Catalogue Search | MBRL
Search Results Heading
Explore the vast range of titles available.
MBRLSearchResults
-
DisciplineDiscipline
-
Is Peer ReviewedIs Peer Reviewed
-
Item TypeItem Type
-
SubjectSubject
-
YearFrom:-To:
-
More FiltersMore FiltersSourceLanguage
Done
Filters
Reset
142
result(s) for
"Tauopathies - drug therapy"
Sort by:
A novel small-molecule PROTAC selectively promotes tau clearance to improve cognitive functions in Alzheimer-like models
by
Li, Shihong
,
Wang, Yipeng
,
Zheng, Jie
in
Alzheimer Disease - drug therapy
,
Alzheimer Disease - metabolism
,
Animals
2021
Intracellular accumulation of tau is a hallmark pathology in Alzheimer disease (AD) and the related tauopathies, thus targeting tau could be promising for drug development. Proteolysis Targeting Chimera (PROTAC) is a novel drug discovery strategy for selective protein degradation from within cells.
A novel small-molecule PROTAC, named as C004019 with a molecular mass of 1,035.29 dalton, was designed to simultaneously recruite tau and E3-ligase (Vhl) and thus to selectively enhance ubiquitination and proteolysis of tau proteins. Western blotting, immunofluoresence and immunohistochemical staining were employed to verify the effects of C004019 in cell models (HEK293 and SH-SY5Y) and mouse models (hTau-transgenic and 3xTg-AD), respectively. The cognitive capacity of the mice was assessed by a suite of behavior experiments. Electrophysiology and Golgi staining were used to evaluate the synaptic plasticity.
C004019 induced a robust tau clearance
promoting its ubiquitination-proteasome-dependent proteolysis in HEK293 cells with stable or transient overexpression of human tau (hTau), and in SH-SY5Y that constitutively overexpress hTau. Furthermore, intracerebral ventricular infusion of C004019 induced a robust tau clearance
. Most importantly, both single-dose and multiple-doses (once per 6 days for a total 5 times) subcutaneous administration of C004019 remarkably decreased tau levels in the brains of wild-type, hTau-transgenic and 3xTg-AD mice with improvement of synaptic and cognitive functions.
The PROTAC (C004019) created in the current study can selectively and efficiently promote tau clearance both
and
, which provides a promising drug candidate for AD and the related tauopathies.
Journal Article
Nanoligomers targeting NF-κB and NLRP3 reduce neuroinflammation and improve cognitive function with aging and tauopathy
by
Osburn, Shelby C.
,
Risen, Sydney J.
,
Nagpal, Prashant
in
Aging
,
Aging - drug effects
,
Animals
2024
Neuroinflammation contributes to impaired cognitive function in brain aging and neurodegenerative disorders like Alzheimer’s disease, which is characterized by the aggregation of pathological tau. One major driver of both age- and tau-associated neuroinflammation is the NF-κB and NLRP3 signaling axis. However, current treatments targeting NF-κB or NLRP3 may have adverse/systemic effects, and most have not been clinically translatable. In this study, we tested the efficacy of a novel, nucleic acid therapeutic (Nanoligomer) cocktail specifically targeting both NF-κB and NLRP3 in the brain for reducing neuroinflammation and improving cognitive function in old (aged 19 months) wildtype mice, and in rTg4510 tau pathology mice (aged 2 months). We found that 4 weeks of NF-κB/NLRP3-targeting Nanoligomer treatment strongly reduced neuro-inflammatory cytokine profiles in the brain and improved cognitive-behavioral function in both old and rTg4510 mice. These effects of NF-κB/NLRP3-targeting Nanoligomers were also associated with reduced glial cell activation and pathology, favorable changes in transcriptome signatures of glia-associated inflammation (reduced) and neuronal health (increased), and positive systemic effects. Collectively, our results provide a basis for future translational studies targeting both NF-κB and NLRP3 in the brain, perhaps using Nanoligomers, to inhibit neuroinflammation and improve cognitive function with aging and neurodegeneration.
Journal Article
Rapamycin Attenuates the Progression of Tau Pathology in P301S Tau Transgenic Mice
by
Skachokova, Zhiva
,
Sinnreich, Michael
,
Tolnay, Markus
in
Alzheimer's disease
,
Analysis
,
Animal models
2013
Altered autophagy contributes to the pathogenesis of Alzheimer's disease and other tauopathies, for which curative treatment options are still lacking. We have recently shown that trehalose reduces tau pathology in a tauopathy mouse model by stimulation of autophagy. Here, we studied the effect of the autophagy inducing drug rapamycin on the progression of tau pathology in P301S mutant tau transgenic mice. Rapamycin treatment resulted in a significant reduction in cortical tau tangles, less tau hyperphosphorylation, and lowered levels of insoluble tau in the forebrain. The favourable effect of rapamycin on tau pathology was paralleled by a qualitative reduction in astrogliosis. These effects were visible with early preventive or late treatment. We further noted an accumulation of the autophagy associated proteins p62 and LC3 in aged tangle bearing P301S mice that was lowered upon rapamycin treatment. Thus, rapamycin treatment defers the progression of tau pathology in a tauopathy animal model and autophagy stimulation may constitute a therapeutic approach for patients suffering from tauopathies.
Journal Article
Advances in tau-focused drug discovery for Alzheimer's disease and related tauopathies
by
Trojanowski, John Q.
,
Lee, Virginia M.-Y.
,
Brunden, Kurt R.
in
Alzheimer Disease - drug therapy
,
Alzheimer Disease - metabolism
,
Alzheimer Disease - pathology
2009
Key Points
A number of neurodegenerative diseases of the brain are characterized by inclusions within neurons that are comprised of aggregated fibrils of hyperphosphorylated tau protein. These disorders, which include Alzheimer's disease, Pick's disease, progressive supranuclear palsy, corticobasal degeneration and certain frontotemporal dementias, are broadly referred to as tauopathies.
Tau is normally a soluble protein that stabilizes microtubules within cells and is particularly enriched in neurons, where microtubules serve as the 'tracks' upon which cellular cargo is transported in axonal projections. The formation of insoluble tau aggregates could cause neurodegeneration through the formation of toxic tau species, or through a loss of tau function owing to its hyperphosphorylation and sequestration in inclusions.
Tau mutations have been shown to cause frontotemporal lobar degeneration with Parkinsonism linked to chromosome 17 (FTLD17), but tau mutations have not been identified in other tauopathies, including Alzheimer's disease. The causes of tau aggregation in these sporadic tauopathies are not fully understood, although tau hyperphosphorylation might be important, as it decreases tau binding to microtubules and increases tau fibrillization.
There is a growing interest in developing therapeutics that target pathological tau, particularly for the treatment of Alzheimer's disease. Most tau-directed drug discovery programmes are in early research stages and are not as advanced as programmes that aim to decrease levels of amyloid-β (Aβ) peptides, which form plaques in the Alzheimer's disease brain.
A number of approaches are being pursued for the treatment of tauopathies, including the development of brain-penetrant compounds that can stabilize microtubules and so compensate for tau loss-of-function; reduce tau hyperphosphorylation; inhibit tau assembly into oligomers and fibrils; or enhance tau intracellular degradative pathways.
As tau-directed therapies move towards clinical testing in Alzheimer's disease and other tauopathies, they will face many of the difficulties that are presently being encountered in trials of Aβ-targeted drugs for Alzheimer's disease. These include drug safety and the challenge of demonstrating clinical efficacy in a population that is likely to have existing neurodegeneration.
Misfolded tau is found in a number of neurodegenerative diseases, including Alzheimer's disease, and could lead to neuronal dysfunction, owing to the formation of toxic species or loss of normal tau function. Given the recent failures of amyloid-β-targeted therapies, the tau-directed drug discovery programmes reviewed here could be an alternative strategy for the treatment of Alzheimer's disease.
Neuronal inclusions comprised of the microtubule-associated protein tau are found in numerous neurodegenerative diseases, commonly known as tauopathies. In Alzheimer's disease — the most prevalent tauopathy — misfolded tau is probably a key pathological agent. The recent failure of amyloid-β-targeted therapeutics in Phase III clinical trials suggests that it is timely and prudent to consider alternative drug discovery strategies for Alzheimer's disease. Here, we focus on strategies directed at reducing misfolded tau and compensating for the loss of normal tau function.
Journal Article
Inhibition of 2-Arachidonoylglycerol Metabolism Alleviates Neuropathology and Improves Cognitive Function in a Tau Mouse Model of Alzheimer’s Disease
by
Hu, Mei
,
Hashem, Jack
,
Gao, Fei
in
2-Arachidonoylglycerol
,
Alzheimer Disease - drug therapy
,
Alzheimer Disease - genetics
2021
Alzheimer’s disease (AD) is the most common cause of dementia, which affects more than 5 million individuals in the USA. Unfortunately, no effective therapies are currently available to prevent development of AD or to halt progression of the disease. It has been proposed that monoacylglycerol lipase (MAGL), the key enzyme degrading the endocannabinoid 2-arachidonoylglycerol (2-AG) in the brain, is a therapeutic target for AD based on the studies using the APP transgenic models of AD. While inhibition of 2-AG metabolism mitigates β-amyloid (Aβ) neuropathology, it is still not clear whether inactivation of MAGL alleviates tauopathies as accumulation and deposition of intracellular hyperphosphorylated tau protein are the neuropathological hallmark of AD. Here we show that JZL184, a potent MAGL inhibitor, significantly reduced proinflammatory cytokines, astrogliosis, phosphorylated GSK3β and tau, cleaved caspase-3, and phosphorylated NF-kB while it elevated PPARγ in P301S/PS19 mice, a tau mouse model of AD. Importantly, tau transgenic mice treated with JZL184 displayed improvements in spatial learning and memory retention. In addition, inactivation of MAGL ameliorates deteriorations in expression of synaptic proteins in P301S/PS19 mice. Our results provide further evidence that MAGL is a promising therapeutic target for AD.
Journal Article
Deferiprone Treatment in Aged Transgenic Tau Mice Improves Y-Maze Performance and Alters Tau Pathology
by
Rao, Shalini S.
,
Shukla, Jay J.
,
Lago, Larissa
in
Aging - drug effects
,
Aging - genetics
,
Aging - pathology
2021
The accumulation of neurofibrillary tangles (NFTs), which is composed of abnormally hyperphosphorylated tau aggregates, is the classic neuropathology associated with cognitive dysfunction in tauopathies such as Alzheimer's disease (AD). However, there is an emerging theory suggesting that dysregulation in cerebral iron may contribute to NFT formation. Iron is speculated to bind to tau and induce conformational changes of the protein, potentially leading to subsequent aggregation and cognitive decline. Deferiprone (DFP) is a clinically available iron chelator, which has demonstrated potential therapeutic advantages of chelating iron in neurodegenerative disorders, and is currently in clinical trials for AD. However, its effect on tau pathology remains unclear. Here, we report the effects of short-term DFP treatment (4 weeks, 100 mg/kg/daily, via oral gavage) in a mixed-gender cohort of the rTg(tauP301L)4510 mouse model of tauopathy. Our results revealed that DFP improved Y-maze and open field performance, accompanied by a 28% decrease in brain iron levels, measured by inductively coupled plasma mass spectrometry (ICP-MS) and reduced AT8-labeled p-tau within the hippocampus in transgenic tau mice. This data supports the notion that iron may play a neurotoxic role in tauopathies and may be a potential therapeutic target for this class of disorders that can be modulated by the clinically available metal chelator DFP.
Journal Article
Efficacy and Safety of A Liposome-Based Vaccine against Protein Tau, Assessed in Tau.P301L Mice That Model Tauopathy
by
Gafner, Valérie
,
López-Deber, María Pilar
,
Adolfsson, Oskar
in
Alzheimer Vaccines - administration & dosage
,
Alzheimer Vaccines - immunology
,
Alzheimer's disease
2013
Progressive aggregation of protein Tau into oligomers and fibrils correlates with cognitive decline and synaptic dysfunction, leading to neurodegeneration in vulnerable brain regions in Alzheimer's disease. The unmet need of effective therapy for Alzheimer's disease, combined with problematic pharmacological approaches, led the field to explore immunotherapy, first against amyloid peptides and recently against protein Tau. Here we adapted the liposome-based amyloid vaccine that proved safe and efficacious, and incorporated a synthetic phosphorylated peptide to mimic the important phospho-epitope of protein Tau at residues pS396/pS404. We demonstrate that the liposome-based vaccine elicited, rapidly and robustly, specific antisera in wild-type mice and in Tau.P301L mice. Long-term vaccination proved to be safe, because it improved the clinical condition and reduced indices of tauopathy in the brain of the Tau.P301L mice, while no signs of neuro-inflammation or other adverse neurological effects were observed. The data corroborate the hypothesis that liposomes carrying phosphorylated peptides of protein Tau have considerable potential as safe and effective treatment against tauopathies, including Alzheimer's disease.
Journal Article
Brain-derived neurotrophic factor protects against tau-related neurodegeneration of Alzheimer’s disease
by
Liu, Y-H
,
Walker, D G
,
Zhou, H-D
in
631/154/436
,
631/378/1595
,
Alzheimer Disease - drug therapy
2016
Reduced expression of brain-derived neurotrophic factor (BDNF) has a crucial role in the pathogenesis of Alzheimer’s disease (AD), which is characterized with the formation of neuritic plaques consisting of amyloid-beta (Aβ) and neurofibrillary tangles composed of hyperphosphorylated tau protein. A growing body of evidence indicates a potential protective effect of BDNF against Aβ-induced neurotoxicity in AD mouse models. However, the direct therapeutic effect of BDNF supplement on tauopathy in AD remains to be established. Here, we found that the BDNF level was reduced in the serum and brain of AD patients and P301L transgenic mice (a mouse model of tauopathy). Intralateral ventricle injection of adeno-associated virus carrying the gene encoding human BDNF (AAV-BDNF) achieved stable expression of BDNF gene and restored the BDNF level in the brains of P301L mice. Restoration of the BDNF level attenuated behavioral deficits, prevented neuron loss, alleviated synaptic degeneration and reduced neuronal abnormality, but did not affect tau hyperphosphorylation level in the brains of P301L mice. Long-term expression of AAV-BDNF in the brain was well tolerated by the mice. These findings suggest that the gene delivery of BDNF is a promising treatment for tau-related neurodegeneration for AD and other neurodegenerative disorders with tauopathy.
Journal Article
The therapeutic landscape of tauopathies: challenges and prospects
by
Toledo-Sherman, Leticia M.
,
Cummings, Jeffrey L.
,
May, Patrick C.
in
Alzheimer Disease - pathology
,
Alzheimer's disease
,
Amyotrophic lateral sclerosis
2023
Tauopathies are a group of neurodegenerative disorders characterized by the aggregation of the microtubule-associated protein tau. Aggregates of misfolded tau protein are believed to be implicated in neuronal death, which leads to a range of symptoms including cognitive decline, behavioral change, dementia, and motor deficits. Currently, there are no effective treatments for tauopathies. There are four clinical candidates in phase III trials and 16 in phase II trials. While no effective treatments are currently approved, there is increasing evidence to suggest that various therapeutic approaches may slow the progression of tauopathies or improve symptoms. This review outlines the landscape of therapeutic drugs (indexed through February 28, 2023) that target tau pathology and describes drug candidates in clinical development as well as those in the discovery and preclinical phases. The review also contains information on notable therapeutic programs that are inactive or that have been discontinued from development.
Journal Article
Current Status of Clinical Trials on Tau Immunotherapies
by
Sigurdsson, Einar M.
,
Ji, Changyi
in
Alzheimer Disease - drug therapy
,
Alzheimer's disease
,
Animal models
2021
Tau immunotherapies have advanced from proof-of-concept studies to over a dozen clinical trials for Alzheimer’s disease (AD) and other tauopathies. Mechanistic studies in animal and culture models have provided valuable insight into how these therapies may work but multiple pathways are likely involved. Different groups have emphasized the importance of intracellular vs extracellular antibody-mediated clearance of the tau protein and there is no consensus on which pool of tau should ideally be targeted. Likewise, various normal and disease-selective epitopes are being targeted, and the antibody isotypes either favor phagocytosis of the tau-antibody complex or are neutral in that aspect. Most of the clinical trials are in early stages, thus their efficacy is not yet known, but all have been without any major adverse effects and some have reported target engagement. A few have been discontinued. One in phase I, presumably because of a poor pharmacokinetic profile, and three in phase II for a lack of efficacy although this trial stage is not well powered for efficacy measures. In these phase II studies, trials with two antibodies in patients with progressive supranuclear palsy or other primary tauopathies were halted but are continuing in patients with AD, and one antibody trial was stopped in early-stage AD but is continuing in moderate AD. These three antibodies have been reported to only work extracellularly and tau is not increased in the cerebrospinal fluid of primary tauopathies, which may explain the failures of two of them. In the discontinued AD trial, there are some concerns about how much of extracellular tau contains the N-terminal epitope that is being targeted. In addition, extracellular tau is only a small part of total tau, compared to intracellular tau. Targeting only the former may not be sufficient for functional benefits. Given these outcomes, decision makers within the pharmaceutical companies who green light these trials should attempt to target tau not only extracellularly but also intracellularly to increase their chances of success. Hopefully, some of the ongoing trials will provide some functional benefits to the large number of patients with tauopathies.
Journal Article