Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Exp Mol Med ; 54(9): 1461-1471, 2022 09.
Article in English | MEDLINE | ID: mdl-36056186

ABSTRACT

Mitochondria in neural progenitors play a crucial role in adult hippocampal neurogenesis by being involved in fate decisions for differentiation. However, the molecular mechanisms by which mitochondria are related to the genetic regulation of neuronal differentiation in neural progenitors are poorly understood. Here, we show that mitochondrial dysfunction induced by amyloid-beta (Aß) in neural progenitors inhibits neuronal differentiation but has no effect on the neural progenitor stage. In line with the phenotypes shown in Alzheimer's disease (AD) model mice, Aß-induced mitochondrial damage in neural progenitors results in deficits in adult hippocampal neurogenesis and cognitive function. Based on hippocampal proteome changes after mitochondrial damage in neural progenitors identified through proteomic analysis, we found that lysine demethylase 5A (KDM5A) in neural progenitors epigenetically suppresses differentiation in response to mitochondrial damage. Mitochondrial damage characteristically causes KDM5A degradation in neural progenitors. Since KDM5A also binds to and activates neuronal genes involved in the early stage of differentiation, functional inhibition of KDM5A consequently inhibits adult hippocampal neurogenesis. We suggest that mitochondria in neural progenitors serve as the checkpoint for neuronal differentiation via KDM5A. Our findings not only reveal a cell-type-specific role of mitochondria but also suggest a new role of KDM5A in neural progenitors as a mediator of retrograde signaling from mitochondria to the nucleus, reflecting the mitochondrial status.


Subject(s)
Alzheimer Disease , Neurons , Proteome , Retinoblastoma-Binding Protein 2/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Cell Differentiation , Lysine/metabolism , Mice , Mitochondria/metabolism , Neurons/cytology , Neurons/metabolism , Proteome/metabolism , Proteomics
2.
Exp Mol Med ; 51(11): 1-17, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31727875

ABSTRACT

Alzheimer's disease (AD), which is the most common type of dementia, is characterized by the deposition of extracellular amyloid plaques. To understand the pathophysiology of the AD brain, the assessment of global proteomic dynamics is required. Since the hippocampus is a major region affected in the AD brain, we performed hippocampal analysis and identified proteins that are differentially expressed between wild-type and 5XFAD model mice via LC-MS methods. To reveal the relationship between proteomic changes and the progression of amyloid plaque deposition in the hippocampus, we analyzed the hippocampal proteome at two ages (5 and 10 months). We identified 9,313 total proteins and 1411 differentially expressed proteins (DEPs) in 5- and 10-month-old wild-type and 5XFAD mice. We designated a group of proteins showing the same pattern of changes as amyloid beta (Aß) as the Aß-responsive proteome. In addition, we examined potential biomarkers by investigating secretory proteins from the Aß-responsive proteome. Consequently, we identified vitamin K-dependent protein S (PROS1) as a novel microglia-derived biomarker candidate in the hippocampus of 5XFAD mice. Moreover, we confirmed that the PROS1 level in the serum of 5XFAD mice increases as the disease progresses. An increase in PROS1 is also observed in the sera of AD patients and shows a close correlation with AD neuroimaging markers in humans. Therefore, our quantitative proteome data obtained from 5XFAD model mice successfully predicted AD-related biological alterations and suggested a novel protein biomarker for AD.


Subject(s)
Alzheimer Disease/metabolism , Hippocampus/metabolism , Proteome/analysis , Alzheimer Disease/blood , Alzheimer Disease/pathology , Animals , Biomarkers/blood , Biomarkers/metabolism , Calcium-Binding Proteins/blood , Calcium-Binding Proteins/metabolism , Cells, Cultured , Chromatography, Liquid , Female , Humans , Mass Spectrometry , Mice , Mice, Transgenic , Microglia/metabolism , Proteomics
3.
Nanomedicine ; 17: 297-307, 2019 04.
Article in English | MEDLINE | ID: mdl-30794963

ABSTRACT

The aggregation and accumulation of amyloid beta (Aß) peptide is believed to be the primary cause of Alzheimer's disease (AD) pathogenesis. Vitamin D-binding protein (DBP) can attenuate Aß aggregation and accumulation. A biocompatible polymer poly (D,L-lactic acid-co-glycolic acid) (PLGA) can be loaded with therapeutic agents and control the rate of their release. In the present study, a PLGA-based drug delivery system was used to examine the therapeutic effects of DBP-PLGA nanoparticles in Aß-overexpressing (5XFAD) mice. DBP was loaded into PLGA nanoparticles and the characteristics of the DBP-PLGA nanoparticles were analyzed. Using a thioflavin-T assay, we observed that DBP-PLGA nanoparticles significantly inhibited Aß aggregation in vitro. In addition, we found that intravenous injection of DBP-PLGA nanoparticles significantly attenuated the Aß accumulation, neuroinflammation, neuronal loss and cognitive dysfunction in the 5XFAD mice. Collectively, our results suggest that DBP-PLGA nanoparticles could be a promising therapeutic candidate for the treatment of AD.


Subject(s)
Alzheimer Disease/drug therapy , Drug Carriers/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Vitamin D-Binding Protein/administration & dosage , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Male , Mice , Mice, Transgenic , Nanoparticles/chemistry , Protein Aggregation, Pathological/drug therapy , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/pathology , Vitamin D-Binding Protein/therapeutic use
4.
Aging Cell ; 18(1): e12866, 2019 02.
Article in English | MEDLINE | ID: mdl-30515963

ABSTRACT

The orphan nuclear receptor Nurr1 (also known as NR4A2) is critical for the development and maintenance of midbrain dopaminergic neurons, and is associated with Parkinson's disease. However, an association between Nurr1 and Alzheimer's disease (AD)-related pathology has not previously been reported. Here, we provide evidence that Nurr1 is expressed in a neuron-specific manner in AD-related brain regions; specifically, it is selectively expressed in glutamatergic neurons in the subiculum and the cortex of both normal and AD brains. Based on Nurr1's expression patterns, we investigated potential functional roles of Nurr1 in AD pathology. Nurr1 expression was examined in the hippocampus and cortex of AD mouse model and postmortem human AD subjects. In addition, we performed both gain-of-function and loss-of-function studies of Nurr1 and its pharmacological activation in 5XFAD mice. We found that knockdown of Nurr1 significantly aggravated AD pathology while its overexpression alleviated it, including effects on Aß accumulation, neuroinflammation, and neurodegeneration. Importantly, 5XFAD mice treated with amodiaquine, a highly selective synthetic Nurr1 agonist, showed robust reduction in typical AD features including deposition of Aß plaques, neuronal loss, microgliosis, and impairment of adult hippocampal neurogenesis, leading to significant improvement of cognitive impairment. These in vivo and in vitro findings suggest that Nurr1 critically regulates AD-related pathophysiology and identify Nurr1 as a novel AD therapeutic target.


Subject(s)
Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Cognition , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Aging/pathology , Alzheimer Disease/complications , Alzheimer Disease/metabolism , Amodiaquine/pharmacology , Animals , Brain/metabolism , Brain/pathology , Cognition/drug effects , Disease Models, Animal , Glutamic Acid/metabolism , Humans , Mice, Inbred C57BL , Mice, Transgenic , Neuroglia/drug effects , Neuroglia/metabolism , Neurons/drug effects , Neurons/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 2/antagonists & inhibitors , Postmortem Changes
5.
Aging Cell ; 18(1): e12872, 2019 02.
Article in English | MEDLINE | ID: mdl-30515991

ABSTRACT

Alzheimer's disease (AD) is an age-related neurodegenerative disease characterized by progressive memory loss resulting from cumulative neuronal cell death. O-linked ß-N-acetyl glucosamine (O-GlcNAc) modification of the proteins reflecting glucose metabolism is altered in the brains of patients with AD. However, the link between altered O-GlcNAc modification and neuronal cell death in AD is poorly understood. Here, we examined the regulation of O-GlcNAcylation of c-Fos and the effects of O-GlcNAcylated c-Fos on neuronal cell death during AD pathogenesis. We found that amyloid beta (Aß)-induced O-GlcNAcylation on serine-56 and 57 of c-Fos was resulted from decreased interaction between c-Fos and O-GlcNAcase and promoted neuronal cell death. O-GlcNAcylated c-Fos increased its stability and potentiated the transcriptional activity through higher interaction with c-Jun, resulting in induction of Bim expression leading to neuronal cell death. Taken together, Aß-induced O-GlcNAcylation of c-Fos plays an important role in neuronal cell death during the pathogenesis of AD.


Subject(s)
Amyloid beta-Peptides/metabolism , Neurons/pathology , Proto-Oncogene Proteins c-fos/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/toxicity , Animals , Bcl-2-Like Protein 11/genetics , Bcl-2-Like Protein 11/metabolism , Cell Death/drug effects , Cell Line , Gene Expression Regulation/drug effects , Glycosylation/drug effects , Humans , Mice, Transgenic , Neurons/drug effects , Protein Stability/drug effects , Proto-Oncogene Proteins c-fos/genetics , Rats, Sprague-Dawley , Transcription, Genetic/drug effects , beta-N-Acetylhexosaminidases/metabolism
6.
PLoS Curr ; 102018 Jul 26.
Article in English | MEDLINE | ID: mdl-30279997

ABSTRACT

OBJECTIVE: Excessive mitochondrial fission has been associated with several neurodegenerative diseases, including Huntington's disease (HD). Consequently, mitochondrial dynamics has been suggested to be a promising therapeutic target for Huntington's disease. Mitochondrial fission depends on recruitment of Drp1 to mitochondria, and Mff (mitochondrial fission factor) is one of the key adaptor proteins for this process. Removal of Mff therefore greatly reduces mitochondrial fission. Here we investigate whether removal of Mff can mitigate HD-associated pathologies in HD transgenic mice (R6/2) expressing mutant Htt. METHOD: We compared the phenotype of HD mice with and without Mff. The mice were monitored for lifespan, neurological phenotypes, Htt aggregate formation, and brain histology. RESULTS: We found that HD mice lacking Mff display more severe neurological phenotypes and have shortened lifespans. Loss of Mff does not affect mutant Htt aggregation, but it accelerates HD pathology, including neuronal loss and neuroinflammation. CONCLUSIONS: Our data indicate a protective role for mitochondrial fission in HD and suggest that more studies are needed before manipulation of mitochondrial dynamics can be applied to HD therapy.

8.
Stem Cells Transl Med ; 6(1): 293-305, 2017 01.
Article in English | MEDLINE | ID: mdl-28170178

ABSTRACT

Transplantation of stem cells into the brain attenuates functional deficits in the central nervous system via cell replacement, the release of specific neurotransmitters, and the production of neurotrophic factors. To identify patient-specific and safe stem cells for treating Alzheimer's disease (AD), we generated induced pluripotent stem cells (iPSCs) derived from mouse skin fibroblasts by treating protein extracts of embryonic stem cells. These reprogrammed cells were pluripotent but nontumorigenic. Here, we report that protein-iPSCs differentiated into glial cells and decreased plaque depositions in the 5XFAD transgenic AD mouse model. We also found that transplanted protein-iPSCs mitigated the cognitive dysfunction observed in these mice. Proteomic analysis revealed that oligodendrocyte-related genes were upregulated in brains injected with protein-iPSCs, providing new insights into the potential function of protein-iPSCs. Taken together, our data indicate that protein-iPSCs might be a promising therapeutic approach for AD. Stem Cells Translational Medicine 2017;6:293-305.


Subject(s)
Alzheimer Disease/physiopathology , Alzheimer Disease/therapy , Amyloid beta-Peptides/metabolism , Cognitive Dysfunction/therapy , Induced Pluripotent Stem Cells/transplantation , Animals , Behavior, Animal , Brain/pathology , Cell Differentiation , Cognitive Dysfunction/complications , Disease Models, Animal , Mice, Inbred C57BL , Mice, Transgenic , Neurons/pathology , Oligodendroglia/metabolism , Plaque, Amyloid/pathology , Proteome/metabolism , Stem Cell Transplantation , Transferrin/metabolism , Up-Regulation/genetics
9.
Elife ; 52016 11 17.
Article in English | MEDLINE | ID: mdl-27852436

ABSTRACT

A defining feature of mitochondria is their maternal mode of inheritance. However, little is understood about the cellular mechanism through which paternal mitochondria, delivered from sperm, are eliminated from early mammalian embryos. Autophagy has been implicated in nematodes, but whether this mechanism is conserved in mammals has been disputed. Here, we show that cultured mouse fibroblasts and pre-implantation embryos use a common pathway for elimination of mitochondria. Both situations utilize mitophagy, in which mitochondria are sequestered by autophagosomes and delivered to lysosomes for degradation. The E3 ubiquitin ligases PARKIN and MUL1 play redundant roles in elimination of paternal mitochondria. The process is associated with depolarization of paternal mitochondria and additionally requires the mitochondrial outer membrane protein FIS1, the autophagy adaptor P62, and PINK1 kinase. Our results indicate that strict maternal transmission of mitochondria relies on mitophagy and uncover a collaboration between MUL1 and PARKIN in this process.


Subject(s)
Mitochondria/genetics , Mitochondrial Proteins/genetics , Mitophagy/genetics , Ubiquitin-Protein Ligases/genetics , Animals , Embryo, Mammalian , Humans , Mice , Paternal Inheritance/genetics , Protein Kinases/genetics , Transcription Factor TFIIH , Transcription Factors/genetics
10.
Autophagy ; 12(5): 784-800, 2016 05 03.
Article in English | MEDLINE | ID: mdl-26963025

ABSTRACT

The secretion of proteins that lack a signal sequence to the extracellular milieu is regulated by their transition through the unconventional secretory pathway. IDE (insulin-degrading enzyme) is one of the major proteases of amyloid beta peptide (Aß), a presumed causative molecule in Alzheimer disease (AD) pathogenesis. IDE acts in the extracellular space despite having no signal sequence, but the underlying mechanism of IDE secretion extracellularly is still unknown. In this study, we found that IDE levels were reduced in the cerebrospinal fluid (CSF) of patients with AD and in pathology-bearing AD-model mice. Since astrocytes are the main cell types for IDE secretion, astrocytes were treated with Aß. Aß increased the IDE levels in a time- and concentration-dependent manner. Moreover, IDE secretion was associated with an autophagy-based unconventional secretory pathway, and depended on the activity of RAB8A and GORASP (Golgi reassembly stacking protein). Finally, mice with global haploinsufficiency of an essential autophagy gene, showed decreased IDE levels in the CSF in response to an intracerebroventricular (i.c.v.) injection of Aß. These results indicate that IDE is secreted from astrocytes through an autophagy-based unconventional secretory pathway in AD conditions, and that the regulation of autophagy is a potential therapeutic target in addressing Aß pathology.


Subject(s)
Alzheimer Disease/metabolism , Astrocytes/metabolism , Autophagy/physiology , Insulin/metabolism , Secretory Pathway/physiology , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Brain/metabolism , Disease Models, Animal , Extracellular Space/metabolism , Humans , Mice
11.
ACS Nano ; 10(2): 2860-70, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26844592

ABSTRACT

Mitochondrial oxidative stress is a key pathologic factor in neurodegenerative diseases, including Alzheimer's disease. Abnormal generation of reactive oxygen species (ROS), resulting from mitochondrial dysfunction, can lead to neuronal cell death. Ceria (CeO2) nanoparticles are known to function as strong and recyclable ROS scavengers by shuttling between Ce(3+) and Ce(4+) oxidation states. Consequently, targeting ceria nanoparticles selectively to mitochondria might be a promising therapeutic approach for neurodegenerative diseases. Here, we report the design and synthesis of triphenylphosphonium-conjugated ceria nanoparticles that localize to mitochondria and suppress neuronal death in a 5XFAD transgenic Alzheimer's disease mouse model. The triphenylphosphonium-conjugated ceria nanoparticles mitigate reactive gliosis and morphological mitochondria damage observed in these mice. Altogether, our data indicate that the triphenylphosphonium-conjugated ceria nanoparticles are a potential therapeutic candidate for mitochondrial oxidative stress in Alzheimer's disease.


Subject(s)
Alzheimer Disease/drug therapy , Antioxidants/administration & dosage , Cerium/administration & dosage , Metal Nanoparticles/chemistry , Mitochondria/drug effects , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Cerium/pharmacology , Cerium/therapeutic use , HeLa Cells , Humans , Mice , Mitochondria/metabolism , Organophosphorus Compounds/chemistry , Oxidative Stress
12.
ACS Cent Sci ; 2(12): 967-975, 2016 Dec 28.
Article in English | MEDLINE | ID: mdl-28058286

ABSTRACT

Monoamine oxidases (MAOs) play an important role in Alzheimer's disease (AD) pathology. We report in vivo comonitoring of MAO activity and amyloid-ß (Aß) plaques dependent on the aging of live mice with AD, using a two-photon fluorescence probe. The probe under the catalytic action of MAO produces a dipolar fluorophore that senses Aß plaques, a general AD biomarker, enabling us to comonitor the enzyme activity and the progress of AD indicated by Aß plaques. The results show that the progress of AD has a close correlation with MAO activity, which can be categorized into three stages: slow initiation stage up to three months, an aggressive stage, and a saturation stage from nine months. Histological analysis also reveals elevation of MAO activity around Aß plaques in aged mice. The close correlation between the MAO activity and AD progress observed by in vivo monitoring for the first time prompts us to investigate the enzyme as a potential biomarker of AD.

13.
Neurobiol Aging ; 36(12): 3214-3227, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26452999

ABSTRACT

Alzheimer's disease (AD) is characterized by impaired cognitive function and memory loss, which are often the result of synaptic pathology. Thrombospondin (TSP) is an astrocyte-secreted protein, well known for its function as a modulator of synaptogenesis and neurogenesis. Here, we investigated the effects of TSP-1 on AD pathogenesis. We found that the level of TSP-1 expression was decreased in AD brains. When we treated astrocytes with amyloid beta (Aß), secreted TSP-1 was decreased in autophagy-dependent manner. In addition, treatment with Aß induced synaptic pathology, such as decreased dendritic spine density and reduced synaptic activity. These effects were prevented by coincubation of TSP-1 with Aß, which acts through the TSP-1 receptor alpha-2-delta-1 in neurons. Finally, intrasubicular injection with TSP-1 into AD model mouse brains mitigated the Aß-mediated reduction of synaptic proteins and related signaling pathways. These results indicate that TSP-1 is a potential therapeutic target in AD pathogenesis.


Subject(s)
Alzheimer Disease/pathology , Alzheimer Disease/prevention & control , Amyloid beta-Peptides/adverse effects , Synapses/pathology , Thrombospondin 1/physiology , Thrombospondin 1/therapeutic use , Alzheimer Disease/genetics , Animals , Astrocytes/metabolism , Autophagy , Brain/metabolism , Disease Models, Animal , Mice, Transgenic , Molecular Targeted Therapy , Neurons/metabolism , Signal Transduction , Synapses/metabolism , Synapses/physiology , Thrombospondin 1/metabolism
14.
Hum Mol Genet ; 24(22): 6492-504, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26358770

ABSTRACT

Glycosylation with O-linked ß-N-acetylglucosamine (O-GlcNAc) is one of the protein glycosylations affecting various intracellular events. However, the role of O-GlcNAcylation in neurodegenerative diseases such as Alzheimer's disease (AD) is poorly understood. Mitochondrial adenosine 5'-triphosphate (ATP) synthase is a multiprotein complex that synthesizes ATP from ADP and Pi. Here, we found that ATP synthase subunit α (ATP5A) was O-GlcNAcylated at Thr432 and ATP5A O-GlcNAcylation was decreased in the brains of AD patients and transgenic mouse model, as well as Aß-treated cells. Indeed, Aß bound to ATP synthase directly and reduced the O-GlcNAcylation of ATP5A by inhibition of direct interaction between ATP5A and mitochondrial O-GlcNAc transferase, resulting in decreased ATP production and ATPase activity. Furthermore, treatment of O-GlcNAcase inhibitor rescued the Aß-induced impairment in ATP production and ATPase activity. These results indicate that Aß-mediated reduction of ATP synthase activity in AD pathology results from direct binding between Aß and ATP synthase and inhibition of O-GlcNAcylation of Thr432 residue on ATP5A.


Subject(s)
Alzheimer Disease/metabolism , Mitochondrial Proton-Translocating ATPases/metabolism , N-Acetylglucosaminyltransferases/metabolism , Oxidative Phosphorylation Coupling Factors/metabolism , Acetylglucosamine/metabolism , Adenosine Triphosphate/metabolism , Alzheimer Disease/enzymology , Alzheimer Disease/genetics , Animals , CHO Cells , Cricetulus , Disease Models, Animal , Glycosylation , HeLa Cells , Humans , Mice , Mice, Transgenic , Mitochondria/enzymology , Mitochondria/metabolism , Mitochondrial Proton-Translocating ATPases/genetics , Oxidative Phosphorylation Coupling Factors/genetics , Protein Processing, Post-Translational , beta-N-Acetylhexosaminidases/metabolism
15.
Exp Mol Med ; 47: e150, 2015 Mar 13.
Article in English | MEDLINE | ID: mdl-25766619

ABSTRACT

Many researchers have reported that oxidative damage to mitochondrial DNA (mtDNA) is increased in several age-related disorders. Damage to mitochondrial constituents and mtDNA can generate additional mitochondrial dysfunction that may result in greater reactive oxygen species production, triggering a circular chain of events. However, the mechanisms underlying this vicious cycle have yet to be fully investigated. In this review, we summarize the relationship of oxidative stress-induced mitochondrial dysfunction with mtDNA mutation in neurodegenerative disorders.


Subject(s)
DNA, Mitochondrial/genetics , Mutation , Neurodegenerative Diseases/genetics , Animals , Humans , Mitochondria/drug effects , Mitochondria/genetics , Mitochondria/metabolism , Molecular Targeted Therapy , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Reactive Oxygen Species/metabolism
16.
Biochemistry ; 54(9): 1831-40, 2015 Mar 10.
Article in English | MEDLINE | ID: mdl-25714795

ABSTRACT

Alzheimer's disease (AD) and Parkinson's disease (PD) are caused by ß-amyloid (Aß) and α-synuclein (αS), respectively. Ample evidence suggests that these two pathogenic proteins are closely linked and have a synergistic effect on eliciting neurodegenerative disorders. However, the pathophysiological consequences of Aß and αS coexistence are still elusive. Here, we show that large-sized αS oligomers, which are normally difficult to form, are readily generated by Aß42-seeding and that these oligomers efficiently hamper neuronal SNARE-mediated vesicle fusion. The direct binding of the Aß-seeded αS oligomers to the N-terminal domain of synaptobrevin-2, a vesicular SNARE protein, is responsible for the inhibition of fusion. In contrast, large-sized Aß42 oligomers (or aggregates) or the products of αS incubated without Aß42 have no effect on vesicle fusion. These results are confirmed by examining PC12 cell exocytosis. Our results suggest that Aß and αS cooperate to escalate the production of toxic oligomers, whose main toxicity is the inhibition of vesicle fusion and consequently prompts synaptic dysfunction.


Subject(s)
Amyloid beta-Peptides/physiology , Cytoplasmic Vesicles/physiology , Membrane Fusion , SNARE Proteins/antagonists & inhibitors , alpha-Synuclein/physiology , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Animals , Exocytosis/genetics , Humans , Membrane Fusion/genetics , PC12 Cells , Protein Binding/genetics , Protein Multimerization/physiology , Rats , SNARE Proteins/metabolism , Synapses/genetics , Synapses/metabolism , Transfection , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
17.
J Alzheimers Dis ; 41(1): 233-41, 2014.
Article in English | MEDLINE | ID: mdl-24583405

ABSTRACT

Alzheimer's disease (AD) is an age-related neurological disorder characterized by the deposition of amyloid-ß (Aß), cognitive deficits, and neuronal loss. The decline in neurogenic capacity could participate in neuronal vulnerability and contribute to memory impairment in AD. In our longitudinal study with AD model mice (5XFAD mice), we found that the number of doublecortin (neurogenesis marker)-positive cells in 5XFAD mice was significantly decreased compared to wild-type littermate mice. Using Aß immunostaining with 4G8 antibody, we observed that impairment in neurogenesis might be associated with the deposits of amyloid plaques. To investigate the effect of the neurogenic hormone ghrelin on defective neurogenesis in the AD brain, 5XFAD mice were administered peripherally with ghrelin. We found that treatment with ghrelin increased the number of doublecortin, HH3, and calretinin-stained cells in the hippocampus of 5XFAD mice. In 5XFAD mice treated with ghrelin, the 4G8-positive area was not significantly different from the saline-treated 5XFAD mice. Together, these findings suggest that hippocampal neurogenesis is impaired in 5XFAD mice and that treatment with ghrelin successfully rescued the abnormality of neurogenesis in 5XFAD mice without affecting Aß pathology.


Subject(s)
Alzheimer Disease/drug therapy , Ghrelin/pharmacology , Hippocampus/drug effects , Neurogenesis/drug effects , Neuroprotective Agents/pharmacology , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Dentate Gyrus/drug effects , Dentate Gyrus/pathology , Dentate Gyrus/physiopathology , Disease Models, Animal , Gliosis/drug therapy , Gliosis/pathology , Gliosis/physiopathology , Hippocampus/pathology , Hippocampus/physiopathology , Humans , Longitudinal Studies , Male , Mice, Transgenic , Microglia/drug effects , Microglia/physiology , Neural Stem Cells/drug effects , Neural Stem Cells/physiology , Neurogenesis/physiology , Plaque, Amyloid/drug therapy , Plaque, Amyloid/pathology , Plaque, Amyloid/physiopathology
18.
Neurobiol Aging ; 35(6): 1286-92, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24485508

ABSTRACT

Immune responses in the brain are thought to play a role in disorders of the central nervous system, but an understanding of the process underlying how immune cells get into the brain and their fate there remains unclear. In this study, we used a 2-photon microscopy to reveal that neutrophils infiltrate brain and migrate toward amyloid plaques in a mouse model of Alzheimer's disease. These findings suggest a new molecular process underlying the pathophysiology of Alzheimer's disease.


Subject(s)
Alzheimer Disease/immunology , Brain/immunology , Cell Movement/immunology , Neutrophils/immunology , Plaque, Amyloid/immunology , Animals , Cells, Cultured , Disease Models, Animal , Humans , Mice , Mice, Transgenic , Microscopy/methods , Molecular Imaging/methods
19.
PLoS One ; 7(4): e34929, 2012.
Article in English | MEDLINE | ID: mdl-22514691

ABSTRACT

Mitochondria are best known as the essential intracellular organelles that host the homeostasis required for cellular survival, but they also have relevance in diverse disease-related conditions, including Alzheimer's disease (AD). Amyloid ß (Aß) peptide is the key molecule in AD pathogenesis, and has been highlighted in the implication of mitochondrial abnormality during the disease progress. Neuronal exposure to Aß impairs mitochondrial dynamics and function. Furthermore, mitochondrial Aß accumulation has been detected in the AD brain. However, the underlying mechanism of how Aß affects mitochondrial function remains uncertain, and it is questionable whether mitochondrial Aß accumulation followed by mitochondrial dysfunction leads directly to neuronal toxicity. This study demonstrated that an exogenous Aß(1-42) treatment, when applied to the hippocampal cell line of mice (specifically HT22 cells), caused a deleterious alteration in mitochondria in both morphology and function. A clathrin-mediated endocytosis blocker rescued the exogenous Aß(1-42)-mediated mitochondrial dysfunction. Furthermore, the mitochondria-targeted accumulation of Aß(1-42) in HT22 cells using Aß(1-42) with a mitochondria-targeting sequence induced the identical morphological alteration of mitochondria as that observed in the APP/PS AD mouse model and exogenous Aß(1-42)-treated HT22 cells. In addition, subsequent mitochondrial dysfunctions were demonstrated in the mitochondria-specific Aß(1-42) accumulation model, which proved indistinguishable from the mitochondrial impairment induced by exogenous Aß(1-42)-treated HT22 cells. Finally, cellular toxicity was directly induced by mitochondria-targeted Aß(1-42) accumulation, which mimics the apoptosis process in exogenous Aß(1-42)-treated HT22 cells. Taken together, these results indicate that mitochondria-targeted Aß(1-42) accumulation is the necessary and sufficient condition for Aß-mediated mitochondria impairments, and leads directly to cellular death rather than along with other Aß-mediated signaling alterations.


Subject(s)
Amyloid beta-Peptides/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Amyloid beta-Peptides/pharmacology , Animals , Apoptosis/drug effects , Cell Line , Chlorpromazine/pharmacology , Endocytosis/drug effects , Mice , Peptide Fragments/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...