Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 62
1.
Biochim Biophys Acta Mol Cell Res ; 1869(12): 119361, 2022 Dec.
Article En | MEDLINE | ID: mdl-36162649

Phospholipase D1 (PLD1) plays a crucial role in cell differentiation of different cell types. However, the involvement of PLD1 in astrocytic differentiation remains uncertain. In the present study, we investigate the possible role of PLD1 and its product phosphatidic acid (PA) in astrocytic differentiation of hippocampal neural stem/progenitor cells (NSPCs) from hippocampi of embryonic day 16.5 rat embryos. We showed that overexpression of PLD1 increased the expression level of glial fibrillary acidic protein (GFAP), an astrocyte marker, and the number of GFAP-positive cells. Knockdown of PLD1 by transfection with Pld1 shRNA inhibited astrocytic differentiation. Moreover, PLD1 deletion (Pld1-/-) suppressed the level of GFAP in the mouse hippocampus. These results indicate that PLD1 plays a crucial role in regulating astrocytic differentiation in hippocampal NSPCs. Interestingly, PA itself was sufficient to promote astrocytic differentiation. PA-induced GFAP expression was decreased by inhibition of signal transducer and activation of transcription 3 (STAT3) using siRNA. Furthermore, PA-induced STAT3 activation and astrocytic differentiation were regulated by the focal adhesion kinase (FAK)/aurora kinase A (AURKA) pathway. Taken together, our findings suggest that PLD1 is an important modulator of astrocytic differentiation in hippocampal NSPCs via the FAK/AURKA/STAT3 signaling pathway.


Aurora Kinase A , Neural Stem Cells , Animals , Aurora Kinase A/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Hippocampus/metabolism , Mice , Phosphatidic Acids/metabolism , Phospholipase D , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Signal Transduction/physiology
2.
FASEB J ; 36(9): e22452, 2022 09.
Article En | MEDLINE | ID: mdl-35916017

House dust mite (HDM) allergens cause inflammatory responses and chronic allergic diseases such as bronchial asthma and atopic dermatitis. Here, we investigate the mechanism by which HDM induces C-C chemokine ligand 20 (CCL20) expression to promote chronic inflammation and airway remodeling in an HDM-induced bronchial asthma mouse model. We showed that HDM increased CCL20 levels via the Akt-ERK1/2-C/EBPß pathway. To investigate the role of CCL20 in chronic airway inflammation and remodeling, we made a mouse model of CCL20-induced bronchial asthma. Treatment of anti-CCL20Ab in this mouse model showed the reduced airway hyper-responsiveness and inflammatory cell infiltration into peribronchial region by neutralizing CCL20. In addition, CCL20 induced the Nod-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome activation through NLRP3 deubiquitination and transcriptional upregulation in BEAS-2B cells. As expected, anti-CCL20Ab markedly suppressed NLRP3 activation induced by CCL20. Moreover, HDM-induced CCL20 leads to epithelial-mesenchymal transition in the lung epithelium which appears to be an important regulator of airway remodeling in allergic asthma. We also found that anti-CCL20Ab attenuates airway inflammation and remodeling in an HDM-induced mouse model of bronchial asthma. Taken together, our results suggest that HDM-induced CCL20 is required for chronic inflammation that contributes airway remodeling in a mouse model of asthma.


Asthma , Pyroglyphidae , Airway Remodeling , Animals , Asthma/metabolism , Disease Models, Animal , Epithelial-Mesenchymal Transition , Inflammation/complications , Ligands , Lung/metabolism , MAP Kinase Signaling System , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Proto-Oncogene Proteins c-akt/metabolism
3.
Int J Mol Sci ; 23(12)2022 Jun 07.
Article En | MEDLINE | ID: mdl-35742844

Neuroinflammation is involved in the pathogenesis of neurodegenerative diseases due to increased levels of pro-inflammatory cytokines in the central nervous system (CNS). Chronic neuroinflammation induced by neurotoxic molecules accelerates neuronal damage. B-cell lymphoma 2 (Bcl-2) is generally accepted to be an important anti-apoptotic factor. However, the role of Bcl-2 in neuroprotection against neuroinflammation remains to be determined. The purpose of this study was to investigate the neuroprotective effect of Bcl-2 on lipopolysaccharide (LPS)-induced neuroinflammation in cortical neural stem cells (NSCs). LPS decreased mRNA and protein levels of Tuj-1, a neuron marker, and also suppressed neurite outgrowth, indicating that LPS results in inhibition of neuronal differentiation of NSCs. Furthermore, LPS treatment inhibited Bcl-2 expression during neuronal differentiation; inhibition of neuronal differentiation by LPS was rescued by Bcl-2 overexpression. LPS-induced pro-inflammatory cytokines, including interleukin (IL)-6 and tumor necrosis factor alpha (TNF-α), were decreased by Bcl-2 overexpression. Conversely, Bcl-2 siRNA increased the LPS-induced levels of IL-6 and TNF-α, and decreased neuronal differentiation of NSCs, raising the possibility that Bcl-2 mediates neuronal differentiation by inhibiting the LPS-induced inflammatory response in NSC. These results suggest that Bcl-2 has a neuroprotective effect by inhibiting the LPS-induced inflammatory response in NSCs.


Neural Stem Cells , Neuroprotective Agents , Cytokines/metabolism , Humans , Interleukin-6/metabolism , Lipopolysaccharides/metabolism , Lipopolysaccharides/toxicity , Microglia/metabolism , Neural Stem Cells/metabolism , Neuroinflammatory Diseases , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacology , Tumor Necrosis Factor-alpha/metabolism
4.
Mol Cell Toxicol ; 18(2): 267-276, 2022.
Article En | MEDLINE | ID: mdl-35069752

Background: Bacterial ghosts (BGs) are empty cell envelopes commonly generated using Gram-negative bacteria; they represent a potential platform for efficient adjuvant and vaccine delivery systems. However, the efficient production of BGs from bacteria in a short period of time is challenging. Objective: The purpose of this study was to investigate the possibility of producing BGs in the Gram-positive Bacillus subtilis using various chemicals, and the potential application of BGs as a novel immunomodulatory agent. Results: In this study, Bacillus subtilis ghosts (BSGs) were generated, for the first time to the best of our knowledge, using the minimum inhibitory concentration (MIC) of hydrochloric acid (HCl; 6.25 mg/mL), sulfuric acid (H2SO4; 3.125 mg/mL), and nitric acid (HNO3; 6.25 mg/mL). Among the BSGs generated using these chemicals, HCl-induced BSGs were completely DNA-free as confirmed by real-time polymerase chain reaction. Scanning electron microscopy showed the formation of transmembrane lysis tunnel structures in HCl-induced BSGs. Murine macrophages exposed to the HCl-induced BSGs at a concentration of 1 × 105 CFU/mL showed a cell viability of 97.8%. Additionally, HCl-induced BSGs upregulated the expression of pro-inflammatory cytokines including interleukin (IL)-1ß, tumor necrosis factor alpha, and IL-6. Furthermore, we found differences in the protein expression profiles between intact live bacteria and BSGs using two-dimensional electrophoresis coupled with peptide mass fingerprinting/matrix-assisted laser desorption/ionization-time of flight mass spectrometry analysis. Conclusion: These data suggest that the HCl-induced BSGs may be potentially safe and effective candidates for inactivated bacterial vaccines and/or immunostimulants. Supplementary Information: The online version contains supplementary material available at 10.1007/s13273-022-00221-5.

5.
FASEB J ; 34(11): 14407-14423, 2020 11.
Article En | MEDLINE | ID: mdl-33000885

Decidualization of the endometrial stroma is an essential differentiation process for embryo implantation and maintenance of pregnancy. We previously reported that protein phosphatase 2A (PP2A) acts as a key mediator during cAMP-induced decidualization of human endometrial stromal cells (hESCs). However, the mechanism underlying its activation has remained obscure in hESCs. In the present study, we aimed to reveal the mechanism that induces the nitration of PP2A catalytic subunit (PP2Ac) during cAMP-induced decidualization of hESCs. First, cAMP-induced PP2Ac nitration was significantly repressed using L-NAME, an inhibitor of nitric oxide synthase (NOS). Among several NOS isoforms, only inducible NOS (iNOS) was highly expressed in hESCs, indicating that iNOS directly induces the nitration of PP2Ac. Second, cAMP-induced iNOS expression and PP2Ac nitration were decreased by treatment with TSA, an inhibitor of histone deacetylase 5 (HDAC5). cAMP-induced phosphorylation of CaMKII and HDAC5 was suppressed by treatment with U73122 (an inhibitor of phospholipase C) or transfection of PLCε siRNA. Finally, small G protein Rap1 and its guanine nucleotide exchange factor Epac1 were found to be involved in cAMP-induced PP2A activation. Taken together, our results suggest that PP2Ac nitration during cAMP-induced decidualization of hESCs is induced through the Epac1-Rap1-PLCε-CaMKII-HDAC5-iNOS signaling pathway.


Decidua/metabolism , Nitric Oxide/metabolism , Protein Phosphatase 2/metabolism , Signal Transduction , Adult , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cell Line , Cells, Cultured , Decidua/cytology , Female , Guanine Nucleotide Exchange Factors/metabolism , Histone Deacetylases/metabolism , Humans , Middle Aged , Nitric Oxide Synthase Type II/metabolism , Phosphoinositide Phospholipase C/metabolism , Shelterin Complex , Stromal Cells/cytology , Stromal Cells/metabolism , Telomere-Binding Proteins/metabolism
6.
Cells ; 9(1)2020 01 17.
Article En | MEDLINE | ID: mdl-31963554

1α,25-dihydroxyvitamin D3 (1,25D3), the most popular drug for osteoporosis treatment, drives osteoblast differentiation and bone mineralization. Wnt/ß-catenin signaling is involved in commitment and differentiation of osteoblasts, but the role of the Dickkopf-related protein 1 (DKK1), a Wnt antagonist, in osteoblasts remains unknown. Here, we demonstrate the molecular mechanism of DKK1 induction by 1,25D3 and its physiological role during osteoblast differentiation. 1,25D3 markedly promoted the expression of both CCAAT/enhancer binding protein beta (C/EBPß) and DKK1 at day 7 during osteoblast differentiation. Interestingly, mRNA and protein levels of C/EBPß and DKK1 in osteoblasts were elevated by 1,25D3. We also found that C/EBPß, in response to 1,25D3, directly binds to the human DKK1 promoter. Knockdown of C/EBPß downregulated the expression of DKK1 in osteoblasts, which was partially reversed by 1,25D3. In contrast, overexpression of C/EBPß upregulated DKK1 expression in osteoblasts, which was enhanced by 1,25D3. Furthermore, 1,25D3 treatment in osteoblasts stimulated secretion of DKK1 protein within the endoplasmic reticulum to extracellular. Intriguingly, blocking DKK1 attenuated calcified nodule formation in mineralized osteoblasts, but not ALP activity or collagen synthesis. Taken together, these observations suggest that 1,25D3 promotes the mineralization of osteoblasts through activation of DKK1 followed by an increase of C/EBPß.


Calcification, Physiologic/drug effects , Calcitriol/pharmacology , Intercellular Signaling Peptides and Proteins/metabolism , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteogenesis/drug effects , Alkaline Phosphatase/metabolism , CCAAT-Binding Factor/genetics , CCAAT-Binding Factor/metabolism , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , Calcification, Physiologic/genetics , Cell Line, Tumor , Chromatin Immunoprecipitation , Collagen/metabolism , Endoplasmic Reticulum/metabolism , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Gene Knockdown Techniques , Humans , Intercellular Signaling Peptides and Proteins/genetics , Oligonucleotide Array Sequence Analysis , Osteoblasts/enzymology , Osteogenesis/genetics
7.
Cell Mol Neurobiol ; 40(1): 153-166, 2020 Jan.
Article En | MEDLINE | ID: mdl-31493044

Bcl-2 is overexpressed in the nervous system during neural development and plays an important role in modulating cell survival. In addition to its anti-apoptotic function, it has been suggested previously that Bcl-2 might act as a mediator of neuronal differentiation. However, the mechanism by which Bcl-2 might influence neurogenesis is not sufficiently understood. In this study, we aimed to determine the non-apoptotic functions of Bcl-2 during neuronal differentiation. First, we used microarrays to analyze the whole-genome expression patterns of rat neural stem cells overexpressing Bcl-2 and found that Bcl-2 overexpression induced the expression of various neurogenic genes. Moreover, Bcl-2 overexpression increased the neurite length as well as expression of Bmp4, Tbx3, and proneural basic helix-loop-helix genes, such as NeuroD1, NeuroD2, and Mash1, in H19-7 rat hippocampal precursor cells. To determine the hierarchy of these molecules, we selectively depleted Bmp4, Tbx3, and NeuroD1 in Bcl-2-overexpressing cells. Bmp4 depletion suppressed the upregulation of Tbx3 and NeuroD1 as well as neurite outgrowth, which was induced by Bcl-2 overexpression. Although Tbx3 knockdown repressed Bcl-2-mediated neurite elaboration and downregulated NeuroD1 expression, it did not affect Bcl-2-induced Bmp4 expression. While the depletion of NeuroD1 had no effect on the expression of Bcl-2, Bmp4, or Tbx3, Bcl-2-mediated neurite outgrowth was suppressed. Taken together, these results demonstrate that Bcl-2 regulates neurite outgrowth through the Bmp4/Tbx3/NeuroD1 cascade in H19-7 cells, indicating that Bcl-2 may have a direct role in neuronal development in addition to its well-known anti-apoptotic function in response to environmental insults.


Basic Helix-Loop-Helix Transcription Factors/metabolism , Bone Morphogenetic Protein 4/metabolism , Neurites/metabolism , Neuronal Outgrowth , Proto-Oncogene Proteins c-bcl-2/metabolism , T-Box Domain Proteins/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation , Gene Expression Regulation , Hippocampus/cytology , Neural Stem Cells/metabolism , Neuronal Outgrowth/genetics , Neuropeptides/genetics , Neuropeptides/metabolism , Rats, Sprague-Dawley , Signal Transduction , Smad Proteins/metabolism , T-Box Domain Proteins/genetics
8.
Cell Mol Life Sci ; 76(22): 4569-4580, 2019 Nov.
Article En | MEDLINE | ID: mdl-31486848

Hippocalcin (HPCA) is a neuron-specific calcium-binding protein predominantly expressed in the nervous system. In the present study, we demonstrate that HPCA regulates neuronal differentiation in SH-SY5Y cells. We observed that the expression level of HPCA was increased during neuronal differentiation. Depletion of HPCA inhibited both neurite outgrowth and synaptophysin (SYP) expression, whereas overexpression of HPCA enhanced neuronal differentiation. Interestingly, we also found that the expression of HPCA mRNA was modulated by miR-24-3p. Using a dual-luciferase assay, we showed that co-transfection of a plasmid containing the miR-24-3p binding site from the 3'-untranslated region (3'UTR) of the HPCA gene and an miR-24-3p mimic effectively reduced luminescence activity. This effect was abolished when miR-24-3p seed sequences in the 3'UTR of the HPCA gene were mutated. miR-24-3p expression was decreased during differentiation, suggesting that the decreased expression level of miR-24-3p might have upregulated mRNA expression of HPCA. As expected, upregulation of miR-24-3p by an miRNA mimic led to reduced HPCA expression, accompanied by diminished neuronal differentiation. In contrast, downregulation of miR-24-3p by an antisense inhibitor promoted neurite outgrowth as well as levels of SYP expression. Taken together, these results suggest that miR-24-3p is an important miRNA that regulates neuronal differentiation by controlling HPCA expression.


Hippocalcin/genetics , MicroRNAs/genetics , Neurons/physiology , 3' Untranslated Regions/genetics , Binding Sites/genetics , Cell Differentiation , Cell Line, Tumor , Down-Regulation/genetics , HeLa Cells , Humans , Neuronal Outgrowth/genetics , RNA, Messenger/genetics , Up-Regulation/genetics
9.
BMB Rep ; 52(6): 391-396, 2019 Jun.
Article En | MEDLINE | ID: mdl-30355436

Receptor activator of nuclear factor kappa B ligand (RANKL) expression in osteoblasts is regulated by 1,25-dihydroxyvitamin D3 (1,25D3). CCAAT/enhancer-binding protein beta (C/EBPß) has been proposed to function as a transcription factor and upregulate RANKL expression, but it is still uncertain how C/EBPß is involved in 1,25D3-induced RANKL expression of osteoblasts. 1,25D3 stimulation increased the expression of RANKL and C/EPBß genes in osteoblasts and enhanced phosphorylation and stability of these proteins. Moreover, induction of RANKL expression by 1,25D3 in osteoblasts was downregulated upon knockdown of C/EBPß. In contrast, C/EBPß overexpression directly upregulated RANKL promoter activity and exhibited a synergistic effect on 1,25D3-induced RANKL expression. In particular, 1,25D3 treatment of osteoblasts increased C/EBPß protein binding to the RANKL promoter. In conclusion, C/EBPß is required for induction of RANKL by 1,25D3. [BMB Reports 2019; 52(6): 391-396].


CCAAT-Enhancer-Binding Protein-beta/metabolism , Calcitriol/pharmacology , Osteoblasts/drug effects , Osteoblasts/metabolism , RANK Ligand/metabolism , Binding Sites , Cell Differentiation/drug effects , Cell Line , Cell Nucleus/metabolism , Humans , NF-kappa B/metabolism , Osteoblasts/cytology , Phosphorylation/drug effects , Promoter Regions, Genetic , Protein Binding , Protein Stability , RANK Ligand/biosynthesis , RANK Ligand/genetics , RNA, Messenger/metabolism
10.
Exp Mol Med ; 50(10): 1-11, 2018 10 30.
Article En | MEDLINE | ID: mdl-30374127

Human endometrium decidualization, a differentiation process involving biochemical and morphological changes, is a prerequisite for embryo implantation and successful pregnancy. Here, we show that the mammalian target of rapamycin (mTOR) is a crucial regulator of 8-bromoadenosine 3',5'-cyclic monophosphate (8-Br-cAMP)-induced decidualization in human endometrial stromal cells. The level of mSin1 in mTOR complex 2 (mTORC2) and DEPTOR in mTOR complex 1 (mTORC1) decreases during 8-Br-cAMP-induced decidualization, resulting in decreased mTORC2 activity and increased mTORC1 activity. Notably, DEPTOR displacement increases the association between raptor and insulin receptor substrate-1 (IRS-1), facilitating IRS-1 phosphorylation at serine 636/639. Finally, both S473 and T308 phosphorylation of Akt are reduced during decidualization, followed by a decrease in forkhead box O1 (FOXO1) phosphorylation and an increase in the mRNA levels of the decidualization markers prolactin (PRL) and insulin-like growth factor-binding protein-1 (IGFBP-1). Taken together, our findings reveal a critical role for mTOR in decidualization, involving the differential regulation of mTORC1 and mTORC2.


8-Bromo Cyclic Adenosine Monophosphate/metabolism , Decidua/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Adult , Female , Forkhead Box Protein O1/metabolism , Humans , Insulin Receptor Substrate Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Middle Aged , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism
11.
Mol Brain ; 11(1): 39, 2018 07 04.
Article En | MEDLINE | ID: mdl-29973222

Pro-inflammatory cytokine interleukin-1 beta (IL-1ß) is a key mediator of inflammation and stress in the central nervous system (CNS), and is highly expressed in the developing brain. In this study, we investigated the possible role of IL-1ß in neuronal differentiation of cortical neural precursor cells (NPCs). We showed that stimulation with IL-1ß increased expression levels of neurotrophin-3 (NT3) and neurogenin 1 (Ngn1) and promoted neurite outgrowth. We also found that IL-1ß increased mRNA and protein levels of Wnt5a. Knockdown of Wnt5a by transfection with Wnt5a siRNA inhibited IL-1ß-induced neuronal differentiation. Moreover, IL-1ß-induced Wnt5a expression was regulated by nuclear factor kappa B (NF-κB) activation, which is involved in IL-1ß-mediated neuronal differentiation. To examine the role of Wnt5a in neuronal differentiation of NPCs, we exogenously added Wnt5a. We found that exogenous Wnt5a promotes neuronal differentiation, and activates the RhoA/Rho-associated kinase (ROCK)/c-jun N-terminal kinase (JNK) pathway. In addition, Wnt5a-induced neuronal differentiation was blocked by RhoA siRNA, as well as by a specific Rho-kinase inhibitor (Y27632) or a SAPK/JNK inhibitor (SP600125). Furthermore, treatment with RhoA siRNA, Y27632, or SP600125 suppressed the IL-1ß-induced neuronal differentiation. Therefore, these results suggest that the sequential Wnt5a/RhoA/ROCK/JNK pathway is involved in IL-1ß-induced neuronal differentiation of NPCs.


Cell Differentiation , Cerebral Cortex/cytology , Interleukin-1beta/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , Neural Stem Cells/enzymology , Neurons/cytology , Wnt-5a Protein/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Cell Differentiation/drug effects , Enzyme Activation/drug effects , NF-kappa B/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neurites/drug effects , Neurites/metabolism , Rats , Signal Transduction/drug effects , rho-Associated Kinases/metabolism
12.
Exp Mol Med ; 50(5): 1-11, 2018 05 01.
Article En | MEDLINE | ID: mdl-29717122

Asthma is a chronic lung disease that causes airflow obstruction due to airway inflammation. However, its therapeutics remain inadequate. We previously reported that phospholipase D1 (PLD1) is a key enzyme involved in the production of pro-inflammatory cytokines in airway inflammation induced by the house dust mite allergen Dermatophagoides farinae 2 (Der f 2). We also revealed that PLD1 is specifically inactivated by AP180 (assembly protein, 180 kDa) and identified the PLD1-specific binding motif (TVTSP) of AP180. Therefore, the aims of this study were to develop a novel anti-asthmatic agent that could suppress airway inflammation by inhibiting PLD1 and examine its acute and chronic toxicity. We designed TAT-TVTSP, a PLD1-inhibitory peptide fused with a cell-penetrating peptide (CPP) delivery system. TAT-TVTSP was efficiently delivered to bronchial epithelial cells and significantly reduced Der f 2-induced PLD activation and Interleukin 13 (IL-13) production. Intranasally administered TAT-TVTSP was also efficiently transferred to airway tissues and ameliorated airway inflammation in a Der f 2-induced allergic asthma mouse model. Moreover, we investigated the safety of TAT-TVTSP as a therapeutic agent through single- and repeated-dose toxicity studies in a mouse model. Taken together, these results indicated that a PLD1-inhibitory peptide fused with a cell-penetrating peptide may be useful for treating allergic inflammatory asthma induced by house dust mites (HDMs).


Anti-Asthmatic Agents/therapeutic use , Antigens, Dermatophagoides/immunology , Asthma/drug therapy , Cell-Penetrating Peptides/therapeutic use , Dermatophagoides farinae/immunology , Inflammation/drug therapy , Lung/pathology , Phospholipase D/antagonists & inhibitors , Administration, Intranasal , Animals , Anti-Asthmatic Agents/toxicity , Asthma/immunology , Asthma/parasitology , Cell Line , Cell-Penetrating Peptides/administration & dosage , Cell-Penetrating Peptides/toxicity , Dermatophagoides farinae/drug effects , Disease Models, Animal , Enzyme Activation/drug effects , Female , Humans , Inflammation/parasitology , Interleukin-13/metabolism , Male , Mice, Inbred BALB C , Phospholipase D/metabolism
13.
J Mol Neurosci ; 64(3): 333-340, 2018 Mar.
Article En | MEDLINE | ID: mdl-29478139

Phospholipase D1 (PLD1) is generally accepted as playing an important role in the regulation of multiple cell functions, such as cell growth, survival, differentiation, membrane trafficking, and cytoskeletal organization. Recent findings suggest that PLD1 also plays an important role in the regulation of neuronal differentiation of neuronal cells. Moreover, PLD1-mediated signaling molecules dynamically regulate the neuronal differentiation of neural stem cells (NSCs). Rho family GTPases and Ca2+-dependent signaling, in particular, are closely involved in PLD1-mediated neuronal differentiation of NSCs. Moreover, PLD1 has a significant effect on the neurogenesis of NSCs via the regulation of SHP-1/STAT3 activation. Therefore, PLD1 has now attracted significant attention as an essential neuronal signaling molecule in the nervous system. In the current review, we summarize recent findings on the regulation of PLD1 in neuronal differentiation and discuss the potential role of PLD1 in the neurogenesis of NSCs.


Neural Stem Cells/metabolism , Neurogenesis , Phospholipase D/metabolism , Animals , Humans , Neural Stem Cells/cytology , Signal Transduction
14.
Mol Cells ; 40(11): 805-813, 2017 Nov 30.
Article En | MEDLINE | ID: mdl-29145720

The role of phospholipase D (PLD) in cancer development and management has been a major area of interest for researchers. The purpose of this mini-review is to explore PLD and its distinct role during chemotherapy including anti-apoptotic function. PLD is an enzyme that belongs to the phospholipase super family and is found in a broad range of organisms such as viruses, yeast, bacteria, animals, and plants. The function and activity of PLD are widely dependent on and regulated by neurotransmitters, hormones, small monomeric GTPases, and lipids. A growing body of research has shown that PLD activity is significantly increased in cancer tissues and cells, indicating that it plays a critical role in signal transduction, cell proliferation, and anti-apoptotic processes. In addition, recent studies show that PLD is a downstream transcriptional target of proteins that contribute to inflammation and carcinogenesis such as Sp1, NFκB, TCF4, ATF-2, NFATc2, and EWS-Fli. Thus, compounds that inhibit expression or activity of PLD in cells can be potentially useful in reducing inflammation and sensitizing resistant cancers during chemotherapy.


Neoplasms/enzymology , Phospholipase D/metabolism , Up-Regulation , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Neoplasms/drug therapy , Phosphatidylcholines/chemistry , Phosphatidylcholines/metabolism , Signal Transduction , Up-Regulation/drug effects
15.
Stem Cell Reports ; 8(1): 95-111, 2017 01 10.
Article En | MEDLINE | ID: mdl-28017654

Hippocalcin (HPCA) is a calcium-binding protein that is restricted to nervous tissue and contributes to neuronal activity. Here we report that, in addition to inducing neurogenesis, HPCA inhibits astrocytic differentiation of neural stem cells. It promotes neurogenesis by regulating protein kinase Cα (PKCα) activation by translocating to the membrane and binding to phosphoinositide-dependent protein kinase 1 (PDK1), which induces PKCα phosphorylation. We also found that phospholipase D1 (PLD1) is implicated in the HPCA-mediated neurogenesis pathway; this enzyme promotes dephosphorylation of signal transducer and activator of transcription 3 (STAT3[Y705]), which is necessary for astrocytic differentiation. Moreover, we found that the SH2-domain-containing tyrosine phosphatase 1 (SHP-1) acts upstream of STAT3. Importantly, this SHP-1-dependent STAT3-inhibitory mechanism is closely involved in neurogenesis and suppression of gliogenesis by HPCA. Taken together, these observations suggest that HPCA promotes neuronal differentiation through activation of the PKCα/PLD1 cascade followed by activation of SHP-1, which dephosphorylates STAT3(Y705), leading to inhibition of astrocytic differentiation.


Astrocytes/cytology , Astrocytes/metabolism , Cell Differentiation/genetics , Hippocalcin/genetics , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Animals , Calcium/metabolism , Gene Expression , Hippocalcin/metabolism , Models, Biological , Neurogenesis , Neurons/cytology , Neurons/metabolism , Phospholipase D/metabolism , Phosphorylation , Protein Kinase C-alpha/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Rats , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , Tubulin/genetics
16.
Mol Neurobiol ; 54(7): 5646-5656, 2017 09.
Article En | MEDLINE | ID: mdl-27624386

We investigated the role of neuronal nitric oxide synthase (nNOS) in the neuronal differentiation of neural progenitor cells (NPCs) from hippocampi of E16.5 rat embryos. The production of nitric oxide (NO) and nNOS expression increased markedly during neuronal differentiation as did the expression of neurotrophin-3 (NT3), neurotrophin-4/5 (NT 4/5), and synapsin I. nNOS siRNA or the nNOS inhibitor, 7-nitroindazole (7-NI), decreased expression of the neurotrophins and synapsin I, and suppressed neurite outgrowth. These results suggest that nNOS plays a critical role in neuronal differentiation of hippocampal NPCs. nNOS-mediated neuronal differentiation is controlled by calcineurin since cyclosporin A (CsA), a calcineurin inhibitor, decreased nNOS activation and NO production, and inhibited neurite outgrowth. We found that inactivation of glycogen synthase kinase-3 beta (GSK3ß) resulting from activation of protein kinase C alpha (PKCα) is involved in the nNOS-mediated neuronal differentiation. Moreover, lithium chloride (LiCl), a GSK3ß inhibitor, increased neuronal differentiation by inhibiting the proliferation of NPCs. Taken together, these results suggest that neuronal differentiation is dependent on calcineurin-mediated activation of nNOS; this induces PKCα-dependent inactivation of GSK3ß, which leads to inhibition of the proliferation of hippocampal NPCs.


Glycogen Synthase Kinase 3 beta/metabolism , Hippocampus/metabolism , Nitric Oxide Synthase Type I/metabolism , Protein Kinase C-alpha/metabolism , Stem Cells/cytology , Animals , Cell Differentiation/drug effects , Enzyme Inhibitors/pharmacology , Neural Stem Cells/cytology , Neurons/metabolism , Rats, Sprague-Dawley , Signal Transduction/drug effects , Stem Cells/drug effects
17.
Front Mol Neurosci ; 9: 110, 2016.
Article En | MEDLINE | ID: mdl-27840601

Hippocalcin (Hpca) is a neuronal calcium sensor protein expressed in the mammalian brain. However, its function in neural stem/precursor cells has not yet been studied. Here, we clarify the function of Hpca in astrocytic differentiation in hippocampal neural precursor cells (HNPCs). When we overexpressed Hpca in HNPCs in the presence or absence of bFGF, expression levels of nerve-growth factors such as neurotrophin-3 (NT-3), neurotrophin-4/5 (NT-4/5), and brain-derived neurotrophic factor (BDNF), together with the proneural basic helix loop helix (bHLH) transcription factors NeuroD and neurogenin 1 (Ngn1), increased significantly. In addition, there was an increase in the number of cells expressing glial fibrillary acidic protein (GFAP), an astrocyte marker, and in branch outgrowth, indicating astrocytic differentiation of the HNPCs. Downregulation of Hpca by transfection with Hpca siRNA reduced expression of NT-3, NT-4/5, BDNF, NeuroD, and Ngn1 as well as levels of GFAP protein. Furthermore, overexpression of Hpca increased the phosphorylation of STAT3 (Ser727), and this effect was abolished by treatment with a STAT3 inhibitor (S3I-201), suggesting that STAT3 (Ser727) activation is involved in Hpca-mediated astrocytic differentiation. As expected, treatment with Stat3 siRNA or STAT3 inhibitor caused a complete inhibition of astrogliogenesis induced by Hpca overexpression. Taken together, this is the first report to show that Hpca, acting through Stat3, has an important role in the expression of neurotrophins and proneural bHLH transcription factors, and that it is an essential regulator of astrocytic differentiation and branch outgrowth in HNPCs.

18.
FEBS J ; 283(22): 4163-4175, 2016 11.
Article En | MEDLINE | ID: mdl-27696687

Decidualization of human endometrial stromal cells (hESCs) is crucial for successful uterine implantation and maintaining pregnancy. We previously reported that phospholipase D1 (PLD1) is required for cAMP-induced decidualization of hESCs. However, the mechanism by which phosphatidic acid (PA), the product of PLD1 action, might regulate decidualization is not known. We confirmed that PA induced decidualization of hESCs by observing morphological changes and measuring increased levels of decidualization markers such as IGFBP1 and prolactin transcripts (P < 0.05). Treatment with PA reduced phosphorylation of Akt and consequently that of FoxO1, which led to the increased IGFBP1 and prolactin mRNA levels (P < 0.05). Conversely, PLD1 knockdown rescued Akt phosphorylation. Binding of PP2A and Akt increased in response to cAMP or PA, suggesting that their binding is directly responsible for the inactivation of Akt during decidualization. Consistent with this observation, treatment with okadaic acid, a PP2A inhibitor, also inhibited cAMP-induced decidualization by blocking Akt dephosphorylation.


Decidua/drug effects , Endometrium/cytology , Phosphatidic Acids/pharmacology , Protein Phosphatase 2/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Stromal Cells/drug effects , Adult , Blotting, Western , Cells, Cultured , Cyclic AMP/pharmacology , Decidua/metabolism , Female , Forkhead Box Protein O1/metabolism , Gene Expression/drug effects , Humans , Insulin-Like Growth Factor Binding Protein 1/genetics , Middle Aged , Okadaic Acid/pharmacology , Phospholipase D/genetics , Phospholipase D/metabolism , Phosphorylation/drug effects , Prolactin/genetics , Protein Binding/drug effects , Protein Phosphatase 2/antagonists & inhibitors , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Stromal Cells/metabolism
19.
Sci Rep ; 6: 31984, 2016 08 18.
Article En | MEDLINE | ID: mdl-27534818

Single-Si-nanowire (NW)-based DNA sensors have been recently developed, but their sensitivity is very limited because of high noise signals, originating from small source-drain current of the single Si NW. Here, we demonstrate that chemical-vapor-deposition-grown large-scale graphene/surface-modified vertical-Si-NW-arrays junctions can be utilized as diode-type biosensors for highly-sensitive and -selective detection of specific oligonucleotides. For this, a twenty-seven-base-long synthetic oligonucleotide, which is a fragment of human DENND2D promoter sequence, is first decorated as a probe on the surface of vertical Si-NW arrays, and then the complementary oligonucleotide is hybridized to the probe. This hybridization gives rise to a doping effect on the surface of Si NWs, resulting in the increase of the current in the biosensor. The current of the biosensor increases from 19 to 120% as the concentration of the target DNA varies from 0.1 to 500 nM. In contrast, such biosensing does not come into play by the use of the oligonucleotide with incompatible or mismatched sequences. Similar results are observed from photoluminescence microscopic images and spectra. The biosensors show very-uniform current changes with standard deviations ranging ~1 to ~10% by ten-times endurance tests. These results are very promising for their applications in accurate, selective, and stable biosensing.


Biosensing Techniques/instrumentation , Nanowires/chemistry , Nucleic Acid Hybridization/methods , DNA Probes , Graphite/chemistry , Guanine Nucleotide Exchange Factors/genetics , Humans , Nanowires/ultrastructure , Silicon/chemistry , Tumor Suppressor Proteins/genetics
20.
J Toxicol Sci ; 41(1): 77-89, 2016 Feb.
Article En | MEDLINE | ID: mdl-26763395

Cigarette smoking is known to be associated with various kinds of diseases, including atherosclerotic cardiovascular disease, cancer, and chronic obstructive pulmonary disease (COPD). Many of the diseases associated with cigarette smoking are also associated with changes in interleukin-6 (IL-6) expression. In this study, we investigated the role of phospholipase D1 (PLD1) in IL-6 expression induced by cigarette smoke extract (CSE). Treatment with CSE increased PLD1 and IL-6 expressions in human bronchial epithelial (BEAS-2B) cells. In addition, CSE treatment activated PLC, PKC, and MAPK pathway through the Gi protein-coupled receptor. Pertussis toxin (PTX, Gi protein-coupled receptor inhibitor), PAO (PLC inhibitor), Go6976 (PKC inhibitor) and SB203580 (p38MAPK inhibitor) decreased CSE-induced PLD1 expression. The results show that Gi protein, PLC, PKC, and p38MAPK act as upstream regulators of PLD1 in CSE-treated BEAS-2B cells. Moreover, PLD1 siRNA transfection decreased CSE-induced ATF2 phosphorylation and IL-6 expression. In addition, inhibitors of Gi protein, PLC, PKC, and p38MAPK, and ATF2 siRNA transfection decreased CSE-induced IL-6 expression, suggesting that CSE-induced IL-6 expression is regulated via Gi protein/PLC/PKC/p38MAPK/PLD1/ATF2 pathway. Taken together, the results suggest that PLD1 is an important regulator of IL-6 expression induced by CSE in BEAS-2B cells.


Bronchi/cytology , Epithelial Cells/metabolism , Gene Expression/drug effects , Interleukin-6/metabolism , Phospholipase D/physiology , Smoke/adverse effects , Smoking/adverse effects , Tobacco Products/adverse effects , Animals , Cattle , Cell Line , Cell Survival/drug effects , Humans , Phospholipase D/metabolism , Reverse Transcriptase Polymerase Chain Reaction
...