.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 11/17/2015] [Accepted: 11/18/2015] [Indexed: 02/07/2023]
Abstract
Aging is the strongest known risk factor for Alzheimer's disease (AD). With the discovery of the mechanistic target of rapamycin (mTOR) as a critical pathway controlling the rate of aging in mice, molecules at the interface between the regulation of aging and the mechanisms of specific age-associated diseases can be identified. We will review emerging evidence that mTOR-dependent brain vascular dysfunction, a universal feature of aging, may be one of the mechanisms linking the regulation of the rate of aging to the pathogenesis of Alzheimer's disease. This article is part of a Special Issue entitled: Vascular Contributions to Cognitive Impairment and Dementia edited by M. Paul Murphy, Roderick A. Corriveau and Donna M. Wilcock.
Collapse
Affiliation(s)
- Veronica Galvan
- Department of Physiology and the Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio.
| | - Matthew J Hart
- Department of Biochemistry, University of Texas Health Science Center at San Antonio
| |
Collapse
|
74
|
Bergen AA, Kaing S, ten Brink JB, Gorgels TG, Janssen SF. Gene expression and functional annotation of human choroid plexus epithelium failure in Alzheimer's disease. BMC Genomics 2015; 16:956. [PMID: 26573292 PMCID: PMC4647590 DOI: 10.1186/s12864-015-2159-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 10/27/2015] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is the most common form of dementia. AD has a multifactorial disease etiology and is currently untreatable. Multiple genes and molecular mechanisms have been implicated in AD, including ß-amyloid deposition in the brain, neurofibrillary tangle accumulation of hyper-phosphorylated Tau, synaptic failure, oxidative stress and inflammation. Relatively little is known about the role of the blood-brain barriers, especially the blood-cerebrospinal fluid barrier (BCSFB), in AD. The BCSFB is involved in cerebrospinal fluid (CSF) production, maintenance of brain homeostasis and neurodegenerative disorders. RESULTS Using an Agilent platform with common reference design, we performed a large scale gene expression analysis and functional annotation of the Choroid Plexus Epithelium (CPE), which forms the BCSFB. We obtained 2 groups of freshly frozen Choroid Plexus (CP) of 7 human donor brains each, with and without AD: Braak stages (0-1) and (5-6). We cut CP cryo-sections and isolated RNA from cresyl-violet stained, laser dissected CPE cells. Gene expression results were analysed with T-tests (R) and the knowledge-database Ingenuity. We found statistically significantly altered gene expression data sets, biological functions, canonical pathways, molecular networks and functionalities in AD-affected CPE. We observed specific cellular changes due to increased oxidative stress, such as the unfolded protein response, E1F2 and NRF2 signalling and the protein ubiquitin pathway. Most likely, the AD-affected BCSFB barrier becomes more permeable due to downregulation of CLDN5. Finally, our data also predicted down regulation of the glutathione mediated detoxification pathway and the urea cycle in the AD CPE, which suggest that the CPE sink action may be impaired. Remarkably, the expression of a number of genes known to be involved in AD, such as APP, PSEN1, PSEN2, TTR and CLU is moderate to high and remains stable in both healthy and affected CPE. Literature labelling of our new functional molecular networks confirmed multiple previous (molecular) observations in the AD literature and revealed many new ones. CONCLUSIONS We conclude that CPE failure in AD exists. Combining our data with those of the literature, we propose the following chronological and overlapping chain of events: increased Aß burden on CPE; increased oxidative stress in CPE; despite continuous high expression of TTR: decreased capability of CPE to process amyloid; (pro-) inflammatory and growth factor signalling by CPE; intracellular ubiquitin involvement, remodelling of CPE tight junctions and, finally, cellular atrophy. Our data corroborates the hypothesis that increased BCSFB permeability, especially loss of selective CLDN5-mediated paracellular transport, altered CSF production and CPE sink action, as well as loss of CPE mediated macrophage recruitment contribute to the pathogenesis of AD.
Collapse
Affiliation(s)
- Arthur A Bergen
- Department of Clinical Genetics, Academic Medical Centre, Amsterdam, AMC, Meibergdreef 9, 1105 AZ AMC, Amsterdam, The Netherlands. .,The Netherlands Institute for Neurosciences (NIN-KNAW), Amsterdam, The Netherlands.
| | - Sovann Kaing
- The Netherlands Institute for Neurosciences (NIN-KNAW), Amsterdam, The Netherlands
| | - Jacoline B ten Brink
- Department of Clinical Genetics, Academic Medical Centre, Amsterdam, AMC, Meibergdreef 9, 1105 AZ AMC, Amsterdam, The Netherlands
| | | | - Theo G Gorgels
- The Netherlands Institute for Neurosciences (NIN-KNAW), Amsterdam, The Netherlands.,University Eye Clinic Maastricht, MUMC, Maastricht, The Netherlands
| | - Sarah F Janssen
- The Netherlands Institute for Neurosciences (NIN-KNAW), Amsterdam, The Netherlands.,Department of Ophthalmology, VUMC, Amsterdam, The Netherlands
| |
Collapse
|
75
|
Zhou P, Zhang N, Nussinov R, Ma B. Defining the Domain Arrangement of the Mammalian Target of Rapamycin Complex Component Rictor Protein. J Comput Biol 2015; 22:876-86. [PMID: 26176550 DOI: 10.1089/cmb.2015.0103] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Mammalian target of rapamycin (mTOR) complexes play a pivotal role in the cell. Raptor and Rictor proteins interact with mTOR to form two distinct complexes, mTORC1 and mTORC2, respectively. While the domain structure of Raptor is known, current bioinformatics tools failed to classify the domains in Rictor. Here we focus on identifying specific domains in Rictor by searching for conserved regions. We scanned the pdb structural database and constructed three protein domain datasets. Next we carried out multiple pairwise sequence alignments of the proteins in the domain dataset. By analyzing the z-scores of Rictor sequence similarity to protein sequences in the dataset, we assigned the structural and functional domains of Rictor. We found that, like Raptor, Rictor also has HEAT and WD40 domains, which could be the common motif binding to mTORC. Rictor may also have pleckstrin homology domains, which mediate cellular localization and transmit signals to downstream targets, as well as a domain that is homologous to 50S protein L17 and human 39S protein L17. This putative ribosome binding domain could mediate mTORC2-ribosome interaction.
Collapse
Affiliation(s)
- Ping Zhou
- 1 Research Center of Basic Medical Sciences and Cancer Institute and Hospital, Tianjin Medical University , Tianjin, China
| | - Ning Zhang
- 1 Research Center of Basic Medical Sciences and Cancer Institute and Hospital, Tianjin Medical University , Tianjin, China
| | - Ruth Nussinov
- 2 Basic Science Program, Leidos Biomedical Research, Inc., Cancer and Inflammation Program, National Cancer Institute , Frederick, Maryland.,3 Department of Human Genetics and Molecular Medicine, Sackler Institute of Molecular Medicine, Sackler School of Medicine, Tel Aviv University , Tel Aviv, Israel
| | - Buyong Ma
- 2 Basic Science Program, Leidos Biomedical Research, Inc., Cancer and Inflammation Program, National Cancer Institute , Frederick, Maryland
| |
Collapse
|
76
|
Hippocampal endosomal, lysosomal, and autophagic dysregulation in mild cognitive impairment: correlation with aβ and tau pathology. J Neuropathol Exp Neurol 2015; 74:345-58. [PMID: 25756588 DOI: 10.1097/nen.0000000000000179] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Endosomal-lysosomal and autophagic dysregulation occurs in the hippocampus in prodromal Alzheimer disease (AD), but its relationship with β-amyloid (Aβ) and tau pathology remains unclear. To investigate this issue, we performed immunoblot analysis of hippocampal homogenates from cases with an antemortem clinical diagnosis of no cognitive impairment, mild cognitive impairment (MCI), and AD. Western blot analysis revealed significant increases in the acid hydrolase cathepsin D and early endosome marker rabaptin5 in the MCI group compared with AD, whereas levels of phosphorylated mammalian target of rapamycin proteins (pmTOR), total mammalian target of rapamycin (mTOR), p62, traf6, and LilrB2 were comparable across clinical groups. Hippocampal Aβ1-40 and Aβ1-42 concentrations and AT8-immunopositive neurofibrillary tangle density were not significantly different across the clinical groups. Greater cathepsin D expression was associated with global cognitive score and episodic memory score but not with mini mental state examination or advanced neuropathology criteria. These results indicate that alterations in hippocampal endosomal-lysosomal proteins in MCI are independent of tau or Aβ pathology.
Collapse
|
77
|
Hussain AR, Al-Romaizan M, Ahmed M, Thangavel S, Al-Dayel F, Beg S, Uddin S, Siraj AK, Al-Kuraya KS. Dual Targeting of mTOR Activity with Torin2 Potentiates Anticancer Effects of Cisplatin in Epithelial Ovarian Cancer. Mol Med 2015; 21:466-78. [PMID: 26023849 DOI: 10.2119/molmed.2014.00238] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 05/25/2015] [Indexed: 12/19/2022] Open
Abstract
Mammalian target of rapamycin (mTOR) and phosphatidylinositol 3-kinase (PI3K) are two key components of PI3K/Akt/mTOR signaling pathway. Dysregulation of these pathways have been found in many cancers, including epithelial ovarian cancer (EOC), however, the role of mTOR has not been fully elucidated in Middle Eastern EOC. Therefore, we investigated the activation of mTOR complexes (mTORC1 and mTORC2) in a cohort of 156 EOC from Saudi Arabia by immunohistochemistry in a tissue microarray format. mTORC1 and mTORC2 were found to be activated in 55 of 146 (37.7%) and 63 of 140 (45%) of EOC samples, respectively. mTORC1 was significantly associated with mTORC2 (p < 0.0001) activation and both mTOR complexes were significantly associated with p-AKT (p = 0.0205 and 0.0298) and p-P70S6 (p < 0.0001 and 0.0035), respectively. Interestingly, mTOR activation incurred a poor progression-free survival (PFS) (p = 0.0188) in EOC. Next, the in vitro effect of inactivation of mTOR complexes was evaluated using a second-generation mTOR inhibitor, Torin2, on a panel of EOC cell lines. Torin2 treatment decreased cell viability and induced apoptosis in a dose-dependent manner via inactivation of mTORC1 and mTORC2 and their downstream targets in EOC cell lines. Furthermore, treatment of EOC cells with a subtoxic dose of Torin2 potentiated a cisplatin-induced apoptotic response in EOC cell lines. Finally, we studied the in vivo effect of a combination of Torin2 and cisplatin and found that this combination synergistically inhibited tumor growth in nude mice. These studies highlight the importance of targeting the mTOR survival pathway and suggest that cotreatment with cisplatin and Torin2 may be beneficial for the management of EOC.
Collapse
Affiliation(s)
- Azhar R Hussain
- Human Cancer Genomic Research, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | | | - Maqbool Ahmed
- Human Cancer Genomic Research, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Saravanan Thangavel
- Human Cancer Genomic Research, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Fouad Al-Dayel
- Department of Pathology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Shaham Beg
- Human Cancer Genomic Research, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Shahab Uddin
- Human Cancer Genomic Research, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Abdul K Siraj
- Human Cancer Genomic Research, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Khawla S Al-Kuraya
- Human Cancer Genomic Research, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia.,AlFaisal University, Riyadh, Saudi Arabia
| |
Collapse
|
78
|
Perluigi M, Di Domenico F, Butterfield DA. mTOR signaling in aging and neurodegeneration: At the crossroad between metabolism dysfunction and impairment of autophagy. Neurobiol Dis 2015; 84:39-49. [PMID: 25796566 DOI: 10.1016/j.nbd.2015.03.014] [Citation(s) in RCA: 230] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 03/09/2015] [Accepted: 03/12/2015] [Indexed: 02/09/2023] Open
Abstract
Compelling evidence indicates that the mammalian target of rapamycin (mTOR) signaling pathway is involved in cellular senescence, organismal aging and age-dependent diseases. mTOR is a conserved serine/threonine kinase that is known to be part of two different protein complexes: mTORC1 and mTORC2, which differ in some components and in upstream and downstream signalling. In multicellular organisms, mTOR regulates cell growth and metabolism in response to nutrients, growth factors and cellular energy conditions. Growing studies highlight that disturbance in mTOR signalling in the brain affects multiple pathways including glucose metabolism, energy production, mitochondrial function, cell growth and autophagy. All these events are key players in age-related cognitive decline such as development of Alzheimer disease (AD). The current review discusses the main regulatory roles of mTOR signalling in the brain, in particular focusing on autophagy, glucose metabolism and mitochondrial functions. Targeting mTOR in the CNS can offer new prospective for drug discovery; however further studies are needed for a comprehensive understanding of mTOR, which lies at the crossroads of multiple signals involved in AD etiology and pathogenesis.
Collapse
Affiliation(s)
- Marzia Perluigi
- Department of Biochemical Sciences, Sapienza University of Rome, Italy.
| | - Fabio Di Domenico
- Department of Biochemical Sciences, Sapienza University of Rome, Italy
| | - D Allan Butterfield
- Sanders-Brown Centre of Aging, University of Kentucky, Lexington KY, USA; Department of Chemistry, University of Kentucky, Lexington KY, USA
| |
Collapse
|
79
|
In vivo AAV1 transduction with hRheb(S16H) protects hippocampal neurons by BDNF production. Mol Ther 2014; 23:445-55. [PMID: 25502903 DOI: 10.1038/mt.2014.241] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 12/06/2014] [Indexed: 01/08/2023] Open
Abstract
Recent evidence has shown that Ras homolog enriched in brain (Rheb) is dysregulated in Alzheimer's disease (AD) brains. However, it is still unclear whether Rheb activation contributes to the survival and protection of hippocampal neurons in the adult brain. To assess the effects of active Rheb in hippocampal neurons in vivo, we transfected neurons in the cornu ammonis 1 (CA1) region in normal adult rats with an adeno-associated virus containing the constitutively active human Rheb (hRheb(S16H)) and evaluated the effects on thrombin-induced neurotoxicity. Transduction with hRheb(S16H) significantly induced neurotrophic effects in hippocampal neurons through activation of mammalian target of rapamycin complex 1 (mTORC1) without side effects such as long-term potentiation impairment and seizures from the alteration of cytoarchitecture, and the expression of hRheb(S16H) prevented thrombin-induced neurodegeneration in vivo, an effect that was diminished by treatment with specific neutralizing antibodies against brain-derived neurotrophic factor (BDNF). In addition, our results showed that the basal mTORC1 activity might be insufficient to mediate the level of BDNF expression, but hRheb(S16H)-activated mTORC1 stimulated BDNF production in hippocampal neurons. These results suggest that viral vector transduction with hRheb(S16H) may have therapeutic value in the treatment of neurodegenerative diseases such as AD.
Collapse
|
80
|
Richardson A, Galvan V, Lin AL, Oddo S. How longevity research can lead to therapies for Alzheimer's disease: The rapamycin story. Exp Gerontol 2014; 68:51-8. [PMID: 25481271 DOI: 10.1016/j.exger.2014.12.002] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 11/24/2014] [Accepted: 12/02/2014] [Indexed: 12/14/2022]
Abstract
The discovery that rapamycin increases lifespan in mice and restores/delays many aging phenotypes has led to the speculation that rapamycin has 'anti-aging' properties. The major question discussed in this review is whether a manipulation that has anti-aging properties can alter the onset and/or progression of Alzheimer's disease, a disease in which age is the major risk factor. Rapamycin has been shown to prevent (and possibly restore in some cases) the deficit in memory observed in the mouse model of Alzheimer's disease (AD-Tg) as well as reduce Aβ and tau aggregation, restore cerebral blood flow and vascularization, and reduce microglia activation. All of these parameters are widely recognized as symptoms central to the development of AD. Furthermore, rapamycin has also been shown to improve memory and reduce anxiety and depression in several other mouse models that show cognitive deficits as well as in 'normal' mice. The current research shows the feasibility of using pharmacological agents that increase lifespan, such as those identified by the National Institute on Aging Intervention Testing Program, to treat Alzheimer's disease.
Collapse
Affiliation(s)
- Arlan Richardson
- Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City VA Medical Center, Oklahoma City, OK 73104, USA.
| | - Veronica Galvan
- Department of Physiology and Barshop Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX 78245, USA
| | - Ai-Ling Lin
- Sanders-Brown Center on Aging, Department of Pharmacology & Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Salvatore Oddo
- Banner Sun Health Research Institute, Sun City, AZ 85351, USA; Department of Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Phoenix, AZ 85004, USA
| |
Collapse
|
81
|
Nacarelli T, Azar A, Sell C. Aberrant mTOR activation in senescence and aging: A mitochondrial stress response? Exp Gerontol 2014; 68:66-70. [PMID: 25449851 DOI: 10.1016/j.exger.2014.11.004] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 10/30/2014] [Accepted: 11/04/2014] [Indexed: 01/18/2023]
Abstract
Unexpected activation of mTOR signaling, measured by ribosomal S6 phosphorylation or ribosomal S6 kinase (p70S6K) activity, has been reported in aging-related settings. Evidence of elevated mTOR activity has been reported in the heart and muscle tissue in aged mice and humans, mouse models of progeria, and senescent human fibroblasts. We explore these reports and the possibility that activation of the mTOR/p70S6K kinase pathway may represent a ROS-mediated response to mitochondrial stress leading to the activation of senescence. This activation is a hallmark of both aged tissue and senescent human cells.
Collapse
Affiliation(s)
- Timothy Nacarelli
- Drexel University College of Medicine, 245N 15th Street, Philadelphia, PA 19102, United States
| | - Ashley Azar
- Drexel University College of Medicine, 245N 15th Street, Philadelphia, PA 19102, United States
| | - Christian Sell
- Drexel University College of Medicine, 245N 15th Street, Philadelphia, PA 19102, United States.
| |
Collapse
|
82
|
Patel S, Grizzell JA, Holmes R, Zeitlin R, Solomon R, Sutton TL, Rohani A, Charry LC, Iarkov A, Mori T, Echeverria Moran V. Cotinine halts the advance of Alzheimer's disease-like pathology and associated depressive-like behavior in Tg6799 mice. Front Aging Neurosci 2014; 6:162. [PMID: 25100990 PMCID: PMC4107855 DOI: 10.3389/fnagi.2014.00162] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 06/24/2014] [Indexed: 12/29/2022] Open
Abstract
Alzheimer's disease (AD) is associated with cognitive and non-cognitive symptoms for which there are currently no effective therapies. We have previously reported that cotinine, a natural product obtained from tobacco leaves, prevented memory loss and diminished amyloid-β (Aβ) plaque pathology in transgenic 6799 mice (Tg6799 mice) when treated prior to the development of the pathology. We have also shown that cotinine reduces depressive-like behavior in normal and chronically stressed C57BL/6 mice. Here, we extend our previous studies by investigating the effects of cotinine on the progression of AD-like pathology, depressive-like behavior, and the mechanisms underlying its beneficial effects in Tg6799 mice when left untreated until after a more advanced stage of the disease's development. The results show that vehicle-treated Tg6799 mice displayed an accentuated loss of working memory and an abundant Aβ plaque pathology that were accompanied by higher levels of depressive-like behavior as compared to control littermates. By contrast, prolonged daily cotinine treatment to Tg6799 mice, withheld until after a mid-level progression of AD-like pathology, reduced Aβ levels/plaques and depressive-like behavior. Moreover, this treatment paradigm dramatically improved working memory as compared to control littermates. The beneficial effects of cotinine were accompanied by an increase in the expression of the active form of protein kinase B and the postsynaptic density protein 95 in the hippocampi and frontal cortices of Tg6799 mice. This suggests that cotinine halts the progression of AD-like pathology while reducing depressive-like behavior by stimulating signaling pathways supporting synaptic plasticity in Tg6799 mice. The potential use of cotinine to treat cognitive and non-cognitive symptoms of AD is discussed.
Collapse
Affiliation(s)
- Sagar Patel
- Research and Development Service, Department of Veterans Affairs, Bay Pines VA Healthcare System Bay Pines, FL, USA
| | - J Alex Grizzell
- Research and Development Service, Department of Veterans Affairs, Bay Pines VA Healthcare System Bay Pines, FL, USA ; Department of Psychiatry and Behavioral Neurosciences, Morsani College of Medicine, University of South Florida Tampa, FL, USA
| | - Rosalee Holmes
- Research and Development Service, Department of Veterans Affairs, Bay Pines VA Healthcare System Bay Pines, FL, USA
| | - Ross Zeitlin
- Research and Development Service, Department of Veterans Affairs, Bay Pines VA Healthcare System Bay Pines, FL, USA
| | - Rosalynn Solomon
- Research and Development Service, Department of Veterans Affairs, Bay Pines VA Healthcare System Bay Pines, FL, USA
| | - Thomas L Sutton
- Research and Development Service, Department of Veterans Affairs, Bay Pines VA Healthcare System Bay Pines, FL, USA
| | - Adeeb Rohani
- Research and Development Service, Department of Veterans Affairs, Bay Pines VA Healthcare System Bay Pines, FL, USA
| | - Laura C Charry
- Research and Development Service, Department of Veterans Affairs, Bay Pines VA Healthcare System Bay Pines, FL, USA
| | - Alexandre Iarkov
- Research and Development Service, Department of Veterans Affairs, Bay Pines VA Healthcare System Bay Pines, FL, USA ; Center of Research in Biomedical Sciences, Universidad Autónoma de Chile Santiago, Chile
| | - Takashi Mori
- Departments of Biomedical Sciences and Pathology, Saitama Medical Center and Saitama Medical University Kawagoe, Saitama, Japan
| | - Valentina Echeverria Moran
- Research and Development Service, Department of Veterans Affairs, Bay Pines VA Healthcare System Bay Pines, FL, USA ; Center of Research in Biomedical Sciences, Universidad Autónoma de Chile Santiago, Chile ; Research Service, Department of Veterans Affairs, Tampa VA Healthcare System Tampa, FL, USA ; Department of Molecular Medicine, Morsani College of Medicine, University of South Florida Tampa, FL, USA
| |
Collapse
|
83
|
Lai EY, Chen ZG, Zhou X, Fan XR, Wang H, Lai PL, Su YC, Zhang BY, Bai XC, Li YF. DEPTOR Expression Negatively Correlates with mTORC1 Activity and Tumor Progression in Colorectal Cancer. Asian Pac J Cancer Prev 2014; 15:4589-94. [DOI: 10.7314/apjcp.2014.15.11.4589] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
84
|
Perluigi M, Pupo G, Tramutola A, Cini C, Coccia R, Barone E, Head E, Butterfield DA, Di Domenico F. Neuropathological role of PI3K/Akt/mTOR axis in Down syndrome brain. Biochim Biophys Acta Mol Basis Dis 2014; 1842:1144-53. [PMID: 24735980 DOI: 10.1016/j.bbadis.2014.04.007] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 03/19/2014] [Accepted: 04/07/2014] [Indexed: 01/11/2023]
Abstract
Down syndrome (DS) is the most frequent genetic cause of intellectual disability characterized by the presence of three copies of chromosome 21 (Chr21). Individuals with DS have sufficient neuropathology for a diagnosis of Alzheimer's disease (AD) after the age of 40years. The aim of our study is to gain new insights in the molecular mechanisms impaired in DS subjects that eventually lead to the development of dementia. We evaluate the PI3K/Akt/mTOR axis in the frontal cortex from DS cases (under the age of 40years) and DS with AD neuropathology compared with age-matched controls (Young and Old). The PI3K/Akt/mTOR axis may control several key pathways involved in AD that, if aberrantly regulated, affect amyloid beta (Aβ) deposition and tau phosphorylation. Our results show a hyperactivation of PI3K/Akt/mTOR axis in individuals with DS, with and without AD pathology, in comparison with respective controls. The PI3K/Akt/mTOR deregulation results in decreased autophagy, inhibition of IRS1 and GSK3β activity. Moreover, our data suggest that aberrant activation of the PI3K/Akt/mTOR axis acts in parallel to RCAN1 in phosphorylating tau, in DS and DS/AD. In conclusion, this study provides insights into the neuropathological mechanisms that may be engaged during the development of AD in DS. We suggest that deregulation of this signaling cascade is already evident in young DS cases and persist in the presence of AD pathology. The impairment of the PI3K/Akt/mTOR axis in DS population might represent a key-contributing factor to the neurodegenerative process that culminates in Alzheimer-like dementia.
Collapse
Affiliation(s)
- Marzia Perluigi
- Department of Biochemical Sciences, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Gilda Pupo
- Department of Biochemical Sciences, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Antonella Tramutola
- Department of Biochemical Sciences, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Chiara Cini
- Department of Biochemical Sciences, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Raffaella Coccia
- Department of Biochemical Sciences, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Eugenio Barone
- Department of Biochemical Sciences, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Elizabeth Head
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506-0055, USA
| | - D Allan Butterfield
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506-0055, USA; Department of Chemistry and Center of Membrane Sciences, University of Kentucky, Lexington, KY 40506-0055, USA
| | - Fabio Di Domenico
- Department of Biochemical Sciences, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy.
| |
Collapse
|
85
|
Li L, Zhang Q, Tan J, Fang Y, An X, Chen B. Autophagy and hippocampal neuronal injury. Sleep Breath 2014; 18:243-9. [DOI: 10.1007/s11325-013-0930-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Revised: 12/11/2013] [Accepted: 12/16/2013] [Indexed: 01/07/2023]
|