Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
J Alzheimers Dis Rep ; 8(1): 615-626, 2024.
Article in English | MEDLINE | ID: mdl-38746631

ABSTRACT

Background: Diabetes mellitus (DM) increases the risk for cognitive impairment and Alzheimer's disease (AD). Diabetic ketoacidosis (DKA), a serious complication of DM, may also cause brain damage and further AD, but the underlying molecular mechanisms remain unclear. Objective: Our objective was to understand how DKA can promote neurodegeneration in AD. Methods: We induced DKA in rats through intraperitoneal injection of streptozotocin, followed by starvation for 48 hours and investigated AD-related brain alterations focusing on tau phosphorylation. Results: We found that DKA induced hyperphosphorylation of tau protein at multiple sites associated with AD. Studies of tau kinases and phosphatases suggest that the DKA-induced hyperphosphorylation of tau was mainly mediated through activation of c-Jun N-terminal kinase and downregulation of protein phosphatase 2A. Disruption of the mTOR-AKT (the mechanistic target of rapamycin-protein kinase B) signaling pathway and increased levels of synaptic proteins were also observed in the brains of rats with DKA. Conclusions: These results shed some light on the mechanisms by which DKA may increase the risk for AD.

2.
Front Aging Neurosci ; 15: 1073774, 2023.
Article in English | MEDLINE | ID: mdl-37091523

ABSTRACT

The regional distribution of neurofibrillary tangles of hyperphosphorylated tau aggregates is associated with the progression of Alzheimer's disease (AD). Misfolded proteopathic tau recruits naïve tau and templates its misfolding and aggregation in a prion-like fashion, which is believed to be the molecular basis of propagation of tau pathology. A practical way to assess tau seeding activity is to measure its ability to recruit/bind other tau molecules and to induce tau aggregation. Based on the properties of proteopathic tau, here we report the development of two simple assays to assess tau seeding activity ----- capture assay in vitro and seeded-tau aggregation assay in cultured cells. In the capture assay, proteopathic tau was applied onto a nitrocellulose membrane and the membrane was incubated with cell lysate containing HA-tagged tau151-391 (HA-tau151-391). The captured tau on the membrane was determined by immuno-blots developed with anti-HA. For the seeded-tau aggregation assay, HEK-293FT cells transiently expressing HA-tau151-391 were treated with proteopathic tau in the presence of Lipofectamine 2000 and then lysed with RIPA buffer. RIPA-insoluble fraction containing aggregated tau was obtained by ultracentrifugation and analyzed by immuno-blot developed with anti-HA. To validate these two assays, we assessed the seeding activity of tau in the middle frontal gyrus, middle temporal gyrus and basal forebrain of AD and control brains and found that AD, but not control, brain extracts effectively captured and seeded tau151-391 aggregation. Basal forebrain contained less phospho-tau and tau seeding activity. The levels of captured tau or seeded-tau aggregates were positively correlated to the levels of phospho-tau, Braak stages and tangle sores. These two assays are specific and sensitive and can be carried out in a regular biomedical laboratory setting by using routine biochemical techniques.

3.
CNS Neurosci Ther ; 28(12): 2245-2259, 2022 12.
Article in English | MEDLINE | ID: mdl-36114722

ABSTRACT

INTRODUCTION: Neurofibrillary tangle (NFT) of hyperphosphorylated tau is a hallmark of Alzheimer's disease (AD) and related tauopathies. Tau lesion starts in the trans-entorhinal cortex, from where it spreads to limbic regions, followed by neocortical areas. The regional distribution of NFTs associates with the progression of AD. Accumulating evidence suggests that proteopathic tau can seed tau aggregation in a prion-like fashion in vitro and in vivo. Inhibition of tau seeding activity could provide a potential therapeutic opportunity to block the propagation of tau pathology in AD and related tauopathies. AIMS: In the present study, we investigated the role of 77G7, a monoclonal tau antibody to the microtubule-binding repeats, in repressing the seeding activity of proteopathic tau. RESULTS: We found that 77G7 had a higher affinity toward aggregated pathological tau fractions than un-aggregated tau derived from AD brain. 77G7 inhibited the internalization of tau aggregates by cells, blocked AD O-tau to capture normal tau, and to seed tau aggregation in vitro and in cultured cells. Tau pathology induced by hippocampal injection of AD O-tau in 3xTg-AD mice was suppressed by mixing 77G7 with AD O-tau. Intravenous administration of 77G7 ameliorated site-specific hyperphosphorylation of tau induced by AD O-tau in the hippocampi of Tg/hTau mice. CONCLUSION: These findings indicate that 77G7 can effectively suppress the seeding activity of AD O-tau and thus could be developed as a potential immunotherapeutic drug to inhibit the propagation of tau pathology in AD and related tauopathies.


Subject(s)
Alzheimer Disease , Tauopathies , Animals , Mice , tau Proteins/metabolism , Tauopathies/metabolism , Tauopathies/pathology , Alzheimer Disease/metabolism , Brain/metabolism , Antibodies, Monoclonal , Microtubules/metabolism , Microtubules/pathology
4.
Acta Neuropathol Commun ; 10(1): 132, 2022 09 05.
Article in English | MEDLINE | ID: mdl-36064460

ABSTRACT

Propagation of tau pathology via the seeding of naive tau aggregation underlies the progression of Alzheimer's disease (AD) and related tauopathies. Individuals with Down syndrome (DS) develop tau pathology at the fourth decade of life, but tau seeding activity in DS brain has not yet been determined. To measure tau seeding activity, we developed capture assay and seeded-tau aggregation assay with truncated tau151-391. By using brain extracts from AD and related tauopathies, we validated these two methods and found that the brain extracts from AD and related tauopathies, but not from controls and the diseases in which tau was not hyperphosphorylated, captured in vitro and seeded 3R-tau151-391 and 4R-tau151-391 to aggregate in cultured cells similarly. Captured tau151-391 levels were strongly correlated with the seeded-tau151-391 aggregation. Employing these two newly developed assays, we analyzed tau seeding activity in the temporal (TC), frontal (FC), and occipital cortex (OC); corpus callosum (CC); and cerebellar cortex (CBC) of DS and control brains. We found that the extracts of TC, FC, or OC, but not the CC or CBC of DS or the corresponding brain regions of control cases, captured tau151-391. Levels of the captured tau151-391 by brain extracts were positively correlated with their levels of phosphorylated tau. Extracts of cerebral cortex and CC, but not CBC of DS with a similar tau level, induced more tau151-391 aggregation than did the corresponding samples from the control cases. Thus, higher tau seeding activity associated with tau hyperphosphorylation was found in the TC, FC, and OC of DS compared with the corresponding control regions as well as with the CBC and CC of DS. Of note, these two assays are sensitive, specific, and repeatable at a low cost and provide a platform for measuring tau seeding activity and for drug screening that targets tau propagation.


Subject(s)
Alzheimer Disease , Down Syndrome , Tauopathies , Alzheimer Disease/pathology , Brain/pathology , Down Syndrome/pathology , Humans , Tauopathies/pathology , tau Proteins/metabolism
5.
Front Mol Neurosci ; 14: 631833, 2021.
Article in English | MEDLINE | ID: mdl-34054426

ABSTRACT

Accumulation of intracellular neurofibrillary tangles (NFTs), which are constituted of abnormally phosphorylated tau, is one of the neuropathological hallmarks of Alzheimer's disease (AD). The oligomeric aggregates of tau in AD brain (AD O-tau) are believed to trigger NFT spreading by seeding normal tau aggregation as toxic seeds, in a prion-like fashion. Here, we revealed the features of AD O-tau by Western blots using antibodies against various epitopes and determined the effect of dephosphorylation on the seeding activity of AD O-tau by capture and seeded aggregation assays. We found that N-terminal truncated and C-terminalhyperphosphorylated tau species were enriched in AD O-tau. Dephosphorylation of AD O-tau by alkaline phosphatasediminished its activity in capturing tau in vitro and ininducing insoluble aggregates in cultured cells. Our resultssuggested that dephosphorylation passivated the seeding activity ofAD O-tau. Inhibition of phosphorylation may be a potentstrategy to prevent the spreading of tau patho3logy.

6.
J Alzheimers Dis ; 82(2): 631-646, 2021.
Article in English | MEDLINE | ID: mdl-34057082

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by impairments in synaptic plasticity and cognitive performance. Cognitive dysfunction and loss of neuronal plasticity are known to begin decades before the clinical diagnosis of the disease. The important influence of congenital genetic mutations on the early development of AD provides a novel opportunity to initiate treatment during early development to prevent the Alzheimer-like behavior and synaptic dysfunction. OBJECTIVE: To explore strategies for early intervention to prevent Alzheimer's disease. METHODS: In the present study, we investigated the effect of treatment during early development with a ciliary neurotrophic factor (CNTF) derived peptidergic compound, P021 (Ac-DGGLAG-NH2) on cognitive function and synaptic plasticity in 3xTg-AD transgenic mouse model of AD. 3xTg-AD and genetic background-matched wild type female mice were treated from birth to postnatal day 120 with P021 in diet or as a control with vehicle diet, and cognitive function and molecular markers of neuroplasticity were evaluated. RESULTS: P021 treatment during early development prevented cognitive impairment and increased expressions of pCREB and BDNF that activated downstream various signaling cascades such as PLC/PKC, MEK/ERK and PI3K/Akt, and ameliorated synaptic protein deficit in 4-month-old 3xTg-AD mice. CONCLUSION: These findings indicate that treatment with the neurotrophic peptide mimetic such as P021 during early development can be an effective therapeutic strategy to rescue synaptic deficit and cognitive impairment in familial AD and related tauopathies.


Subject(s)
Adamantane/analogs & derivatives , Alzheimer Disease , Cognitive Dysfunction , Neuronal Plasticity , Oligopeptides/pharmacology , Adamantane/pharmacology , Alzheimer Disease/genetics , Alzheimer Disease/prevention & control , Alzheimer Disease/psychology , Animals , Cognition/drug effects , Cognitive Dysfunction/diagnosis , Cognitive Dysfunction/etiology , Cognitive Dysfunction/prevention & control , Disease Models, Animal , Early Medical Intervention/methods , Maze Learning , Mice , Mice, Transgenic , Nerve Growth Factors/pharmacology , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Postpartum Period , Signal Transduction/drug effects , Treatment Outcome
7.
Acta Neuropathol Commun ; 9(1): 28, 2021 02 17.
Article in English | MEDLINE | ID: mdl-33597014

ABSTRACT

Neurofibrillary tangles (NFTs) made of abnormally hyperphosphorylated tau are a hallmark of Alzheimer's disease (AD) and related tauopathies. Regional distribution of NFTs is associated with the progression of the disease and has been proposed to be a result of prion-like propagation of misfolded tau. Tau in AD brain is heterogenous and presents in various forms. In the present study, we prepared different tau fractions by sedimentation combined with sarkosyl solubility from AD brains and analyzed their biochemical and pathological properties. We found that tau in oligomeric fraction (O-tau), sarkosyl-insoluble fractions 1 and 2 (SI1-tau and SI2-tau) and monomeric heat-stable fraction (HS-tau) showed differences in truncation, hyperphosphorylation, and resistance to proteinase K. O-tau, SI1-tau, and SI2-tau, but not HS-tau, were hyperphosphorylated at multiple sites and contained SDS- and ß-mercaptoethanol-resistant high molecular weight aggregates, which lacked the N-terminal portion of tau. O-tau and SI2-tau displayed more truncation and less hyperphosphorylation than SI1-tau. Resistance to proteinase K was increased from O-tau to SI1-tau to SI2-tau. O-tau and SI1-tau, but not SI2-tau or HS-tau, captured tau from cell lysates and seeded tau aggregation in cultured cells. Heat treatment could not kill the prion-like activity of O-tau to capture normal tau. Hippocampal injection of O-tau into 18-month-old FVB mice induced significant tau aggregation in both ipsilateral and contralateral hippocampi, but SI1-tau only induced tau pathology in the ipsilateral hippocampus, and SI2-tau and HS-tau failed to induce any detectable tau aggregation. These findings suggest that O-tau and SI1-tau have prion-like activities and may serve as seeds to recruit tau and template tau to aggregate, resulting in the propagation of tau pathology. Heterogeneity of tau pathology within AD brain results in different fractions with different biological and prion-like properties, which may pose a major challenge in targeting tau for development of effective therapeutic treatments.


Subject(s)
Alzheimer Disease/metabolism , Brain/metabolism , Prions/metabolism , tau Proteins/isolation & purification , tau Proteins/metabolism , Alzheimer Disease/pathology , Animals , Brain/pathology , Fluorescent Antibody Technique , HEK293 Cells , HeLa Cells , Hippocampus/pathology , Humans , Mice , Neurofibrillary Tangles/pathology , Phosphorylation
8.
J Alzheimers Dis ; 79(4): 1647-1659, 2021.
Article in English | MEDLINE | ID: mdl-33459649

ABSTRACT

BACKGROUND: Neurofibrillary pathology of abnormally hyperphosphorylated tau spreads along neuroanatomical connections, underlying the progression of Alzheimer's disease (AD). The propagation of tau pathology to axonally connected brain regions inevitably involves trafficking of seeding-competent tau within the axonal compartment of the neuron. OBJECTIVE: To determine the seeding activity of tau in cerebral gray and white matters of AD. METHODS: Levels of total tau, hyperphosphorylation of tau, and SDS- and ß-mercaptoethanol-resistant high molecular weight tau (HMW-tau) in crude extracts from gray and white matters of AD frontal lobes were analyzed by immuno-blots. Tau seeding activity was quantitatively assessed by measuring RIPA buffer-insoluble tau in HEK-293FT/tau151-391 cells treated with brain extracts. RESULTS: We found a comparable level of soluble tau in gray matter versus white matter of control brains, but a higher level of soluble tau in gray matter than white matter of AD brains. In AD brains, tau is hyperphosphorylated in both gray and white matters, with a higher level in the former. The extracts of both gray and white matters of AD brains seeded tau aggregation in HEK-293FT/tau151-391 cells but the white matter showed less potency. Seeding activity of tau in brain extracts was positively correlated with the levels of tau hyperphosphorylation and HMW-tau. RIPA-insoluble tau, but not RIPA-soluble tau, was hyperphosphorylated tau at multiple sites. CONCLUSION: Both gray and white matters of AD brain contain seeding-competent tau that can template aggregation of hyperphosphorylated tau, but the seeding potency is markedly higher in gray matter than in white matter.


Subject(s)
Alzheimer Disease/pathology , Brain/pathology , Gray Matter/pathology , White Matter/pathology , tau Proteins/metabolism , Alzheimer Disease/metabolism , Brain/metabolism , Disease Progression , Gray Matter/metabolism , Humans , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/pathology , White Matter/metabolism
9.
Alzheimers Res Ther ; 12(1): 102, 2020 08 27.
Article in English | MEDLINE | ID: mdl-32854771

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is a progressive neurodegenerative disorder of middle-aged to old individuals. The pathophysiological process of AD is believed to begin many years before the emergence of clinical symptoms. The important influence of congenital genetic aberrations on the development of AD provides a novel opportunity to initiate prenatal to early postnatal pharmacological treatment to address the role of this critical period of brain development in the disease. METHODS: We investigated for the first time the effect of oral treatment during prenatal to early postnatal development with a neurotrophic compound, P021 (Ac-DGGLAG-NH2), on neurobehavior and AD-like pathology in 3xTg-AD, a transgenic mouse model of AD. The transgenic and control wild-type female mice were treated from prenatal day 8 to postnatal day 21 with a custom-made diet containing P021 or a vehicle diet, followed by a standard diet. AD-type cognitive function and pathological features were studied during adulthood and old age. RESULTS: The P021 treatment rescued cognitive deficits at 4 months, reduced abnormal hyperphosphorylation and accumulation of tau at known major AD neurofibrillary pathology-associated sites, and decreased Aß plaque load at 22 months in 3xTg-AD mice. Prenatal to early postnatal treatment with P021 also ameliorated certain markers of postsynaptic deficits, including PSD-95 levels and CREB activity, and decreased one measure of neuroinflammation, GFAP level in the brain at 4 and 22 months in 3xTg mice. CONCLUSIONS: These findings suggest that neurotrophic impairment during early development can be one of the etiopathogenic factors of AD and that the neurotrophic peptide mimetic is a potential early prevention strategy for this disease.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor , Animals , Brain/metabolism , Disease Models, Animal , Female , Mice , Mice, Inbred C57BL , Mice, Transgenic , Plaque, Amyloid , Pregnancy , tau Proteins/metabolism
10.
Front Aging Neurosci ; 11: 34, 2019.
Article in English | MEDLINE | ID: mdl-30890929

ABSTRACT

Neurofibrillary tangles (NFTs) made up of hyperphosphorylated tau are a histopathological hallmark of Alzheimer's disease (AD) and related tauopathies. Hyperphosphorylation of tau is responsible for its loss of normal physiological function, gain of toxicity and its aggregation to form NFTs. Injection of misfolded tau seeds into mouse brain induces tau aggregation, but the nature of tau phosphorylation in pathologic tau seeded pathology is unclear. In the present study, we injected hyperphosphorylated and oligomeric tau isolated from AD brain (AD P-tau) into hippocampus of human tau transgenic mice and found that in addition to tau aggregation/pathology, tau was hyperphosphorylated at Ser202/Thr205, Thr212, Ser214, Thr217, Ser262, and Ser422 in AD P-tau injected hippocampus and at Ser422 in the contralateral hippocampus and in the ipsilateral cortex. AD P-tau-induced AD-like high molecular weight aggregation of tau that was SDS- and reducing agent-resistant and site-specifically hyperphosphorylated in the ipsilateral hippocampus. There were no detectable alterations in levels of tau phosphatases or tau kinases in AD P-tau-injected brains. Furthermore, we found that hyperphosphorylated tau was easier to be captured by AD P-tau and that aggregated tau was more difficult to be dephosphorylated than the non-aggregated tau by protein phosphatase 2A (PP2A). Based on these findings, we speculate that AD P-tau seeds hyperphosphorylated tau to form aggregates, which resist to the dephosphorylation by PP2A, resulting in hyperphosphorylation and pathology of tau.

11.
Mol Neurobiol ; 56(9): 6168-6183, 2019 Sep.
Article in English | MEDLINE | ID: mdl-30734228

ABSTRACT

Microtubule-associated protein tau in Alzheimer's disease (AD) brain is hyperphosphorylated, truncated, and aggregated into neurofibrillary tangles. Oligomeric and hyperphosphorylated tau (Oligo-tau) isolated from AD brain captures and templates normal tau into filaments both in vitro and in vivo; this prion-like activity is believed to be responsible for the progression of neurofibrillary pathology in AD. The 3xTg-AD mouse model develops both Aß and tau pathologies and thus gains popularity in preclinical studies of AD. Despite the histopathological similarity of the 3xTg-AD model to AD, biochemical authenticity of tau alterations in this model remains elusive. To investigate the biochemical basis of tau pathology in 3xTg-AD brain, we here compared pathological alterations of tau in the aged 3xTg-AD brain to those in AD brain. We found that in contrast to substantial high molecular weight smear tau (HMW-tau) lacking the N-terminal portion and hyperphosphorylated at multiple sites in AD brain, tau in 3xTg-AD mouse brain showed no detectable HMW-tau or truncation but slightly increased phosphorylation when normalized with total tau. In addition, AT8 immunostaining exhibited filamentous tau inclusions in AD brain, but predominantly truffle-like morphology in aged 3xTg-AD mouse brain. Further, Oligo-tau isolated from 3xTg-AD mice showed minimal potency in capturing tau in vitro and seeding tau aggregation in cultured cells when compared to AD Oligo-tau. These findings suggest that the alterations of tau in 3xTg-AD mouse brain differ from those in AD brain. In 3xTg-AD mice, the lack of N-terminal truncation, scarce SDS/reducing reagent-resistant HMW-tau, and minimal hyperphosphorylation may collectively result in low potency in prion-like activity of the Oligo-tau.


Subject(s)
Alzheimer Disease/pathology , Brain/pathology , tau Proteins/metabolism , Aged , Aged, 80 and over , Animals , Antibodies/pharmacology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microtubules/drug effects , Microtubules/metabolism , Molecular Weight , Phosphorylation/drug effects , Prions/metabolism , Protein Aggregates , Protein Multimerization , tau Proteins/chemistry
12.
J Alzheimers Dis ; 64(3): 709-722, 2018.
Article in English | MEDLINE | ID: mdl-29889065

ABSTRACT

Traumatic brain injury (TBI) is an established risk factor for the development of neurodegeneration and dementia late in life. Repetitive mild TBI (r-mTBI) is directly associated with chronic traumatic encephalopathy (CTE), a progressive neurodegenerative disorder characterized by focal perivascular to widespread Alzheimer-type neurofibrillary pathology of hyperphosphorylated tau. Studies in animal models have shown hyperphosphorylation of tau after TBI. However, the molecular mechanisms by which TBI leads to tau pathology are not understood. In this study, we employed western blots and immunohistochemistry to test, in triple-transgenic mouse model of Alzheimer's disease (3xTg-AD), the effect of r-mTBI on tau hyperphosphorylation and activation of asparaginyl endopeptidase (AEP), a cysteine proteinase which is known to be involved in tau hyperphosphorylation. We found that the level of active AEP was increased and correlated with the level of tau hyperphosphorylation following r-mTBI, and that fimbria showed increased immunoreactivity to phospho-tau. In addition, inhibitor 2 of protein phosphatase 2A (I2PP2A) was translocated from neuronal nucleus to the cytoplasm and colocalized with hyperphosphorylated tau. These data suggest the involvement of AEP-I2PP2A-PP2A-ptau pathway in tau pathology in TBI.


Subject(s)
Brain Injuries, Traumatic/pathology , Brain/enzymology , Cysteine Endopeptidases/metabolism , tau Proteins/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Brain/pathology , Brain Injuries, Traumatic/complications , Cytoplasm/metabolism , Disease Models, Animal , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , Neurons/metabolism , Neurons/pathology , Phosphorylation/physiology , Presenilin-1/genetics , Protein Transport/genetics , Synapsins/metabolism , tau Proteins/genetics
13.
J Alzheimers Dis ; 63(2): 821-833, 2018.
Article in English | MEDLINE | ID: mdl-29689717

ABSTRACT

Dementias including Alzheimer's disease (AD) are multifactorial disorders that involve several different etiopathogenic mechanisms. Cerebral ischemia has been suspected in the altered regulation of protein kinases and phosphatases that leads to hyperphosphorylation of tau and further neurofibrillary pathology, a key hallmark of AD and related neurodegenerative diseases. However, the deregulation of these enzymes and their relationship with ischemia and AD remain unclear. Previously, we reported a mechanism by which the lysosomal enzyme asparagine endopeptidase (AEP) is associated with brain acidosis and AD. In this study, we subjected mice to middle cerebral artery occlusion and found that compared with wild type mice, the ischemia-induced brain injury and motor deficit in AEP-knockout mice are reduced, probably because ischemia activates AEP. AEP cleaves inhibitor 2 of protein phosphatase 2A (I2PP2A), which translocates from the neuronal nucleus to the cytoplasm and produces hyperphosphorylation of tau through inhibition of PP2A. These findings suggest a possible mechanism of tau pathology associated with ischemia.


Subject(s)
Brain Ischemia/metabolism , Cysteine Endopeptidases/metabolism , Lysosomes/metabolism , tau Proteins/metabolism , Animals , Apoptosis/physiology , Brain/metabolism , Brain/pathology , Brain Ischemia/pathology , Cysteine Endopeptidases/genetics , DNA-Binding Proteins , Female , Histone Chaperones , Lysosomes/pathology , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/physiology , Neurons/metabolism , Neurons/pathology , Oncogene Proteins/metabolism , Phosphorylation/physiology
14.
Alzheimers Res Ther ; 10(1): 13, 2018 01 31.
Article in English | MEDLINE | ID: mdl-29386065

ABSTRACT

BACKGROUND: Accumulating evidence indicates that Tau pathology can spread from neuron to neuron by intake and coaggregation of the hyperphosphorylated Tau (p-Tau) seeds with the host neuron protein. Thus, clearance of Tau seeds by immunization with Tau antibodies could provide a potential therapeutic opportunity to block the spread of the pathology in Alzheimer's disease (AD) and other tauopathies. We report prevention of the seeding and spread of tau pathology with mouse monoclonal antibody 43D against the N-terminal projection domain of Tau (Tau 6-18) in triple-transgenic AD (3 × Tg-AD) mice. METHODS: Female 11- to 12-month-old 3 × Tg-AD mice were intravenously immunized weekly for 6 weeks with 15 µg/injection of mouse monoclonal antibody 43D or with mouse immunoglobulin G as a control. AD p-Tau isolated from a frozen autopsied AD brain was unilaterally injected into the right hippocampus on the day of the second dose of immunization. Tau pathology and its effect on Aß pathology were assessed by immunohistochemical staining. RESULTS: We found that the injection of AD p-Tau into the hippocampus of 11- to 12-month-old 3 × Tg-AD mice time-dependently induced Tau aggregation in the hippocampus and promoted the spread of Tau pathology to the contralateral hippocampus. Tau pathology was observed as early as 6 weeks after AD p-Tau injection. Tau pathology templated by AD p-Tau was thioflavin-S-positive and was about two-fold greater than that seen in naive 18-month-old 3 × Tg-AD mice; Tau pathology in the latter was thioflavin-S-negative. Immunization with Tau antibody 43D dramatically blocked AD p-Tau seeding in the ipsilateral hippocampus and inhibited its propagation to the contralateral side in 3 × Tg-AD mice. Furthermore, AD p-Tau injection enhanced the amyloid plaque load in the ipsilateral side, and immunization with 43D showed a tendency to attenuate it. CONCLUSIONS: These findings indicate that AD p-Tau-injected 3 × Tg-AD mice represent a practical model to study the seeding and spread of Tau pathology, their effect on Aß pathology, and the effect of Tau immunotherapy on both Tau and Aß pathologies. Immunization with Tau antibody 43D to Tau 6-18 can prevent the seeding and spread of Tau pathology, making it a potential therapeutic treatment for AD and related tauopathies.


Subject(s)
Alzheimer Disease/pathology , Alzheimer Disease/prevention & control , Antibodies, Monoclonal/administration & dosage , Brain/pathology , Immunization, Passive , tau Proteins/immunology , Acyltransferases/genetics , Acyltransferases/metabolism , Administration, Intravenous , Alzheimer Disease/immunology , Animals , Brain/immunology , Disease Models, Animal , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Female , Humans , Immunoglobulin G/administration & dosage , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Phosphorylation , Plaque, Amyloid/immunology , Plaque, Amyloid/pathology , Plaque, Amyloid/prevention & control , Presenilin-1/genetics , Presenilin-1/metabolism , Random Allocation , tau Proteins/administration & dosage , tau Proteins/genetics
15.
Alzheimers Res Ther ; 9(1): 1, 2017 Jan 10.
Article in English | MEDLINE | ID: mdl-28073379

ABSTRACT

BACKGROUND: Accumulation of hyperphosphorylated tau protein is a histopathological hallmark of Alzheimer's disease (AD) and related tauopathies. Currently, there is no effective treatment available for these progressive neurodegenerative diseases. In recent years, tau immunotherapy has shown great potential in animal models. We report the effect of immunization with tau antibodies 43D against tau 6-18 and 77E9 against tau 184-195 on tau and amyloid-ß (Aß) pathologies and cognition in triple-transgenic (3×Tg)-AD mice at mild to moderate stages of the disease. METHODS: We immunized 12-month-old female 3×Tg-AD mice with two to six or seven intravenous weekly doses of 15 µg of mouse monoclonal antibody 43D, 77E9, a combination of one-half dose each of 43D and 77E9, or as control of mouse immunoglobulin G (IgG). Age-matched wild-type mice treated with mouse IgG or a mixture of 43D and 77E9 were also used as controls. The effect of immunization with tau antibodies on tau and Aß pathologies was assessed by Western blot and immunofluorescence analysis, and the effect on cognition was analyzed by using Morris water maze, one-trial novel object recognition, and novel object location tasks. RESULTS: We found that two doses of 43D and 77E9 reduced total tau but had no significant impact on hyperphosphorylation of tau. However, six doses of 43D reduced levels of both total tau and tau hyperphosphorylated at Ser262/356 and Ser396/404 sites in the hippocampus. Importantly, both 43D and 77E9 antibodies rescued spatial memory and short-term memory impairments in 3×Tg-AD mice. The beneficial effect of 43D and 77E9 antibodies on cognitive performance was sustained up to 3 months after the last dose. Six doses of immunization with 43D also decreased amyloid precursor protein (APP) level in CA1 and amyloid plaques in subiculum, and showed a trend toward reducing Aß40 and Aß42 in the forebrain. Immunization with 43D increased levels of complement components C1 and C9 and resulted in activation of microglia, especially surrounding Aß plaques. CONCLUSIONS: These findings suggest the potential of passive immunization targeting proximal N-terminal domain tau 6-18 as a disease-modifying approach to AD and related tauopathies.


Subject(s)
Amyloid beta-Peptides/metabolism , Brain/pathology , Immunization, Passive , tau Proteins/immunology , tau Proteins/metabolism , Alzheimer Disease/immunology , Alzheimer Disease/pathology , Alzheimer Disease/psychology , Alzheimer Disease/therapy , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Brain/immunology , Cell Line , Disease Models, Animal , Female , Humans , Immunoglobulin G/immunology , Mice, 129 Strain , Mice, Inbred BALB C , Mice, Transgenic , Phosphorylation , Plaque, Amyloid/immunology , Plaque, Amyloid/pathology , Plaque, Amyloid/therapy , Random Allocation , Recombinant Proteins , Spleen/cytology , tau Proteins/genetics
16.
Alzheimers Dement ; 12(10): 1066-1077, 2016 10.
Article in English | MEDLINE | ID: mdl-27133892

ABSTRACT

INTRODUCTION: Neurofibrillary pathology of abnormally hyperphosphorylated tau (P-tau) is a hallmark of Alzheimer's disease (AD) and other tauopathies. Tau pathology can be experimentally induced and propagated. However, what induces the prion-like transmission character to tau and produces morphologically distinct tau lesions remains elusive. METHODS: We investigated the role of hyperphosphorylation in the spread of tau pathology in hTau transgenic mice. RESULTS: We found that intrahippocampal injection with AD P-tau, but not nonphosphorylated tau, produced numerous P-tau tangles and neuropil threads locally and in neocortex lateral to injection and upstream to the hippocampus. Dephosphorylation of AD P-tau with protein phosphatase-2A dramatically reduced and switched tau pathology from neurofibrillary tangles to argyrophilic grain-like morphology. CONCLUSIONS: Our findings show that abnormal hyperphosphorylation of tau determines the spread and morphology of tau lesions and that the propagation of tau pathology takes place both locally and in axonally connected areas and highlight tau hyperphosphorylation as a potential drug target.


Subject(s)
Neocortex/pathology , Neurofibrillary Tangles/pathology , Tauopathies , Alzheimer Disease/pathology , Animals , Hippocampus/pathology , Humans , Immunohistochemistry , Mice , Mice, Transgenic , Phosphorylation , Protein Phosphatase 2 , tau Proteins/metabolism
17.
J Alzheimers Dis ; 45(2): 423-35, 2015.
Article in English | MEDLINE | ID: mdl-25589718

ABSTRACT

Protein phosphatase-2A (PP2A) deficiency is a cause of the abnormal hyperphosphorylation of tau, which composes neurofibrillary tangles (NFTs) in Alzheimer's disease (AD) brain. We previously reported that both mRNA and protein expression of inhibitor I of PP2A (I(1)(PP2A)) are elevated in AD brain and that this inhibitor induces a dose-dependent inhibition of PP2A activity and tau hyperphosphorylation in NIH3T3 cells. However, whether I(1)(PP2A) can induce AD neurofibrillary degeneration and cognitive impairment was not known. In the present study, we infected the brains of rat pups within 24 hours of birth with adeno-associated virus serotype 1 (AAV1) carrying I(1)(PP2A). In the adult AAV1-I(1)(PP2A) rats, we found a decrease in PP2A activity and abnormal hyperphosphorylation of tau in the brain. Immunohistochemistry showed a significant reduction of MAP2 and synapsin 1 in AAV1- I(1)(PP2A) animals, suggesting that I(1)(PP2A) can induce a loss of dendritic and synaptic plasticity markers. Behavioral tests revealed that infection with AAV1- I(1)(PP2A) induced deficits in exploratory activity, spatial reference memory, and memory consolidation in adult rats. These studies suggest that I(1)(PP2A) can inhibit PP2A activity, and in turn induce AD neurofibrillary degeneration and cognitive deficits in rats.


Subject(s)
Cognition Disorders/etiology , Intracellular Signaling Peptides and Proteins/metabolism , Neurodegenerative Diseases/etiology , Protein Phosphatase 2/metabolism , tau Proteins/metabolism , Animals , Animals, Newborn , Brain/metabolism , Brain/pathology , Cognition Disorders/genetics , Cognition Disorders/metabolism , Dependovirus/genetics , Disease Models, Animal , Exploratory Behavior/physiology , Intracellular Signaling Peptides and Proteins/genetics , Maze Learning/physiology , Mice , NIH 3T3 Cells , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurofibrillary Tangles , Neurologic Examination , Nuclear Proteins , Phosphorylation/genetics , Protein Phosphatase 2/genetics , RNA-Binding Proteins , Rats , Rats, Wistar , Synapsins/metabolism
18.
Neurobiol Aging ; 35(12): 2701-2712, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25034344

ABSTRACT

A minor component of coffee unrelated to caffeine, eicosanoyl-5-hydroxytryptamide (EHT), provides protection in a rat model for Alzheimer's disease (AD). In this model, viral expression of the phosphoprotein phosphatase 2A (PP2A) endogenous inhibitor, the I2(PP2A), or SET protein in the brains of rats leads to several characteristic features of AD including cognitive impairment, tau hyperphosphorylation, and elevated levels of cytoplasmic amyloid-ß protein. Dietary supplementation with EHT for 6-12 months resulted in substantial amelioration of all these defects. The beneficial effects of EHT could be associated with its ability to increase PP2A activity by inhibiting the demethylation of its catalytic subunit PP2Ac. These findings raise the possibility that EHT may make a substantial contribution to the apparent neuroprotective benefits associated with coffee consumption as evidenced by numerous epidemiologic studies indicating that coffee drinkers have substantially lowered risk of developing AD.


Subject(s)
Alzheimer Disease/prevention & control , Coffee/chemistry , Disease Models, Animal , Neuroprotective Agents , Serotonin/analogs & derivatives , Animals , Female , Methylation/drug effects , Protein Phosphatase 2/metabolism , Rats , Rats, Transgenic , Serotonin/pharmacology , Serotonin/therapeutic use
19.
Neurobiol Dis ; 71: 110-30, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25046994

ABSTRACT

Besides the presence of amyloid beta (Aß) plaques and neurofibrillary tangles, neurogenesis and synaptic plasticity are markedly impaired in Alzheimer's disease (AD) possibly contributing to cognitive impairment. In this context, neurotrophic factors serve as a promising therapeutic approach via utilization of regenerative capacity of brain to shift the balance from neurodegeneration to neural regeneration. However, besides more conventional "bystander" effect, to what extent can neurotrophic compounds affect underlying AD pathology remains questionable. Here we investigated the effect of chronic oral treatment with a ciliary neurotrophic factor (CNTF) derived peptidergic compound, P021 (Ac-DGGL(A)G-NH2), on disease pathology both at moderate and severe stages in a transgenic mouse model of AD. 3xTg-AD and wild type female mice were treated for 12months with P021 or vehicle diet starting at 9-10months of age. A significant reduction in abnormal hyperphosphorylation and accumulation of tau at known major AD neurofibrillary pathology associated sites was observed. The effect of P021 on Aß pathology was limited to a significant decrease in soluble Aß levels and a trend towards reduction in Aß plaque load in CA1 region of hippocampus, consistent with reduction in Aß generation and not clearance. This disease modifying effect was probably via increased brain derived neurotrophic factor (BDNF) expression mediated decrease in glycogen synthase kinase-3-ß (GSK3ß) activity we found in P021 treated 3xTg-AD mice. P021 treatment also rescued deficits in cognition, neurogenesis, and synaptic plasticity in 3xTg-AD mice. These findings demonstrate the potential of the neurotrophic peptide mimetic as a disease modifying therapy for AD.


Subject(s)
Alzheimer Disease/drug therapy , Antipsychotic Agents/administration & dosage , Brain/drug effects , Ciliary Neurotrophic Factor/administration & dosage , Administration, Oral , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Animals , Antipsychotic Agents/blood , Antipsychotic Agents/chemistry , Blood-Testis Barrier/drug effects , Blood-Testis Barrier/physiology , Cells, Cultured , Ciliary Neurotrophic Factor/blood , Ciliary Neurotrophic Factor/chemistry , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/drug effects , Presenilin-1/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Time Factors , tau Proteins/genetics
20.
J Alzheimers Dis ; 37(3): 469-74, 2013.
Article in English | MEDLINE | ID: mdl-23948903

ABSTRACT

Alzheimer's disease is multifactorial and involves several different mechanisms. The sporadic form of the disease accounts for over 99% of the cases. As of yet, there is no practical and widely available animal model of the sporadic form of the disease. In the Alzheimer's disease brain, the lysosomal enzyme asparaginyl endopeptidase is activated and translocated from the neuronal lysosomes to the cytoplasm, probably due to brain acidosis caused by ischemic changes associated with age-associated microinfarcts. The activated asparaginyl endopeptidase cleaves inhibitor-2 of protein phosphatase-2A, I2(PP2A), into I(2NTF) and I(2CTF) which translocate to the neuronal cytoplasm and inhibit the protein phosphatase activity and consequently the abnormal hyperphosphorylation of tau. Employing adeno-associated virus serotype 1 (AAV1) vector containing I(2NTF-CTF) and transduction of the brains of newborn rat pups with this virus, an animal model has been generated. The AAV1-I(2NTF-CTF) rats show neurodegeneration and cognitive impairment at 4 months and abnormal hyperphosphorylation and aggregation of tau and intraneuronal accumulation of amyloid-ß at 13 months. The AAV1-I(2NTF-CTF) rats not only offer a disease-relevant model of the sporadic form of Alzheimer's disease but also represent a practical and widely available animal model. This short perspective on the need to focus on and develop the disease-relevant models of the sporadic form of Alzheimer's disease very much reflects the thinking of Inge Grundke-Iqbal who passed away on September 22, 2012 and to whom this article is dedicated.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Brain/metabolism , Brain/pathology , Disease Models, Animal , Alzheimer Disease/genetics , Animals , Humans , Protein Phosphatase 2/metabolism , Rats , tau Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...