Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 20
1.
Cell Death Dis ; 15(4): 287, 2024 Apr 23.
Article En | MEDLINE | ID: mdl-38654003

This study aimed to elucidate the role of O-GlcNAc cycling in 6-hydroxydopamine (6-OHDA)-induced Parkinson's disease (PD)-like neurodegeneration and the underlying mechanisms. We observed dose-dependent downregulation of O-GlcNAcylation, accompanied by an increase in O-GlcNAcase following 6-OHDA treatment in both mouse brain and Neuro2a cells. Interestingly, elevating O-GlcNAcylation through glucosamine (GlcN) injection provided protection against PD pathogenesis induced by 6-OHDA. At the behavioral level, GlcN mitigated motor deficits induced by 6-OHDA, as determined using the pole, cylinder, and apomorphine rotation tests. Furthermore, GlcN attenuated 6-OHDA-induced neuroinflammation and mitochondrial dysfunction. Notably, augmented O-GlcNAcylation, achieved through O-GlcNAc transferase (OGT) overexpression in mouse brain, conferred protection against 6-OHDA-induced PD pathology, encompassing neuronal cell death, motor deficits, neuroinflammation, and mitochondrial dysfunction. These collective findings suggest that O-GlcNAcylation plays a crucial role in the normal functioning of dopamine neurons. Moreover, enhancing O-GlcNAcylation through genetic and pharmacological means could effectively ameliorate neurodegeneration and motor impairment in an animal model of PD. These results propose a potential strategy for safeguarding against the deterioration of dopamine neurons implicated in PD pathogenesis.


Mice, Inbred C57BL , N-Acetylglucosaminyltransferases , Oxidopamine , Parkinson Disease , Animals , Oxidopamine/pharmacology , Mice , N-Acetylglucosaminyltransferases/metabolism , Parkinson Disease/metabolism , Parkinson Disease/pathology , Male , Glucosamine/pharmacology , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/pathology , Mitochondria/metabolism , Mitochondria/drug effects , Acetylglucosamine/metabolism , Acetylglucosamine/pharmacology , Brain/metabolism , Brain/pathology , Brain/drug effects , beta-N-Acetylhexosaminidases/metabolism , Disease Models, Animal
2.
Am J Physiol Cell Physiol ; 326(3): C978-C989, 2024 Mar 01.
Article En | MEDLINE | ID: mdl-38314722

Sleep deprivation (SD) is widely acknowledged as a significant risk factor for cognitive impairment. In this study, intraperitoneal caffeine administration significantly ameliorated the learning and memory (L/M) deficits induced by SD and reduced aggressive behaviors in adult zebrafish. SD led to a reduction in protein kinase A (PKA) phosphorylation, phosphorylated-cAMP response element-binding protein (p-CREB), and c-Fos expression in zebrafish brain. Notably, these alterations were effectively reversed by caffeine. In addition, caffeine mitigated neuroinflammation induced by SD, as evident from suppression of the SD-mediated increase in glial fibrillary acidic protein (GFAP) and nuclear factor-κB (NF-κB) activation. Caffeine restored normal O-GlcNAcylation and O-GlcNAc transferase (OGT) levels while reversing the increased expression of O-GlcNAcase (OGA) in zebrafish brain after SD. Intriguingly, rolipram, a selective phosphodiesterase 4 (PDE4) inhibitor, effectively mitigated cognitive deficits, restored p-CREB and c-Fos levels, and attenuated the increase in GFAP in brain induced by SD. In addition, rolipram reversed the decrease in O-GlcNAcylation and OGT expression as well as elevation of OGA expression following SD. Treatment with H89, a PKA inhibitor, significantly impaired the L/M functions of zebrafish compared with the control group, inducing a decrease in O-GlcNAcylation and OGT expression and, conversely, an increase in OGA expression. The H89-induced changes in O-GlcNAc cycling and L/M dysfunction were effectively reversed by glucosamine treatment. H89 suppressed, whereas caffeine and rolipram promoted O-GlcNAc cycling in Neuro2a cells. Our collective findings underscore the interplay between PKA signaling and O-GlcNAc cycling in the regulation of cognitive function in the brain, offering potential therapeutic targets for cognitive deficits associated with SD.NEW & NOTEWORTHY Our observation highlights the intricate interplay between cAMP/PKA signaling and O-GlcNAc cycling, unveiling a novel mechanism that potentially governs the regulation of learning and memory functions. The dynamic interplay between these two pathways provides a novel and nuanced perspective on the molecular foundation of learning and memory regulation. These insights open avenues for the development of targeted interventions to treat conditions that impact cognitive function, including SD.


Cognitive Dysfunction , Isoquinolines , Sleep Deprivation , Sulfonamides , Animals , Sleep Deprivation/drug therapy , Zebrafish/metabolism , Caffeine/pharmacology , Rolipram , Acetylglucosamine/metabolism , Protein Processing, Post-Translational , Cognition , Cognitive Dysfunction/drug therapy , Cyclic AMP-Dependent Protein Kinases/metabolism , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/metabolism
3.
Biochem Pharmacol ; 221: 116032, 2024 Mar.
Article En | MEDLINE | ID: mdl-38281601

Repeated sublethal hypoxia exposure induces brain inflammation and affects the initiation and progression of cognitive dysfunction. Experiments from the current study showed that hypoxic exposure downregulates PKA/CREB signaling, which is restored by forskolin (FSK), an adenylate cyclase activator, in both Neuro2a (N2a) cells and zebrafish brain. FSK significantly protected N2a cells from hypoxia-induced cell death and neurite shrinkage. Intraperitoneal administration of FSK for 5 days on zebrafish additionally led to significant recovery from hypoxia-induced social interaction impairment and learning and memory (L/M) deficit. FSK suppressed hypoxia-induced neuroinflammation, as indicated by the observed decrease in NF-κB activation and GFAP expression. We further investigated the potential effect of FSK on O-GlcNAcylation changes induced by hypoxia. Intriguingly FSK induced marked upregulation of the protein level of O-GlcNAc transferase catalyzing addition of the GlcNAc group to target proteins, accompanied by elevated O-GlcNAcylation of nucleocytoplasmic proteins. The hypoxia-induced O-GlcNAcylation decrease in the brain of zebrafish was considerably restored following FSK treatment. Based on the collective results, we propose that FSK rescues hypoxia-induced cognitive dysfunction, potentially through regulation of HBP/O-GlcNAc cycling.


Cognitive Dysfunction , Zebrafish , Animals , Colforsin/pharmacology , Cognition , Hypoxia/complications , Memory Disorders
4.
BMB Rep ; 57(2): 92-97, 2024 Feb.
Article En | MEDLINE | ID: mdl-37964636

Elevated blood glucose is associated with an increased risk of atherosclerosis. Data from the current study showed that glucosamine (GlcN), a normal glucose metabolite of the hexosamine biosynthetic pathway (HBP), promoted lipid accumulation in RAW264.7 macrophage cells. Oleic acid- and lipopolysaccharide (LPS)-induced lipid accumulation was further enhanced by GlcN in RAW264.7 cells, although there was no a significant change in the rate of fatty acid uptake. GlcN increased acetyl CoA carboxylase (ACC), fatty acid synthase (FAS), scavenger receptor class A, liver X receptor, and sterol regulatory elementbinding protein-1c (SREBP-1c) mRNA expression, and; conversely, suppressed ATP-binding cassette transporter A1 (ABCA-1) and ABCG-1 expression. Additionally, GlcN promoted O-GlcNAcylation of nuclear SREBP-1 but did not affect its DNA binding activity. GlcN stimulated phosphorylation of mammalian target of rapamycin (mTOR) and S6 kinase. Rapamycin, a mTOR-specific inhibitor, suppressed GlcN-induced lipid accumulation in RAW264.7 cells. The GlcN-mediated increase in ACC and FAS mRNA was suppressed, while the decrease in ABCA-1 and ABCG-1 by GlcN was not significantly altered by rapamycin. Together, our results highlight the importance of the mTOR signaling pathway in GlcN-induced macrophage lipid accumulation and further support a potential link between mTOR and HBP signaling in lipogenesis. [BMB Reports 2024; 57(2): 92-97].


Glucosamine , Signal Transduction , Animals , Mice , Glucosamine/pharmacology , Lipopolysaccharides , Macrophages , RAW 264.7 Cells , RNA, Messenger , Sirolimus , TOR Serine-Threonine Kinases , Transcription Factors
5.
J Neuroinflammation ; 20(1): 257, 2023 Nov 09.
Article En | MEDLINE | ID: mdl-37946213

This study investigated chronic and repeated sleep deprivation (RSD)-induced neuronal changes in hexosamine biosynthetic pathway/O-linked N-acetylglucosamine (HBP/O-GlcNAc) cycling of glucose metabolism and further explored the role of altered O-GlcNAc cycling in promoting neurodegeneration using an adult zebrafish model. RSD-triggered degenerative changes in the brain led to impairment of memory, neuroinflammation and amyloid beta (Aß) accumulation. Metabolite profiling of RSD zebrafish brain revealed a significant decrease in glucose, indicating a potential association between RSD-induced neurodegeneration and dysregulated glucose metabolism. While RSD had no impact on overall O-GlcNAcylation levels in the hippocampus region, changes were observed in two O-GlcNAcylation-regulating enzymes, specifically, a decrease in O-GlcNAc transferase (OGT) and an increase in O-GlcNAcase (OGA). Glucosamine (GlcN) treatment induced an increase in O-GlcNAcylation and recovery of the OGT level that was decreased in the RSD group. In addition, GlcN reversed cognitive impairment by RSD. GlcN reduced neuroinflammation and attenuated Aß accumulation induced by RSD. Repeated treatment of zebrafish with diazo-5-oxo-l-norleucine (DON), an inhibitor of HBP metabolism, resulted in cognitive dysfunction, neuroinflammation and Aß accumulation, similar to the effects of RSD. The pathological changes induced by DON were restored to normal upon treatment with GlcN. Both the SD and DON-treated groups exhibited a common decrease in glutamate and γ-aminobutyric acid compared to the control group. Overexpression of OGT in zebrafish brain rescued RSD-induced neuronal dysfunction and neurodegeneration. RSD induced a decrease in O-GlcNAcylation of amyloid precursor protein and increase in ß-secretase activity, which were reversed by GlcN treatment. Based on the collective findings, we propose that dysregulation of HBP and O-GlcNAc cycling in brain plays a crucial role in RSD-mediated progression of neurodegeneration and Alzheimer's disease pathogenesis. Targeting of this pathway may, therefore, offer an effective regulatory approach for treatment of sleep-associated neurodegenerative disorders.


Alzheimer Disease , Animals , Alzheimer Disease/pathology , Hexosamines , Zebrafish/metabolism , Sleep Deprivation , Amyloid beta-Peptides/metabolism , Neuroinflammatory Diseases , Biosynthetic Pathways , Glucose
6.
Proc Natl Acad Sci U S A ; 120(37): e2221929120, 2023 09 12.
Article En | MEDLINE | ID: mdl-37669380

The endoplasmic reticulum (ER) is selectively degraded by ER-phagy to maintain cell homeostasis. α-synuclein accumulates in the ER, causing ER stress that contributes to neurodegeneration in Parkinson's disease (PD), but the role of ER-phagy in α-synuclein modulation is largely unknown. Here, we investigated the mechanisms by which ER-phagy selectively recognizes α-synuclein for degradation in the ER. We found that ER-phagy played an important role in the degradation of α-synuclein and recovery of ER function through interaction with FAM134B, where calnexin is required for the selective FAM134B-mediated α-synuclein clearance via ER-phagy. Overexpression of α-synuclein in the ER of the substantia nigra (SN) resulted in marked loss of dopaminergic neurons and motor deficits, mimicking PD characteristics. However, enhancement of ER-phagy using FAM134B overexpression in the SN exerted neuroprotective effects on dopaminergic neurons and recovered motor performance. These data suggest that ER-phagy represents a specific ER clearance mechanism for the degradation of α-synuclein.


Neuroprotective Agents , Parkinson Disease , Humans , alpha-Synuclein , Endoplasmic Reticulum , Autophagy
7.
Am J Physiol Cell Physiol ; 325(4): C981-C998, 2023 10 01.
Article En | MEDLINE | ID: mdl-37602414

Impaired brain glucose metabolism is considered a hallmark of brain dysfunction and neurodegeneration. Disruption of the hexosamine biosynthetic pathway (HBP) and subsequent O-linked N-acetylglucosamine (O-GlcNAc) cycling has been identified as an emerging link between altered glucose metabolism and defects in the brain. Myriads of cytosolic and nuclear proteins in the nervous system are modified at serine or threonine residues with a single N-acetylglucosamine (O-GlcNAc) molecule by O-GlcNAc transferase (OGT), which can be removed by ß-N-acetylglucosaminidase (O-GlcNAcase, OGA). Homeostatic regulation of O-GlcNAc cycling is important for the maintenance of normal brain activity. Although significant evidence linking dysregulated HBP metabolism and aberrant O-GlcNAc cycling to induction or progression of neuronal diseases has been obtained, the issue of whether altered O-GlcNAcylation is causal in brain pathogenesis remains uncertain. Elucidation of the specific functions and regulatory mechanisms of individual O-GlcNAcylated neuronal proteins in both normal and diseased states may facilitate the identification of novel therapeutic targets for various neuronal disorders. The information presented in this review highlights the importance of HBP/O-GlcNAcylation in the neuronal system and summarizes the roles and potential mechanisms of O-GlcNAcylated neuronal proteins in maintaining normal brain function and initiation and progression of neurological diseases.


Acetylglucosamine , Biosynthetic Pathways , Acetylglucosamine/metabolism , Hexosamines/metabolism , Proteins/metabolism , Glucose/metabolism , Brain/metabolism , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/metabolism , Protein Processing, Post-Translational
8.
Stem Cell Res Ther ; 13(1): 483, 2022 09 24.
Article En | MEDLINE | ID: mdl-36153562

BACKGROUND: Mesenchymal stem cells (MSCs) may be one of candidates for disease-modifying therapy in Parkinsonian diseases. As knowledge regarding the therapeutic properties of MSCs accumulates, some obstacles still remain to be overcome, especially, successful clinical translation requires the development of culture systems that mimic the natural MSC niche, while allowing clinical-scale cell expansion without compromising quality and function of the cells. In recent years, priming approaches using bioactive peptide or complement components have been investigated to enhance the therapeutic potential of MSCs. METHODS: We investigated an innovative priming strategy by conditioning the MSCs with α-synuclein (α-syn). To induce priming, MSCs were treated with different concentrations of α-syn and various time course. We evaluated whether α-syn enhances stemness properties of MSCs and priming MSCs with α-syn would modulate autophagy-related gene expression profiles. RESULTS: Treatment of naïve MSCs with α-syn upregulated transcriptional factors responsible for regulation of stemness, which was associated with the elevated expression of genes involved in glycolysis and cell re-programming. Primed MSCs with α-syn enhanced the expression of autophagy-regulating miRNA, and exosomes derived from primed MSCs were packed with autophagy-associated miRNA. In α-syn-overexpressing neuronal cells, primed MSCs with α-syn enhanced neuronal viability relative to naïve MSCs, through the induction of autophagy and lysosome activity. Animal study using an α-syn-overexpressing mice showed that the pro-survival effect of MSCs on dopaminergic neurons was more prominent in primed MSC-treated mice compared with that in naïve MSC-treated mice. CONCLUSIONS: The present data suggest that MSC priming with α-syn exerts neuroprotective effects through augmented stemness and possibly the enhancement of autophagy-mediated α-syn modulation in Parkinsonian models.


Mesenchymal Stem Cells , MicroRNAs , Neuroprotective Agents , Animals , Autophagy/genetics , Dopaminergic Neurons/metabolism , Mesenchymal Stem Cells/metabolism , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Neuroprotective Agents/pharmacology , alpha-Synuclein/genetics , alpha-Synuclein/metabolism , alpha-Synuclein/pharmacology
9.
Front Aging Neurosci ; 14: 851711, 2022.
Article En | MEDLINE | ID: mdl-35721028

Background: Adult neurogenesis is the process of generating new neurons to enter neural circuits and differentiate into functional neurons. However, it is significantly reduced in Parkinson's disease (PD). Uric acid (UA), a natural antioxidant, has neuroprotective properties in patients with PD. This study aimed to investigate whether UA would enhance neurogenesis in PD. Methods: We evaluated whether elevating serum UA levels in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced parkinsonian mouse model would restore neurogenesis in the subventricular zone (SVZ). For a cellular model, we primary cultured neural precursor cells (NPCs) from post-natal day 1 rat and evaluated whether UA treatment promoted cell proliferation against 1-methyl-4-phenylpyridinium (MPP+). Results: Uric acid enhanced neurogenesis in both in vivo and in vitro parkinsonian model. UA-elevating therapy significantly increased the number of bromodeoxyuridine (BrdU)-positive cells in the SVZ of PD animals as compared to PD mice with normal UA levels. In a cellular model, UA treatment increased the expression of Ki-67. In the process of modulating neurogenesis, UA elevation up-regulated the expression of mitochondrial fusion markers. Conclusion: In MPTP-induced parkinsonian model, UA probably enhanced neurogenesis via regulating mitochondrial dynamics, promoting fusion machinery, and inhibiting fission process.

10.
Exp Neurol ; 344: 113810, 2021 10.
Article En | MEDLINE | ID: mdl-34270920

Ample evidence has demonstrated that α-Synuclein can propagate from one area of the brain to others via cell-to-cell transmission, which might be the underlying mechanism for pathological propagation and the disease progression of Parkinson's disease (PD). Recent reports have demonstrated cell surface receptor-mediated cell-to-cell transmission of α-synuclein. Memantine decreased the levels of internalized cytosolic α-synuclein and led to attenuation in α-synuclein-induced cell death. Specifically, memantine attenuated α-synuclein-induced expression of clathrin and EEA1, and increased expression of NR2A subunits. Moreover, memantine inhibited propagation of extracellular α-synuclein and thus, decreased the expression of the phosphorylated form of α-synuclein in dopaminergic neurons of the substantia nigra, which was accompanied by increased survival of dopaminergic neurons with functional improvement of motor deficits. The present study demonstrated that memantine modulates extracellular α-synuclein propagation by inhibiting interactions between α-synuclein and NR2A subunits, which leads to neuroprotective effects on nigral dopaminergic neurons against α-synuclein-enriched conditions. The repositioning use of memantine in α-synuclein propagation needs to be further evaluated in patients with α-synucleinopathies as an effective therapeutic approach.


Memantine/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Parkinsonian Disorders/pathology , alpha-Synuclein/drug effects , Animals , Cell Line , Humans , Mice, Inbred C57BL , Neurons/metabolism , Neurons/pathology , Parkinsonian Disorders/metabolism , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , alpha-Synuclein/metabolism
11.
J Tissue Eng ; 12: 20417314211004816, 2021.
Article En | MEDLINE | ID: mdl-33854750

Mesenchymal stem cells (MSCs) are a potential source of cell-based disease-modifying therapy in Parkinsonian disorders. A promising approach to develop in vitro culture methods that mimic natural MSC niche is cell priming. Uric acid (UA), a powerful antioxidant, scavenges reactive oxygen species, which has a vital role in maintaining self-renewal and differentiation potential of MSCs. Here, we demonstrated that UA treatment in naïve MSCs stimulated glycolysis and upregulated transcriptional factors responsible for regulation of stemness, leading to increase in the expression levels of osteogenesis-, adipogenesis-, and chondrogenesis-related genes. UA-primed MSCs had more enhanced neuroprotective properties in cellular and parkinsonian animal models compared to naïve MSCs by inhibiting apoptotic signaling pathways. Additionally, expression of miR-137 and miR-145 was decreased in UA-treated MSCs. Our data demonstrated that priming MSCs with UA augment neuroprotective properties through enhanced self-renewal and differentiation potential, suggesting a practical strategy for improving the application of MSCs in parkinsonian disorders.

12.
J Alzheimers Dis ; 76(4): 1281-1296, 2020.
Article En | MEDLINE | ID: mdl-32597802

Mesenchymal stem cells (MSCs) promote functional recoveries in pathological experimental models of the central nervous system and are currently being tested in clinical trials for neurological disorders. However, no studies have examined the various roles of embryonic stem cell derived (ES)-MSCs in eliciting therapeutic effects for Alzheimer's disease (AD). In the present study, we investigated the neuroprotective effect of ES-MSCs in cellular and animal models of AD, as well as the safety of the intra-arterial administration of ES-MSCs in an AD animal model. ES-MSCs displayed higher cell viability than that of bone marrow (BM)-MSCs in amyloid-ß (Aß)-induced cellular models. Moreover, the efficacy of autophagy induction in ES-MSCs was comparable to that of BM-MSCs; however, intracellular Aß levels were more significantly reduced in ES-MSCs than in BM-MSCs. In a rat model of AD, ES-MSCs significantly inhibited Aß-induced cell death in the hippocampus and promoted autophagolysosomal clearance of Aß, which was concomitantly followed by decreased levels of Aß in the hippocampus. Furthermore, ES-MSC treatment in Aß-treated rats featured a higher memory performance than that of rats injected solely with Aß. Finally, intra-arterial administration of an appropriate cell density of ES-MSCs was safe and free from in situ occlusion or cerebral ischemia. These data support the therapeutic potential of ES-MSCs and clinical applications of the intra-arterial route of ES-MSC administration in AD.


Alzheimer Disease/pathology , Alzheimer Disease/therapy , Embryonic Stem Cells/cytology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Amyloid beta-Peptides/metabolism , Animals , Disease Models, Animal , Embryonic Stem Cells/pathology , Feasibility Studies , Female , Hippocampus/pathology , Humans , Male , Mesenchymal Stem Cell Transplantation/methods , Neuroprotective Agents , Rats, Sprague-Dawley
13.
Stem Cells ; 35(8): 1934-1947, 2017 08.
Article En | MEDLINE | ID: mdl-28580639

Genome-wide association studies have identified two loci, SNCA and the microtubule (MT)-associated protein tau, as common risk factors for Parkinson's disease (PD). Specifically, α-synuclein directly destabilizes MT via tau phosphorylation and induces axonal transport deficits that are the primary events leading to an abnormal accumulation of α-synuclein that causes nigral dopaminergic cell loss. In this study, we demonstrated that mesenchymal stem cells (MSCs) could modulate cytoskeletal networks and trafficking to exert neuroprotective properties in wild-type or A53T α-synuclein overexpressing cells and mice. Moreover, we found that eukaryotic elongation factor 1A-2, a soluble factor derived from MSCs, stabilized MT assembly by decreasing calcium/calmodulin-dependent tau phosphorylation and induced autophagolysosome fusion, which was accompanied by an increase in the axonal motor proteins and increased neuronal survival. Our data suggest that MSCs have beneficial effects on axonal transports via MT stability by controlling α-synuclein-induced tau phosphorylation, indicating that MSCs may exert a protective role in the early stages of axonal transport defects in α-synucleinopathies. Stem Cells 2017;35:1934-1947.


Autophagy , Axonal Transport , Mesenchymal Stem Cells/metabolism , Models, Biological , Parkinson Disease/metabolism , Parkinson Disease/pathology , alpha-Synuclein/metabolism , Animals , Calcium/metabolism , Calmodulin/metabolism , Cell Line , Dependovirus/metabolism , Humans , Male , Mesenchymal Stem Cells/ultrastructure , Mice, Inbred C57BL , Microtubules/metabolism , Mutation/genetics , Transfection
14.
Stem Cells Transl Med ; 6(3): 949-961, 2017 03.
Article En | MEDLINE | ID: mdl-28297586

Ample evidence has suggested that extracellular α-synuclein aggregates would play key roles in the pathogenesis and progression of Parkinsonian disorders (PDs). In the present study, we investigated whether mesenchymal stem cells (MSCs) and their derived soluble factors could exert neuroprotective effects via proteolysis of extracellular α-synuclein. When preformed α-synuclein aggregates were incubated with MSC-conditioned medium, α-synuclein aggregates were disassembled, and insoluble and oligomeric forms of α-synuclein were markedly decreased, thus leading to a significant increase in neuronal viability. In an animal study, MSC or MSC-conditioned medium treatment decreased the expression of α-synuclein oligomers and the induction of pathogenic α-synuclein with an attenuation of apoptotic cell death signaling. Furthermore, we identified that matrix metalloproteinase-2 (MMP-2), a soluble factor derived from MSCs, played an important role in the degradation of extracellular α-synuclein. Our data demonstrated that MSCs and their derived MMP-2 exert neuroprotective properties through proteolysis of aggregated α-synuclein in PD-related microenvironments. Stem Cells Translational Medicine 2017;6:949-961.


Extracellular Space/chemistry , Matrix Metalloproteinase 2/metabolism , Mesenchymal Stem Cells/metabolism , Parkinson Disease/enzymology , Parkinson Disease/pathology , Protein Aggregates , alpha-Synuclein/metabolism , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Cell Line , Cell Survival/drug effects , Culture Media, Conditioned/pharmacology , Disease Models, Animal , Humans , Male , Mesenchymal Stem Cells/drug effects , Mice, Inbred C57BL , alpha-Synuclein/ultrastructure
15.
J Pediatr Orthop B ; 26(3): 274-276, 2017 May.
Article En | MEDLINE | ID: mdl-26569429

Solitary osteochondromas originating from the carpal bones are very uncommon; when they occur, they usually arise from the scaphoid or capitate. We report a solitary, unilobed osteochondroma arising from the hamate that was excised, with no evidence of recurrence at the 3-year follow-up.


Bone Neoplasms/surgery , Carpal Bones/surgery , Osteochondroma/surgery , Wrist/surgery , Child , Follow-Up Studies , Hand/surgery , Humans , Male , Scaphoid Bone , Tomography, X-Ray Computed , Treatment Outcome
16.
Ann Plast Surg ; 77(1): 80-4, 2016 Jan.
Article En | MEDLINE | ID: mdl-26418806

PURPOSE: We prospectively analyzed the differences in the preoperative status and final outcomes between patients with or without the motivation for prompt surgery after a recent diagnosis of carpal tunnel syndrome (CTS). METHODS: One hundred fifty-six patients were enrolled and followed up from a cohort of 220 patients who were diagnosed with CTS between 2011 and 2013. Basic demographic factors, including the occupational features, were investigated in group 1 (n = 52, conservative treatment followed by surgery) and group 2 (n = 100, surgery immediately after diagnosis). The preoperative electrodiagnosis, clinical items by Graham, Disabilities of the Arm, Shoulder, and Hand (DASH) outcomes questionnaire, Boston Carpal Tunnel Questionnaire scores, and grip/pinch strengths were evaluated, and the degree of improvements was compared. RESULTS: The onset period of the symptoms or signs, as well as the time from the diagnosis to surgery, was significantly longer in group 1 than in group 2. In group 1, nonprofessional, simple, and repetitive jobs were more prevalent; however, the professional category was more common in group 2. The preoperative distributions among the electrodiagnostic grades were not different in both groups. Most of the clinical items by Graham were more definitively improved in group 2. DASH improvement at the final follow-up was meaningfully more in group 2 (34 ± 5.3) than in group 1 (29 ± 6.5). Boston Carpal Tunnel Questionnaire showed a similar trend regarding the DASH score (preoperative, improvement at the final follow-up) with statistical significance. The increase in grip/pinch strength was also greater in group 2. CONCLUSIONS: Most patients, who refused/delayed surgery as the initial treatment for CTS, were not improved by conservative options. Eventually, carpal tunnel release was performed; however, the overall outcomes were inferior compared with those of the groups who agreed to operative treatment as the initial option.


Carpal Tunnel Syndrome/surgery , Conservative Treatment , Orthopedic Procedures , Patient Preference , Adult , Carpal Tunnel Syndrome/diagnosis , Carpal Tunnel Syndrome/therapy , Female , Follow-Up Studies , Humans , Male , Middle Aged , Preoperative Period , Prospective Studies , Treatment Outcome
17.
Clin Orthop Surg ; 7(4): 443-8, 2015 Dec.
Article En | MEDLINE | ID: mdl-26640626

BACKGROUND: Painful hip following hip dislocation or acetabular fracture can be an important signal for early degeneration and progression to osteoarthritis due to intraarticular pathology. However, there is limited literature discussing the use of arthroscopy for the treatment of painful hip. The purpose of this retrospective study was to analyze the effectiveness and benefit of arthroscopic treatment for patients with a painful hip after major trauma. METHODS: From July 2003 to February 2013, we reviewed 13 patients who underwent arthroscopic treatment after acetabular fracture or hip dislocation and were followed up for a minimum of 2 postoperative years. The degree of osteoarthritis based on the Tonnis classification pre- and postoperatively at final follow-up was determined. Clinical outcomes were evaluated using visual analogue scale for pain (VAS) and modified Harris hip score (MHHS), and range of motion (ROM) of the hip pre- and postoperatively at final follow-up. RESULTS: There were nine male and four female patients with a mean age at surgery of 28 years (range, 20 to 50 years). The mean follow-up period of the patients was 59.8 months (range, 24 to 115 months), and the mean interval between initial trauma and arthroscopic treatment was 40.8 months (range, 1 to 144 months). At the final follow-up, VAS and MHHS improved significantly from 6.3 and 53.4 to 3.0 and 88.3, respectively (p = 0.002 and p < 0.001, respectively). However, there were no significant differences in hip flexion, abduction, adduction, external rotation, and internal rotation as minor improvements from 113.1°, 38.5°, 28.5°, 36.5°, and 22.7° to 118.5°, 39.0°, 29.2°, 38.9°, and 26.5° were observed, respectively (p = 0.070, p = 0.414, p = 0.317, p = 0.084, and p = 0.136, respectively). None of the patients exhibited progression of osteoarthritis of the hip at the final follow-up. CONCLUSIONS: Arthroscopic treatment after acetabular fracture or hip dislocation is effective and delays the progression of traumatic osteoarthritis.


Arthroscopy/methods , Hip Dislocation/surgery , Hip Fractures/surgery , Hip/physiopathology , Hip/surgery , Pain/surgery , Adult , Female , Hip/diagnostic imaging , Humans , Male , Middle Aged , Osteoarthritis , Pain/physiopathology , Pain Measurement , Radiography , Range of Motion, Articular , Retrospective Studies , Young Adult
18.
Mol Cells ; 38(2): 145-50, 2015.
Article En | MEDLINE | ID: mdl-25537860

Continuous intra- and extracellular stresses induce disorder of Ca(2+) homeostasis and accumulation of unfolded protein in the endoplasmic reticulum (ER), which results in ER stress. Severe long-term ER stress triggers apoptosis signaling pathways, resulting in cell death. Neural epidermal growth factor-like like protein 2 (NELL2) has been reported to be important in protection of cells from cell death-inducing environments. In this study, we investigated the cytoprotective effect of NELL2 in the context of ER stress induced by thapsigargin, a strong ER stress inducer, in Cos7 cells. Overexpression of NELL2 prevented ER stress-mediated apoptosis by decreasing expression of ER stress-induced C/EBP homologous protein (CHOP) and increasing ER chaperones. In this context, expression of anti-apoptotic Bcl-xL was increased by NELL2, whereas NELL2 decreased expression of pro-apoptotic proteins, such as cleaved caspases 3 and 7. This anti-apoptotic effect of NELL2 is likely mediated by extracellular signal-regulated kinase (ERK) signaling, because its inhibitor, U0126, inhibited effects of NELL2 on the expression of anti- and pro-apoptotic proteins and on the protection from ER stress-induced cell death.


Endoplasmic Reticulum Stress , Endoplasmic Reticulum/physiology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Animals , Apoptosis , COS Cells , Chlorocebus aethiops , Enzyme Inhibitors/pharmacology , Gene Expression Regulation , Thapsigargin/pharmacology , Transcription Factor CHOP/metabolism
19.
PLoS One ; 9(1): e85898, 2014.
Article En | MEDLINE | ID: mdl-24465772

NELL2 was first identified as a mammalian homolog of chick NEL (Neural EGF-like) protein. It is almost exclusively expressed in neurons of the rat brain and has been suggested to play a role in neural differentiation. However, there is still no clear evidence for the detailed function of NELL2 in the differentiation of neurons. In this study, we identified NELL2 function during neural differentiation of mouse embryonic carcinoma P19 cells. Endogenous expression of NELL2 in the P19 cells increased in parallel with the neuronal differentiation induced by retinoic acid (RA). We found that the mouse NELL2 promoter contains RA response elements (RAREs) and that treatment with RA increased NELL2 promoter activity. Transfection of P19 cells with NELL2 expression vectors induced a dramatic increase in cell aggregation, resulting in the facilitation of neural differentiation. Moreover, NELL2 significantly increased N-cadherin expression in the P19 cell. These data suggest that NELL2 plays an important role in the regulation of neuronal differentiation via control of N-cadherin expression and cell aggregation.


Cadherins/metabolism , Embryonal Carcinoma Stem Cells/metabolism , Embryonal Carcinoma Stem Cells/pathology , Nerve Tissue Proteins/metabolism , 5' Flanking Region/genetics , Animals , Cell Aggregation/drug effects , Cell Aggregation/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line, Tumor , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , Gene Expression Regulation, Neoplastic/drug effects , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Mice , Models, Biological , Nerve Tissue Proteins/genetics , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Protein Binding/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Receptors, Retinoic Acid/metabolism , Response Elements/genetics , Tretinoin/pharmacology
20.
J Biol Chem ; 285(32): 25074-84, 2010 Aug 06.
Article En | MEDLINE | ID: mdl-20538601

NELL2 (neural tissue-specific epidermal growth factor-like repeat domain-containing protein) is a secreted glycoprotein that is predominantly expressed in neural tissues. We reported previously that NELL2 mRNA abundance in brain is increased by estrogen (E2) treatment and that NELL2 is involved in the E2-dependent organization of a sexually dimorphic nucleus in the preoptic area. In this study we cloned the mouse NELL2 promoter and found it to contain two half-E2 response elements. Electrophoretic mobility shift assays and promoter assays showed that E2 and its receptors (ERalpha and ERbeta) stimulated NELL2 transcription by binding to the two half-E2 response elements. Hippocampal neuroprogenitor HiB5 cells expressing recombinant NELL2 showed increased cell survival under cell death-inducing conditions. Blockade of endogenous synthesis of NELL2 in HiB5 cells abolished the cell survival effect of E2 and resulted in a decrease in phosphorylation of extracellular signal-regulated kinase 1 and 2 (ERK1/2). These data suggest that the NELL2 gene is trans-activated by E2 and contributes to mediating the survival promoting effects of E2 via intracellular signaling pathway of ERK.


Estrogens/metabolism , Gene Expression Regulation , Nerve Tissue Proteins/metabolism , Animals , Apoptosis , Cell Death , DNA Primers/genetics , Hippocampus/metabolism , Mice , Mitogen-Activated Protein Kinase 3/metabolism , Models, Biological , Promoter Regions, Genetic , Protein Structure, Tertiary , Response Elements , Signal Transduction
...