Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 300
1.
Transl Neurodegener ; 11(1): 34, 2022 07 01.
Article En | MEDLINE | ID: mdl-35773715

BACKGROUND: Neuronal dysfunction and degeneration linked to α-synuclein (αS) pathology is thought to be responsible for the progressive nature of Parkinson's disease and related dementia with Lewy bodies. Studies have indicated bidirectional pathological relationships between αS pathology and tau abnormalities. We recently showed that A53T mutant human αS (HuαS) can cause post-synaptic and cognitive deficits that require microtubule-associated protein tau expression. However, the role of tau in the development of αS pathology and subsequent neuronal dysfunction has been controversial. Herein, we set out to determine the role of tau in the onset and progression of αS pathology (α-synucleinopathy) using a transgenic mouse model of α-synucleinopathy lacking mouse tau expression. METHODS: Transgenic mice expressing A53T mutant HuαS (TgA53T) were crossed with mTau-/- mice to generate TgA53T/mTau-/-. To achieve more uniform induction of α-synucleinopathy in mice, we used intramuscular injections of αS preformed fibrils (PFF) in non-transgenic (nTg), TgA53T, TgA53T/mTau-/-, and mTau-/- mice. Motor behavior was analyzed at 70 days post inoculation (dpi) of PFF and tissues for biochemical and neuropathological analysis were collected at 40 dpi, 70 dpi, and end stage. RESULTS: Loss of tau expression significantly delayed the onset of motor deficits in the TgA53T model and the progression of α-synucleinopathy disease, as evidenced by a significant reduction in histopathological and behavioral markers of neurodegeneration and disease, and a significant improvement in survival. In vitro application of PFF to primary mouse hippocampal neurons demonstrated no changes in PFF uptake and processing or pS129 αS aggregation as a function of tau expression. However, PFF-induced neurotoxicity, including morphological deficits in nTg neurons, was prevented with tau removal. CONCLUSIONS: Collectively, our data suggest that tau is likely acting downstream of αS pathology to affect neuronal homeostasis and survival. This work further supports the investigation of tau in α-synucleinopathies to identify novel disease-modifying therapeutic strategies.


Parkinson Disease , Synucleinopathies , tau Proteins , Animals , Disease Models, Animal , Hippocampus/metabolism , Hippocampus/pathology , Mice , Mice, Transgenic , Parkinson Disease/genetics , Parkinson Disease/pathology , Synucleinopathies/genetics , Synucleinopathies/metabolism , Synucleinopathies/pathology , alpha-Synuclein/genetics , alpha-Synuclein/metabolism , tau Proteins/biosynthesis , tau Proteins/deficiency , tau Proteins/genetics
2.
Brain Res Bull ; 175: 136-149, 2021 10.
Article En | MEDLINE | ID: mdl-34284074

Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder characterized by irreversible cognitive deficits and memory dysfunction. Dopamine is the most abundant catecholaminergic neurotransmitter in the brain which regulates motivation, reward, movement, and cognition. Recently, increasing evidences have shown that dopaminergic system is disturbed in AD conditions, and pharmacological interventions targeting dopamine D1 receptor (DRD1) exhibit certain therapeutic benefits in AD models. However, the underlying link between DRD1 and AD remains elusive. This study sought to test whether the selective DRD1 agonist A-68930 could improve streptozotocin (STZ)-induced cognitive impairment in mice. Here we found that A-68930 treatment through intraperitoneal injection efficiently alleviated STZ-induced cognitive deficits in mice. Moreover, our mechanism researches revealed that the DRD1 signaling induced by A-68930 significantly rescued STZ-induced mitochondrial biogenesis deficit, mitochondrial dysfunction, Aß overexpression, and tau phosphorylation in mice hippocampus and cortex and SH-SY5Y cells, which may be mediated through stimulating AMPK/PGC-1α pathway. This study indicates that DRD1 agonist A-68930 can improve STZ-induced cognitive deficits and mitochondrial dysfunction in vivo and in vitro, and DRD1 may represent an appropriate target candidate for AD drug development.


Alzheimer Disease/drug therapy , Chromans/therapeutic use , Cognition Disorders/prevention & control , Cognition Disorders/psychology , Diabetes Mellitus, Experimental/drug therapy , Dopamine Agonists/therapeutic use , Mitochondria/metabolism , Receptors, Dopamine D1/agonists , Amyloid beta-Protein Precursor/biosynthesis , Amyloid beta-Protein Precursor/genetics , Animals , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/psychology , Humans , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, Inbred ICR , Organelle Biogenesis , Receptors, Dopamine D1/genetics , tau Proteins/biosynthesis , tau Proteins/genetics
3.
Aging (Albany NY) ; 13(13): 17097-17117, 2021 07 12.
Article En | MEDLINE | ID: mdl-34252884

Aberrant expression and denaturation of Tau, amyloid-beta and TDP-43 can lead to cell death and is a major component of pathologies such as Alzheimer's Disease (AD). AD neurons exhibit a reduced ability to form autophagosomes and degrade proteins via autophagy. Using genetically manipulated colon cancer cells we determined whether drugs that directly inhibit the chaperone ATPase activity or cause chaperone degradation and endoplasmic reticulum stress signaling leading to macroautophagy could reduce the levels of these proteins. The antiviral chaperone ATPase inhibitor AR12 reduced the ATPase activities and total expression of GRP78, HSP90, and HSP70, and of Tau, Tau 301L, APP, APP692, APP715, SOD1 G93A and TDP-43. In parallel, it increased the phosphorylation of ATG13 S318 and eIF2A S51 and caused eIF2A-dependent autophagosome formation and autophagic flux. Knock down of Beclin1 or ATG5 prevented chaperone, APP and Tau degradation. Neratinib, used to treat HER2+ breast cancer, reduced chaperone levels and expression of Tau and APP via macroautophagy, and neratinib interacted with AR12 to cause further reductions in protein levels. The autophagy-regulatory protein ATG16L1 is expressed as two isoforms, T300 or A300: Africans trend to express T300 and Europeans A300. We observed higher basal expression of Tau in T300 cells when compared to isogenic A300 cells. ATG16L1 isoform expression did not alter basal levels of HSP90, HSP70 or HSP27, however, basal levels of GRP78 were reduced in A300 cells. The abilities of both AR12 and neratinib to stimulate ATG13 S318 and eIF2A S51 phosphorylation and autophagic flux was also reduced in A300 cells. Our data support further evaluation of AR12 and neratinib in neuronal cells as repurposed treatments for AD.


Autophagosomes/drug effects , Autophagy/drug effects , Heat-Shock Proteins/antagonists & inhibitors , Signal Transduction/drug effects , Adenosine Triphosphatases/antagonists & inhibitors , Amyloid beta-Protein Precursor/biosynthesis , Amyloid beta-Protein Precursor/genetics , Autophagy-Related Protein 5/genetics , Beclin-1/genetics , Black People , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Endoplasmic Reticulum Chaperone BiP , Gene Knockdown Techniques , Humans , Quinolines/pharmacology , Superoxide Dismutase-1/biosynthesis , Superoxide Dismutase-1/genetics , White People , tau Proteins/biosynthesis , tau Proteins/genetics
4.
Sci Rep ; 11(1): 12946, 2021 06 21.
Article En | MEDLINE | ID: mdl-34155306

The deposition of tau aggregates throughout the brain is a pathological characteristic within a group of neurodegenerative diseases collectively termed tauopathies, which includes Alzheimer's disease. While recent findings suggest the involvement of unconventional secretory pathways driving tau into the extracellular space and mediating the propagation of the disease-associated pathology, many of the mechanistic details governing this process remain elusive. In the current study, we provide an in-depth characterization of the unconventional secretory pathway of tau and identify novel molecular determinants that are required for this process. Here, using Drosophila models of tauopathy, we correlate the hyperphosphorylation and aggregation state of tau with the disease-related neurotoxicity. These newly established systems recapitulate all the previously identified hallmarks of tau secretion, including the contribution of tau hyperphosphorylation as well as the requirement for PI(4,5)P2 triggering the direct translocation of tau. Using a series of cellular assays, we demonstrate that both the sulfated proteoglycans on the cell surface and the correct orientation of the protein at the inner plasma membrane leaflet are critical determinants of this process. Finally, we identify two cysteine residues within the microtubule binding repeat domain as novel cis-elements that are important for both unconventional secretion and trans-cellular propagation of tau.


Gene Expression Regulation , Regulatory Sequences, Nucleic Acid , tau Proteins/biosynthesis , tau Proteins/genetics , Animals , CHO Cells , Cell Membrane/metabolism , Cell Membrane Permeability , Chromatography, Liquid , Cricetulus , Cysteine/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Fluorescent Antibody Technique , Gene Expression Profiling , Phosphorylation , Protein Transport , Recombinant Proteins , Retina/metabolism , Tandem Mass Spectrometry
5.
Nature ; 594(7861): 117-123, 2021 06.
Article En | MEDLINE | ID: mdl-34012113

The human genome expresses thousands of natural antisense transcripts (NAT) that can regulate epigenetic state, transcription, RNA stability or translation of their overlapping genes1,2. Here we describe MAPT-AS1, a brain-enriched NAT that is conserved in primates and contains an embedded mammalian-wide interspersed repeat (MIR), which represses tau translation by competing for ribosomal RNA pairing with the MAPT mRNA internal ribosome entry site3. MAPT encodes tau, a neuronal intrinsically disordered protein (IDP) that stabilizes axonal microtubules. Hyperphosphorylated, aggregation-prone tau forms the hallmark inclusions of tauopathies4. Mutations in MAPT cause familial frontotemporal dementia, and common variations forming the MAPT H1 haplotype are a significant risk factor in many tauopathies5 and Parkinson's disease. Notably, expression of MAPT-AS1 or minimal essential sequences from MAPT-AS1 (including MIR) reduces-whereas silencing MAPT-AS1 expression increases-neuronal tau levels, and correlate with tau pathology in human brain. Moreover, we identified many additional NATs with embedded MIRs (MIR-NATs), which are overrepresented at coding genes linked to neurodegeneration and/or encoding IDPs, and confirmed MIR-NAT-mediated translational control of one such gene, PLCG1. These results demonstrate a key role for MAPT-AS1 in tauopathies and reveal a potentially broad contribution of MIR-NATs to the tightly controlled translation of IDPs6, with particular relevance for proteostasis in neurodegeneration.


Protein Biosynthesis/genetics , Proteostasis/genetics , RNA, Antisense/genetics , Tauopathies/genetics , Tauopathies/metabolism , tau Proteins/genetics , tau Proteins/metabolism , Aged , Animals , Binding Sites , Brain/metabolism , Brain/pathology , Case-Control Studies , Cell Differentiation , Disease Progression , Female , Humans , Internal Ribosome Entry Sites/genetics , Male , Mice , Mice, Transgenic , Middle Aged , Neurons/metabolism , Neurons/pathology , Ribosomes/metabolism , tau Proteins/biosynthesis
6.
Acta Neuropathol Commun ; 9(1): 86, 2021 05 12.
Article En | MEDLINE | ID: mdl-33980303

Chronic traumatic encephalopathy (CTE) is a progressive neurodegenerative disease, characterized by hyperphosphorylated tau, found in individuals with a history of exposure to repetitive head impacts. While the neuropathologic hallmark of CTE is found in the cortex, hippocampal tau has proven to be an important neuropathologic feature to examine the extent of disease severity. However, the hippocampus is also heavily affected in many other tauopathies, such as Alzheimer's disease (AD). How CTE and AD differentially affect the hippocampus is unclear. Using immunofluorescent analysis, a detailed histologic characterization of 3R and 4R tau isoforms and their differential accumulation in the temporal cortex in CTE and AD was performed. CTE and AD were both observed to contain mixed 3R and 4R tau isoforms, with 4R predominating in mild disease and 3R increasing proportionally as pathological severity increased. CTE demonstrated high levels of tau in hippocampal subfields CA2 and CA3 compared to CA1. There were also low levels of tau in the subiculum compared to CA1 in CTE. In contrast, AD had higher levels of tau in CA1 and subiculum compared to CA2/3. Direct comparison of the tau burden between AD and CTE demonstrated that CTE had higher tau densities in CA4 and CA2/3, while AD had elevated tau in the subiculum. Amyloid beta pathology did not contribute to tau isoform levels. Finally, it was demonstrated that higher levels of 3R tau correlated to more severe extracellular tau (ghost tangles) pathology. These findings suggest that mixed 3R/4R tauopathies begin as 4R predominant then transition to 3R predominant as pathological severity increases and ghost tangles develop. Overall, this work demonstrates that the relative deposition of tau isoforms among hippocampal subfields can aid in differential diagnosis of AD and CTE, and might help improve specificity of biomarkers for in vivo diagnosis.


Alzheimer Disease/metabolism , Chronic Traumatic Encephalopathy/metabolism , Hippocampus/metabolism , tau Proteins/biosynthesis , Adult , Aged , Aged, 80 and over , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Chronic Traumatic Encephalopathy/genetics , Chronic Traumatic Encephalopathy/pathology , Female , Gene Expression , Hippocampus/pathology , Humans , Male , Middle Aged , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , tau Proteins/genetics
7.
Exp Neurol ; 343: 113766, 2021 09.
Article En | MEDLINE | ID: mdl-34029610

Tauopathies, including Alzheimer's disease, are characterized by progressive accumulation of hyperphosphorylated and pathologic tau protein in association with onset of cognitive and behavioral impairment. Tau pathology is also associated with increased susceptibility to seizures and epilepsy, with tau-/- mice showing seizure resistance in some epilepsy models. To better understand how tau pathology is related to neuronal excitability, we performed whole-cell patch-clamp electrophysiology in dentate gyrus granule cells of tau-/- and human-tau expressing, htau mice. The htau mouse is unique from other transgenic tau models in that the endogenous murine tau gene has been and replaced with readily phosphorylated human tau. We assessed several measures of neuronal excitability, including evoked action potential frequency and excitatory synaptic responses in dentate granule cells from tau-/-, htau, and non-transgenic control mice at 1.5, 4, and 9 months of age. Compared to age matched controls, dentate granule cells from both tau-/- and htau mice had a lower peak frequency of evoked action potentials and greater paired pulse facilitation, suggesting reduced neuronal excitability. Our results suggest that neuronal excitability is more strongly influenced by the absence of functional tau than by the presence of pathologic tau. These results also suggest that tau's effect on neuronal excitability is more complex than previously understood.


Dentate Gyrus/cytology , Dentate Gyrus/metabolism , Excitatory Postsynaptic Potentials/physiology , tau Proteins/biosynthesis , Age Factors , Animals , Female , Gene Expression , Humans , Male , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Organ Culture Techniques , tau Proteins/genetics
8.
Acta Neuropathol Commun ; 9(1): 90, 2021 05 17.
Article En | MEDLINE | ID: mdl-34001284

Tauopathies are a group of neurodegenerative diseases characterized by the alteration/aggregation of TAU protein, for which there is still no effective treatment. Therefore, new pharmacological targets are being sought, such as elements of the endocannabinoid system (ECS). We analysed the occurrence of changes in the ECS in tauopathies and their implication in the pathogenesis. By integrating gene expression analysis, immunofluorescence, genetic and adeno-associated virus expressing TAU mouse models, we found a TAU-dependent increase in CB2 receptor expression in hippocampal neurons, that occurs as an early event in the pathology and was maintained until late stages. These changes were accompanied by alterations in the endocannabinoid metabolism. Remarkably, CB2 ablation in mice protects from neurodegeneration induced by hTAUP301L overexpression, corroborated at the level of cognitive behaviour, synaptic plasticity, and aggregates of insoluble TAU. At the level of neuroinflammation, the absence of CB2 did not produce significant changes in concordance with a possible neuronal location rather than its classic glial expression in these models. These findings were corroborated in post-mortem samples of patients with Alzheimer's disease, the most common tauopathy. Our results show that neurons with accumulated TAU induce the expression of the CB2 receptor, which enhances neurodegeneration. These results are important for our understanding of disease mechanisms, providing a novel therapeutic strategy to be investigated in tauopathies.


Brain/metabolism , Neuroprotection/physiology , Receptor, Cannabinoid, CB2/biosynthesis , Tauopathies/metabolism , tau Proteins/biosynthesis , Adult , Aged , Aged, 80 and over , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Brain/pathology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Middle Aged , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Receptor, Cannabinoid, CB2/genetics , Tauopathies/genetics , Tauopathies/pathology , tau Proteins/genetics
9.
J Med Chem ; 64(3): 1497-1509, 2021 02 11.
Article En | MEDLINE | ID: mdl-33499592

For the first time, the in silico design, screening, and in vitro validation of potent GSK-3ß type-II inhibitors are presented. In the absence of crystallographic evidence for a DFG-out GSK-3ß activation loop conformation, computational models were designed using an adapted DOLPHIN approach and a method consisting of Prime loop refinement, induced-fit docking, and molecular dynamics. Virtual screening of the Biogenics subset from the ZINC database led to an initial selection of 20 Phase I compounds revealing two low micromolar inhibitors in an isolated enzyme assay. Twenty more analogues (Phase II compounds) related to the hit [pyrimidin-2-yl]amino-furo[3,2-b]furyl-urea scaffold were selected for structure-activity relationship analysis. The Phase II studies led to five highly potent nanomolar inhibitors, with compound 23 (IC50 =0.087 µM) > 100 times more potent than the best Phase I inhibitor, and selectivity for GSK-3ß inhibition compared to homologous kinases was observed. Ex vivo experiments (SH-SY5Y cell lines) for tau hyperphosphorylation revealed promising neuroprotective effects at low micromolar concentrations. The type-II inhibitor design has been unraveled as a potential route toward more clinically effective GSK-3ß inhibitors.


Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/pharmacology , Cell Line, Tumor , Drug Design , Humans , Models, Molecular , Molecular Conformation , Molecular Docking Simulation , Molecular Dynamics Simulation , Phosphorylation , Structure-Activity Relationship , Substrate Specificity , tau Proteins/biosynthesis , tau Proteins/genetics
10.
J Neurochem ; 157(3): 727-751, 2021 05.
Article En | MEDLINE | ID: mdl-33264426

Parkinson disease (PD) is the second most common neurodegenerative disorder, whose prevalence is 2~3% in the population over 65. α-Synuclein aggregation is the major pathological hallmark of PD. However, recent studies have demonstrated enhancing evidence of tau pathology in PD. Despite extensive considerations, thus far, the actual spreading mechanism of neurodegeneration has remained elusive in a PD brain. This study aimed to further investigate the development of α-synuclein and tau pathology. We employed various PD models, including cultured neurons treated with either 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) or with recombinant α-synuclein. Also, we studied dopaminergic neurons of cytokine Interferon-ß knock-out. Moreover, we examined rats treated with 6-hydroxydopamine, Rhesus monkeys administrated with MPTP neurotoxin, and finally, human post-mortem brains. We found the α-synuclein phosphorylation triggers tau pathogenicity. Also, we observed more widespread phosphorylated tau than α-synuclein with prion-like nature in various brain areas. We optionally removed P-tau or P-α-synuclein from cytokine interferon-ß knock out with respective monoclonal antibodies. We found that tau immunotherapy suppressed neurodegeneration more than α-synuclein elimination. Our findings indicate that the pathogenic tau could be one of the leading causes of comprehensive neurodegeneration triggered by PD. Thus, we can propose an efficient therapeutic target to fight the devastating disorder.


Brain/pathology , Parkinson Disease/pathology , Tauopathies/pathology , alpha-Synuclein/genetics , Animals , Autopsy , Behavior, Animal , Cell Survival/drug effects , Dopaminergic Neurons/drug effects , Female , Humans , Interferon-beta/genetics , MPTP Poisoning/pathology , Macaca mulatta , Male , Mice , Mice, Knockout , Parkinson Disease/psychology , Pregnancy , Rats , Rats, Wistar , Recombinant Proteins , tau Proteins/biosynthesis , tau Proteins/genetics
11.
Neuropharmacology ; 180: 108305, 2020 12 01.
Article En | MEDLINE | ID: mdl-32931815

The NOD-like receptor pyrin domain-containing protein 3 (NLRP3) inflammasome has been implicated as a crucial component in both neurodegeneration and diabetes. However, the role of metabolic signalling pathways and the NLRP3 inflammasome in frontotemporal dementia remain largely elusive. We therefore investigated the effects of an NLRP3 inhibitor (MCC950) in a murine tau knock-in (PLB2TAU) model vs. wild-type (PLBWT) control mice. In male PLB2TAU mice (4 months at start of study), MCC950 treatment (20 mg/kg, for 12 weeks) improved insulin sensitivity and reduced circulating plasma insulin levels. Further molecular analysis suggested normalisation in insulin signalling pathways in both liver and muscle tissue. Treatment also resulted in improvements in inflammation and ER stress signalling, both peripherally and centrally, alongside a partial normalisation of phospho-tau levels. Overall, we provide evidence that MCC950 improved metabolic, inflammatory and frontotemporal dementia (FTD) relevant phenotypes in multiple tissues. NLRP3 inhibition may therefore offer a therapeutic approach to ameliorate FTD pathology.


Disease Models, Animal , Frontotemporal Dementia/drug therapy , Frontotemporal Dementia/metabolism , Furans/therapeutic use , Indenes/therapeutic use , Insulin Resistance/physiology , Receptors, Cell Surface/antagonists & inhibitors , Sulfonamides/therapeutic use , Animals , Frontotemporal Dementia/genetics , Furans/pharmacology , Humans , Indenes/pharmacology , Inflammation/drug therapy , Inflammation/genetics , Inflammation/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/genetics , Sulfonamides/pharmacology , tau Proteins/biosynthesis , tau Proteins/genetics
12.
J Alzheimers Dis ; 77(3): 1339-1351, 2020.
Article En | MEDLINE | ID: mdl-32804095

BACKGROUND: The risk of developing Alzheimer's disease as well as its progression and severity are known to be different in men and women, and cognitive decline is greater in women than in men at the same stage of disease and could be correlated at least in part on estradiol levels. OBJECTIVE: In our work we found that biological sex influences the effect of amyloid-ß42 (Aß42) monomers on pathological tau conformational change. METHODS: In this study we used transgenic mice expressing the wild-type human tau (hTau) which were subjected to intraventricular (ICV) injections of Aß peptides in nanomolar concentration. RESULTS: We found that Aß42 produces pathological conformational changes and hyperphosphorylation of tau protein in male or ovariectomized female mice but not in control females. The treatment of ovariectomized females with estradiol replacement protects against the pathological conformation of tau and seems to be mediated by antioxidant activity as well as the ability to modulate the expression of miRNA 218 linked to tau phosphorylation. CONCLUSION: Our study indicates that factors as age, reproductive stage, hormone levels, and the interplay with other risk factors should be considered in women, in order to identify the best appropriate therapeutic approach in prevention of cognitive impairment.


Amyloid beta-Peptides/toxicity , Antioxidants/administration & dosage , Estradiol/administration & dosage , MicroRNAs/biosynthesis , Peptide Fragments/toxicity , tau Proteins/biosynthesis , tau Proteins/chemistry , Animals , Estrogens/deficiency , Female , Humans , Injections, Intraventricular , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Ovariectomy , Protein Conformation
13.
Aging (Albany NY) ; 12(14): 14819-14829, 2020 07 23.
Article En | MEDLINE | ID: mdl-32701063

Previously, we revealed that brain Ang-(1-7) deficiency was involved in the pathogenesis of sporadic Alzheimer's disease (AD). We speculated that restoration of brain Ang-(1-7) levels might have a therapeutic effect against AD. However, the relatively short duration of biological effect limited the application of Ang-(1-7) in animal experiments. Since Ang-(1-7) is generated by its metabolic enzyme ACE2, we then tested the efficacy of an ACE2 activator diminazene aceturate (DIZE) on AD-like neuropathology and cognitive impairment in senescence-accelerated mouse prone substrain 8 (SAMP8) mice, an animal model of sporadic AD. Eight-month-old SAMP8 mice were injected intraperitoneally with vehicle or DIZE once a day for 30 consecutive days. DIZE markedly elevated brain Ang-(1-7) and MAS1 levels. Meanwhile, DIZE significantly reduced the levels of Aß1-42, hyperphosphorylated tau and pro-inflammatory cytokines in the brain. The synaptic and neuronal losses in the brain were ameliorated by DIZE. Importantly, DIZE improved spatial cognitive functions in the Morris water maze test. In conclusion, this study demonstrates that DIZE ameliorates AD-like neuropathology and rescues cognitive impairment in SAMP8 mice. These beneficial effects of DIZE may be achieved by activating brain ACE2/Ang-(1-7)/MAS1 axis. These findings highlight brain ACE2/Ang-(1-7)/MAS1 axis as a potential target for the treatment of sporadic AD.


Alzheimer Disease/drug therapy , Angiotensin-Converting Enzyme 2/drug effects , Cognitive Dysfunction/drug therapy , Diminazene/analogs & derivatives , Alzheimer Disease/pathology , Alzheimer Disease/psychology , Amyloid beta-Peptides/biosynthesis , Amyloid beta-Peptides/genetics , Angiotensin I/metabolism , Animals , Brain Chemistry/drug effects , Brain Chemistry/genetics , Cognitive Dysfunction/etiology , Cytokines/biosynthesis , Diminazene/therapeutic use , Infusions, Parenteral , Male , Maze Learning , Mice , Mice, Neurologic Mutants , Peptide Fragments/biosynthesis , Peptide Fragments/genetics , Peptide Fragments/metabolism , Proto-Oncogene Mas , Proto-Oncogene Proteins/genetics , Receptors, G-Protein-Coupled/genetics , tau Proteins/biosynthesis
14.
Drug Des Devel Ther ; 14: 2295-2306, 2020.
Article En | MEDLINE | ID: mdl-32606599

BACKGROUND: Subarachnoid hemorrhage (SAH) is a cerebral hemorrhage disease that severely damages the brain and causes cognitive impairment (CI). Therefore, accurate and appropriate treatment strategies are urgently needed. The application of nimodipine can not only improve blood circulation in patients with SAH but also repair ischemic neuron injury. PURPOSE: To investigate the effects of nimodipine and lncRNA nuclear paraspeckle assembly transcript 1 (NEAT1)/miR-27a/microtubule-associated protein tau (MAPT) axis on CI after SAH. METHODS: One hundred and twenty healthy male rats were selected and equally divided into control group, sham operation group, model group, PBS group, nimodipine group (drug group), NC siRNA group, NC mimics group, NEAT1 siRNA, miR-27a mimics, MAPT siRNA, drug + NEAT1-ad, and drug + NC-ad groups by random number table. Rats in the model group were constructed by double-hemorrhage model, and expression vectors were injected into the tail to regulate the expression of lncRNA NEAT1, miR-27a and MAPT. In addition, Western blot was employed to detect brain tissue protein, flow cytometry was applied to measure brain tissue apoptosis, and MTT was utilized to determine cell activity, so as to evaluate brain damage and cognitive function in each group. RESULTS: Nimodipine, down-regulated lncRNA NEAT1, up-regulated miR-27a and down-regulated MAPT all improved brain damage and CI, inhibited brain tissue cell apoptosis, and enhanced brain cell activity. The common binding sites of lncRNA NEAT1 and MAPT were found on the miR-27a sequence fragment, and miR-27a could be paired with the former two. Nimodipine was found to cause the down-regulation of lncRNA NEAT1 and MAPT, as well as the up-regulation of miR-27a. CONCLUSION: Nimodipine can improve CI after SAH in rats through the lncRNA NEAT1/miR-27a/MAPT axis.


Antihypertensive Agents/pharmacology , Cognitive Dysfunction/drug therapy , MicroRNAs/biosynthesis , Nimodipine/pharmacology , RNA, Long Noncoding/biosynthesis , Subarachnoid Hemorrhage/drug therapy , tau Proteins/biosynthesis , Animals , Cognitive Dysfunction/metabolism , Disease Models, Animal , Down-Regulation/drug effects , Male , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , Rats , Rats, Sprague-Dawley , Subarachnoid Hemorrhage/metabolism , Up-Regulation/drug effects , tau Proteins/metabolism
15.
Exp Eye Res ; 198: 108153, 2020 09.
Article En | MEDLINE | ID: mdl-32710889

Photoreceptor cells undergo changes with aging. It is unknown if their microtubules are stable or not with aging. This study examined photoreceptor cell ultrastructure from 18 human donor retinas (32 eyes; age: 45-94 years) and quantified the photoreceptors with altered microtubules over six to ninth decades in four defined retinal regions. In addition, immunoreactivity (IR) to microtubule-associated protein-2 (MAP-2), tau and hyperphophorylated tau was performed in retinal sections from companion eyes. In young donor retinas below 75 years of age, microtubules appeared straight in photoreceptor inner segments and axons. With age, they appeared bent or misaligned in macular and mid-peripheral photoreceptors. In addition, dense granular materials were present in photoreceptor axons and synaptic terminals in advanced ages. In all decades, rod microtubules were affected more than their cone counterparts (28% vs 15%, p < 0.005). Both rods and cones were significantly affected in mid-peripheral retina (5-8 mm outside the macular border) in eighth decade, compared to other decades or retinal regions (parafoveal, perifoveal and nasal) examined (p < 0.005). IR showed a steady expression of MAP-2 in inner segments, and tau in inner segments to axons below 75 years of age, but was absent for both markers in scattered macular and mid-peripheral photoreceptors in advanced ages (>75 years). IR to hyperphosphorylated tau was present mainly in inner retina and increased with aging. Markers of oxidative stress, e.g., lipid peroxidation (4-hydroxy 2-nonenal) and nitrosative stress (nitrotyrosine) were immunopositive in aged photoreceptors. The sporadic loss of MAP-2 and tau-IR in photoreceptors may be due to microtubule changes; all these changes may affect intracellular transport and be partly responsible for photoreceptor death in aged human retina.


Gene Expression Regulation , Microtubule-Associated Proteins/genetics , Microtubules/ultrastructure , Retinal Cone Photoreceptor Cells/metabolism , tau Proteins/genetics , Aged , Aged, 80 and over , Cellular Senescence , Female , Humans , Immunohistochemistry , Male , Microscopy, Electron, Transmission , Microtubule-Associated Proteins/biosynthesis , Middle Aged , RNA/genetics , Retinal Cone Photoreceptor Cells/ultrastructure , tau Proteins/biosynthesis
16.
J Alzheimers Dis ; 76(2): 553-569, 2020.
Article En | MEDLINE | ID: mdl-32538846

BACKGROUND: Neurofibrillary tangles (NFTs) and amyloid plaques are the neuropathological hallmarks in brains with Alzheimer's disease (AD). Post-translational modifications of tau, such as phosphorylation and truncation, have been proposed as initiators in the assembly of the abnormal paired helical filaments that constitute the NFTs. Neurons and NFTs are sites of matrix metalloproteinases (MMPs). OBJECTIVE: The aim of this study was to analyze the relationship of MMP-9 and tau protein in brain samples with AD. METHODS: This study was performed on brain tissue samples from patients with early, moderate, and late AD. MMPs and tau levels were analyzed by western blot and gelatin-substrate zymography. Immunofluorescence techniques and confocal microscopy were used to analyze the presence of both proteins in NFTs. Further, molecular dynamics simulations (MDS) and protein-protein docking were conducted to predict interaction between MMP-9 and tau protein. RESULTS: MMP-9 expression was greatest in moderate and late AD, whereas MMP-2 expression was only increased in late-stage AD. Interestingly, confocal microscopy revealed NFTs in which there was co-localization of MMP-9 and tau protein. MDS and protein-protein docking predictions indicate that a high-affinity complex can be formed between MMP-9 and full-length tau protein. CONCLUSION: These observations provide preliminary evidence of an interaction between these two proteins. Post-translational modifications of tau protein, such as C-terminal truncation or phosphorylation of amino acid residues in the MMP-9 recognition site and conformational changes in the protein, such as folding of the N-terminal sequence over the three-repeat domain, could preclude the interaction between MMP-9 and tau protein during stages of NFT development.


Alzheimer Disease/metabolism , Entorhinal Cortex/metabolism , Matrix Metalloproteinase 9/biosynthesis , tau Proteins/biosynthesis , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Entorhinal Cortex/pathology , Female , Humans , Male , Matrix Metalloproteinase 9/chemistry , Middle Aged , Molecular Docking Simulation , Protein Binding/physiology , Protein Structure, Secondary , tau Proteins/chemistry
17.
Int J Mol Sci ; 21(8)2020 Apr 15.
Article En | MEDLINE | ID: mdl-32326589

The brain is an organ in which energy metabolism occurs most intensively and glucose is an essential and dominant energy substrate. There have been many studies in recent years suggesting a close relationship between type 2 diabetes mellitus (T2DM) and Alzheimer's disease (AD) as they have many pathophysiological features in common. The condition of hyperglycemia exposes brain cells to the detrimental effects of glucose, increasing protein glycation and is the cause of different non-psychiatric complications. Numerous observational studies show that not only hyperglycemia but also blood glucose levels near lower fasting limits (72 to 99 mg/dL) increase the incidence of AD, regardless of whether T2DM will develop in the future. As the comorbidity of these diseases and earlier development of AD in T2DM sufferers exist, new AD biomarkers are being sought for etiopathogenetic changes associated with early neurodegenerative processes as a result of carbohydrate disorders. The S100B protein seem to be interesting in this respect as it may be a potential candidate, especially important in early diagnostics of these diseases, given that it plays a role in both carbohydrate metabolism disorders and neurodegenerative processes. It is therefore necessary to clarify the relationship between the concentration of the S100B protein and glucose and insulin levels. This paper draws attention to a valuable research objective that may in the future contribute to a better diagnosis of early neurodegenerative changes, in particular in subjects with T2DM and may be a good basis for planning experiments related to this issue as well as a more detailed explanation of the relationship between the neuropathological disturbances and changes of glucose and insulin concentrations in the brain.


Alzheimer Disease/blood , Alzheimer Disease/diagnosis , Amyloid beta-Peptides/metabolism , Brain/metabolism , Diabetes Mellitus, Type 2/metabolism , Hyperglycemia/metabolism , tau Proteins/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Biomarkers/blood , Biomarkers/cerebrospinal fluid , Carbohydrates/blood , Humans , S100 Calcium Binding Protein beta Subunit/blood , tau Proteins/biosynthesis
18.
Urol Oncol ; 38(6): 605.e9-605.e17, 2020 06.
Article En | MEDLINE | ID: mdl-32139291

INTRODUCTION: Microtubule-associated protein tau (MAPT) overexpression has been linked to poor prognosis in several cancers. MAPT-AS1 is a long noncoding RNA existing at the antisense strand of the MAPT promoter region. The clinical significance of MAPT and MAPT-AS-1 in clear cell renal cell carcinoma (ccRCC) is unknown. This study aimed to assess the expression and function of MAPT and MAPT-AS1 in ccRCC. METHODS: The expression of MAPT was determined using immunohistochemistry in ccRCC. The effects of MAPT knockdown on cell growth and invasion were evaluated and the interaction between MAPT and microtubule-associated protein tau antisense (MAPT-AS1) were analyzed. The expression of MAPT-AS1 was determined using quantitative reverse transcription polymerase chain reaction in ccRCC tissues. We investigated the effect of MAPT-AS1 knockdown on cell growth and invasion. We analyzed the regulation of MAPT and MAPT-AS1. RESULTS: Immunohistochemistry in 135 ccRCC cases showed that 61% of the cases were positive for MAPT. Kaplan-Meier analysis showed that the low expression of MAPT was associated with poor overall survival after nephrectomy. Knockdown of MAPT enhanced cell growth and invasion. quantitative reverse transcription polymerase chain reaction revealed a positive correlation between MAPT and MAPT-AS1. The expression of MAPT-AS1 was higher in ccRCC tissue than in nonneoplastic kidney tissue. Kaplan-Meier analysis showed that the low expression of MAPT-AS1 was associated with poor overall survival after nephrectomy by in silico analysis. MAPT-AS1 knockdown promoted cell growth and invasion activity. P53 knockout suppressed the expression of MAPT and MAPT-AS1. CONCLUSION: These results suggest that MAPT and MAPT-AS1 may be promising predictive biomarkers for survival and play a tumor-suppressive role in ccRCC.


Biomarkers, Tumor/physiology , Carcinoma, Renal Cell/etiology , Kidney Neoplasms/etiology , Tumor Suppressor Proteins/physiology , tau Proteins/physiology , Aged , Biomarkers, Tumor/biosynthesis , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/mortality , Female , Humans , Kidney Neoplasms/metabolism , Kidney Neoplasms/mortality , Male , Middle Aged , Prognosis , RNA, Long Noncoding , Survival Rate , Tumor Suppressor Proteins/biosynthesis , Tumor Suppressor Proteins/genetics , tau Proteins/biosynthesis , tau Proteins/genetics
19.
J Alzheimers Dis ; 74(2): 637-647, 2020.
Article En | MEDLINE | ID: mdl-32065789

Alzheimer's disease (AD) is neuropathologically defined by two key hallmarks: extracellular senile plaques composed primarily of amyloid-ß (Aß) peptide and intraneuronal neurofibrillary tangles, containing abnormally hyperphosphorylated tau protein. The tau protein is encoded by the MAPT gene. Recently, the H1 and H2 haplotypes of the MAPT gene were associated with AD risk. The minor MAPT H2 haplotype has been linked with a decreased risk of developing late-onset AD (LOAD). MAPT haplotypes show different levels of MAPT/Tau expression with H1 being ∼1.5-fold more expressed than H2, suggesting that MAPT expression level could be related to LOAD risk. In this study, we investigated whether this moderate difference in MAPT/Tau expression could influence Aß-induced toxicity in vivo. We show that modest overexpression of tau protein in Drosophila exacerbates neuronal phenotypes in AßPP/BACE1 flies. The exacerbation of neuronal defects correlates with the accumulation of insoluble dTau oligomers, suggesting that the moderate difference in level of tau expression observed between H1 and H2 haplotypes could influence Aß toxicity through the production of oligomeric tau insoluble species.


Amyloid beta-Peptides/toxicity , Neurons/metabolism , Phenotype , tau Proteins/biosynthesis , Animals , Animals, Genetically Modified , Drosophila , Gene Expression , Neurons/drug effects , tau Proteins/genetics
20.
J Neurosci ; 40(1): 220-236, 2020 01 02.
Article En | MEDLINE | ID: mdl-31685653

Tau is a microtubule-associated protein that becomes dysregulated in a group of neurodegenerative diseases called tauopathies. Differential tau isoforms, expression levels, promoters, and disruption of endogenous genes in transgenic mouse models of tauopathy make it difficult to draw definitive conclusions about the biological role of tau in these models. We addressed this shortcoming by characterizing the molecular and cognitive phenotypes associated with the pathogenic P301L tau mutation (rT2 mice) in relation to a genetically matched transgenic mouse overexpressing nonmutant (NM) 4-repeat (4R) human tau (rT1 mice). Both male and female mice were included in this study. Unexpectedly, we found that 4R NM human tau (hTau) exhibited abnormal dynamics in young mice that were lost with the P301L mutation, including elevated protein stability and hyperphosphorylation, which were associated with cognitive impairment in 5-month-old rT1 mice. Hyperphosphorylation of NM hTau was observed as early as 4 weeks of age, and transgene suppression for the first 4 or 12 weeks of life prevented abnormal molecular and cognitive phenotypes in rT1, demonstrating that NM hTau pathogenicity is specific to postnatal development. We also show that NM hTau exhibits stronger binding to microtubules than P301L hTau, and is associated with mitochondrial abnormalities. Overall, our genetically matched mice have revealed that 4R NM hTau overexpression is pathogenic in a manner distinct from classical aging-related tauopathy, underlining the importance of assaying the effects of transgenic disease-related proteins at appropriate stages in life.SIGNIFICANCE STATEMENT Due to differences in creation of transgenic lines, the pathological properties of the P301L mutation confers to the tau protein in vivo have remained elusive, perhaps contributing to the lack of disease-modifying therapies for tauopathies. In an attempt to characterize P301L-specific effects on tau biology and cognition in novel genetically matched transgenic mouse models, we surprisingly found that nonmutant human tau has development-specific pathogenic properties of its own. Our findings indicate that overexpression of 4-repeat human tau during postnatal development is associated with excessive microtubule binding, which may disrupt important cellular processes, such as mitochondrial dynamics, leading to elevated stability and hyperphosphorylation of tau, and eventual cognitive impairments.


Memory Disorders/genetics , Mitochondrial Diseases/genetics , tau Proteins/genetics , Animals , Cells, Cultured , Female , Genes, Synthetic , Hippocampus/cytology , Humans , INDEL Mutation , Male , Maze Learning , Memory Disorders/physiopathology , Mice , Mice, Transgenic , Microtubules/physiology , Mitochondria/metabolism , Mitochondria/ultrastructure , Mitochondrial Diseases/physiopathology , Mutation, Missense , Oxidative Stress , Phenotype , Phosphorylation , Point Mutation , Prosencephalon/physiology , Protein Processing, Post-Translational , Recombinant Proteins , Repetitive Sequences, Amino Acid , Species Specificity , Up-Regulation , tau Proteins/biosynthesis
...