151
|
Zare-Shahabadi A, Masliah E, Johnson GVW, Rezaei N. Autophagy in Alzheimer's disease. Rev Neurosci 2015; 26:385-95. [PMID: 25870960 DOI: 10.1515/revneuro-2014-0076] [Citation(s) in RCA: 147] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 02/20/2015] [Indexed: 12/12/2022]
Abstract
Autophagy is a vesicle and lysosome-mediated degradative pathway that is essential for protein homeostasis and cell health. In particular, compared to nonneuronal cells, neurons are dependent on high basal autophagy for survival. There is emerging agreement that defects in autophagy are likely to contribute to the neurodegenerative processes in numerous diseases, including Alzheimer's disease (AD). Autophagy-lysosome defects occur early in the pathogenesis of AD and have been proposed to be a significant contributor to the disease process. Given the fact that autophagy deficits are likely major contributors to the etiology of AD, the focus of this review will be on recent studies that support a role for autophagy deficits in AD.
Collapse
|
152
|
Yang SH, Li W, Sumien N, Forster M, Simpkins JW, Liu R. Alternative mitochondrial electron transfer for the treatment of neurodegenerative diseases and cancers: Methylene blue connects the dots. Prog Neurobiol 2015; 157:273-291. [PMID: 26603930 DOI: 10.1016/j.pneurobio.2015.10.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 09/10/2015] [Accepted: 10/20/2015] [Indexed: 12/21/2022]
Abstract
Brain has exceptional high requirement for energy metabolism with glucose as the exclusive energy source. Decrease of brain energy metabolism and glucose uptake has been found in patients of Alzheimer's, Parkinson's and other neurodegenerative diseases, providing a clear link between neurodegenerative disorders and energy metabolism. On the other hand, cancers, including glioblastoma, have increased glucose uptake and rely on aerobic glycolysis for energy metabolism. The switch of high efficient oxidative phosphorylation to low efficient aerobic glycolysis pathway (Warburg effect) provides macromolecule for biosynthesis and proliferation. Current research indicates that methylene blue, a century old drug, can receive electron from NADH in the presence of complex I and donates it to cytochrome c, providing an alternative electron transfer pathway. Methylene blue increases oxygen consumption, decrease glycolysis, and increases glucose uptake in vitro. Methylene blue enhances glucose uptake and regional cerebral blood flow in rats upon acute treatment. In addition, methylene blue provides protective effect in neuron and astrocyte against various insults in vitro and in rodent models of Alzheimer's, Parkinson's, and Huntington's disease. In glioblastoma cells, methylene blue reverses Warburg effect by enhancing mitochondrial oxidative phosphorylation, arrests glioma cell cycle at s-phase, and inhibits glioma cell proliferation. Accordingly, methylene blue activates AMP-activated protein kinase, inhibits downstream acetyl-coA carboxylase and cyclin-dependent kinases. In summary, there is accumulating evidence providing a proof of concept that enhancement of mitochondrial oxidative phosphorylation via alternative mitochondrial electron transfer may offer protective action against neurodegenerative diseases and inhibit cancers proliferation.
Collapse
Affiliation(s)
- Shao-Hua Yang
- Center for Neuroscience Discovery, University of North Texas Health Science Center, Fort Worth, TX 76107, USA.
| | - Wenjun Li
- Center for Neuroscience Discovery, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Nathalie Sumien
- Center for Neuroscience Discovery, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Michael Forster
- Center for Neuroscience Discovery, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - James W Simpkins
- Department of Physiology and Pharmacology, Center for Neuroscience, Health Science Center, West Virginia University, Medical Center Drive, Morgantown, WV 26506, USA
| | - Ran Liu
- Center for Neuroscience Discovery, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| |
Collapse
|
153
|
Zhao M, Liang F, Xu H, Yan W, Zhang J. Methylene blue exerts a neuroprotective effect against traumatic brain injury by promoting autophagy and inhibiting microglial activation. Mol Med Rep 2015; 13:13-20. [PMID: 26572258 PMCID: PMC4686104 DOI: 10.3892/mmr.2015.4551] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Accepted: 05/21/2015] [Indexed: 11/12/2022] Open
Abstract
Traumatic brain injury (TBI) leads to permanent neurological impairment, and methylene blue (MB) exerts central nervous system neuroprotective effects. However, only one previous study has investigated the effectiveness of MB in a controlled cortical impact injury model of TBI. In addition, the specific mechanisms underlying the effect of MB against TBI remain to be elucidated. Therefore, the present study investigated the neuroprotective effect of MB on TBI and the possible mechanisms involved. In a mouse model of TBI, the animals were randomly divided into sham, vehicle (normal saline) or MB groups. The treatment time-points were 24 and 72 h (acute phase of TBI), and 14 days (chronic phase of TBI) post-TBI. The brain water content (BWC), and levels of neuronal death, and autophagy were determined during the acute phase, and neurological deficit, injury volume and microglial activation were assessed at all time-points. The injured hemisphere BWC was significantly increased 24 h post-TBI, and this was attenuated following treatment with MB. There was a significantly higher number of surviving neurons in the MB group, compared with the Vehicle group at 24 and 72 h post-TBI. In the acute phase, the MB-treated animals exhibited significantly upregulated expression of Beclin 1 and increased LC3-II to LC3-I ratios, compared with the vehicle group, indicating an increased rate of autophagy. Neurological functional deficits, measured using the modified neurological severity score, were significantly lower in the acute phase in the MB-treated animals and cerebral lesion volumes in the MB-treated animals were significantly lower, compared with the other groups at all time-points. Microglia were activated 24 h after TBI, peaked at 72 h and persisted until 14 days after TBI. Although the number of Iba-1-positive cells in the vehicle and MB groups 24 h post-TBI were not significantly different, marked microglial inhibition was observed in the MB group 72 h and 14 days after-TBI. These results indicated that MB exerts a neuroprotective effect by increasing autophagy, decreasing brain edema and inhibiting microglial activation.
Collapse
Affiliation(s)
- Mingfei Zhao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Feng Liang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Hangdi Xu
- Department of Respiratory Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China
| | - Wei Yan
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Jianmin Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| |
Collapse
|
154
|
Siman R, Cocca R, Dong Y. The mTOR Inhibitor Rapamycin Mitigates Perforant Pathway Neurodegeneration and Synapse Loss in a Mouse Model of Early-Stage Alzheimer-Type Tauopathy. PLoS One 2015; 10:e0142340. [PMID: 26540269 PMCID: PMC4634963 DOI: 10.1371/journal.pone.0142340] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 10/20/2015] [Indexed: 11/19/2022] Open
Abstract
The perforant pathway projection from layer II of the entorhinal cortex to the hippocampal dentate gyrus is especially important for long-term memory formation, and is preferentially vulnerable to developing a degenerative tauopathy early in Alzheimer's disease (AD) that may spread over time trans-synaptically. Despite the importance of the perforant pathway to the clinical onset and progression of AD, a therapeutic has not been identified yet that protects it from tau-mediated toxicity. Here, we used an adeno-associated viral vector-based mouse model of early-stage AD-type tauopathy to investigate effects of the mTOR inhibitor and autophagy stimulator rapamycin on the tau-driven loss of perforant pathway neurons and synapses. Focal expression of human tau carrying a P301L mutation but not eGFP as a control in layer II of the lateral entorhinal cortex triggered rapid degeneration of these neurons, loss of lateral perforant pathway synapses in the dentate gyrus outer molecular layer, and activation of neuroinflammatory microglia and astroglia in the two locations. Chronic systemic rapamycin treatment partially inhibited phosphorylation of a mechanistic target of rapamycin substrate in brain and stimulated LC3 cleavage, a marker of autophagic flux. Compared with vehicle-treated controls, rapamycin protected against the tau-induced neuronal loss, synaptotoxicity, reactive microgliosis and astrogliosis, and activation of innate neuroimmunity. It did not alter human tau mRNA or total protein levels. Finally, rapamycin inhibited trans-synaptic transfer of human tau expression to the dentate granule neuron targets for the perforant pathway, likely by preventing the synaptic spread of the AAV vector in response to pathway degeneration. These results identify systemic rapamycin as a treatment that protects the entorhinal cortex and perforant pathway projection from tau-mediated neurodegeneration, axonal and synapse loss, and neuroinflammatory reactive gliosis. The findings support the potential for slowing the progression of AD by abrogating tau-mediated neurotoxicity at its earliest neuropathological stages.
Collapse
Affiliation(s)
- Robert Siman
- Department of Neurosurgery, Center for Brain Injury and Repair, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Ryan Cocca
- Department of Neurosurgery, Center for Brain Injury and Repair, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Yina Dong
- Department of Neurosurgery, Center for Brain Injury and Repair, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
155
|
Li YQ, Tan MS, Yu JT, Tan L. Frontotemporal Lobar Degeneration: Mechanisms and Therapeutic Strategies. Mol Neurobiol 2015; 53:6091-6105. [PMID: 26537902 DOI: 10.1007/s12035-015-9507-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 10/20/2015] [Indexed: 12/11/2022]
Abstract
Frontotemporal lobar degeneration (FTLD) is characterized by progressive deterioration of frontal and anterior temporal lobes of the brain and often exhibits frontotemporal dementia (FTD) on clinic, in <65-year-old patients at the time of diagnosis. Interdisciplinary approaches combining genetics, molecular and cell biology, and laboratory animal science have revealed some of its potential molecular mechanisms. Although there is still no effective treatment to delay, prevent, and reverse the progression of FTD, emergence of agents targeting molecular mechanisms has been beginning to promote potential pharmaceutical development. Our review summarizes the latest new findings of FTLD and challenges in FTLD therapy.
Collapse
Affiliation(s)
- Ya-Qing Li
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, China
| | - Meng-Shan Tan
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, China
| | - Jin-Tai Yu
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, China. .,Memory and Aging Center, Department of Neurology, University of California, San Francisco, CA, USA.
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, China.
| |
Collapse
|
156
|
Wagner J, Krauss S, Shi S, Ryazanov S, Steffen J, Miklitz C, Leonov A, Kleinknecht A, Göricke B, Weishaupt JH, Weckbecker D, Reiner AM, Zinth W, Levin J, Ehninger D, Remy S, Kretzschmar HA, Griesinger C, Giese A, Fuhrmann M. Reducing tau aggregates with anle138b delays disease progression in a mouse model of tauopathies. Acta Neuropathol 2015; 130:619-31. [PMID: 26439832 PMCID: PMC4612332 DOI: 10.1007/s00401-015-1483-3] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 09/24/2015] [Accepted: 09/24/2015] [Indexed: 12/29/2022]
Abstract
Pathological tau aggregation leads to filamentous tau inclusions and characterizes neurodegenerative tauopathies such as Alzheimer’s disease and frontotemporal dementia and parkinsonism linked to chromosome 17. Tau aggregation coincides with clinical symptoms and is thought to mediate neurodegeneration. Transgenic mice overexpressing mutant human P301S tau exhibit many neuropathological features of human tauopathies including behavioral deficits and increased mortality. Here, we show that the di-phenyl-pyrazole anle138b binds to aggregated tau and inhibits tau aggregation in vitro and in vivo. Furthermore, anle138b treatment effectively ameliorates disease symptoms, increases survival time and improves cognition of tau transgenic PS19 mice. In addition, we found decreased synapse and neuron loss accompanied by a decreased gliosis in the hippocampus. Our results suggest that reducing tau aggregates with anle138b may represent an effective and promising approach for the treatment of human tauopathies.
Collapse
|
157
|
Reducing Aβ load and tau phosphorylation: Emerging perspective for treating Alzheimer's disease. Eur J Pharmacol 2015. [DOI: 10.1016/j.ejphar.2015.07.043] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
158
|
Mattson MP. Late-onset dementia: a mosaic of prototypical pathologies modifiable by diet and lifestyle. NPJ Aging Mech Dis 2015. [PMID: 28642821 PMCID: PMC5478237 DOI: 10.1038/npjamd.2015.3] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Idiopathic late-onset dementia (ILOD) describes impairments of memory, reasoning and/or social abilities in the elderly that compromise their daily functioning. Dementia occurs in several major prototypical neurodegenerative disorders that are currently defined by neuropathological criteria, most notably Alzheimer’s disease (AD), Lewy body dementia (LBD), frontotemporal dementia (FTD) and hippocampal sclerosis of aging (HSA). However, people who die with ILOD commonly exhibit mixed pathologies that vary within and between brain regions. Indeed, many patients diagnosed with probable AD exhibit only modest amounts of disease-defining amyloid β-peptide plaques and p-Tau tangles, and may have features of FTD (TDP-43 inclusions), Parkinson’s disease (α-synuclein accumulation), HSA and vascular lesions. Here I argue that this ‘mosaic neuropathological landscape’ is the result of commonalities in aging-related processes that render neurons vulnerable to the entire spectrum of ILODs. In this view, all ILODs involve deficits in neuronal energy metabolism, neurotrophic signaling and adaptive cellular stress responses, and associated dysregulation of neuronal calcium handling and autophagy. Although this mosaic of neuropathologies and underlying mechanisms poses major hurdles for development of disease-specific therapeutic interventions, it also suggests that certain interventions would be beneficial for all ILODs. Indeed, emerging evidence suggests that the brain can be protected against ILOD by lifelong intermittent physiological challenges including exercise, energy restriction and intellectual endeavors; these interventions enhance cellular stress resistance and facilitate neuroplasticity. There is also therapeutic potential for interventions that bolster neuronal bioenergetics and/or activate one or more adaptive cellular stress response pathways in brain cells. A wider appreciation that all ILODs share age-related cellular and molecular alterations upstream of aggregated protein lesions, and that these upstream events can be mitigated, may lead to implementation of novel intervention strategies aimed at reversing the rising tide of ILODs.
Collapse
Affiliation(s)
- Mark P Mattson
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD 21224.,Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| |
Collapse
|
159
|
Pakavathkumar P, Sharma G, Kaushal V, Foveau B, LeBlanc AC. Methylene Blue Inhibits Caspases by Oxidation of the Catalytic Cysteine. Sci Rep 2015; 5:13730. [PMID: 26400108 PMCID: PMC4585840 DOI: 10.1038/srep13730] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 08/03/2015] [Indexed: 12/20/2022] Open
Abstract
Methylene blue, currently in phase 3 clinical trials against Alzheimer Disease, disaggregates the Tau protein of neurofibrillary tangles by oxidizing specific cysteine residues. Here, we investigated if methylene blue can inhibit caspases via the oxidation of their active site cysteine. Methylene blue, and derivatives, azure A and azure B competitively inhibited recombinant Caspase-6 (Casp6), and inhibited Casp6 activity in transfected human colon carcinoma cells and in serum-deprived primary human neuron cultures. Methylene blue also inhibited recombinant Casp1 and Casp3. Furthermore, methylene blue inhibited Casp3 activity in an acute mouse model of liver toxicity. Mass spectrometry confirmed methylene blue and azure B oxidation of the catalytic Cys163 cysteine of Casp6. Together, these results show a novel inhibitory mechanism of caspases via sulfenation of the active site cysteine. These results indicate that methylene blue or its derivatives could (1) have an additional effect against Alzheimer Disease by inhibiting brain caspase activity, (2) be used as a drug to prevent caspase activation in other conditions, and (3) predispose chronically treated individuals to cancer via the inhibition of caspases.
Collapse
Affiliation(s)
- Prateep Pakavathkumar
- Bloomfield Center for Research in Aging, Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Gyanesh Sharma
- Bloomfield Center for Research in Aging, Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Vikas Kaushal
- Bloomfield Center for Research in Aging, Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Bénédicte Foveau
- Bloomfield Center for Research in Aging, Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Andrea C. LeBlanc
- Bloomfield Center for Research in Aging, Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
160
|
Violet M, Chauderlier A, Delattre L, Tardivel M, Chouala MS, Sultan A, Marciniak E, Humez S, Binder L, Kayed R, Lefebvre B, Bonnefoy E, Buée L, Galas MC. Prefibrillar Tau oligomers alter the nucleic acid protective function of Tau in hippocampal neurons in vivo. Neurobiol Dis 2015; 82:540-551. [PMID: 26385829 DOI: 10.1016/j.nbd.2015.09.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2015] [Revised: 07/09/2015] [Accepted: 09/13/2015] [Indexed: 01/05/2023] Open
Abstract
The accumulation of DNA and RNA oxidative damage is observed in cortical and hippocampal neurons from Alzheimer's disease (AD) brains at early stages of pathology. We recently reported that Tau is a key nuclear player in the protection of neuronal nucleic acid integrity in vivo under physiological conditions and hyperthermia, a strong inducer of oxidative stress. In a mouse model of tauopathy (THY-Tau22), we demonstrate that hyperthermia selectively induces nucleic acid oxidative damage and nucleic acid strand breaks in the nucleus and cytoplasm of hippocampal neurons that display early Tau phosphorylation but no Tau fibrils. Nucleic acid-damaged neurons were exclusively immunoreactive for prefibrillar Tau oligomers. A similar association between prefibrillar Tau oligomers and nucleic acid oxidative damage was observed in AD brains. Pretreatment with Methylene Blue (MB), a Tau aggregation inhibitor and a redox cycler, reduced hyperthermia-induced Tau oligomerization as well as nucleic acid damage. This study clearly highlights the existence of an early and critical time frame for hyperthermia-induced Tau oligomerization, which most likely occurs through increased oxidative stress, and nucleic acid vulnerability during the progression of Tau pathology. These results suggest that at early stages of AD, Tau oligomerization triggers the loss of the nucleic acid protective function of monomeric Tau. This study highlights the existence of a short therapeutic window in which to prevent the formation of pathological forms of Tau and their harmful consequences on nucleic acid integrity during the progression of Tau pathology.
Collapse
Affiliation(s)
- Marie Violet
- Inserm, UMRS1172, JPArc, Alzheimer & Tauopathies, 1 rue Polonovski, 59045 Lille, France; Université de Lille, Faculté de Médecine, Lille, France; CHRU, Memory Clinic, Lille, France
| | - Alban Chauderlier
- Inserm, UMRS1172, JPArc, Alzheimer & Tauopathies, 1 rue Polonovski, 59045 Lille, France; Université de Lille, Faculté de Médecine, Lille, France; CHRU, Memory Clinic, Lille, France
| | - Lucie Delattre
- Inserm, UMRS1172, JPArc, Alzheimer & Tauopathies, 1 rue Polonovski, 59045 Lille, France; Université de Lille, Faculté de Médecine, Lille, France; CHRU, Memory Clinic, Lille, France
| | - Meryem Tardivel
- Inserm, UMRS1172, JPArc, Alzheimer & Tauopathies, 1 rue Polonovski, 59045 Lille, France; Université de Lille, Faculté de Médecine, Lille, France; CHRU, Memory Clinic, Lille, France
| | - Meliza Sendid Chouala
- Inserm, UMRS1172, JPArc, Alzheimer & Tauopathies, 1 rue Polonovski, 59045 Lille, France; Université de Lille, Faculté de Médecine, Lille, France; CHRU, Memory Clinic, Lille, France
| | - Audrey Sultan
- Inserm, UMRS1172, JPArc, Alzheimer & Tauopathies, 1 rue Polonovski, 59045 Lille, France; Université de Lille, Faculté de Médecine, Lille, France; CHRU, Memory Clinic, Lille, France
| | - Elodie Marciniak
- Inserm, UMRS1172, JPArc, Alzheimer & Tauopathies, 1 rue Polonovski, 59045 Lille, France; Université de Lille, Faculté de Médecine, Lille, France; CHRU, Memory Clinic, Lille, France
| | - Sandrine Humez
- Inserm, UMRS1172, JPArc, Alzheimer & Tauopathies, 1 rue Polonovski, 59045 Lille, France; Université de Lille, Faculté de Médecine, Lille, France; CHRU, Memory Clinic, Lille, France
| | - Lester Binder
- Department of Translational Science & Molecular Medicine, College of Human Medicine, Michigan State University, 333 Bostwick Ave. NE, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Rakez Kayed
- Department of Neurology, George and Cynthia Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, 301 University Blvd, Medical Research Building, Room 10.138C, Galveston, TX 77555-1045, USA; Department of Neuroscience & Cell Biology, George and Cynthia Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, 301 University Blvd, Medical Research Building, Room 10.138C, Galveston, TX 77555-1045, USA
| | - Bruno Lefebvre
- Inserm, UMRS1172, JPArc, Alzheimer & Tauopathies, 1 rue Polonovski, 59045 Lille, France; Université de Lille, Faculté de Médecine, Lille, France; CHRU, Memory Clinic, Lille, France
| | - Eliette Bonnefoy
- Inserm UMRS 1007, Université Paris Descartes, 45 rue des Saints Pères, 75006 Paris Cedex 06, France
| | - Luc Buée
- Inserm, UMRS1172, JPArc, Alzheimer & Tauopathies, 1 rue Polonovski, 59045 Lille, France; Université de Lille, Faculté de Médecine, Lille, France; CHRU, Memory Clinic, Lille, France
| | - Marie-Christine Galas
- Inserm, UMRS1172, JPArc, Alzheimer & Tauopathies, 1 rue Polonovski, 59045 Lille, France; Université de Lille, Faculté de Médecine, Lille, France; CHRU, Memory Clinic, Lille, France.
| |
Collapse
|
161
|
The Role of MAPT in Neurodegenerative Diseases: Genetics, Mechanisms and Therapy. Mol Neurobiol 2015; 53:4893-904. [PMID: 26363795 DOI: 10.1007/s12035-015-9415-8] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 09/01/2015] [Indexed: 12/11/2022]
Abstract
Microtubule-associated protein tau (MAPT) is a gene responsible for encoding tau protein, which is tightly implicated in keeping the function of microtubules and axonal transport. Hyperphosphorylated tau protein participates in the formation of neurofibrillary tangles (NFTs), which characterize many neurodegenerative disorders termed tauopathies. Genome-wide association studies (GWAS) have demonstrated numerous single nucleotide polymorphisms (SNPs) located in MAPT associated with various neurodegenerative diseases. Thus, it has been presumed that MAPT plays a crucial role in pathogenesis of neurodegeneration via affecting the structure and function of tau. Here, we review the advanced studies to summarize the biochemical properties of MAPT and its encoded protein, as well as the genetics and epigenetics of MAPT in neurodegeneration. Finally, given the potential mechanisms of MAPT to neurodegeneration pathogenesis, targeting MAPT and tau might present significant treatments of MAPT mutation-related neurodegeneration. Affirmatively, the identification of MAPT is extremely beneficial for improving our understanding of the pathogenesis of various neurodegenerative diseases and developing the mechanism-based therapies.
Collapse
|
162
|
Jiang Z, Watts LT, Huang S, Shen Q, Rodriguez P, Chen C, Zhou C, Duong TQ. The Effects of Methylene Blue on Autophagy and Apoptosis in MRI-Defined Normal Tissue, Ischemic Penumbra and Ischemic Core. PLoS One 2015; 10:e0131929. [PMID: 26121129 PMCID: PMC4488003 DOI: 10.1371/journal.pone.0131929] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Accepted: 06/09/2015] [Indexed: 12/11/2022] Open
Abstract
Methylene blue (MB) USP, which has energy-enhancing and antioxidant properties, is currently used to treat methemoglobinemia and cyanide poisoning in humans. We recently showed that MB administration reduces infarct volume and behavioral deficits in rat models of ischemic stroke and traumatic brain injury. This study reports the underlying molecular mechanisms of MB neuroprotection following transient ischemic stroke in rats. Rats were subjected to transient (60-mins) ischemic stroke. Multimodal MRI during the acute phase and at 24 hrs were used to define three regions of interest (ROIs): i) the perfusion-diffusion mismatch salvaged by reperfusion, ii) the perfusion-diffusion mismatch not salvaged by reperfusion, and iii) the ischemic core. The tissues from these ROIs were extracted for western blot analyses of autophagic and apoptotic markers. The major findings were: 1) MB treatment reduced infarct volume and behavioral deficits, 2) MB improved cerebral blood flow to the perfusion-diffusion mismatch tissue after reperfusion and minimized harmful hyperperfusion 24 hrs after stroke, 3) MB inhibited apoptosis and enhanced autophagy in the perfusion-diffusion mismatch, 4) MB inhibited apoptotic signaling cascades (p53-Bax-Bcl2-Caspase3), and 5) MB enhanced autophagic signaling cascades (p53-AMPK-TSC2-mTOR). MB induced neuroprotection, at least in part, by enhancing autophagy and reducing apoptosis in the perfusion-diffusion mismatch tissue following ischemic stroke.
Collapse
Affiliation(s)
- Zhao Jiang
- Department of Anatomy and Embryology, Peking University Health Science Center, Beijing, China
- Research Imaging Institute, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Lora Talley Watts
- Research Imaging Institute, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Shiliang Huang
- Research Imaging Institute, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Qiang Shen
- Research Imaging Institute, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Pavel Rodriguez
- Research Imaging Institute, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Chunhua Chen
- Department of Anatomy and Embryology, Peking University Health Science Center, Beijing, China
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Changman Zhou
- Department of Anatomy and Embryology, Peking University Health Science Center, Beijing, China
| | - Timothy Q. Duong
- Research Imaging Institute, University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| |
Collapse
|
163
|
Sheik Mohideen S, Yamasaki Y, Omata Y, Tsuda L, Yoshiike Y. Nontoxic singlet oxygen generator as a therapeutic candidate for treating tauopathies. Sci Rep 2015; 5:10821. [PMID: 26027742 PMCID: PMC4450544 DOI: 10.1038/srep10821] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Accepted: 04/30/2015] [Indexed: 11/09/2022] Open
Abstract
Methylene blue (MB) inhibits the aggregation of tau, a main constituent of neurofibrillary tangles. However, MB's mode of action in vivo is not fully understood. MB treatment reduced the amount of sarkosyl-insoluble tau in Drosophila that express human wild-type tau. MB concurrently ameliorated the climbing deficits of transgenic tau flies to a limited extent and diminished the climbing activity of wild-type flies. MB also decreased the survival rate of wild-type flies. Based on its photosensitive efficacies, we surmised that singlet oxygen generated through MB under light might contribute to both the beneficial and toxic effects of MB in vivo. We identified rose bengal (RB) that suppressed tau accumulation and ameliorated the behavioral deficits to a lesser extent than MB. Unlike MB, RB did not reduce the survival rate of flies. Our findings indicate that singlet oxygen generators with little toxicity may be suitable drug candidates for treating tauopathies.
Collapse
Affiliation(s)
- Sahabudeen Sheik Mohideen
- Alzheimer's Disease Project Team, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, 7-430 Morioka, Obu, Aichi, Japan 474-8511
| | - Yasutoyo Yamasaki
- Department of Drug Discovery, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, 7-430 Morioka, Obu, Aichi, Japan 474-8511
| | - Yasuhiro Omata
- Department of Occupational and Environmental Health, Graduate School of Medicine, Nagoya University, 65 Tsurumai-cho, Showa-ku, Nagoya, Japan 466-8550
| | - Leo Tsuda
- Animal Models of Aging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, 7-430 Morioka, Obu, Aichi, Japan 474-8511
| | - Yuji Yoshiike
- Alzheimer's Disease Project Team, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, 7-430 Morioka, Obu, Aichi, Japan 474-8511
| |
Collapse
|
164
|
Di Y, He YL, Zhao T, Huang X, Wu KW, Liu SH, Zhao YQ, Fan M, Wu LY, Zhu LL. Methylene Blue Reduces Acute Cerebral Ischemic Injury via the Induction of Mitophagy. Mol Med 2015; 21:420-9. [PMID: 25998511 DOI: 10.2119/molmed.2015.00038] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2015] [Accepted: 05/19/2015] [Indexed: 01/19/2023] Open
Abstract
The treatment of stroke is limited by a short therapeutic window and a lack of effective clinical drugs. Methylene blue (MB) has been used in laboratories and clinics since the 1890s. Few studies have reported the neuroprotective role of MB in cerebral ischemia-reperfusion injury. However, whether and how MB protects against acute cerebral ischemia (ACI) injury was unclear. In this study, we investigated the effect of MB on this injury and revealed that MB protected against ACI injury by augmenting mitophagy. Using a rat middle cerebral artery occlusion (MCAO) model, we demonstrated that MB improved neurological function and reduced the infarct volume and necrosis after ACI injury. These improvements depended on the effect of MB on mitochondrial structure and function. ACI caused the disorder and disintegration of mitochondrial structure, while MB ameliorated the destruction of mitochondria. In addition, mitophagy was inhibited at 24 h after stroke and MB augmented mitophagy. In an oxygen-glucose deprivation (OGD) model in vitro, we further revealed that the elevation of mitochondrial membrane potential (MMP) by MB under OGD conditions mediated the augmented mitophagy. In contrast, exacerbating the decline of MMP during OGD abolished the MB-induced activation of mitophagy. Taken together, MB promotes mitophagy by maintaining the MMP at a relatively high level, which contributes to a decrease in necrosis and an improvement in neurological function, thereby protecting against ACI injury.
Collapse
Affiliation(s)
- Yao Di
- Department of Cognitive Science, Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China
| | - Yun-Ling He
- Department of Cognitive Science, Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China
| | - Tong Zhao
- Department of Cognitive Science, Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China
| | - Xin Huang
- Department of Cognitive Science, Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China
| | - Kui-Wu Wu
- Department of Cognitive Science, Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China
| | - Shu-Hong Liu
- Department of Cognitive Science, Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China
| | - Yong-Qi Zhao
- Department of Cognitive Science, Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China
| | - Ming Fan
- Department of Cognitive Science, Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, People's Republic of China.,Beijing Institute for Brain Disorders, Beijing, People's Republic of China
| | - Li-Ying Wu
- Department of Cognitive Science, Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China
| | - Ling-Ling Zhu
- Department of Cognitive Science, Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, People's Republic of China
| |
Collapse
|
165
|
Hochgräfe K, Sydow A, Matenia D, Cadinu D, Könen S, Petrova O, Pickhardt M, Goll P, Morellini F, Mandelkow E, Mandelkow EM. Preventive methylene blue treatment preserves cognition in mice expressing full-length pro-aggregant human Tau. Acta Neuropathol Commun 2015; 3:25. [PMID: 25958115 PMCID: PMC4425867 DOI: 10.1186/s40478-015-0204-4] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 04/16/2015] [Indexed: 11/21/2022] Open
Abstract
Introduction Neurofibrillary tangles (NFT) composed of Tau are hallmarks of neurodegeneration in Alzheimer disease. Transgenic mice expressing full-length pro-aggregant human Tau (2N4R Tau-ΔK280, termed TauΔK) or its repeat domain (TauRD-ΔK280, TauRDΔK) develop a progressive Tau pathology with missorting, phosphorylation, aggregation of Tau, loss of synapses and functional deficits. Whereas TauRDΔK assembles into NFT concomitant with neuronal death, TauΔK accumulates into Tau pretangles without overt neuronal loss. Both forms cause a comparable cognitive decline (with onset at 10mo and 12mo, respectively), which is rescued upon switch-off of transgene expression. Since methylene blue (MB) is able to inhibit Tau aggregation in vitro, we investigated whether MB can prevent or rescue Tau-induced cognitive impairments in our mouse models. Both types of mice received MB orally using different preventive and therapeutic treatment protocols, initiated either before or after disease onset. The cognitive status of the mice was assessed by behavior tasks (open field, Morris water maze) to determine the most successful conditions for therapeutic intervention. Results Preventive and therapeutic MB application failed to avert or recover learning and memory deficits of TauRDΔK mice. Similarly, therapeutic MB treatment initiated after onset of cognitive impairments was ineffective in TauΔK mice. In contrast, preventive MB application starting before onset of functional deficits preserved cognition of TauΔK mice. Beside improved learning and memory, MB-treated TauΔK mice showed a strong decrease of insoluble Tau, a reduction of conformationally changed (MC1) and phosphorylated Tau species (AT180, PHF1) as well as an upregulation of protein degradation systems (autophagy and proteasome). This argues for additional pleiotropic effects of MB beyond its properties as Tau aggregation inhibitor. Conclusions Our data support the use of Tau aggregation inhibitors as potential drugs for the treatment of AD and other tauopathies and highlights the need for preventive treatment before onset of cognitive impairments. Electronic supplementary material The online version of this article (doi:10.1186/s40478-015-0204-4) contains supplementary material, which is available to authorized users.
Collapse
|
166
|
Martin MD, Calcul L, Smith C, Jinwal UK, Fontaine SN, Darling A, Seeley K, Wojtas L, Narayan M, Gestwicki JE, Smith GR, Reitz AB, Baker BJ, Dickey CA. Synthesis, stereochemical analysis, and derivatization of myricanol provide new probes that promote autophagic tau clearance. ACS Chem Biol 2015; 10:1099-109. [PMID: 25588114 DOI: 10.1021/cb501013w] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
We previously discovered that one specific scalemic preparation of myricanol (1), a constituent of Myrica cerifera (bayberry/southern wax myrtle) root bark, could lower the levels of the microtubule-associated protein tau (MAPT). The significance is that tau accumulates in a number of neurodegenerative diseases, the most common being Alzheimer's disease (AD). Herein, a new synthetic route to prepare myricanol using a suitable boronic acid pinacol ester intermediate is reported. An X-ray crystal structure of the isolated myricanol (1) was obtained and showed a co-crystal consisting of (+)-aR,11S-myricanol (2) and (-)-aS,11R-myricanol (3) coformers. Surprisingly, 3, obtained from chiral separation from 1, reduced tau levels in both cultured cells and ex vivo brain slices from a mouse model of tauopathy at reasonable mid-to-low micromolar potency, whereas 2 did not. SILAC proteomics and cell assays revealed that 3 promoted tau degradation through an autophagic mechanism, which was in contrast to that of other tau-lowering compounds previously identified by our group. During the course of structure-activity relationship (SAR) development, we prepared compound 13 by acid-catalyzed dehydration of 1. 13 had undergone an unexpected structural rearrangement through the isomyricanol substitution pattern (e.g., 16), as verified by X-ray structural analysis. Compound 13 displayed robust tau-lowering activity, and, importantly, its enantiomers reduced tau levels similarly. Therefore, the semisynthetic analogue 13 provides a foundation for further development as a tau-lowering agent without its SAR being based on chirality.
Collapse
Affiliation(s)
- Mackenzie D. Martin
- Department
of Molecular Medicine and Alzheimer’s Institute, University of South Florida, Tampa, Florida 33613, United States
| | - Laurent Calcul
- Department
of Chemistry and Center for Drug Discovery and Innovation, University of South Florida, Tampa, Florida 33620, United States
| | - Courtney Smith
- Department
of Chemistry and Center for Drug Discovery and Innovation, University of South Florida, Tampa, Florida 33620, United States
| | - Umesh K. Jinwal
- Department
of Molecular Medicine and Alzheimer’s Institute, University of South Florida, Tampa, Florida 33613, United States
| | - Sarah N. Fontaine
- Department
of Molecular Medicine and Alzheimer’s Institute, University of South Florida, Tampa, Florida 33613, United States
| | - April Darling
- Department
of Molecular Medicine and Alzheimer’s Institute, University of South Florida, Tampa, Florida 33613, United States
| | - Kent Seeley
- Department
of Chemistry and Center for Drug Discovery and Innovation, University of South Florida, Tampa, Florida 33620, United States
| | - Lukasz Wojtas
- Department
of Chemistry and Center for Drug Discovery and Innovation, University of South Florida, Tampa, Florida 33620, United States
| | - Malathi Narayan
- Department
of Molecular Medicine and Alzheimer’s Institute, University of South Florida, Tampa, Florida 33613, United States
| | - Jason E. Gestwicki
- Department
of Pharmaceutical Chemistry, University of California, San Francisco, San
Francisco, California 94158, United States
| | - Garry R. Smith
- ALS Biopharma, LLC, 3805
Old Easton Road, Doylestown, Pennsylvania 18902, United States
| | - Allen B. Reitz
- ALS Biopharma, LLC, 3805
Old Easton Road, Doylestown, Pennsylvania 18902, United States
| | - Bill J. Baker
- Department
of Chemistry and Center for Drug Discovery and Innovation, University of South Florida, Tampa, Florida 33620, United States
| | - Chad A. Dickey
- Department
of Molecular Medicine and Alzheimer’s Institute, University of South Florida, Tampa, Florida 33613, United States
- Department
of Chemistry and Center for Drug Discovery and Innovation, University of South Florida, Tampa, Florida 33620, United States
- James A. Haley Veteran’s Hospital, 13000 Bruce B. Downs Boulevard, Tampa, Florida 33612, United States
| |
Collapse
|
167
|
Correia SC, Resende R, Moreira PI, Pereira CM. Alzheimer's Disease-Related Misfolded Proteins and Dysfunctional Organelles on Autophagy Menu. DNA Cell Biol 2015; 34:261-73. [DOI: 10.1089/dna.2014.2757] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Affiliation(s)
- Sónia C. Correia
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Rosa Resende
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Paula I. Moreira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Laboratory of Physiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Cláudia M. Pereira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Laboratory of Biochemistry, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
168
|
Okuda M, Hijikuro I, Fujita Y, Wu X, Nakayama S, Sakata Y, Noguchi Y, Ogo M, Akasofu S, Ito Y, Soeda Y, Tsuchiya N, Tanaka N, Takahashi T, Sugimoto H. PE859, a novel tau aggregation inhibitor, reduces aggregated tau and prevents onset and progression of neural dysfunction in vivo. PLoS One 2015; 10:e0117511. [PMID: 25659102 PMCID: PMC4319983 DOI: 10.1371/journal.pone.0117511] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 12/29/2014] [Indexed: 12/16/2022] Open
Abstract
In tauopathies, a neural microtubule-associated protein tau (MAPT) is abnormally aggregated and forms neurofibrillary tangle. Therefore, inhibition of the tau aggregation is one of the key approaches for the treatment of these diseases. Here, we have identified a novel tau aggregation inhibitor, PE859. An oral administration of PE859 resulted in the significant reduction of sarkosyl-insoluble aggregated tau along with the prevention of onset and progression of the motor dysfunction in JNPL3 P301L-mutated human tau transgenic mice. These results suggest that PE859 is useful for the treatment of tauopathies.
Collapse
MESH Headings
- Amino Acid Substitution
- Animals
- Heterocyclic Compounds, 4 or More Rings/chemical synthesis
- Heterocyclic Compounds, 4 or More Rings/chemistry
- Heterocyclic Compounds, 4 or More Rings/pharmacology
- Humans
- Indoles/chemical synthesis
- Indoles/chemistry
- Indoles/pharmacology
- Male
- Mice
- Mice, Inbred ICR
- Mice, Transgenic
- Mutation, Missense
- Protein Aggregation, Pathological/drug therapy
- Protein Aggregation, Pathological/genetics
- Protein Aggregation, Pathological/metabolism
- Protein Aggregation, Pathological/pathology
- Pyrazoles/chemical synthesis
- Pyrazoles/chemistry
- Pyrazoles/pharmacology
- Tauopathies/drug therapy
- Tauopathies/genetics
- Tauopathies/metabolism
- Tauopathies/pathology
- tau Proteins/antagonists & inhibitors
- tau Proteins/genetics
- tau Proteins/metabolism
Collapse
Affiliation(s)
- Michiaki Okuda
- Pharma Eight Co., Ltd., Kyoto, Japan
- Graduate School of Brain Science, Doshisha University, Kyoto, Japan
- * E-mail:
| | | | - Yuki Fujita
- Pharma Eight Co., Ltd., Kyoto, Japan
- Graduate School of Brain Science, Doshisha University, Kyoto, Japan
| | - Xiaofeng Wu
- Key Lab of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | | | | | | | - Makoto Ogo
- Tsukuba Research Laboratories, Eisai Co., Ltd., Tsukuba, Japan
| | - Shigeru Akasofu
- Tsukuba Research Laboratories, Eisai Co., Ltd., Tsukuba, Japan
| | - Yoshimasa Ito
- Tsukuba Research Laboratories, Eisai Co., Ltd., Tsukuba, Japan
| | - Yoshiyuki Soeda
- Department of Aging Neurobiology, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Nobuhiko Tsuchiya
- Department of Biomolecular Engineering, Kyoto Institute of Technology, Kyoto, Japan
| | - Naoki Tanaka
- Department of Biomolecular Engineering, Kyoto Institute of Technology, Kyoto, Japan
| | - Takashi Takahashi
- Natural Product Chemistry & Pharmaceutical Research Center, Yokohama College of Pharmacy, Yokohama, Japan
| | - Hachiro Sugimoto
- Pharma Eight Co., Ltd., Kyoto, Japan
- Graduate School of Brain Science, Doshisha University, Kyoto, Japan
| |
Collapse
|
169
|
Methylene Blue Improves Brain Mitochondrial ABAD Functions and Decreases Aβ in a Neuroinflammatory Alzheimer’s Disease Mouse Model. Mol Neurobiol 2015; 53:1220-1228. [DOI: 10.1007/s12035-014-9088-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2014] [Accepted: 12/29/2014] [Indexed: 10/24/2022]
|
170
|
Abstract
Most neurodegenerative diseases that afflict humans are associated with the intracytoplasmic deposition of aggregate-prone proteins in neurons. Autophagy is a powerful process for removing such proteins. In this Review, we consider how certain neurodegenerative diseases may be associated with impaired autophagy and how this may affect pathology. We also discuss how autophagy induction may be a plausible therapeutic strategy for some conditions and review studies in various models that support this hypothesis. Finally, we briefly describe some of the signaling pathways that may be amenable to therapeutic targeting for these goals.
Collapse
|
171
|
Friedman LG, Qureshi YH, Yu WH. Promoting autophagic clearance: viable therapeutic targets in Alzheimer's disease. Neurotherapeutics 2015; 12:94-108. [PMID: 25421002 PMCID: PMC4322072 DOI: 10.1007/s13311-014-0320-z] [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] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Many neurodegenerative disorders are characterized by the aberrant accumulation of aggregate-prone proteins. Alzheimer's disease (AD) is associated with the buildup of β-amyloid peptides and tau, which aggregate into extracellular plaques and neurofibrillary tangles, respectively. Multiple studies have linked dysfunctional intracellular degradation mechanisms with AD pathogenesis. One such pathway is the autophagy-lysosomal system, which involves the delivery of large protein aggregates/inclusions and organelles to lysosomes through the formation, trafficking, and degradation of double-membrane structures known as autophagosomes. Converging data suggest that promoting autophagic degradation, either by inducing autophagosome formation or enhancing lysosomal digestion, provides viable therapeutic strategies. In this review, we discuss compounds that can augment autophagic clearance and may ameliorate disease-related pathology in cell and mouse models of AD. Canonical autophagy induction is associated with multiple signaling cascades; on the one hand, the best characterized is mammalian target of rapamycin (mTOR). Accordingly, multiple mTOR-dependent and mTOR-independent drugs that stimulate autophagy have been tested in preclinical models. On the other hand, there is a growing list of drugs that can enhance the later stages of autophagic flux by stabilizing microtubule-mediated trafficking, promoting lysosomal fusion, or bolstering lysosomal enzyme function. Although altering the different stages of autophagy provides many potential targets for AD therapeutic interventions, it is important to consider how autophagy drugs might also disturb the delicate balance between autophagosome formation and lysosomal degradation.
Collapse
Affiliation(s)
- Lauren G. Friedman
- Department of Pathology and Cell Biology, Taub Institute for Alzheimer’s Disease Research, Columbia University, 630 West 168th St., New York, NY 10032 USA
| | - Yasir H. Qureshi
- Department of Pathology and Cell Biology, Taub Institute for Alzheimer’s Disease Research, Columbia University, 630 West 168th St., New York, NY 10032 USA
| | - Wai Haung Yu
- Department of Pathology and Cell Biology, Taub Institute for Alzheimer’s Disease Research, Columbia University, 630 West 168th St., New York, NY 10032 USA
| |
Collapse
|
172
|
Cárdenas-Aguayo MDC, Gómez-Virgilio L, DeRosa S, Meraz-Ríos MA. The role of tau oligomers in the onset of Alzheimer's disease neuropathology. ACS Chem Neurosci 2014; 5:1178-91. [PMID: 25268947 DOI: 10.1021/cn500148z] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Most neurodegenerative diseases are characterized by the presence of protein aggregates. Alzheimer's disease (AD) is the most common cause of dementia in people over age 60. One of the histopathological hallmarks of AD is the presence of tau protein aggregates. Historically, it has been thought that paired helical filaments (PHFs) were the toxic form of tau that assembled to form neurofibrillary tangles (NFTs), but recently there has been evidence that tau oligomers, which form before PHFs and NFTs, could be the structures mediating neurodegeneration even before the fibrillary tau is deposited. Here, we discuss the recent advances in tau oligomer research, their implications on AD and other tauopathies, the mechanisms of tau turnover by the principal protein clearance systems (the proteasome and autophagy), and the potential use of tau oligomers as drug targets for the development of new therapeutic approaches.
Collapse
Affiliation(s)
- María del Carmen Cárdenas-Aguayo
- Molecular
Biomedicine Department, CINVESTAV-IPN, Ave. Politécnico 2508, Colonia
San Pedro Zacatenco, México City, D.F. 07360, México
| | - Laura Gómez-Virgilio
- Molecular
Biomedicine Department, CINVESTAV-IPN, Ave. Politécnico 2508, Colonia
San Pedro Zacatenco, México City, D.F. 07360, México
| | - Steven DeRosa
- Center
for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, 140 Old Orangeburg Road, Orangeburg, New York 10962, United States
| | - Marco Antonio Meraz-Ríos
- Molecular
Biomedicine Department, CINVESTAV-IPN, Ave. Politécnico 2508, Colonia
San Pedro Zacatenco, México City, D.F. 07360, México
| |
Collapse
|
173
|
Lonskaya I, Hebron M, Chen W, Schachter J, Moussa C. Tau deletion impairs intracellular β-amyloid-42 clearance and leads to more extracellular plaque deposition in gene transfer models. Mol Neurodegener 2014; 9:46. [PMID: 25384392 PMCID: PMC4247762 DOI: 10.1186/1750-1326-9-46] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 10/21/2014] [Indexed: 01/02/2023] Open
Abstract
Background Tau is an axonal protein that binds to and regulates microtubule function. Hyper-phosphorylation of Tau reduces its binding to microtubules and it is associated with β-amyloid deposition in Alzheimer’s disease. Paradoxically, Tau reduction may prevent β-amyloid pathology, raising the possibility that Tau mediates intracellular Aβ clearance. The current studies investigated the role of Tau in autophagic and proteasomal intracellular Aβ1-42 clearance and the subsequent effect on plaque deposition. Results Tau deletion impaired Aβ clearance via autophagy, but not the proteasome, while introduction of wild type human Tau into Tau−/− mice partially restored autophagic clearance of Aβ1-42, suggesting that exogenous Tau expression can support autophagic Aβ1-42 clearance. Tau deletion impaired autophagic flux and resulted in Aβ1-42 accumulation in pre-lysosomal autophagic vacuoles, affecting Aβ1-42 deposition into the lysosome. This autophagic defect was associated with decreased intracellular Aβ1-42 and increased plaque load in Tau−/− mice, which displayed less cell death. Nilotinib, an Abl tyrosine kinase inhibitor that promotes autophagic clearance mechanisms, reduced Aβ1-42 only when exogenous human Tau was expressed in Tau−/− mice. Conclusions These studies demonstrate that Tau deletion affects intracellular Aβ1-42 clearance, leading to extracellular plaque. Electronic supplementary material The online version of this article (doi:10.1186/1750-1326-9-46) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | | | - Charbel Moussa
- Department of Neuroscience, Laboratory for Dementia and Parkinsonism, Georgetown University Medical Center, 3970 Reservoir RD, Washington, DC 20057, USA.
| |
Collapse
|
174
|
Hyttinen JM, Amadio M, Viiri J, Pascale A, Salminen A, Kaarniranta K. Clearance of misfolded and aggregated proteins by aggrephagy and implications for aggregation diseases. Ageing Res Rev 2014; 18:16-28. [PMID: 25062811 DOI: 10.1016/j.arr.2014.07.002] [Citation(s) in RCA: 126] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 07/02/2014] [Accepted: 07/14/2014] [Indexed: 12/19/2022]
Abstract
Processing of misfolded proteins is important in order for the cell to maintain its normal functioning and homeostasis. Three systems control the quality of proteins: chaperone-mediated refolding, proteasomal degradation of ubiquitinated proteins, and finally, when the two others fail, aggrephagy, as selective form of autophagy, degrades ubiquitin-labelled aggregated cargos. In this route misfolded proteins gradually form larger aggregates, aggresomes and they eventually become double membrane-wrapped organelles called autophagosomes, which become degraded when they fuse to lysosomes, for reuse by the cell. The stages, the main molecules participating in the process, and the regulation of aggrephagy are discussed here, as is the role of protein aggregation in protein accumulation diseases. In particular, we emphasize that both Alzheimer's disease and age-related macular degeneration, two of the most common pathologies in the aged, are characterized by altered protein clearance and deposits. Based on the hypothesis that manipulations of autophagy may be potentially useful in these and other aggregation-related diseases, we will discuss some promising therapeutic strategies to counteract protein aggregates-induced cellular toxicity.
Collapse
|
175
|
Targeting autophagy in neurodegenerative diseases. Trends Pharmacol Sci 2014; 35:583-91. [DOI: 10.1016/j.tips.2014.09.002] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 08/28/2014] [Accepted: 09/03/2014] [Indexed: 12/14/2022]
|
176
|
Xie L, Choudhury GR, Wang J, Park Y, Liu R, Yuan F, Zhang CL, Yorio T, Jin K, Yang SH. Methylene blue promotes quiescence of rat neural progenitor cells. Front Cell Neurosci 2014; 8:315. [PMID: 25339866 PMCID: PMC4188130 DOI: 10.3389/fncel.2014.00315] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 09/18/2014] [Indexed: 12/03/2022] Open
Abstract
Neural stem cell-based treatment holds a new therapeutic opportunity for neurodegenerative disorders. Here, we investigated the effect of methylene blue on proliferation and differentiation of rat neural progenitor cells (NPCs) both in vitro and in vivo. We found that methylene blue inhibited proliferation and promoted quiescence of NPCs in vitro without affecting committed neuronal differentiation. Consistently, intracerebroventricular infusion of methylene blue significantly inhibited NPC proliferation at the subventricular zone (SVZ). Methylene blue inhibited mTOR signaling along with down-regulation of cyclins in NPCs in vitro and in vivo. In summary, our study indicates that methylene blue may delay NPC senescence through enhancing NPCs quiescence.
Collapse
Affiliation(s)
- Luokun Xie
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center Fort Worth, TX, USA
| | - Gourav R Choudhury
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center Fort Worth, TX, USA
| | - Jixian Wang
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center Fort Worth, TX, USA
| | - Yong Park
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center Fort Worth, TX, USA
| | - Ran Liu
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center Fort Worth, TX, USA
| | - Fang Yuan
- Department of Neurosurgery, Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University Beijing, China
| | - Chun-Li Zhang
- Department of Molecular Biology, University of Texas Southwestern Medical Center Dallas, TX, USA
| | - Thomas Yorio
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center Fort Worth, TX, USA
| | - Kunlin Jin
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center Fort Worth, TX, USA
| | - Shao-Hua Yang
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center Fort Worth, TX, USA ; Department of Neurosurgery, Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University Beijing, China
| |
Collapse
|
177
|
Adler G, Mautes AE. Improvement in behavioral symptoms, motor impairment and activities of daily living in a patient with the behavioral variant of frontotemporal dementia under treatment with methylene blue. ACTA ACUST UNITED AC 2014. [DOI: 10.1016/j.gmhc.2014.06.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
178
|
Kumar A, Singh A, Ekavali. A review on Alzheimer's disease pathophysiology and its management: an update. Pharmacol Rep 2014; 67:195-203. [PMID: 25712639 DOI: 10.1016/j.pharep.2014.09.004] [Citation(s) in RCA: 967] [Impact Index Per Article: 96.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 08/26/2014] [Accepted: 09/08/2014] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease acknowledged as progressive multifarious neurodegenerative disorder, is the leading cause of dementia in late adult life. Pathologically it is characterized by intracellular neurofibrillary tangles and extracellular amyloidal protein deposits contributing to senile plaques. Over the last two decades, advances in the field of pathogenesis have inspired the researchers for the investigation of novel pharmacological therapeutics centered more towards the pathophysiological events of the disease. Currently available treatments i.e. acetylcholinesterase inhibitors (rivastigmine, galantamine, donepezil) and N-methyl d-aspartate receptor antagonist (memantine) contribute minimal impact on the disease and target late aspects of the disease. These drugs decelerate the progression of the disease, provide symptomatic relief but fail to achieve a definite cure. While the neuropathological features of Alzheimer's disease are recognized but the intricacies of the mechanism have not been clearly defined. This lack of understanding regarding the pathogenic process may be the likely reason for the non-availability of effective treatment which can prevent onset and progression of the disease. Owing to the important progress in the field of pathophysiology in the last couple of years, new therapeutic targets are available that should render the underlying disease process to be tackled directly. In this review, authors will discusses the different aspects of pathophysiological mechanisms behind Alzheimer's disease and its management through conventional drug therapy, including modern investigational therapeutic strategies, recently completed and ongoing.
Collapse
Affiliation(s)
- Anil Kumar
- Pharmacology Division, University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Study, Panjab University, Chandigarh 160014, India.
| | - Arti Singh
- Pharmacology Division, University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Study, Panjab University, Chandigarh 160014, India
| | - Ekavali
- Pharmacology Division, University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Study, Panjab University, Chandigarh 160014, India
| |
Collapse
|
179
|
Gerson JE, Castillo-Carranza DL, Kayed R. Advances in therapeutics for neurodegenerative tauopathies: moving toward the specific targeting of the most toxic tau species. ACS Chem Neurosci 2014; 5:752-69. [PMID: 25075869 DOI: 10.1021/cn500143n] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Neurodegenerative disease is one of the greatest health concerns today and with no effective treatment in sight, it is crucial that researchers find a safe and successful therapeutic. While neurofibrillary tangles are considered the primary tauopathy hallmark, more evidence continues to come to light to suggest that soluble, intermediate tau aggregates--tau oligomers--are the most toxic species in disease. These intermediate tau species may also be responsible for the spread of pathology, suggesting that oligomeric tau may be the best therapeutic target. Here, we summarize results for the modulation of tau by molecular chaperones, small molecules and aggregation inhibitors, post-translational modifications, immunotherapy, other techniques, and future directions.
Collapse
Affiliation(s)
- Julia E. Gerson
- Department
of Neurology, George and Cynthia Mitchell
Center for Alzheimer’s Disease Research, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Diana L. Castillo-Carranza
- Department
of Neurology, George and Cynthia Mitchell
Center for Alzheimer’s Disease Research, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Rakez Kayed
- Department
of Neurology, George and Cynthia Mitchell
Center for Alzheimer’s Disease Research, University of Texas Medical Branch, Galveston, Texas 77555, United States
| |
Collapse
|
180
|
Specific calpain inhibition by calpastatin prevents tauopathy and neurodegeneration and restores normal lifespan in tau P301L mice. J Neurosci 2014; 34:9222-34. [PMID: 25009256 DOI: 10.1523/jneurosci.1132-14.2014] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Tau pathogenicity in Alzheimer's disease and other tauopathies is thought to involve the generation of hyperphosphorylated, truncated, and oligomeric tau species with enhanced neurotoxicity, although the generative mechanisms and the implications for disease therapy are not well understood. Here, we report a striking rescue from mutant tau toxicity in the JNPL3 mouse model of tauopathy. We show that pathological activation of calpains gives rise to a range of potentially toxic forms of tau, directly, and by activating cdk5. Calpain overactivation in brains of these mice is accelerated as a result of the marked depletion of the endogenous calpain inhibitor, calpastatin. When levels of this inhibitor are restored in neurons of JNPL3 mice by overexpressing calpastatin, tauopathy is prevented, including calpain-mediated breakdown of cytoskeletal proteins, cdk5 activation, tau hyperphosphorylation, formation of potentially neurotoxic tau fragments by either calpain or caspase-3, and tau oligomerization. Calpastatin overexpression also prevents loss of motor axons, delays disease onset, and extends survival of JNPL3 mice by 3 months to within the range of normal lifespan. Our findings support the therapeutic promise of highly specific calpain inhibition in the treatment of tauopathies and other neurodegenerative states.
Collapse
|
181
|
Rodriguez P, Jiang Z, Huang S, Shen Q, Duong TQ. Methylene blue treatment delays progression of perfusion-diffusion mismatch to infarct in permanent ischemic stroke. Brain Res 2014; 1588:144-9. [PMID: 25218555 DOI: 10.1016/j.brainres.2014.09.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 07/12/2014] [Accepted: 09/02/2014] [Indexed: 11/29/2022]
Abstract
Stroke is a leading cause of morbidity and mortality in the world. Low-dose methylene blue (MB), which has been used safely to treat methemoglobinemia and cyanide poisoning in humans, has energy enhancing and antioxidant properties. We tested the hypothesis that methylene blue treatment delays progression of at-risk tissue (ca. perfusion-diffusion mismatch) to infarct in permanent middle cerebral artery occlusion in rats at two MB treatment doses. Serial MRI was used to evaluate MB treatment efficacy. The major findings were: (i) MB significantly prolonged the perfusion-diffusion mismatch, (ii) MB mildly increased the CBF in the hypoperfused tissue, (iii) MB did not change the final infarct volume in permanent ischemic stroke, and (iv) there were no dose-dependent effects on mismatch progression for the 1 and 3mg/kg doses studied. This neuroprotective effect is likely the result of sustained ATP production and increased CBF to tissue at risk. This work has the potential to readily lead to clinical stroke trials given MB's excellent safety profile.
Collapse
Affiliation(s)
- Pavel Rodriguez
- Research Imaging Institute, Radiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Zhao Jiang
- Research Imaging Institute, Radiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States; Department of Anatomy and Embryology, Peking University Health Science Center, Beijing, China
| | - Shiliang Huang
- Research Imaging Institute, Radiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Qiang Shen
- Research Imaging Institute, Radiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| | - Timothy Q Duong
- Research Imaging Institute, Radiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States.
| |
Collapse
|
182
|
Mori T, Koyama N, Segawa T, Maeda M, Maruyama N, Kinoshita N, Hou H, Tan J, Town T. Methylene blue modulates β-secretase, reverses cerebral amyloidosis, and improves cognition in transgenic mice. J Biol Chem 2014; 289:30303-30317. [PMID: 25157105 DOI: 10.1074/jbc.m114.568212] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Amyloid precursor protein (APP) proteolysis is required for production of amyloid-β (Aβ) peptides that comprise β-amyloid plaques in the brains of patients with Alzheimer disease (AD). Here, we tested whether the experimental agent methylene blue (MB), used for treatment of methemoglobinemia, might improve AD-like pathology and behavioral deficits. We orally administered MB to the aged transgenic PSAPP mouse model of cerebral amyloidosis and evaluated cognitive function and cerebral amyloid pathology. Beginning at 15 months of age, animals were gavaged with MB (3 mg/kg) or vehicle once daily for 3 months. MB treatment significantly prevented transgene-associated behavioral impairment, including hyperactivity, decreased object recognition, and defective spatial working and reference memory, but it did not alter nontransgenic mouse behavior. Moreover, brain parenchymal and cerebral vascular β-amyloid deposits as well as levels of various Aβ species, including oligomers, were mitigated in MB-treated PSAPP mice. These effects occurred with inhibition of amyloidogenic APP proteolysis. Specifically, β-carboxyl-terminal APP fragment and β-site APP cleaving enzyme 1 protein expression and activity were attenuated. Additionally, treatment of Chinese hamster ovary cells overexpressing human wild-type APP with MB significantly decreased Aβ production and amyloidogenic APP proteolysis. These results underscore the potential for oral MB treatment against AD-related cerebral amyloidosis by modulating the amyloidogenic pathway.
Collapse
Affiliation(s)
- Takashi Mori
- Departments of Biomedical Sciences and Saitama Medical Center and University, Kawagoe, Saitama 350-8550, Japan; Departments of Pathology, Saitama Medical Center and University, Kawagoe, Saitama 350-8550, Japan,.
| | - Naoki Koyama
- Departments of Biomedical Sciences and Saitama Medical Center and University, Kawagoe, Saitama 350-8550, Japan
| | - Tatsuya Segawa
- Immuno-Biological Laboratories Co., Ltd., Fujioka, Gunma 375-0005, Japan
| | - Masahiro Maeda
- Immuno-Biological Laboratories Co., Ltd., Fujioka, Gunma 375-0005, Japan
| | - Nobuhiro Maruyama
- Immuno-Biological Laboratories Co., Ltd., Fujioka, Gunma 375-0005, Japan
| | - Noriaki Kinoshita
- Immuno-Biological Laboratories Co., Ltd., Fujioka, Gunma 375-0005, Japan
| | - Huayan Hou
- Rashid Laboratory for Developmental Neurobiology, Silver Child Development Center and University of South Florida, Tampa, Florida 33613
| | - Jun Tan
- Rashid Laboratory for Developmental Neurobiology, Silver Child Development Center and University of South Florida, Tampa, Florida 33613; Neuroimmunology Laboratory, Department of Psychiatry and Behavioral Neurosciences, Morsoni College of Medicine, University of South Florida, Tampa, Florida 33613, and
| | - Terrence Town
- Zilkha Neurogenetic Institute, Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California 90089-2821.
| |
Collapse
|
183
|
Protective effects of humanin on okadaic Acid-induced neurotoxicities in cultured cortical neurons. Neurochem Res 2014; 39:2150-9. [PMID: 25142935 DOI: 10.1007/s11064-014-1410-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2014] [Revised: 08/05/2014] [Accepted: 08/06/2014] [Indexed: 12/30/2022]
Abstract
Neurofibrillary tangles are pathological hallmarks of Alzheimer's disease (AD), which are mostly composed of hyperphosphorylated tau and directly correlate with dementia in AD patients. Okadaic acid (OA), a toxin extracted from marine life, can specifically inhibit protein phosphatases (PPs), including PP1 and Protein phosphatase 2A (PP2A), resulting in tau hyperphosphorylation. Humanin (HN), a peptide of 24 amino acids, was initially reported to protect neurons from AD-related cell toxicities. The present study was designed to test if HN could attenuate OA-induced neurotoxicities, including neural insults, apoptosis, autophagy, and tau hyperphosphorylation. We found that administration of OA for 24 h induced neuronal insults, including lactate dehydrogenase released, decreased of cell viability and numbers of living cells, neuronal apoptosis, cells autophagy and tau protein hyperphosphorylation. Pretreatment of cells with HN produced significant protective effects against OA-induced neural insults, apoptosis, autophagy and tau hyperphosphorylation. We also found that OA treatment inhibited PP2A activity and HN pretreatment significantly attenuated the inhibitory effects of OA. This study demonstrated for the first time that HN protected cortical neurons against OA-induced neurotoxicities, including neuronal insults, apoptosis, autophagy, and tau hyperphosphorylation. The mechanisms underlying the protections of HN may involve restoration of PP2A activity.
Collapse
|
184
|
Franco R, Cedazo-Minguez A. Successful therapies for Alzheimer's disease: why so many in animal models and none in humans? Front Pharmacol 2014; 5:146. [PMID: 25009496 PMCID: PMC4070393 DOI: 10.3389/fphar.2014.00146] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Accepted: 06/03/2014] [Indexed: 11/29/2022] Open
Abstract
Peering into the field of Alzheimer’s disease (AD), the outsider realizes that many of the therapeutic strategies tested (in animal models) have been successful. One also may notice that there is a deficit in translational research, i.e., to take a successful drug in mice and translate it to the patient. Efforts are still focused on novel projects to expand the therapeutic arsenal to “cure mice.” Scientific reasons behind so many successful strategies are not obvious. This article aims to review the current approaches to combat AD and to open a debate on common mechanisms of cognitive enhancement and neuroprotection. In short, either the rodent models are not good and should be discontinued, or we should extract the most useful information from those models. An example of a question that may be debated for the advancement in AD therapy is: In addition to reducing amyloid and tau pathologies, would it be necessary to boost synaptic strength and cognition? The debate could provide clues to turn around the current negative output in generating effective drugs for patients. Furthermore, discovery of biomarkers in human body fluids, and a clear distinction between cognitive enhancers and disease modifying strategies, should be instrumental for advancing in anti-AD drug discovery.
Collapse
Affiliation(s)
- Rafael Franco
- Division of Neurosciences, Centro de Investigación Médica Aplicada, Universidad de Navarra Pamplona, Spain ; Department of Biochemistry and Molecular Biology, Faculty of Biology, Universitat de Barcelona Barcelona, Spain
| | - Angel Cedazo-Minguez
- Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet Huddinge, Sweden
| |
Collapse
|
185
|
Mishra AK, ur Rasheed MS, Shukla S, Tripathi MK, Dixit A, Singh MP. Aberrant Autophagy and Parkinsonism: Does Correction Rescue from Disease Progression? Mol Neurobiol 2014; 51:893-908. [DOI: 10.1007/s12035-014-8744-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Accepted: 05/05/2014] [Indexed: 12/29/2022]
|
186
|
Tenreiro S, Eckermann K, Outeiro TF. Protein phosphorylation in neurodegeneration: friend or foe? Front Mol Neurosci 2014; 7:42. [PMID: 24860424 PMCID: PMC4026737 DOI: 10.3389/fnmol.2014.00042] [Citation(s) in RCA: 176] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 04/22/2014] [Indexed: 12/15/2022] Open
Abstract
Protein misfolding and aggregation is a common hallmark in neurodegenerative disorders, including Alzheimer's disease (AD), Parkinson's disease (PD), and fronto-temporal dementia (FTD). In these disorders, the misfolding and aggregation of specific proteins occurs alongside neuronal degeneration in somewhat specific brain areas, depending on the disorder and the stage of the disease. However, we still do not fully understand the mechanisms governing protein aggregation, and whether this constitutes a protective or detrimental process. In PD, alpha-synuclein (aSyn) forms protein aggregates, known as Lewy bodies, and is phosphorylated at serine 129. Other residues have also been shown to be phosphorylated, but the significance of phosphorylation in the biology and pathophysiology of the protein is still controversial. In AD and in FTD, hyperphosphorylation of tau protein causes its misfolding and aggregation. Again, our understanding of the precise consequences of tau phosphorylation in the biology and pathophysiology of the protein is still limited. Through the use of a variety of model organisms and technical approaches, we are now gaining stronger insight into the effects of phosphorylation in the behavior of these proteins. In this review, we cover recent findings in the field and discuss how targeting phosphorylation events might be used for therapeutic intervention in these devastating diseases of the nervous system.
Collapse
Affiliation(s)
- Sandra Tenreiro
- Cell and Molecular Neuroscience Unit, Instituto de Medicina Molecular Lisboa, Portugal
| | - Katrin Eckermann
- Department of Neurology, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen Göttingen, Germany
| | - Tiago F Outeiro
- Cell and Molecular Neuroscience Unit, Instituto de Medicina Molecular Lisboa, Portugal ; Instituto de Fisiologia, Faculdade de Medicina da Universidade de Lisboa Lisboa, Portugal ; Department of NeuroDegeneration and Restorative Research, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen Göttingen, Germany
| |
Collapse
|
187
|
Godoy JA, Rios JA, Zolezzi JM, Braidy N, Inestrosa NC. Signaling pathway cross talk in Alzheimer's disease. Cell Commun Signal 2014; 12:23. [PMID: 24679124 PMCID: PMC3977891 DOI: 10.1186/1478-811x-12-23] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Accepted: 03/11/2014] [Indexed: 01/11/2023] Open
Abstract
Numerous studies suggest energy failure and accumulative intracellular waste play a causal role in the pathogenesis of several neurodegenerative disorders and Alzheimer's disease (AD) in particular. AD is characterized by extracellular amyloid deposits, intracellular neurofibrillary tangles, cholinergic deficits, synaptic loss, inflammation and extensive oxidative stress. These pathobiological changes are accompanied by significant behavioral, motor, and cognitive impairment leading to accelerated mortality. Currently, the potential role of several metabolic pathways associated with AD, including Wnt signaling, 5' adenosine monophosphate-activated protein kinase (AMPK), mammalian target of rapamycin (mTOR), Sirtuin 1 (Sirt1, silent mating-type information regulator 2 homolog 1), and peroxisome proliferator-activated receptor gamma co-activator 1-α (PGC-1α) have widened, with recent discoveries that they are able to modulate several pathological events in AD. These include reduction of amyloid-β aggregation and inflammation, regulation of mitochondrial dynamics, and increased availability of neuronal energy. This review aims to highlight the involvement of these new set of signaling pathways, which we have collectively termed "anti-ageing pathways", for their potentiality in multi-target therapies against AD where cellular metabolic processes are severely impaired.
Collapse
Affiliation(s)
- Juan A Godoy
- Centro de Envejecimiento y Regeneración (CARE); Departamento de Biología Celular y Molecular; Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Av. Alameda 340, Santiago, Chile
| | - Juvenal A Rios
- Centro de Envejecimiento y Regeneración (CARE); Departamento de Biología Celular y Molecular; Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Av. Alameda 340, Santiago, Chile
| | - Juan M Zolezzi
- Departamento de Biología, Facultad de Ciencias, Universidad de Tarapacá, Arica, Chile
| | - Nady Braidy
- Center for Healthy Brain Ageing, School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Nibaldo C Inestrosa
- Centro de Envejecimiento y Regeneración (CARE); Departamento de Biología Celular y Molecular; Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Av. Alameda 340, Santiago, Chile
- Center for Healthy Brain Ageing, School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, Australia
| |
Collapse
|
188
|
Fant X, Durieu E, Chicanne G, Payrastre B, Sbrissa D, Shisheva A, Limanton E, Carreaux F, Bazureau JP, Meijer L. cdc-like/dual-specificity tyrosine phosphorylation-regulated kinases inhibitor leucettine L41 induces mTOR-dependent autophagy: implication for Alzheimer's disease. Mol Pharmacol 2014; 85:441-50. [PMID: 24366666 PMCID: PMC6067634 DOI: 10.1124/mol.113.090837] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 12/23/2013] [Indexed: 12/26/2022] Open
Abstract
Leucettines, a family of pharmacological inhibitors of dual-specificity tyrosine phosphorylation regulated kinases and cdc-like kinases (CLKs), are currently under investigation for their potential therapeutic application to Down syndrome and Alzheimer's disease. We here report that leucettine L41 triggers bona fide autophagy in osteosarcoma U-2 OS cells and immortalized mouse hippocampal HT22 cells, characterized by microtubule-associated protein light chain 3 membrane translocation and foci formation. Leucettine L41-triggered autophagy requires the Unc-51-like kinase and is sensitive to the phosphatidylinositol 3-kinase (PI3K) inhibitors wortmannin and 3-methyladenine, suggesting that it acts through the mammalian target of rapamycin (mTOR)/PI3K-dependent pathway. Leucettine L41 does not act by modifying the autophagic flux of vesicles. Leucettine L41-induced autophagy correlates best with inhibition of CLKs. Leucettine L41 modestly inhibited phosphatidylinositol-3-phosphate 5-kinase, FYVE domain-containing activity as tested both in vitro and in vivo, which may also contribute to autophagy induction. Altogether these results demonstrate that leucettines can activate the autophagic mTOR/PI3K pathway, a characteristic that may turn advantageous in the context of Alzheimer's disease treatment.
Collapse
Affiliation(s)
- Xavier Fant
- Centre National de la Recherche Scientifique (CNRS), USR3151, "Protein Phosphorylation and Human Disease," Station Biologique, Roscoff cedex, France (X.F., E.D.); Institut National de la Santé et de la Recherche Médicale/Université Paul Sabatier Unité Mixte de Recherche (UMR) 1048, "Production et fonctions plaquettaires: signalisation et phosphoinositides" group, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Toulouse cedex, France (G.C., B.P.); Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan (D.S., A.S.); Laboratoire Sciences Chimiques de Rennes, UMR CNRS 6226, Groupe Ingénierie Chimique et Molécules pour le Vivant (ICMV), Université de Rennes, Campus de Beaulieu, Rennes cedex, France (E.L., F.C., J.-P.B.); and ManRos Therapeutics, Perharidy Research Center, Roscoff, Bretagne, France (L.M.)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
189
|
Golbe LI. The tau of PSP: a long road to treatment. Mov Disord 2014; 29:431-4. [PMID: 24585428 DOI: 10.1002/mds.25855] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2014] [Accepted: 02/03/2014] [Indexed: 11/10/2022] Open
Affiliation(s)
- Lawrence I Golbe
- Department of Neurology, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA
| |
Collapse
|
190
|
Stack C, Jainuddin S, Elipenahli C, Gerges M, Starkova N, Starkov AA, Jové M, Portero-Otin M, Launay N, Pujol A, Kaidery NA, Thomas B, Tampellini D, Beal MF, Dumont M. Methylene blue upregulates Nrf2/ARE genes and prevents tau-related neurotoxicity. Hum Mol Genet 2014; 23:3716-32. [PMID: 24556215 DOI: 10.1093/hmg/ddu080] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Methylene blue (MB, methylthioninium chloride) is a phenothiazine that crosses the blood brain barrier and acts as a redox cycler. Among its beneficial properties are its abilities to act as an antioxidant, to reduce tau protein aggregation and to improve energy metabolism. These actions are of particular interest for the treatment of neurodegenerative diseases with tau protein aggregates known as tauopathies. The present study examined the effects of MB in the P301S mouse model of tauopathy. Both 4 mg/kg MB (low dose) and 40 mg/kg MB (high dose) were administered in the diet ad libitum from 1 to 10 months of age. We assessed behavior, tau pathology, oxidative damage, inflammation and numbers of mitochondria. MB improved the behavioral abnormalities and reduced tau pathology, inflammation and oxidative damage in the P301S mice. These beneficial effects were associated with increased expression of genes regulated by NF-E2-related factor 2 (Nrf2)/antioxidant response element (ARE), which play an important role in antioxidant defenses, preventing protein aggregation, and reducing inflammation. The activation of Nrf2/ARE genes is neuroprotective in other transgenic mouse models of neurodegenerative diseases and it appears to be an important mediator of the neuroprotective effects of MB in P301S mice. Moreover, we used Nrf2 knock out fibroblasts to show that the upregulation of Nrf2/ARE genes by MB is Nrf2 dependent and not due to secondary effects of the compound. These findings provide further evidence that MB has important neuroprotective effects that may be beneficial in the treatment of human neurodegenerative diseases with tau pathology.
Collapse
Affiliation(s)
- Cliona Stack
- Department of Neurology and Neuroscience, Weill Cornell Medical College, New York, NY 10065, USA
| | - Shari Jainuddin
- Department of Neurology and Neuroscience, Weill Cornell Medical College, New York, NY 10065, USA
| | - Ceyhan Elipenahli
- Department of Neurology and Neuroscience, Weill Cornell Medical College, New York, NY 10065, USA
| | - Meri Gerges
- Department of Neurology and Neuroscience, Weill Cornell Medical College, New York, NY 10065, USA
| | - Natalia Starkova
- Department of Neurology and Neuroscience, Weill Cornell Medical College, New York, NY 10065, USA
| | - Anatoly A Starkov
- Department of Neurology and Neuroscience, Weill Cornell Medical College, New York, NY 10065, USA
| | - Mariona Jové
- Department de Medicina Experimental, Universitat de Lleida-IRBLLEIDA, Spain
| | | | - Nathalie Launay
- Neurometabolic Diseases Laboratory-IDIBELL, Hospital Duran i Reynals, 08908 L'Hospitalet de Llobregat, Barcelona, Spain, CIBERER, Spanish Network for Rare Diseases, ISCIII, Spain
| | - Aurora Pujol
- Neurometabolic Diseases Laboratory-IDIBELL, Hospital Duran i Reynals, 08908 L'Hospitalet de Llobregat, Barcelona, Spain, CIBERER, Spanish Network for Rare Diseases, ISCIII, Spain, ICREA, Catalan Institution for Research and Advanced Studies, Spain
| | - Navneet Ammal Kaidery
- Department of Pharmacology and Toxicology and Department of Neurology, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912, USA
| | - Bobby Thomas
- Department of Pharmacology and Toxicology and Department of Neurology, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912, USA
| | - Davide Tampellini
- Hospital Kremlin Bicêtre, UMR 788, Institut National de la Santé et de la Recherche Médicale (INSERM), Université Paris Sud, Le Kremlin Bicêtre, France and
| | - M Flint Beal
- Department of Neurology and Neuroscience, Weill Cornell Medical College, New York, NY 10065, USA
| | - Magali Dumont
- Department of Neurology and Neuroscience, Weill Cornell Medical College, New York, NY 10065, USA, IHU-A-ICM, Hospital Pitié-Salpêtrière, 75013 Paris, France
| |
Collapse
|
191
|
Tricoire H, Palandri A, Bourdais A, Camadro JM, Monnier V. Methylene blue rescues heart defects in a Drosophila model of Friedreich's ataxia. Hum Mol Genet 2014; 23:968-79. [PMID: 24105471 DOI: 10.1093/hmg/ddt493] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Friedreich's ataxia (FRDA), the most common hereditary ataxia, is characterized by progressive degeneration of the central and peripheral nervous system, hypertrophic cardiomyopathy and a high risk of diabetes. FRDA is caused by abnormally low levels of frataxin, a highly conserved mitochondrial protein. Drosophila has been previously successfully used to model FRDA in various cell types, including neurons and glial cells. Here, we report the development of a Drosophila cardiac model of FRDA. In vivo heart imaging revealed profound impairments in heart function in frataxin-depleted Drosophila, including a strong increase in end-systolic and end-diastolic diameters and a decrease in fractional shortening (FS). These features, reminiscent of pathological phenotypes in humans, are fully rescued by complementation with human frataxin, suggesting conserved cardiac functions of frataxin between the two organisms. Oxidative stress is not a major factor of heart impairment in frataxin-depleted flies, suggesting the involvement of other pathological mechanisms notably mitochondrial respiratory chain (MRC) dysfunction. Accordingly, we report that methylene blue (MB), a compound known to act as an alternative electron carrier that bypasses mitochondrial complexes I-III, was able to prevent heart dysfunction. MB also partially rescued the phenotype when administered post-symptomatically. Analysis of MB derivatives demonstrates that only compounds with electron carrier properties are able to prevent the heart phenotype. Thus MB, a compound already used for several clinical applications, appears promising for the treatment of the heart dysfunctions that are a major cause of death of FRDA patients. This work provides the grounds for further evaluation of MB action in mammals.
Collapse
Affiliation(s)
- Hervé Tricoire
- Unité de Biologie Fonctionnelle et Adaptative (BFA) EAC4413 CNRS, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | | | | | | | | |
Collapse
|
192
|
Spires-Jones TL, Friedman T, Pitstick R, Polydoro M, Roe A, Carlson GA, Hyman BT. Methylene blue does not reverse existing neurofibrillary tangle pathology in the rTg4510 mouse model of tauopathy. Neurosci Lett 2014; 562:63-8. [PMID: 24462887 DOI: 10.1016/j.neulet.2014.01.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 12/18/2013] [Accepted: 01/10/2014] [Indexed: 10/25/2022]
Abstract
Alzheimer's disease is characterized pathologically by aggregation of amyloid beta into senile plaques and aggregation of pathologically modified tau into neurofibrillary tangles. While changes in amyloid processing are strongly implicated in disease initiation, the recent failure of amyloid-based therapies has highlighted the importance of tau as a therapeutic target. "Tangle busting" compounds including methylene blue and analogous molecules are currently being evaluated as therapeutics in Alzheimer's disease. Previous studies indicated that methylene blue can reverse tau aggregation in vitro after 10 min, and subsequent studies suggested that high levels of drug reduce tau protein levels (assessed biochemically) in vivo. Here, we tested whether methylene blue could remove established neurofibrillary tangles in the rTg4510 model of tauopathy, which develops robust tangle pathology. We find that 6 weeks of methylene blue dosing in the water from 16 months to 17.5 months of age decreases soluble tau but does not remove sarkosyl insoluble tau, or histologically defined PHF1 or Gallyas positive tangle pathology. These data indicate that methylene blue treatment will likely not rapidly reverse existing tangle pathology.
Collapse
Affiliation(s)
- Tara L Spires-Jones
- Massachusetts General Hospital, 114 16th Street, Charlestown, MA 02129, USA.
| | - Taylor Friedman
- Massachusetts General Hospital, 114 16th Street, Charlestown, MA 02129, USA
| | | | - Manuela Polydoro
- Massachusetts General Hospital, 114 16th Street, Charlestown, MA 02129, USA
| | - Allyson Roe
- Massachusetts General Hospital, 114 16th Street, Charlestown, MA 02129, USA
| | | | - Bradley T Hyman
- Massachusetts General Hospital, 114 16th Street, Charlestown, MA 02129, USA
| |
Collapse
|
193
|
Guerrero-Muñoz MJ, Castillo-Carranza DL, Kayed R. Therapeutic approaches against common structural features of toxic oligomers shared by multiple amyloidogenic proteins. Biochem Pharmacol 2014; 88:468-78. [PMID: 24406245 DOI: 10.1016/j.bcp.2013.12.023] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 12/18/2013] [Accepted: 12/19/2013] [Indexed: 02/03/2023]
Abstract
Impaired proteostasis is one of the main features of all amyloid diseases, which are associated with the formation of insoluble aggregates from amyloidogenic proteins. The aggregation process can be caused by overproduction or poor clearance of these proteins. However, numerous reports suggest that amyloid oligomers are the most toxic species, rather than insoluble fibrillar material, in Alzheimer's, Parkinson's, and Prion diseases, among others. Although the exact protein that aggregates varies between amyloid disorders, they all share common structural features that can be used as therapeutic targets. In this review, we focus on therapeutic approaches against shared features of toxic oligomeric structures and future directions.
Collapse
Affiliation(s)
- Marcos J Guerrero-Muñoz
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA; Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Diana L Castillo-Carranza
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA; Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA; Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
194
|
|
195
|
Wischik CM, Harrington CR, Storey JMD. Tau-aggregation inhibitor therapy for Alzheimer's disease. Biochem Pharmacol 2013; 88:529-39. [PMID: 24361915 DOI: 10.1016/j.bcp.2013.12.008] [Citation(s) in RCA: 183] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2013] [Revised: 12/08/2013] [Accepted: 12/09/2013] [Indexed: 12/22/2022]
Abstract
Many trials of drugs aimed at preventing or clearing β-amyloid pathology have failed to demonstrate efficacy in recent years and further trials continue with drugs aimed at the same targets and mechanisms. The Alzheimer neurofibrillary tangle is composed of tau and the core of its constituent filaments are made of a truncated fragment from the repeat domain of tau. This truncated tau can catalyse the conversion of normal soluble tau into aggregated oligomeric and fibrillar tau which, in turn, can spread to neighbouring neurons. Tau aggregation is not a late-life process and onset of Braak stage 1 peaks in people in their late 40s or early 50s. Tau aggregation pathology at Braak stage 1 or beyond affects 50% of the population over the age of 45. The initiation of tau aggregation requires its binding to a non-specific substrate to expose a high affinity tau-tau binding domain and it is self-propagating thereafter. The initiating substrate complex is most likely formed as a consequence of a progressive loss of endosomal-lysosomal processing of neuronal proteins, particularly of membrane proteins from mitochondria. Mutations in the APP/presenilin membrane complex may simply add to the age-related endosomal-lysosomal processing failure, bringing forward, but not directly causing, the tau aggregation cascade in carriers. Methylthioninium chloride (MTC), the first identified tau aggregation inhibitor (TAI), offers an alternative to the amyloid approach. Phase 3 trials are underway with a novel stabilized reduced form of methylthioninium (LMTX) that has improved tolerability and absorption.
Collapse
Affiliation(s)
- Claude M Wischik
- TauRx Therapeutics Ltd., Singapore; School of Medicine and Dentistry, University of Aberdeen, Scotland, United Kingdom.
| | - Charles R Harrington
- TauRx Therapeutics Ltd., Singapore; School of Medicine and Dentistry, University of Aberdeen, Scotland, United Kingdom
| | - John M D Storey
- TauRx Therapeutics Ltd., Singapore; Department of Chemistry, University of Aberdeen, Scotland, United Kingdom
| |
Collapse
|
196
|
François A, Terro F, Janet T, Rioux Bilan A, Paccalin M, Page G. Involvement of interleukin-1β in the autophagic process of microglia: relevance to Alzheimer's disease. J Neuroinflammation 2013; 10:151. [PMID: 24330807 PMCID: PMC3878742 DOI: 10.1186/1742-2094-10-151] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Accepted: 11/24/2013] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Autophagy is a major pathway of protein and organelle degradation in the lysosome. Autophagy exists at basal constitutive level and can be induced as a defense mechanism under stress conditions. Molecular relationships between autophagy and inflammation at the periphery were recently evidenced, highlighting a role of autophagy in the regulation of inflammation. Impairment of autophagy (with accumulation of autophagic vacuoles) and substantial inflammation are found in neurodegenerative diseases such as Alzheimer's Disease (AD). However, the links between autophagy and inflammation in AD remain to be determined. METHODS Here, we examined the inflammatory reaction and autophagy in murine tri-cultures of neurons, astrocytes, and microglia. Tri-cultures were exposed to various inflammatory stresses (lipopolysaccharide (LPS), amyloid peptide (Aβ42) with or without cytokines) for 48 hours. Furthermore, the relationships between inflammation and autophagy were also analyzed in astrocyte- and microglia-enriched cultures. Data for multiple variable comparisons were analyzed by a one-way ANOVA followed by a Newman-keuls' test. RESULTS Aβ42 induced a low inflammation without accumulation of acidic vesicles contrary to moderate or severe inflammation induced by LPS or the cytokine cocktail (IL-1β, TNF-α, and IL-6) or IL-1β alone which led to co-localization of p62 and LC3, two markers of autophagy, with acidic vesicles stained with Lyso-ID Red dye. Moreover, the study reveals a major role of IL-1β in the induction of autophagy in tri-cultures in the presence or absence of Aβ42. However, the vulnerability of the autophagic process in purified microglia to IL-1β was prevented by Aβ42. CONCLUSION These findings show a close relationship between inflammation and autophagy, in particular a major role of IL-1β in the induction of the microglial autophagy which could be the case in AD. New therapeutic strategies could target inflammasome and autophagy in microglia to maintain its role in the amyloid immunosurveillance.
Collapse
Affiliation(s)
| | | | | | | | | | - Guylène Page
- EA3808 Molecular Targets and Therapeutic of Alzheimer's Disease, University of Poitiers, Poitiers F-86022, France.
| |
Collapse
|
197
|
Potential synergy between tau aggregation inhibitors and tau chaperone modulators. ALZHEIMERS RESEARCH & THERAPY 2013; 5:41. [PMID: 24041111 PMCID: PMC3979086 DOI: 10.1186/alzrt207] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Tau is a soluble, microtubule-associated protein known to aberrantly form
amyloid-positive aggregates. This pathology is characteristic for more than 15
neuropathies, the most common of which is Alzheimer’s disease. Finding
therapeutics to reverse or remove this non-native tau state is of great
interest; however, at this time only one drug is entering phase III clinical
trials for treating tauopathies. Generally, tau manipulation by therapeutics can
either directly or indirectly alter tau aggregation and stability. Drugs that
bind and change the conformation of tau itself are largely classified as
aggregation inhibitors, while drugs that alter the activity of a tau-effector
protein fall into several categories, such as kinase inhibitors, microtubule
stabilizers, or chaperone modulators. Chaperone inhibitors that have proven
effective in tau models include heat shock protein 90 inhibitors, heat shock
protein 70 inhibitors and activators, as well as inducers of heat shock
proteins. While many of these compounds can alter tau levels and/or aggregation
states, it is possible that combining these approaches may produce the most
optimal outcome. However, because many of these compounds have multiple
off-target effects or poor blood–brain barrier permeability, the
development of this synergistic therapeutic strategy presents significant
challenges. This review will summarize many of the drugs that have been
identified to alter tau biology, with special focus on therapeutics that prevent
tau aggregation and regulate chaperone-mediated clearance of tau.
Collapse
|
198
|
Chesser AS, Pritchard SM, Johnson GVW. Tau clearance mechanisms and their possible role in the pathogenesis of Alzheimer disease. Front Neurol 2013; 4:122. [PMID: 24027553 PMCID: PMC3759803 DOI: 10.3389/fneur.2013.00122] [Citation(s) in RCA: 148] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 08/15/2013] [Indexed: 12/24/2022] Open
Abstract
One of the defining pathological features of Alzheimer disease (AD) is the intraneuronal accumulation of tau. The tau that forms these accumulations is altered both posttranslationally and conformationally, and there is now significant evidence that soluble forms of these modified tau species are the toxic entities rather than the insoluble neurofibrillary tangles. However there is still noteworthy debate concerning which specific pathological forms of tau are the contributors to neuronal dysfunction and death in AD. Given that increases in aberrant forms of tau play a role in the neurodegeneration process in AD, there is growing interest in understanding the degradative pathways that remove tau from the cell, and the selectivity of these different pathways for various forms of tau. Indeed, one can speculate that deficits in a pathway that selectively removes certain pathological forms of tau could play a pivotal role in AD. In this review we will discuss the different proteolytic and degradative machineries that may be involved in removing tau from the cell. How deficits in these different degradative pathways may contribute to abnormal accumulation of tau in AD will also be considered. In addition, the issue of the selective targeting of specific tau species to a given degradative pathway for clearance from the cell will be addressed.
Collapse
Affiliation(s)
- Adrianne S Chesser
- Neuroscience Graduate Program, Department of Anesthesiology, University of Rochester , Rochester, NY , USA
| | | | | |
Collapse
|
199
|
Abisambra J, Jinwal UK, Miyata Y, Rogers J, Blair L, Li X, Seguin SP, Wang L, Jin Y, Bacon J, Brady S, Cockman M, Guidi C, Zhang J, Koren J, Young ZT, Atkins CA, Zhang B, Lawson LY, Weeber EJ, Brodsky JL, Gestwicki JE, Dickey CA. Allosteric heat shock protein 70 inhibitors rapidly rescue synaptic plasticity deficits by reducing aberrant tau. Biol Psychiatry 2013; 74:367-74. [PMID: 23607970 PMCID: PMC3740016 DOI: 10.1016/j.biopsych.2013.02.027] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Revised: 02/05/2013] [Accepted: 02/22/2013] [Indexed: 02/03/2023]
Abstract
BACKGROUND The microtubule-associated protein tau accumulates in neurodegenerative diseases known as tauopathies, the most common being Alzheimer's disease. One way to treat these disorders may be to reduce abnormal tau levels through chaperone manipulation, thus subverting synaptic plasticity defects caused by tau's toxic accretion. METHODS Tauopathy models were used to study the impact of YM-01 on tau. YM-01 is an allosteric promoter of triage functions of the most abundant variant of the heat shock protein 70 (Hsp70) family in the brain, heat shock cognate 70 protein (Hsc70). The mechanisms by which YM-01 modified Hsc70 activity and tau stability were evaluated with biochemical methods, cell cultures, and primary neuronal cultures from tau transgenic mice. YM-01 was also administered to acute brain slices of tau mice; changes in tau stability and electrophysiological correlates of learning and memory were measured. RESULTS Tau levels were rapidly and potently reduced in vitro and ex vivo upon treatment with nanomolar concentrations of YM-01. Consistent with Hsc70 having a key role in this process, overexpression of heat shock protein 40 (DNAJB2), an Hsp70 co-chaperone, suppressed YM-01 activity. In contrast to its effects in pathogenic tauopathy models, YM-01 had little activity in ex vivo brain slices from normal, wild-type mice unless microtubules were disrupted, suggesting that Hsc70 acts preferentially on abnormal pools of free tau. Finally, treatment with YM-01 increased long-term potentiation in tau transgenic brain slices. CONCLUSIONS Therapeutics that exploit the ability of chaperones to selectively target abnormal tau can rapidly and potently rescue the synaptic dysfunction that occurs in Alzheimer's disease and other tauopathies.
Collapse
Affiliation(s)
- Jose Abisambra
- Department of Molecular Medicine, USF Health Byrd Alzheimer’s Institute, University of South Florida; Tampa, FL, 33613, USA
| | - Umesh K. Jinwal
- Department of Pharmaceutical Sciences, USF Health Byrd Alzheimer’s Institute, University of South Florida; Tampa, FL, 33613, USA
| | - Yoshinari Miyata
- Life Sciences Institute and Departments of Pathology and Biological Chemistry, University of Michigan; Ann Arbor, MI, 48109, USA
| | - Justin Rogers
- Department of Molecular Pharmacology and Physiology, USF Health Byrd Alzheimer’s Institute, University of South Florida; Tampa, FL, 33613, USA
| | - Laura Blair
- Department of Molecular Medicine, USF Health Byrd Alzheimer’s Institute, University of South Florida; Tampa, FL, 33613, USA
| | - Xiaokai Li
- Life Sciences Institute and Departments of Pathology and Biological Chemistry, University of Michigan; Ann Arbor, MI, 48109, USA
| | - Sandlin P. Seguin
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260
| | - Li Wang
- Department of Molecular Medicine, USF Health Byrd Alzheimer’s Institute, University of South Florida; Tampa, FL, 33613, USA
| | - Ying Jin
- Department of Molecular Medicine, USF Health Byrd Alzheimer’s Institute, University of South Florida; Tampa, FL, 33613, USA
| | - Justin Bacon
- Department of Molecular Medicine, USF Health Byrd Alzheimer’s Institute, University of South Florida; Tampa, FL, 33613, USA
| | - Sarah Brady
- Department of Molecular Medicine, USF Health Byrd Alzheimer’s Institute, University of South Florida; Tampa, FL, 33613, USA
| | - Matthew Cockman
- Department of Molecular Medicine, USF Health Byrd Alzheimer’s Institute, University of South Florida; Tampa, FL, 33613, USA
| | - Chantal Guidi
- Department of Molecular Medicine, USF Health Byrd Alzheimer’s Institute, University of South Florida; Tampa, FL, 33613, USA
| | - Juan Zhang
- Department of Molecular Medicine, USF Health Byrd Alzheimer’s Institute, University of South Florida; Tampa, FL, 33613, USA
| | - John Koren
- Department of Molecular Medicine, USF Health Byrd Alzheimer’s Institute, University of South Florida; Tampa, FL, 33613, USA
| | - Zapporah T. Young
- Life Sciences Institute and Departments of Pathology and Biological Chemistry, University of Michigan; Ann Arbor, MI, 48109, USA
| | - Christopher A. Atkins
- Department of Molecular Medicine, USF Health Byrd Alzheimer’s Institute, University of South Florida; Tampa, FL, 33613, USA
| | - Bo Zhang
- Department of Molecular Medicine, USF Health Byrd Alzheimer’s Institute, University of South Florida; Tampa, FL, 33613, USA
| | - Lisa Y. Lawson
- Department of Molecular Medicine, USF Health Byrd Alzheimer’s Institute, University of South Florida; Tampa, FL, 33613, USA
| | - Edwin J. Weeber
- Department of Molecular Pharmacology and Physiology, USF Health Byrd Alzheimer’s Institute, University of South Florida; Tampa, FL, 33613, USA
| | - Jeffrey L. Brodsky
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260
| | - Jason E. Gestwicki
- Life Sciences Institute and Departments of Pathology and Biological Chemistry, University of Michigan; Ann Arbor, MI, 48109, USA
| | - Chad A. Dickey
- Department of Molecular Medicine, USF Health Byrd Alzheimer’s Institute, University of South Florida; Tampa, FL, 33613, USA
| |
Collapse
|
200
|
Appleby BS, Nacopoulos D, Milano N, Zhong K, Cummings JL. A review: treatment of Alzheimer's disease discovered in repurposed agents. Dement Geriatr Cogn Disord 2013; 35:1-22. [PMID: 23307039 DOI: 10.1159/000345791] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/13/2012] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND/AIMS Many compounds that have already been approved for alternate diagnoses have been studied in relation to Alzheimer's disease (AD). The purpose of this review is to summarize these studies and discuss the rationale and benefits of repurposing drugs for AD treatment. METHODS Studies of drugs related to AD treatment that were relevant to a disease-modifying mechanism of action (MOA) and are already approved by the Food and Drug Administration for non-AD diagnoses were collected from PubMed. RESULTS Many drugs already approved for the treatment of other diseases have been studied in relation to AD treatment. Numerous drugs with known toxicity profiles have the potential to be repurposed as a treatment for AD. CONCLUSION Known MOA, toxicology, and pharmacodynamic profiles would accelerate the process and increase the odds of finding a more timely disease-modifying treatment for AD.
Collapse
Affiliation(s)
- Brian S Appleby
- Cleveland Clinic Lou Ruvo Center for Brain Health, Cleveland, OH, USA
| | | | | | | | | |
Collapse
|