Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
Yonago Acta Med ; 67(1): 1-8, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38380436

ABSTRACT

In recent years, microglia have attracted attention owing to their roles in various neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. Microglia, which are brain-resident macrophages, not only act as immune cells but also perform other functions in the body. Interestingly, they exert contrasting effects on different neurodegenerative diseases. In addition to the previously reported M1 (toxic) and M2 (protective) types, microglia now also include disease-associated microglia owing to a more elaborate classification. Understanding this detailed classification is necessary to elucidate the association between microglia and neurodegenerative diseases. In this review, we discuss the diverse roles of microglia in neurodegenerative diseases and highlight their potential as therapeutic targets.

2.
Anticancer Res ; 43(11): 4843-4853, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37909958

ABSTRACT

BACKGROUND/AIM: Persistent hyperglycemia caused by diabetes mellitus is a risk factor for pancreatic cancer (PC). We have previously reported that aberrant activation of atypical protein kinase C (aPKC) enhances PC cell progression. However, no reports have elucidated whether hyperglycemia promotes PC cell progression and whether aPKC activation is related to PC cell progression mechanisms. MATERIALS AND METHODS: We examined whether high-glucose stimulation accelerates PC cell proliferation, migration, and invasion. Furthermore, to determine whether PC cells activate aPKC upon high-glucose stimulation, we measured the phosphorylation of aPKC at T560 in PC cells. RESULTS: High-glucose stimulation accelerated PC cell proliferation, migration, and invasion. High-glucose treatment increased aPKC's activated form, with T560 phosphorylation, in PC cells. However, aPKC knockdown attenuated these effects. aPKC reportedly induces cell transformation through Yes-associated protein (YAP) activation. YAP expression was increased in high glucose-treated PC cells but not in aPKC-knockdown cells. aPKC interacts with partitioning defective 3 (Par-3), which aids in establishing cell polarity and inhibits aPKC by binding as a substrate. In Par-3-knockdown PC cells, YAP expression increased independently of high-glucose treatment. Over-expression of Par-3 and aPKC-dominant negative mutants prevented the high glucose-stimulated nuclear localization of YAP. YAP forms a complex with the zinc finger E-box binding homeobox 1 protein (ZEB1), an activator of epithelial-mesenchymal transition. ZEB1 expression was increased by high glucose treatment or Par-3 knockdown, but aPKC knockdown suppressed this increase. CONCLUSION: High glucose-induced aPKC activation promotes PC progression by enhancing the YAP signaling pathway.


Subject(s)
Hyperglycemia , Pancreatic Neoplasms , Humans , Glucose/pharmacology , Pancreatic Neoplasms/genetics , Signal Transduction , Pancreatic Neoplasms
3.
Transl Oncol ; 28: 101608, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36577166

ABSTRACT

Pancreatic cancer (PC) is an aggressive malignancy with few treatment options, and improved treatment strategies are urgently required. TYRO3, a member of the TAM receptor tyrosine kinase family, is a known oncogene; however, the relationship between TYRO3 expression and PC chemoresistance remains to be elucidated. We performed gain- and loss-of-function experiments on TYRO3 to examine whether it is involved in chemoresistance in PC cells. TYRO3 knockdown decreased cell viability and enhanced apoptosis following treatment of PC cells with gemcitabine and 5-fluorouracil (5-FU). In contrast, no such effects were observed in TYRO3-overexpressing PC cells. It is known that autophagy is associated with cancer chemoresistance. We then examined effects of TYRO3 on autophagy in PC cells. TYRO3 overexpression increased LC3 mRNA levels and induced LC3 puncta in PC cells. Inhibition of autophagy by chloroquine mitigated cell resistance to gemcitabine and 5-FU. In a xenograft mouse model, TYRO3 silencing significantly increased sensitivity of the cells to gemcitabine and 5-FU. To further investigate the involvement of autophagy in patients with PC, we immunohistochemically analyzed LC3 expression in the tissues of patients who underwent pancreatectomy and compared it with disease prognosis and TYRO3 expression. LC3 expression was negatively and positively correlated with prognosis and TYRO3 expression, respectively. Furthermore, LC3- and TYRO3-positive patients had a significantly worse prognosis among patients with PC who received chemotherapy after recurrence. These results indicated that the TYRO3-autophagy signaling pathway confers PC resistance to gemcitabine and 5-FU, and could be a novel therapeutic target to resolve PC chemoresistance.

4.
J Clin Biochem Nutr ; 70(3): 222-230, 2022 May.
Article in English | MEDLINE | ID: mdl-35692678

ABSTRACT

Coenzyme Q10 (CoQ10) promotes wound healing in vitro and in vivo. However, the molecular mechanisms underlying the promoting effects of CoQ10 on wound repair remain unknown. In the present study, we investigated the molecular mechanisms through which CoQ10 induces wound repair using a cellular wound-healing model. CoQ10 promoted wound closure in a dose-dependent manner and wound-mediated cell polarization after wounding in HaCaT cells. A comparison with other CoQ homologs, benzoquinone derivatives, and polyisoprenyl compounds suggested that the whole structure of CoQ10 is required for potent wound repair. The phosphorylation of Akt after wounding and the plasma membrane translocation of Akt were elevated in CoQ10-treated cells. The promoting effect of CoQ10 on wound repair was abrogated by co-treatment with a phosphatidylinositol 3-kinase (PI3K) inhibitor. Immuno-histochemical and biochemical analyses showed that CoQ10 increased the localization of caveolin-1 (Cav-1) to the apical membrane domains of the cells and the Cav-1 content in the membrane-rich fractions. Depletion of Cav-1 suppressed CoQ10-mediated wound repair and PI3K/Akt signaling activation in HaCaT cells. These results indicated that CoQ10 increases the translocation of Cav-1 to the plasma membranes, activating the downstream PI3K/Akt signaling pathway, and resulting in wound closure in HaCaT cells.

5.
Cancer Lett ; 470: 149-160, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31765735

ABSTRACT

The expression and functions of TYRO3, a member of the TAM receptor tyrosine kinase family, in pancreatic cancer (PC) have not been specifically elucidated. In this study, we confirmed TYRO3 expression in five human PC cell lines (PANC-1, MIA PaCa-2, BxPC-3, AsPC-1, and PK-9) using Western blotting. TYRO3 silencing and overexpression studies have revealed that TYRO3 promotes cell proliferation and invasion in PC via phosphorylation of protein kinase B (Akt) and extracellular signal-regulated kinase (ERK). Using a mouse xenograft model, we showed that tumor growth was significantly suppressed in mice subcutaneously inoculated with TYRO3-knockdown PC cells compared with mice inoculated with control PC cells. Furthermore, TYRO3 expression was examined in PC tissues obtained from 106 patients who underwent pancreatic resection for invasive ductal carcinoma through immunohistochemical staining. TYRO3-positive patients had poor prognoses for overall survival and disease-specific survival compared with TYRO3-negative patients. Multivariate analysis revealed that TYRO3 expression is an independent prognostic factor for overall survival. Our study demonstrates the critical role of TYRO3 in PC progression through Akt and ERK activation and suggests TYRO3 as a novel promising target for therapeutic strategies against PC.


Subject(s)
Pancreatic Neoplasms/pathology , Receptor Protein-Tyrosine Kinases/metabolism , Aged , Animals , Carcinogenesis , Cell Line, Tumor , Cell Proliferation , Disease Progression , Female , Gene Knockdown Techniques , Humans , Kaplan-Meier Estimate , MAP Kinase Signaling System , Male , Mice , Middle Aged , Neoplasm Invasiveness/pathology , Neoplasm Staging , Pancreas/pathology , Pancreatic Neoplasms/mortality , Phosphorylation , Prognosis , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Xenograft Model Antitumor Assays
6.
Sci Rep ; 9(1): 7562, 2019 05 17.
Article in English | MEDLINE | ID: mdl-31101857

ABSTRACT

Post-septic neurological and psychiatric illness (PSNPI) including dementia and depression may be observed after sepsis. However, the etiology of PSNPI and therapeutic treatment of PSNPI are unclear. We show that glutamate produced from microglia through the activity of system xc- plays a role in PSNPI. We established a mouse model of PSNPI by lipopolysaccharide (LPS) treatment that shows a disturbance of short/working memory and depression-like hypoactivity. Glutamate receptor antagonists (MK801 and DNQX) reduced these phenotypes, and isolated microglia from LPS-treated mice released abundant glutamate. We identified system xc- as a source of the extracellular glutamate. xCT, a component of system xc-, was induced and expressed in microglia after LPS treatment. In xCT knockout mice, PSNPI were decreased compared to those in wildtype mice. Moreover, TNF-α and IL-1ß expression in wildtype mice was increased after LPS treatment, but inhibited in xCT knockout mice. Thus, system xc- in microglia may be a therapeutic target for PSNPI. The administration of sulfasalazine, an inhibitor of xCT, in symptomatic and post-symptomatic mice improved PSNPI. Our results suggest that glutamate released from microglia through system xc- plays a critical role in the manifestations of PSNPI and that system xc- may be a therapeutic target for PSNPI.


Subject(s)
Amino Acid Transport System y+/genetics , Glutamic Acid/metabolism , Mental Disorders/etiology , Microglia/metabolism , Nervous System Diseases/etiology , Amino Acid Transport System y+/antagonists & inhibitors , Amino Acid Transport System y+/metabolism , Animals , Disease Models, Animal , Dizocilpine Maleate/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Female , Interleukin-1beta/metabolism , Lipopolysaccharides/toxicity , Male , Mental Disorders/drug therapy , Mice , Mice, Inbred C57BL , Mice, Knockout , Nervous System Diseases/drug therapy , Quinoxalines/pharmacology , Sepsis/psychology , Sulfasalazine/pharmacology , Tumor Necrosis Factor-alpha/metabolism
7.
Biofactors ; 44(2): 180-191, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29399897

ABSTRACT

In many developed countries including Japan, how to care the bedridden elderly people with chronic wounds such as decubitus becomes one of the most concerned issues. Although antioxidant micronutrients including vitamin E, especially α-tocopherol (α-Toc), are reported to shorten a period of wound closure, the promoting effect of α-Toc on wound healing independent of its antioxidant activity remains to be fully elucidated. The aim of this study was to examine whether α-Toc affects wound-mediated HaCaT keratinocyte polarization process including the recruitment of polarity regulating proteins, leading to wound repair independently of its antioxidant activity. We investigated the effects of α-Toc and other antioxidants such as Trolox, a cell-permeable α-Toc analog on the migration, proliferation, and cell polarization of HaCaT keratinocytes after wounding. We analyzed the localization and complex formation of polarity proteins, partitioning defective 3 (Par3), and atypical protein kinase C (aPKC), and aPKC activity by immunohistochemistry, immunoprecipitation analyses, and in vitro kinase assays, respectively. α-Toc but not other antioxidants enhanced the wound closure and cell polarization in HaCaT keratinocytes after wounding. α-Toc regulated the localization and complex formation of Par3 and aPKC during wound healing. Knockdown of aPKC or Par3 abrogated α-Toc-mediated promotion of the wound closure and cell polarization in HaCaT keratinocytes. Furthermore, aPKC kinase activity was significantly increased in α-Toc-treated cells through activation of phosphatidylinositol 3-kinase/Akt signaling pathway. These results suggest that α-Toc promotes HaCaT keratinocyte wound repair by regulating the aPKC kinase activity and the formation of aPKC-Par3 complex. © 2017 BioFactors, 44(2):180-191, 2018.


Subject(s)
Cell Movement/drug effects , Cell Polarity/drug effects , Gene Expression Regulation/drug effects , Keratinocytes/drug effects , alpha-Tocopherol/pharmacology , Adaptor Proteins, Signal Transducing , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Transformed , Cell Polarity/genetics , Cell Proliferation/drug effects , Chromans/pharmacology , Humans , Keratinocytes/cytology , Keratinocytes/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Models, Biological , Phosphatidylinositol 3-Kinase/genetics , Phosphatidylinositol 3-Kinase/metabolism , Protein Kinase C/genetics , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Wound Healing/drug effects , Wound Healing/genetics
8.
Yonago Acta Med ; 59(3): 188-195, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27708533

ABSTRACT

BACKGROUND: Vitamin E (VE, α-tocopherol) is a fat-soluble vitamin and is well known as an antioxidant. A deficiency in VE induces oxidative stress in the brain and causes motor and memory dysfunction. The consumption of a VE-rich diet has been given much attention in recent years, in regards to anti-aging and the prevention of age-related neuronal disorders. METHODS: A VE-deficient mouse model was prepared by feeding the animals a diet lacking VE. In addition, to evaluate the effect of VE-containing rice bran (RB) on VE deficiency, a diet including RB was also provided. VE levels in the brain tissue, as well as in the RB, were measured using an HPLC system. Behavioral tests, including rotarod, wheel running activity, Y-maze, and elevated plus maze were performed. To clarify the effect of VE deficiency and RB, we investigated the induction of heme oxygenase-1 (HO-1). Histological studies were performed using HE staining and immunohistochemical studies were performed using antibodies against glial fibrillary acidic protein (GFAP) and ionized calcium binding adaptor molecule 1 (Iba1). RESULTS: VE in the mouse brain under a VE-deficient diet was decreased, and recovered α-tocopherol levels were observed in the brain of mice fed an RB diet. Motor behavioral scores were decreased in VE-deficient conditions, while the supplementation of RB improved motor function. HO-1, a marker of oxidative stress, was upregulated in the mouse brain under VE deficiency, however, RB supplementation inhibited the increase of HO-1. Histological analyses showed neuronal degeneration of Purkinje cells and decreased GFAP-immunoreactivity of Bergmann glia in the cerebellum. In addition, activated astrocytes and microglia were observed in mice fed the VE-deficient diet. Mice fed the RB diet showed improvement in these histological abnormalities. CONCLUSION: A VE-deficient diet induced motor dysfunction in mice due to the degeneration of Purkinje cells in the cerebellum. Oral supplementation of RB increases VE in the brain and improved the motor dysfunction caused by VE deficiency. Thus, RB or unpolished rice may be a promising VE supplement.

9.
Yonago Acta Med ; 59(3): 223-231, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27708538

ABSTRACT

BACKGROUND: Sepsis is a syndrome triggered by endotoxin lipopolysaccharide (LPS) during bacterial infection. Sepsis sometimes recurs, with the second sepsis giving rise to a different phenotype because of disease modification by the preceding sepsis. Such a protective modification is called a preconditioning (PC) effect. PC is an endogenous protective mechanism by which sublethal damage confers tolerance to a subsequent lethal load. Oxidative stress is one of the important pathogenetic mechanisms that occur in sepsis. The nuclear factor erythroid 2 (NF-E2)-related factor-2 (Nrf2) system is a key regulatory transcription factor that protects organs and cells against oxidative stress and may be associated with the PC effect in repeated sepsis. METHODS: The effect of PC induced by low-dose LPS on survival rate and liver injury against subsequent high-dose LPS stimulation was examined using a mouse model of sepsis. In order to understand the detailed mechanism(s) involved in the PC effect within the liver, gene expression array was performed. As a candidate mechanism of PC, the activation of the Nrf2 system was analyzed using Nrf2 reporter mice. Furthermore, the induction of heme oxygenase-1 (HO-1), one of the main targets of Nrf2, in the liver was examined by immunoblotting and immunohistochemistry. The PC effect on liver injury induced by LPS was further examined using Nrf2-deficient mice. RESULTS: PC by LPS (1.7 or 5.0 mg/kg body weight, intraperitoneally) increased the survival rate of mice and decreased liver injury in response to a subsequent injection of a lethal level of LPS (20 mg/kg body weight). DNA array revealed that the gene ontology term "antioxidant activity" as one of the candidate mechanisms of the PC effect by LPS. In Nrf2 reporter mice, PC immediately and intensely enhanced luminescence that indicated Nrf2 activation after subsequent LPS injection. The induction of HO-1 by LPS was also enhanced by preceding PC, and its induction was observed mainly in Kupffer cells of the liver. In Nrf2-deficient mice, the induction of HO-1 in Kupffer cells and the hepatoprotective effect of PC were decreased as compared with wild-type mice. CONCLUSION: Our results suggest that activation of the Nrf2 system is, at least in part, one of the mechanisms of a PC effect in the mouse liver in the case of repeated LPS stimulation.

10.
Biochim Biophys Acta ; 1860(11 Pt A): 2404-2415, 2016 11.
Article in English | MEDLINE | ID: mdl-27424921

ABSTRACT

BACKGROUND: Pancreatic cancer (PC) is the most lethal malignancy among solid tumors, and the most common risk factor for its development is cigarette smoking. Atypical protein kinase C (aPKC) isozymes function in cell polarity, proliferation, and survival, and have also been implicated in carcinogenesis. However, the involvement of aPKC in PC progression and the effect of nicotine, a major component of cigarette smoke, on the biological activities of aPKC remain to be fully elucidated. METHODS: We investigated the effects of nicotine on the proliferation, migration and invasion of the human PC cell lines Panc1 and BxPC3. We analyzed aPKC localization and activity by immunohistochemistry and in vitro kinase assays, respectively, to assess their involvement in the regulation of PC progression. Moreover, we examined the effect of nicotine on implanted peritoneal tumors of PC cells in mice. RESULTS: Nicotine enhanced cell proliferation, migration and invasion in Panc1 and BxPC3 cells. In nicotine-treated PC cells, the aPKC was significantly activated. We also found that nicotine induced phosphatidylinositol 3-kinase (PI3K) signal activation, and a specific inhibitor of the nicotine acetylcholine receptor (nAChR) as well as knockdown of nAChR prevented nicotine-mediated Akt phosphorylation and aPKC activation. In a peritoneal dissemination model of PC, nicotine-treated mice had larger tumors and increased numbers of nodules. Immunohistochemistry showed enhanced expression levels of aPKC and phosphorylated Akt in nodules from nicotine-treated mice. CONCLUSIONS AND GENERAL SIGNIFICANCE: Nicotine induces aberrant activation of aPKC via nAChR/PI3K signaling in PC cells, resulting in enhancement of cellular proliferation, migration and invasion.


Subject(s)
Nicotine/pharmacology , Pancreatic Neoplasms/metabolism , Protein Kinase C/metabolism , Animals , Cell Line , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Humans , Male , Mice , Mice, Inbred BALB C , Neoplasm Invasiveness , Nicotine/toxicity , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , Smoking/adverse effects
11.
Hum Mol Genet ; 25(12): 2514-2524, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27206983

ABSTRACT

p53 has been implicated in the pathophysiology of Huntington's disease (HD). Nonetheless, the molecular mechanism of how p53 may play a unique role in the pathology remains elusive. To address this question at the molecular and cellular biology levels, we initially screened differentially expressed molecules specifically dependent on p53 in a HD animal model. Among the candidate molecules, wild-type p53-induced gene 1 (Wig1) is markedly upregulated in the cerebral cortex of HD patients. Wig1 preferentially upregulates the level of mutant Huntingtin (Htt) compared with wild-type Htt. This allele-specific characteristic of Wig1 is likely to be explained by higher affinity binding to mutant Htt transcripts than normal counterpart for the stabilization. Knockdown of Wig1 level significantly ameliorates mutant Htt-elicited cytotoxicity and aggregate formation. Together, we propose that Wig1, a key p53 downstream molecule in HD condition, play an important role in stabilizing mutant Htt mRNA and thereby accelerating HD pathology in the mHtt-p53-Wig1 positive feedback manner.


Subject(s)
Carrier Proteins/biosynthesis , Huntingtin Protein/genetics , Huntington Disease/genetics , Nuclear Proteins/biosynthesis , Tumor Suppressor Protein p53/genetics , Adult , Aged , Aged, 80 and over , Alleles , Animals , Autopsy , Carrier Proteins/genetics , Cerebral Cortex/metabolism , Cerebral Cortex/physiology , Disease Models, Animal , Female , Gene Expression Regulation , Gene Knockdown Techniques , Humans , Huntington Disease/pathology , Male , Mice , Middle Aged , Mutant Proteins/genetics , Nuclear Proteins/genetics , RNA, Messenger/genetics , RNA-Binding Proteins
12.
Neurosci Lett ; 610: 117-22, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26523792

ABSTRACT

Neuroprotection following signal transduction has been investigated recently as a strategy for Parkinson's disease (PD) therapy. While oxidative stress is important in the pathogenesis of PD, neuroprotection using antioxidants such as α-tocopherol have not been successful. δ-tocotrienol (δT3), a member of the vitamin E family, has received attention because of activities other than its antioxidative effects. In the present study, we examined the estrogen receptor-ß (ERß)-mediated neuroprotective effects of δT3 in a mouse model of PD. ERß is expressed in neuronal cells, including dopaminergic neurons in the substantia nigra. Daily forced oral administration of δT3 inhibited the loss of dopaminergic neurons in the substantia nigra. In addition, the ER inhibitor tamoxifen canceled the neuroprotective effects of δT3. Moreover, δT3 administration improved the performance of the PD mice in the wheel running activity, while tamoxifen inhibited this improved performance. These results suggest that the oral administration of δT3 may be useful in the treatment of PD patients, and ERß may be a candidate target for the neuroprotection activity of δT3.


Subject(s)
1-Methyl-4-phenylpyridinium , Estrogen Receptor beta/metabolism , Neuroprotective Agents/therapeutic use , Parkinson Disease/drug therapy , Vitamin E/analogs & derivatives , Animals , Estrogen Receptor alpha/metabolism , Female , Male , Mice, Inbred C57BL , Motor Skills , Parkinson Disease/etiology , Parkinson Disease/physiopathology , Vitamin E/therapeutic use
13.
Hum Mol Genet ; 24(4): 1092-105, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25305080

ABSTRACT

Huntington's disease (HD) is a dominantly inherited genetic disease caused by mutant huntingtin (htt) protein with expanded polyglutamine (polyQ) tracts. A neuropathological hallmark of HD is the presence of neuronal inclusions of mutant htt. p62 is an important regulatory protein in selective autophagy, a process by which aggregated proteins are degraded, and it is associated with several neurodegenerative disorders including HD. Here, we investigated the effect of p62 depletion in three HD model mice: R6/2, HD190QG and HD120QG mice. We found that loss of p62 in these models led to longer life spans and reduced nuclear inclusions, although cytoplasmic inclusions increased with polyQ length. In mouse embryonic fibroblasts (MEFs) with or without p62, mutant htt with a nuclear localization signal (NLS) showed no difference in nuclear inclusion between the two MEF types. In the case of mutant htt without NLS, however, p62 depletion increased cytoplasmic inclusions. Furthermore, to examine the effect of impaired autophagy in HD model mice, we crossed R6/2 mice with Atg5 conditional knockout mice. These mice also showed decreased nuclear inclusions and increased cytoplasmic inclusions, similar to HD mice lacking p62. These data suggest that the genetic ablation of p62 in HD model mice enhances cytoplasmic inclusion formation by interrupting autophagic clearance of polyQ inclusions. This reduces polyQ nuclear influx and paradoxically ameliorates disease phenotypes by decreasing toxic nuclear inclusions.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Huntington Disease/genetics , Intranuclear Inclusion Bodies/genetics , Phenotype , Animals , Autophagy , Corpus Striatum/metabolism , Corpus Striatum/pathology , Disease Models, Animal , Female , Hippocampus/metabolism , Hippocampus/pathology , Huntington Disease/mortality , Huntington Disease/pathology , Intracellular Space/metabolism , Longevity/genetics , Mice , Mice, Knockout , Peptides/genetics , Proteolysis
14.
Biochim Biophys Acta ; 1842(9): 1303-12, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24768803

ABSTRACT

Tocotrienols (T3s) are members of the vitamin E family, have antioxidant properties, and are promising candidates for neuroprotection in the pathogenesis of neurodegenerative disorders such as Parkinson's disease (PD). However, whether their antioxidant capacities are required for their cytoprotective activity remains unclear. In this regard, the antioxidant-independent cytoprotective activity of T3s has received considerable attention. Here, we investigated the signaling pathways that are induced during T3-dependent cytoprotection of human neuroblastoma SH-SY5Y cells, as these cells are used to model certain elements of PD. T3s were cytoprotective against 1-methyl-4-phenylpyridinium ion (MPP(+)) and other PD-related toxicities. γT3 and δT3 treatments led to marked activation of the PI3K/Akt signaling pathway. Furthermore, we identified estrogen receptor (ER) ß as an upstream mediator of PI3K/Akt signaling following γT3/δT3 stimulation. Highly purified γT3/δT3 bound to ERß directly in vitro, and knockdown of ERß in SH-SY5Y cells abrogated both γT3/δT3-dependent cytoprotection and Akt phosphorylation. Since membrane-bound ERß was important for the signal-related cytoprotective effects of γT3/δT3, we investigated receptor-mediated caveola formation as a candidate for the early events of signal transduction. Knockdown of caveolin-1 and/or caveolin-2 prevented the cytoprotective effects of γT3/δT3, but did not affect Akt phosphorylation. This finding suggests that T3s and, in particular, γT3/δT3, exhibit not only antioxidant effects but also a receptor signal-mediated protective action following ERß/PI3K/Akt signaling. Furthermore, receptor-mediated caveola formation is an important event during the early steps following T3 treatment.


Subject(s)
Apoptosis/drug effects , Cytoprotection/drug effects , Estrogen Receptor beta/metabolism , Neuroblastoma/drug therapy , Parkinson Disease/drug therapy , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Tocotrienols/pharmacology , Antioxidants/pharmacology , Blotting, Western , Cell Proliferation/drug effects , Estrogen Receptor beta/antagonists & inhibitors , Estrogen Receptor beta/genetics , Fluorescent Antibody Technique , Humans , Neuroblastoma/metabolism , Neuroblastoma/pathology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Protein Array Analysis , RNA, Small Interfering/genetics , Signal Transduction/drug effects , Tumor Cells, Cultured
15.
J Neurosci ; 33(37): 14767-77, 2013 Sep 11.
Article in English | MEDLINE | ID: mdl-24027277

ABSTRACT

The cytoplasmic regulatory protein p62 (Sequestosome 1/A170) is known to modulate various receptor-mediated intracellular signaling pathways. p62 deficiency was shown to result in mature-onset obesity in mice, but the mechanisms underlying this abnormality remained unclear. Here we report that hyperphagia due to central leptin resistance is the cause of obesity in p62(-/-) mice. We found that these mice show hyperphagia. Restriction of food to the amount eaten by wild-type mice prevented excess body weight gain and fat accumulation, suggesting that overfeeding is the primary cause of obesity in p62(-/-) mice. Brain-specific p62 deficiency caused mature-onset obesity to the same extent as in p62(-/-) mice, further supporting a neuronal mechanism as the major cause of obesity in these mice. Immunohistochemical analysis revealed that p62 is highly expressed in hypothalamic neurons, including POMC neurons in the arcuate nucleus. Central leptin resistance was observed even in young preobese p62(-/-) mice. We found a defect in intracellular distribution of the transcription factor Stat3, which is essential for the action of leptin, in p62(-/-) mice. These results indicate that brain p62 plays an important role in bodyweight control by modulating the central leptin-signaling pathway and that lack of p62 in the brain causes leptin resistance, leading to hyperphagia. Thus, p62 could be a clinical target for treating obesity and metabolic syndrome.


Subject(s)
Brain/drug effects , Hyperphagia/genetics , Hyperphagia/pathology , Leptin/pharmacology , Transcription Factors/deficiency , Animals , Body Weight/drug effects , Body Weight/genetics , Brain/cytology , Brain/metabolism , Eating/drug effects , Eating/genetics , Embryo, Mammalian , Food Deprivation , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , In Vitro Techniques , Leptin/blood , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nestin/genetics , Nestin/metabolism , Neuropeptide Y/genetics , Neuropeptide Y/pharmacology , Oxygen Consumption/genetics , Pro-Opiomelanocortin/genetics , Receptors, Leptin/genetics , Receptors, Leptin/metabolism , STAT3 Transcription Factor/metabolism , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism , Transcription Factor TFIIH
16.
PLoS One ; 8(2): e55068, 2013.
Article in English | MEDLINE | ID: mdl-23457458

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disorder characterized by the selective loss of dopaminergic neurons and the presence of Lewy bodies. Many recent studies focused on the interaction between α-synuclein (α-syn) and dopamine in the pathogenesis of PD, and fluorescent anisotropy suggested that the C-terminal region of α-syn may be a target for modification by dopamine. However, it is not well understood why PD-related pathogenesis occurs selectively in dopaminergic neurons. We investigated the interaction between dopamine and α-syn with regard to cytotoxicity. A soluble oligomer was formed by co-incubating α-syn and dopamine in vitro. To clarify the effect of dopamine on α-syn in cells, we generated PC12 cells expressing human α-syn, as well as the α-syn mutants, M116A, Y125D, M127A, S129A, and M116A/M127A, in a tetracycline-inducible manner (PC12-TetOFF-α-syn). Overexpression of wildtype α-syn in catecholaminergic PC12 cells decreased cell viability in long-term cultures, while a competitive inhibitor of tyrosine hydroxylase blocked this vulnerability, suggesting that α-syn-related cytotoxicity is associated with dopamine metabolism. The vulnerabilities of all mutant cell lines were lower than that of wildtype α-syn-expressing cells. Moreover, α-syn containing dopamine-mediated oxidized methionine (Met(O)) was detected in PC12-TetOFF-α-syn. Met(O) was lower in methionine mutant cells, especially in the M127A or M116A/M127A mutants, but also in the Y125D and S129A mutants. Co-incubation of dopamine and the 125YEMPS129 peptide enhanced the production of H2O2, which may oxidize methionine residues and convert them to Met(O). Y125- or S129-lacking peptides did not enhance the dopamine-related production of H2O2. Our results suggest that M127 is the major target for oxidative modification by dopamine, and that Y125 and S129 may act as enhancers of this modification. These results may describe a mechanism of dopaminergic neuron-specific toxicity of α-syn in the pathogenesis of PD.


Subject(s)
Dopamine/metabolism , Methionine/metabolism , alpha-Synuclein/metabolism , Amino Acid Sequence , Animals , Gene Expression , Humans , Hydrogen Peroxide/metabolism , Methionine/analogs & derivatives , Methionine/chemistry , Methionine/genetics , Methionine/toxicity , Oxidation-Reduction , PC12 Cells , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinson Disease/pathology , Point Mutation , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/toxicity , alpha-Synuclein/chemistry , alpha-Synuclein/genetics , alpha-Synuclein/toxicity
17.
J Clin Neurol ; 8(3): 230-4, 2012 Sep.
Article in English | MEDLINE | ID: mdl-23091534

ABSTRACT

BACKGROUND: Leber's hereditary optic neuropathy (LHON) is a mitochondrial disorder with optic nerve atrophy. Although there are no other associated neurological abnormalities in most cases of LHON, cases of "LHON plus" have been reported. CASE REPORT: The proband was a 37-year-old man who had visual and gait disturbances that had first appeared at 10 years of age. He showed horizontal gaze palsy, gaze-evoked nystagmus, dysarthria, and cerebellar ataxia. Brain and orbit MRI disclosed atrophy of the optic nerve and cerebellum, and degenerative changes in the bilateral inferior olivary nucleus. Mutational analyses of mitochondrial DNA identified the coexistence of heteroplasmic G11778A and homoplasmic T3394C mutations. CONCLUSIONS: These results suggest that the combination of G11778A and T3394C mutations leads to an atypical LHON phenotype.

18.
J Clin Biochem Nutr ; 51(2): 122-7, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22962530

ABSTRACT

Parkinson's disease is a major neurodegenerative disease involving the selective degeneration of dopaminergic neurons and α-synuclein containing Lewy bodies formation in the substantia nigra. Although α-synuclein is a key molecule for both dopaminergic neuron death and the formation of inclusion bodies, the mechanism of α-synuclein induction of Parkinson's disease-related pathogenesis is not understood. In the present study, we found that the interaction between dopamine and α-synuclein requires the oxidation of dopamine. Furthermore, we examined the protective effect of chlorogenic acid, a major polyphenol contained in coffee, against α-syn and dopamine-related toxicity. Chlorogenic acid inhibits several DA/α-synuclein-related phenomenon, including the oxidation of dopamine, the interaction of oxidized dopamine with α-synuclein, and the oligomerization of α-synuclein under dopamine existing conditions in vitro. Finally, we showed that the cytoprotective effect against α-synuclein-related toxicity in PC12 cells that can be controlled by the Tet-Off system. Although the induction of α-synuclein in catecholaminergic PC12 cells causes a decrease in cell viability, chlorogenic acid rescued this cytotoxicity significantly in a dose dependent manner. These results suggest that the interaction of oxidized DA with α-synuclein may be a novel therapeutic target for Parkinson's disease, and polyphenols, including chlorogenic acid, are candidates as protective and preventive agents for Parkinson's disease onset.

19.
Nat Commun ; 3: 668, 2012 Feb 07.
Article in English | MEDLINE | ID: mdl-22314364

ABSTRACT

Parkinson's disease (PD) is defined by the degeneration of nigral dopaminergic (DA) neurons and can be caused by monogenic mutations of genes such as parkin. The lack of phenotype in parkin knockout mice suggests that human nigral DA neurons have unique vulnerabilities. Here we generate induced pluripotent stem cells from normal subjects and PD patients with parkin mutations. We demonstrate that loss of parkin in human midbrain DA neurons greatly increases the transcription of monoamine oxidases and oxidative stress, significantly reduces DA uptake and increases spontaneous DA release. Lentiviral expression of parkin, but not its PD-linked mutant, rescues these phenotypes. The results suggest that parkin controls dopamine utilization in human midbrain DA neurons by enhancing the precision of DA neurotransmission and suppressing dopamine oxidation. Thus, the study provides novel targets and a physiologically relevant screening platform for disease-modifying therapies of PD.


Subject(s)
Brain/embryology , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Gene Expression Regulation , Induced Pluripotent Stem Cells/cytology , Parkinson Disease/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Binding Sites , Fibroblasts/metabolism , Humans , Mice , Mitochondria/metabolism , Models, Biological , Monoamine Oxidase/biosynthesis , Mutation , Neurons/metabolism , Oxidative Stress , Oxygen/chemistry , Phenotype , Skin/metabolism , Time Factors
20.
Yonago Acta Med ; 55(1): 11-20, 2012 Mar.
Article in English | MEDLINE | ID: mdl-24031135

ABSTRACT

The present study was performed by using selective inhibitors of caspase-8 and caspase-3 functioning upstream and downstream from mitochondria, respectively to determine whether mitochondria are involved in the mechanisms underlying production and externalization of oxidized phosphatidylserine (PSox) during Fas-mediated apoptosis. Treatment with anti-Fas antibody induced caspase-3 activation, chromatin condensation, release of cytochrome c (cyt c) from mitochondria into the cytosol as well as production of PSox and its exposure to the cell surface in Jurkat cells. Inhibition of caspase-8 by pretreatment with Z-IETD-FMK, a membrane permeable selective caspase-8 inhibitor reduced mitochondrial cyt c release, the amount of PSox not only within but also on the surface of Jurkat cells, caspase-3 activation, and apoptotic cell number after treatment with anti-Fas antibody. In contrast, Z-DEVD-FMK, a membrane permeable selective caspase-3 inhibitor was unable to inhibit cyt c release, and the amount of PSox both within and on the surface of the cells after anti-Fas antibody, although it suppressed caspase-3 activation and apoptosis. Thus, these results strongly suggest that mitochondria play an important role in production of PSox and subsequent its externalization during apoptosis.

SELECTION OF CITATIONS
SEARCH DETAIL
...