Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 318
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Proc Natl Acad Sci U S A ; 120(51): e2316823120, 2023 Dec 19.
Article in English | MEDLINE | ID: mdl-38091289

ABSTRACT

Mitochondrial dysfunction plays a critical role in the pathogenesis of Alzheimer's disease (AD). Mitochondrial proteostasis regulated by chaperones and proteases in each compartment of mitochondria is critical for mitochondrial function, and it is suspected that mitochondrial proteostasis deficits may be involved in mitochondrial dysfunction in AD. In this study, we identified LONP1, an ATP-dependent protease in the matrix, as a top Aß42 interacting mitochondrial protein through an unbiased screening and found significantly decreased LONP1 expression and extensive mitochondrial proteostasis deficits in AD experimental models both in vitro and in vivo, as well as in the brain of AD patients. Impaired METTL3-m6A signaling contributed at least in part to Aß42-induced LONP1 reduction. Moreover, Aß42 interaction with LONP1 impaired the assembly and protease activity of LONP1 both in vitro and in vivo. Importantly, LONP1 knockdown caused mitochondrial proteostasis deficits and dysfunction in neurons, while restored expression of LONP1 in neurons expressing intracellular Aß and in the brain of CRND8 APP transgenic mice rescued Aß-induced mitochondrial deficits and cognitive deficits. These results demonstrated a critical role of LONP1 in disturbed mitochondrial proteostasis and mitochondrial dysfunction in AD and revealed a mechanism underlying intracellular Aß42-induced mitochondrial toxicity through its impact on LONP1 and mitochondrial proteostasis.


Subject(s)
Alzheimer Disease , Mitochondrial Diseases , Mice , Animals , Humans , Proteostasis , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Alzheimer Disease/metabolism , Mitochondria/metabolism , Mice, Transgenic , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Mitochondrial Diseases/metabolism , Methyltransferases/metabolism , ATP-Dependent Proteases/metabolism
2.
Bioinformatics ; 40(8)2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39153205

ABSTRACT

SUMMARY: Recent methodology advances in computational signal deconvolution have enabled bulk transcriptome data analysis at a finer cell-type level. Through deconvolution, identifying cell-type-specific differentially expressed (csDE) genes is drawing increasing attention in clinical applications. However, researchers still face a number of difficulties in adopting csDE genes detection methods in practice, especially in their experimental design. Here we present cypress, the first experimental design and statistical power analysis tool in csDE genes identification. This tool can reliably model purified cell-type-specific (CTS) profiles, cell-type compositions, biological and technical variations, offering a high-fidelity simulator for bulk RNA-seq convolution and deconvolution. cypress conducts simulation and evaluates the impact of multiple influencing factors, by various statistical metrics, to help researchers optimize experimental design and conduct power analysis. AVAILABILITY AND IMPLEMENTATION: cypress is an open-source R/Bioconductor package at https://bioconductor.org/packages/cypress/.


Subject(s)
Gene Expression Profiling , Software , Humans , Gene Expression Profiling/methods , Transcriptome
3.
Hum Mol Genet ; 31(20): 3494-3503, 2022 10 10.
Article in English | MEDLINE | ID: mdl-35661211

ABSTRACT

Parkinson's disease (PD) is the second most common neurodegenerative disorder, and aging and genetic and environmental exposure can contribute to its pathogenesis. DNA methylation has been suggested to play a pivotal role in neurodevelopment and neurodegenerative diseases. 5-hydroxymethylcytosine (5hmC) is generated through 5-methylcytosine (5mC) oxidization by ten-eleven translocation proteins and is particularly enriched in the brain. Although 5hmC has been linked to multiple neurological disorders, little is known about 5hmC alterations in the substantia nigra of patients with PD. To determine the specific alterations in DNA methylation and hydroxymethylation in PD brain samples, we examined the genome-wide profiles of 5mC and 5hmC in the substantia nigra of patients with PD and Alzheimer's disease (ad). We identified 4119 differentially hydroxymethylated regions (DhMRs) and no differentially methylated regions (DMRs) in the postmortem brains of patients with PD compared with those of controls. These DhMRs were PD-specific when compared with the results of AD. Gene ontology analysis revealed that several signaling pathways, such as neurogenesis and neuronal differentiation, were significantly enriched in PD DhMRs. KEGG enrichment analysis revealed substantial alterations in multiple signaling pathways, including phospholipase D (PLD), cAMP and Rap1. In addition, using a PD Drosophila model, we found that one of the 5hmC-modulated genes, PLD1, modulated α-synuclein toxicity. Our analysis suggested that 5hmC may act as an independent epigenetic marker and contribute to the pathogenesis of PD.


Subject(s)
Parkinson Disease , Phospholipase D , 5-Methylcytosine/metabolism , DNA Methylation/genetics , Epigenesis, Genetic , Humans , Parkinson Disease/genetics , Phospholipase D/genetics , Phospholipase D/metabolism , Substantia Nigra/metabolism , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
4.
J Cell Sci ; 134(13)2021 07 01.
Article in English | MEDLINE | ID: mdl-34110411

ABSTRACT

Mitochondria-endoplasmic reticulum contacts (MERCs) play an essential role in multiple cell physiological processes. Although Mfn2 was the first protein implicated in the formation of MERCs, there is debate as to whether it acts as a tether or antagonizer, largely based on in vitro studies. To understand the role of Mfn2 in MERCs in vivo, we characterized ultrastructural and biochemical changes of MERCs in pyramidal neurons of hippocampus in Mfn2 conditional knockout mice and in Mfn2 overexpressing mice, and found that Mfn2 ablation caused reduced close contacts, whereas Mfn2 overexpression caused increased close contacts between the endoplasmic reticulum (ER) and mitochondria in vivo. Functional studies on SH-SY5Y cells with Mfn2 knockout or overexpression demonstrating similar biochemical changes found that mitochondrial calcium uptake along with IP3R3-Grp75 interaction was decreased in Mfn2 knockout cells but increased in Mfn2 overexpressing cells. Lastly, we found Mfn2 knockout decreased and Mfn2 overexpression increased the interaction between the ER-mitochondria tethering pair of VAPB-PTPIP51. In conclusion, our study supports the notion that Mfn2 plays a critical role in ER-mitochondrial tethering and the formation of close contacts in neuronal cells in vivo.


Subject(s)
Endoplasmic Reticulum , Mitochondrial Proteins , Animals , Endoplasmic Reticulum/metabolism , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Hippocampus/metabolism , Mice , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Neurons/metabolism , Protein Tyrosine Phosphatases/metabolism
5.
Acta Neuropathol ; 146(1): 31-50, 2023 07.
Article in English | MEDLINE | ID: mdl-37154939

ABSTRACT

Tau neurofibrillary tangles are a hallmark of Alzheimer's disease neuropathological change. However, it remains largely unclear how distinctive Alzheimer's disease tau seeds (i.e. 3R/4R) correlate with histological indicators of tau accumulation. Furthermore, AD tau co-pathology is thought to influence features and progression of other neurodegenerative diseases including Lewy body disease; yet measurements of different types of tau seeds in the setting of such diseases is an unmet need. Here, we use tau real-time quaking-induced conversion (RT-QuIC) assays to selectively quantitate 3R/4R tau seeds in the frontal lobe which accumulates histologically identifiable tau pathology at late disease stages of AD neuropathologic change. Seed quantitation across a spectrum of neurodegenerative disease cases and controls indicated tau seeding activity can be detected well before accompanying histopathological indication of tau deposits, and even prior to the earliest evidence of Alzheimer's-related tau accumulation anywhere in the brain. In later stages of AD, 3R/4R tau RT-QuIC measures correlated with immunohistochemical tau burden. In addition, Alzheimer's tau seeds occur in the vast majority of cases evaluated here inclusive of primary synucleinopathies, frontotemporal lobar degeneration and even controls albeit at multi-log lower levels than Alzheimer's cases. α-synuclein seeding activity confirmed synucleinopathy cases and further indicated the co-occurrence of α-synuclein seeds in some Alzheimer's disease and primary tauopathy cases. Our analysis indicates that 3R/4R tau seeds in the mid-frontal lobe correlate with the overall Braak stage and Alzheimer's disease neuropathologic change, supporting the quantitative predictive value of tau RT-QuIC assays. Our data also indicate 3R/4R tau seeds are elevated in females compared to males at high (≥ IV) Braak stages. This study suggests 3R/4R tau seeds are widespread even prior to the earliest stages of Alzheimer's disease changes, including in normal, and even young individuals, with prevalence across multiple neurodegenerative diseases to further define disease subtypes.


Subject(s)
Alzheimer Disease , Neurodegenerative Diseases , Synucleinopathies , Tauopathies , Female , Humans , Male , alpha-Synuclein , Alzheimer Disease/pathology , tau Proteins , Tauopathies/pathology
6.
Int J Neuropsychopharmacol ; 26(6): 415-425, 2023 Jun 23.
Article in English | MEDLINE | ID: mdl-37208298

ABSTRACT

BACKGROUND: Phosphodiesterase 2A (PDE2A) represents a novel target for new therapies addressing psychiatric disorders. To date, the development of PDE2A inhibitors suitable for human clinical evaluation has been hampered by the poor brain accessibility and metabolic stability of the available compounds. METHODS: Corticosterone (CORT)-induced neuronal cell lesion and restraint stress mouse model were used to measure the neuroprotective effect in cells and antidepressant-like behavior in mice. RESULTS: The cell-based assay showed that both Hcyb1 and PF were potent in protecting cells against stress hormone CORT insults by stimulating cAMP and cGMP signaling in hippocampal cells (HT-22). Administration of both compounds before treatment of CORT to cells increased cAMP/cGMP, VASP phosphorylation at Ser239 and Ser157, cAMP response element binding protein phosphorylation at Ser133, and brain derived neurotrophic factor BDNF expression. Further in vivo study showed that both Hcyb1 and PF displayed -antidepressant- and anxiolytic-like effects against restraint stress as indicated by reduced immobility time in the forced swimming and tail suspension tasks as well as increased open arm entries and time spent in open arms and holes visit in elevated plus maze and hole-board tests, respectively. The biochemical study confirmed that these antidepressant- and anxiolytic-like effects of Hcyb1 and PF were related to cAMP and cGMP signaling in the hippocampus. CONCLUSIONS: The results extend the previous studies and validate that PDE2A is a tractable target for drug development in the treatment of emotional disorders such as depression and anxiety.


Subject(s)
Anti-Anxiety Agents , Phosphodiesterase Inhibitors , Mice , Humans , Animals , Phosphodiesterase Inhibitors/pharmacology , Depression/psychology , Anti-Anxiety Agents/pharmacology , Antidepressive Agents/therapeutic use , Anxiety/drug therapy , Anxiety/chemically induced , Hippocampus , Phosphoric Diester Hydrolases/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Behavior, Animal , Disease Models, Animal
7.
Hum Mol Genet ; 29(1): 149-158, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31814020

ABSTRACT

5-Methylcytosine (5mC), generated through the covalent addition of a methyl group to the fifth carbon of cytosine, is the most prevalent DNA modification in humans and functions as a critical player in the regulation of tissue and cell-specific gene expression. 5mC can be oxidized to 5-hydroxymethylcytosine (5hmC) by ten-eleven translocation (TET) enzymes, which is enriched in brain. Alzheimer's disease (AD) is the most common neurodegenerative disorder, and several studies using the samples collected from Caucasian cohorts have found that epigenetics, particularly cytosine methylation, could play a role in the etiological process of AD. However, little research has been conducted using the samples of other ethnic groups. Here we generated genome-wide profiles of both 5mC and 5hmC in human frontal cortex tissues from late-onset Chinese AD patients and cognitively normal controls. We identified both Chinese-specific and overlapping differentially hydroxymethylated regions (DhMRs) with Caucasian cohorts. Pathway analyses revealed specific pathways enriched among Chinese-specific DhMRs, as well as the shared DhMRs with Caucasian cohorts. Furthermore, two important transcription factor-binding motifs, hypoxia-inducible factor 2α (HIF2α) and hypoxia-inducible factor 1α (HIF1α), were enriched in the DhMRs. Our analyses provide the first genome-wide profiling of DNA hydroxymethylation of the frontal cortex of AD patients from China, emphasizing an important role of 5hmC in AD pathogenesis and highlighting both ethnicity-specific and overlapping changes of brain hydroxymethylome in AD.


Subject(s)
Alzheimer Disease/metabolism , Brain/metabolism , 5-Methylcytosine/analogs & derivatives , 5-Methylcytosine/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain/pathology , Computational Biology , DNA Methylation/genetics , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , RNA-Seq
8.
Proc Natl Acad Sci U S A ; 116(50): 25322-25328, 2019 12 10.
Article in English | MEDLINE | ID: mdl-31767755

ABSTRACT

Loss-of-function mutations in DJ-1 are associated with autosomal recessive early onset Parkinson's disease (PD), yet the underlying pathogenic mechanism remains elusive. Here we demonstrate that DJ-1 localized to the mitochondria-associated membrane (MAM) both in vitro and in vivo. In fact, DJ-1 physically interacts with and is an essential component of the IP3R3-Grp75-VDAC1 complexes at MAM. Loss of DJ-1 disrupted the IP3R3-Grp75-VDAC1 complex and led to reduced endoplasmic reticulum (ER)-mitochondria association and disturbed function of MAM and mitochondria in vitro. These deficits could be rescued by wild-type DJ-1 but not by the familial PD-associated L166P mutant which had demonstrated reduced interaction with IP3R3-Grp75. Furthermore, DJ-1 ablation disturbed calcium efflux-induced IP3R3 degradation after carbachol treatment and caused IP3R3 accumulation at the MAM in vitro. Importantly, similar deficits in IP3R3-Grp75-VDAC1 complexes and MAM were found in the brain of DJ-1 knockout mice in vivo. The DJ-1 level was reduced in the substantia nigra of sporadic PD patients, which was associated with reduced IP3R3-DJ-1 interaction and ER-mitochondria association. Together, these findings offer insights into the cellular mechanism in the involvement of DJ-1 in the regulation of the integrity and calcium cross-talk between ER and mitochondria and suggests that impaired ER-mitochondria association could contribute to the pathogenesis of PD.


Subject(s)
Endoplasmic Reticulum/metabolism , HSP70 Heat-Shock Proteins/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Membrane Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Parkinson Disease/metabolism , Voltage-Dependent Anion Channel 1/metabolism , Animals , Calcium/metabolism , Endoplasmic Reticulum/genetics , HSP70 Heat-Shock Proteins/genetics , Humans , Inositol 1,4,5-Trisphosphate Receptors/genetics , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mitochondria/genetics , Parkinson Disease/genetics , Protein Binding , Voltage-Dependent Anion Channel 1/genetics
9.
Proc Natl Acad Sci U S A ; 115(45): 11567-11572, 2018 11 06.
Article in English | MEDLINE | ID: mdl-30348779

ABSTRACT

Whole-exome sequencing has been successful in identifying genetic factors contributing to familial or sporadic Parkinson's disease (PD). However, this approach has not been applied to explore the impact of de novo mutations on PD pathogenesis. Here, we sequenced the exomes of 39 early onset patients, their parents, and 20 unaffected siblings to investigate the effects of de novo mutations on PD. We identified 12 genes with de novo mutations (MAD1L1, NUP98, PPP2CB, PKMYT1, TRIM24, CEP131, CTTNBP2, NUS1, SMPD3, MGRN1, IFI35, and RUSC2), which could be functionally relevant to PD pathogenesis. Further analyses of two independent case-control cohorts (1,852 patients and 1,565 controls in one cohort and 3,237 patients and 2,858 controls in the other) revealed that NUS1 harbors significantly more rare nonsynonymous variants (P = 1.01E-5, odds ratio = 11.3) in PD patients than in controls. Functional studies in Drosophila demonstrated that the loss of NUS1 could reduce the climbing ability, dopamine level, and number of dopaminergic neurons in 30-day-old flies and could induce apoptosis in fly brain. Together, our data suggest that de novo mutations could contribute to early onset PD pathogenesis and identify NUS1 as a candidate gene for PD.


Subject(s)
Brain/metabolism , Dopaminergic Neurons/metabolism , Mutation , Nerve Tissue Proteins/genetics , Parkinson Disease/genetics , Receptors, Cell Surface/genetics , Adult , Age of Onset , Animals , Apoptosis/genetics , Aryl Hydrocarbon Receptor Nuclear Translocator/antagonists & inhibitors , Aryl Hydrocarbon Receptor Nuclear Translocator/genetics , Aryl Hydrocarbon Receptor Nuclear Translocator/metabolism , Base Sequence , Brain/pathology , Case-Control Studies , Cohort Studies , Disease Models, Animal , Dopamine/metabolism , Dopaminergic Neurons/pathology , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Early Diagnosis , Female , Gene Expression , Gene Regulatory Networks , Humans , Male , Nerve Tissue Proteins/metabolism , Parents , Parkinson Disease/diagnosis , Parkinson Disease/metabolism , Parkinson Disease/pathology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Cell Surface/metabolism , Siblings
10.
Int J Mol Sci ; 22(2)2021 Jan 09.
Article in English | MEDLINE | ID: mdl-33435331

ABSTRACT

Mitochondrial dysfunction represents a critical event in the pathogenesis of Parkinson's disease (PD). Increasing evidence demonstrates that disturbed mitochondrial dynamics and quality control play an important role in mitochondrial dysfunction in PD. Our previous study demonstrated that MPP+ induces mitochondrial fragmentation in vitro. In this study, we aimed to assess whether blocking MPTP-induced mitochondrial fragmentation by overexpressing Mfn2 affords neuroprotection in vivo. We found that the significant loss of dopaminergic neurons in the substantia nigra (SN) induced by MPTP treatment, as seen in wild-type littermate control mice, was almost completely blocked in mice overexpressing Mfn2 (hMfn2 mice). The dramatic reduction in dopamine neuronal fibers and dopamine levels in the striatum caused by MPTP administration was also partially inhibited in hMfn2 mice. MPTP-induced oxidative stress and inflammatory response in the SN and striatum were significantly alleviated in hMfn2 mice. The impairment of motor function caused by MPTP was also blocked in hMfn2 mice. Overall, our work demonstrates that restoration of mitochondrial dynamics by Mfn2 overexpression protects against neuronal toxicity in an MPTP-based PD mouse model, which supports the modulation of mitochondrial dynamics as a potential therapeutic target for PD treatment.


Subject(s)
GTP Phosphohydrolases/genetics , Mitochondrial Proteins/genetics , Parkinsonian Disorders/genetics , Up-Regulation , Animals , Corpus Striatum/metabolism , Corpus Striatum/pathology , Disease Models, Animal , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Female , Male , Mice , Mice, Transgenic , Mitochondrial Dynamics , Oxidative Stress , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/pathology
11.
Mol Psychiatry ; 24(9): 1369-1382, 2019 09.
Article in English | MEDLINE | ID: mdl-30899091

ABSTRACT

Alzheimer's disease (AD) is characterized by the presence of neuritic plaques in which dystrophic neurites (DNs) are typical constituents. We recently showed that DNs labeled by antibodies to the tubular endoplasmic reticulum (ER) protein reticulon-3 (RTN3) are enriched with clustered tubular ER. However, multi-vesicle bodies are also found in DNs, suggesting that different populations of DNs exist in brains of AD patients. To understand how different DNs evolve to surround core amyloid plaques, we monitored the growth of DNs in AD mouse brains (5xFAD and APP/PS1ΔE9 mice) by multiple approaches, including two-dimensional and three-dimensional (3D) electron microscopy (EM). We discovered that a pre-autophagosome protein ATG9A was enriched in DNs when a plaque was just beginning to develop. ATG9A-positive DNs were often closer to the core amyloid plaque, whereas RTN3 immunoreactive DNs were mostly located in the outer layers of ATG9A-positive DNs. Proteins such as RAB7 and LC3 appeared in DNs at later stages during plaque growth, likely accumulated as a part of large autophagy vesicles, and were distributed relatively furthest from the core amyloid plaque. Reconstructing the 3D structure of different morphologies of DNs revealed that DNs in AD mouse brains were constituted in three layers that are distinct by enriching different types of vesicles, as validated by immune-EM methods. Collectively, our results provide the first evidence that DNs evolve from dysfunctions of pre-autophagosomes, tubular ER, mature autophagosomes, and the ubiquitin proteasome system during plaque growth.


Subject(s)
Alzheimer Disease/metabolism , Neurites/pathology , Neuroaxonal Dystrophies/pathology , Alzheimer Disease/physiopathology , Amyloid beta-Protein Precursor/metabolism , Animals , Autophagy-Related Proteins/metabolism , Brain/metabolism , Disease Models, Animal , Endoplasmic Reticulum/metabolism , Humans , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Neuroaxonal Dystrophies/classification , Neuroaxonal Dystrophies/diagnostic imaging , Plaque, Amyloid/metabolism , Vesicular Transport Proteins/metabolism
12.
Hum Mol Genet ; 26(4): 781-789, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28040727

ABSTRACT

Impaired mitochondria dynamics and quality control are involved in mitochondrial dysfunction and pathogenesis of Parkinson's disease (PD). VPS35 mutations cause autosomal dominant PD and we recently demonstrated that fPD-associated VPS35 mutants can cause mitochondrial fragmentation through enhanced VPS35-DLP1 interaction. In this study, we focused on the specific sites on DLP1 responsible for the VPS35-DLP1 interaction. A highly conserved FLV motif was identified in the C-terminus of DLP1, mutation of which significantly reduced VPS35-DLP1 interaction. A decoy peptide design based on this FLV motif could block the VPS35-DLP1 interaction and inhibit the recycling of mitochondrial DLP1 complexes. Importantly, VPS35 D620N mutant-induced mitochondrial fragmentation and respiratory deficits could be rescued by the treatment of this decoy peptide in both M17 cells overexpressing D620N or PD fibroblasts bearing this mutation. Overall, our results lend further support to the notion that VPS35-DLP1 interaction is key to the retromer-dependent recycling of mitochondrial DLP1 complex during mitochondrial fission and provide a novel therapeutic target to control excessive fission and associated mitochondrial deficits.


Subject(s)
GTP Phosphohydrolases/metabolism , Microtubule-Associated Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Dynamics , Mitochondrial Proteins/metabolism , Mutation, Missense , Parkinson Disease/metabolism , Vesicular Transport Proteins/metabolism , Amino Acid Motifs , Amino Acid Substitution , Cell Line, Tumor , Dynamins , GTP Phosphohydrolases/genetics , Humans , Microtubule-Associated Proteins/genetics , Mitochondria/genetics , Mitochondrial Proteins/genetics , Parkinson Disease/genetics , Protein Domains , Vesicular Transport Proteins/genetics
13.
Hum Mol Genet ; 26(21): 4118-4131, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28973308

ABSTRACT

Mitochondrial dysfunction is an early prominent feature in susceptible neurons in the brain of patients with Alzheimer's disease, which likely plays a critical role in the pathogenesis of disease. Increasing evidence suggests abnormal mitochondrial dynamics as important underlying mechanisms. In this study, we characterized marked mitochondrial fragmentation and abnormal mitochondrial distribution in the pyramidal neurons along with mitochondrial dysfunction in the brain of Alzheimer's disease mouse model CRND8 as early as 3 months of age before the accumulation of amyloid pathology. To establish the pathogenic significance of these abnormalities, we inhibited mitochondrial fragmentation by the treatment of mitochondrial division inhibitor 1 (mdivi-1), a mitochondrial fission inhibitor. Mdivi-1 treatment could rescue both mitochondrial fragmentation and distribution deficits and improve mitochondrial function in the CRND8 neurons both in vitro and in vivo. More importantly, the amelioration of mitochondrial dynamic deficits by mdivi-1 treatment markedly decreased extracellular amyloid deposition and Aß1-42/Aß1-40 ratio, prevented the development of cognitive deficits in Y-maze test and improved synaptic parameters. Our findings support the notion that abnormal mitochondrial dynamics plays an early and causal role in mitochondrial dysfunction and Alzheimer's disease-related pathological and cognitive impairments in vivo and indicate the potential value of restoration of mitochondrial dynamics as an innovative therapeutic strategy for Alzheimer's disease.


Subject(s)
Alzheimer Disease/metabolism , Mitochondria/physiology , Mitochondrial Dynamics/drug effects , Alzheimer Disease/prevention & control , Amyloid beta-Peptides/metabolism , Amyloidogenic Proteins/drug effects , Amyloidogenic Proteins/metabolism , Animals , Brain/metabolism , Cognition Disorders/physiopathology , Disease Models, Animal , Mice , Mitochondrial Proteins/metabolism , Neurons/metabolism , Pyramidal Cells/metabolism , Pyramidal Cells/physiology , Quinazolinones/pharmacology
14.
J Neuroinflammation ; 16(1): 263, 2019 Dec 12.
Article in English | MEDLINE | ID: mdl-31831012

ABSTRACT

BACKGROUND: The mechanisms underlying the pathogenesis and progression of Parkinson's disease (PD) remain elusive, but recent opinions and perspectives have focused on whether the inflammation process induced by microglia contributes to α-synuclein-mediated toxicity. Migration of microglia to the substantia nigra (SN) could precede neurodegeneration in A53T mice. We hypothesized that CXCL12 could be a mediator in the α-synuclein-induced migration of microglia. METHODS: After establishing appropriate animal and cell culture models, we explored the relationship between α-synuclein and CXCL12 in A53T mice, primary microglia, and BV-2 cell lines. We also explored the mechanisms of these interactions and the signaling processes involved in neuroinflammation. RESULTS: We confirmed the positive correlation between α-synuclein and CXCL12 in the postmortem brain tissue of PD patients and the upregulated CXCR4 expression in SN microglia of A53T mice. In addition, as expected, α-synuclein increased the production of CXCL12 in microglia via TLR4/IκB-α/NF-κB signaling. Importantly, CXCL12/CXCR4/FAK/Src/Rac1 signaling was shown to be involved in α-synuclein-induced microglial accumulation. CONCLUSIONS: Our study suggests that CXCL12 could be a novel target for the prevention of α-synuclein-triggered ongoing microglial responses. Blocking CXCL12/CXCR4 may be a potential therapeutic approach for PD progression.


Subject(s)
Chemokine CXCL12/metabolism , Parkinson Disease/metabolism , alpha-Synuclein/metabolism , alpha-Synuclein/toxicity , Aged , Aged, 80 and over , Animals , Cell Line, Transformed , Cell Movement/drug effects , Cell Movement/physiology , Chemokine CXCL12/genetics , Female , Humans , Inflammation/chemically induced , Inflammation/genetics , Inflammation/metabolism , Male , Mice , Mice, Transgenic , Middle Aged , Parkinson Disease/genetics , Parkinson Disease/pathology , RAW 264.7 Cells , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Substantia Nigra/pathology , alpha-Synuclein/genetics
15.
J Neurochem ; 147(5): 580-583, 2018 12.
Article in English | MEDLINE | ID: mdl-30474860

ABSTRACT

ε4 allele of ApoE is the strongest genetic risk factor for late onset Alzheimer's disease (AD). Supplementation of ApoE proteins or mimetics has been pursued for drug developments against AD. A very low-density lipoprotein (HDL) mimetic peptide 4F was shown to alleviate AD-related deficits in APP transgenic mice, and this editorial highlights a study by Chernick et al. who use both mouse and human neuroglial cells to explore the mechanism underlying beneficial effects of this peptide. The authors demonstrate that 4F peptide significantly increased the secretion and lipidation of ApoE in the absence and presence of Aß independent of de novo transcription/translation, but requiring ABCA1 and the integrity of the secretory pathway between ER and Golgi. This study reveals a novel mechanism of HDL mimetic peptide as a functional ApoE enhancer and support further development of ApoA-I 4F peptide as effective ApoE modulating agents against AD.


Subject(s)
Alzheimer Disease , Astrocytes , Amyloid beta-Peptides , Animals , Apolipoproteins E , Brain , Dietary Supplements , Humans , Lipoproteins, HDL , Mice , Mice, Transgenic , Microglia , Peptides
16.
Hum Mol Genet ; 24(5): 1322-35, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25355420

ABSTRACT

Parkinson's disease (PD) is characterized by selective degeneration of dopaminergic neurons. Although the etiology of PD remains incompletely understood, oxidative stress has been implicated as an important contributor in the development of PD. Oxidative stress can lead to oxidation and functional perturbation of proteins critical to neuronal survival. Glutaredoxin 1 (Grx1) is an evolutionally conserved antioxidant enzyme that repairs protein oxidation by reversing the oxidative modification of cysteine known as S-glutathionylation. We aimed to explore the regulatory role of Grx1 in PD. We first examined the levels of Grx1 in postmortem midbrain samples from PD patients, and observed that Grx1 content is decreased in PD, specifically within the dopaminergic neurons. We subsequently investigated the potential role of Grx1 deficiency in PD pathogenesis by examining the consequences of loss of the Caenorhabditis elegans Grx1 homolog in well-established worm models of familial PD caused by overexpression of pathogenic human LRRK2 mutants G2019S or R1441C. We found that loss of the Grx1 homolog led to significant exacerbation of the neurodegenerative phenotype in C. elegans overexpressing the human LRRK2 mutants. Re-expression in the dopaminergic neurons of the active, but not a catalytically inactive form of the Grx1 homolog rescued the exacerbated phenotype. Loss of the Grx1 homolog also exacerbated the neurodegenerative phenotype in other C. elegans models, including overexpression of human α-synuclein and overexpression of tyrosine hydroxylase (a model of sporadic PD). Therefore, our results reveal a novel neuroprotective role of glutaredoxin against dopaminergic neurodegeneration in models of familial and sporadic PD.


Subject(s)
Caenorhabditis elegans/genetics , Glutaredoxins/genetics , Helminth Proteins/metabolism , Parkinson Disease/genetics , Animals , Cell Survival , Cysteine/metabolism , Disease Models, Animal , Dopaminergic Neurons/metabolism , Evolution, Molecular , Gene Expression Regulation , Glutaredoxins/deficiency , Glutaredoxins/metabolism , Helminth Proteins/genetics , Homeostasis , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Mesencephalon/metabolism , Oxidative Stress , Phenotype , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
17.
Biochim Biophys Acta Mol Basis Dis ; 1863(11): 2791-2795, 2017 11.
Article in English | MEDLINE | ID: mdl-28765075

ABSTRACT

Defect in the complex I of the mitochondrial electron-transport chain is a characteristic of Parkinson's disease (PD) which is thought to play a critical role in the disease pathogenesis. Mutations in vacuolar protein sorting 35 (VPS35) cause autosomal dominant PD and we recently demonstrated that pathogenic VPS35 mutations cause mitochondrial damage through enhanced mitochondrial fragmentation. In this study, we aimed to determine whether pathogenic VPS35 mutation impacts the activity of complex I and its underlying mechanism. Indeed, VPS35 D620N mutation led to decreased enzymatic activity and respiratory defects in complex I and II in patient fibroblasts. While no changes in the expression of the complex I and II subunits were noted, the level of assembled complex I and II as well as the supercomplex was significantly reduced in D620N fibroblasts. Importantly, inhibition of mitochondrial fission rescued the contents of assembled complexes as well as the functional defects in complex I and II. Overall, these results suggest that VPS35 D620N mutation-induced excessive mitochondrial fission leads to the defects in the assembled complex I and supercomplex and causes bioenergetics deficits.


Subject(s)
Electron Transport Complex I/deficiency , Fibroblasts/metabolism , Mutation, Missense , Parkinson Disease/metabolism , Vesicular Transport Proteins/metabolism , Amino Acid Substitution , Electron Transport Complex II/genetics , Electron Transport Complex II/metabolism , Fibroblasts/pathology , Humans , Parkinson Disease/genetics , Parkinson Disease/pathology , Vesicular Transport Proteins/genetics
18.
Biochim Biophys Acta Mol Basis Dis ; 1863(6): 1359-1370, 2017 06.
Article in English | MEDLINE | ID: mdl-28215578

ABSTRACT

Mitochondrial dynamics and quality control play a critical role in the maintenance of mitochondrial homeostasis and function. Pathogenic mutations of many genes associated with familial Parkinson's disease (PD) caused abnormal mitochondrial dynamics, suggesting a likely involvement of disturbed mitochondrial fission/fusion in the pathogenesis of PD. In this study, we focused on the potential role of mitochondrial fission/fusion in idiopathic PD patients and in neuronal cells and animals exposed to paraquat (PQ), a commonly used herbicide and PD-related neurotoxin, as models for idiopathic PD. Significantly increased expression of dynamin-like protein 1 (DLP1) and a trend towards reduced expression of Mfn1 and Mfn2 were noted in the substantia nigra tissues from idiopathic PD cases. Interestingly, PQ treatment led to similar changes in the expression of fission/fusion proteins both in vitro and in vivo which was accompanied by extensive mitochondrial fragmentation and mitochondrial dysfunction. Blockage of PQ-induced mitochondrial fragmentation by Mfn2 overexpression protected neurons against PQ-induced mitochondrial dysfunction in vitro. More importantly, PQ-induced oxidative damage and stress signaling as well as selective loss of dopaminergic (DA) neurons in the substantia nigra and axonal terminals in striatum was also inhibited in transgenic mice overexpressing hMfn2. Overall, our study demonstrated that disturbed mitochondrial dynamics mediates PQ-induced mitochondrial dysfunction and neurotoxicity both in vitro and in vivo and is also likely involved in the pathogenesis of idiopathic PD which make them a promising therapeutic target for PD treatment.


Subject(s)
Dopaminergic Neurons/metabolism , GTP Phosphohydrolases/metabolism , Mitochondrial Proteins/metabolism , Nerve Tissue Proteins/metabolism , Paraquat/adverse effects , Parkinson Disease, Secondary/metabolism , Substantia Nigra/metabolism , Animals , Cell Line, Tumor , Dopaminergic Neurons/pathology , GTP Phosphohydrolases/genetics , Humans , Mice , Mice, Transgenic , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Proteins/genetics , Nerve Tissue Proteins/genetics , Oxidative Stress/drug effects , Paraquat/pharmacology , Parkinson Disease, Secondary/chemically induced , Parkinson Disease, Secondary/genetics , Parkinson Disease, Secondary/pathology , Substantia Nigra/pathology
19.
Biochem Soc Trans ; 45(5): 1053-1066, 2017 Oct 15.
Article in English | MEDLINE | ID: mdl-28778984

ABSTRACT

Unlike DNA, oxidative damage to RNA has received little attention presumably due to the assumed transient nature of RNA. However, RNAs including mRNA can persist for several hours to days in certain tissues and are demonstrated to sustain greater oxidative damage than DNA. Because neuronal cells in the brain are continuously exposed to reactive oxygen species due to a high oxygen consumption rate, it is not surprising that neuronal RNA oxidation is observed as a common feature at an early stage in a series of neurodegenerative disorders. A recent study on a well-defined bacterial translation system has revealed that mRNA containing 8-oxo-guanosine (8-oxoGuo) has little effect on fidelity despite the anticipated miscoding. Indeed, 8-oxoGuo-containing mRNA leads to ribosomal stalling with a reduced rate of peptide-bond formation by 3-4 orders of magnitude and is subject to no-go decay, a ribosome-based mRNA surveillance mechanism. Another study demonstrates that transfer RNA oxidation catalyzed by cytochrome c (cyt c) leads to its depurination and cross-linking, which may facilitate cyt c release from mitochondria and subsequently induce apoptosis. Even more importantly, a discovery of oxidized microRNA has been recently reported. The oxidized microRNA causes misrecognizing the target mRNAs and subsequent down-regulation in the protein synthesis. It is noteworthy that oxidative modification to RNA not only interferes with the translational machinery but also with regulatory mechanisms of noncoding RNAs that contribute toward the biological complexity of the mammalian brain. Oxidative RNA damage might be a promising therapeutic target potentially useful for an early intervention of diverse neuropsychiatric disorders.


Subject(s)
Neurodegenerative Diseases/genetics , Oxidation-Reduction , Protein Biosynthesis , RNA/chemistry , Animals , Guanosine/analogs & derivatives , Guanosine/metabolism , Humans , MicroRNAs/chemistry , Neurons/chemistry , RNA, Messenger/chemistry , RNA, Messenger/metabolism , RNA, Transfer/chemistry , Ribosomes/genetics , Ribosomes/metabolism
20.
Biochemistry ; 55(32): 4519-32, 2016 08 16.
Article in English | MEDLINE | ID: mdl-26894491

ABSTRACT

Parkinson's disease (PD) is the second most common neurodegenerative disease worldwide, caused by the degeneration of the dopaminergic neurons in the substantia nigra. Mutations in PARK7 (DJ-1) result in early onset autosomal recessive PD, and oxidative modification of DJ-1 has been reported to regulate the protective activity of DJ-1 in vitro. Glutathionylation is a prevalent redox modification of proteins resulting from the disulfide adduction of the glutathione moiety to a reactive cysteine-SH, and glutathionylation of specific proteins has been implicated in regulation of cell viability. Glutaredoxin 1 (Grx1) is the principal deglutathionylating enzyme within cells, and it has been reported to mediate protection of dopaminergic neurons in Caenorhabditis elegans; however many of the functional downstream targets of Grx1 in vivo remain unknown. Previously, DJ-1 protein content was shown to decrease concomitantly with diminution of Grx1 protein content in cell culture of model neurons (SH-SY5Y and Neuro-2A lines). In the current study we aimed to investigate the regulation of DJ-1 by Grx1 in vivo and characterize its glutathionylation in vitro. Here, with Grx(-/-) mice we provide show that Grx1 regulates protein levels of DJ-1 in vivo. Furthermore, with model neuronal cells (SH-SY5Y) we observed decreased DJ-1 protein content in response to treatment with known glutathionylating agents, and with isolated DJ-1 we identified two distinct sites of glutathionylation. Finally, we found that overexpression of DJ-1 in the dopaminergic neurons partly compensates for the loss of the Grx1 homologue in a C. elegans in vivo model of PD. Therefore, our results reveal a novel redox modification of DJ-1 and suggest a novel regulatory mechanism for DJ-1 content in vivo.


Subject(s)
Glutaredoxins/metabolism , Parkinson Disease/metabolism , Protein Deglycase DJ-1/metabolism , Amino Acid Sequence , Animals , Caenorhabditis elegans , Cell Line, Tumor , Cysteine/metabolism , Glutathione/metabolism , Humans , Mice , Protein Deglycase DJ-1/chemistry , Protein Deglycase DJ-1/deficiency , Protein Processing, Post-Translational
SELECTION OF CITATIONS
SEARCH DETAIL