Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 39
1.
Chemosphere ; 359: 142120, 2024 Jul.
Article En | MEDLINE | ID: mdl-38670503

Volatile organic compounds (VOCs) are crucial air pollutants in indoor environments, emitted from building materials, furniture, consumer products, cleaning products, smoking, fuel combustion, cooking, and other sources. VOCs are also emitted from human beings via breath and whole-body skin. Some VOCs cause dermal/ocular irritation as well as gastrointestinal, neurological, cardiovascular, and/or carcinogenic damage to human health. Because people spend most of their time indoors, active control of indoor VOCs has garnered attention. Phytoremediation and microbial remediation, based on plant and microorganism activities, are deemed sustainable, cost-effective, and public-friendly technologies for mitigating indoor VOCs. This study presents the major sources of VOCs in indoor environments and their compositions. Various herbaceous and woody plants used to mitigate indoor VOCs are summarized and their VOCs removal performance is compared. Moreover, this paper reviews the current state of active phytoremediation and microbial remediation for the control of indoor VOCs, and discusses future directions.


Air Pollutants , Air Pollution, Indoor , Biodegradation, Environmental , Volatile Organic Compounds , Volatile Organic Compounds/analysis , Volatile Organic Compounds/metabolism , Air Pollution, Indoor/analysis , Air Pollutants/analysis , Humans , Plants/metabolism
2.
Environ Pollut ; 348: 123834, 2024 May 01.
Article En | MEDLINE | ID: mdl-38518971

Particulate matter with an aerodynamic diameter of 2.5 µm or less (PM2.5) harbors a diverse microbial community. To assess the ecological dynamics and potential health risks associated with airborne microorganisms, it is crucial to understand the factors influencing microbial communities within PM2.5. This study investigated the influence of abiotic parameters, including air pollutants, PM2.5 chemical composition (water-soluble ions and organics), and meteorological variables, on microbial communities in PM2.5 samples collected in Seoul during the spring season. Results revealed a significant correlation between air pollutants and water-soluble ions of PM2.5 with microbial α-diversity indices. Additionally, air pollutants exerted a dominant effect on the microbial community structure, with stronger correlations observed for fungi than bacteria, whereas meteorological variables including temperature, pressure, wind speed, and humidity exerted a limited influence on fungal α-diversity. Furthermore, the results revealed specific water-soluble ions, such as SO42-, NO3-, and NH4+, as important factors influencing fungal α-diversity, whereas K+ negatively correlated with both microbial α-diversity. Moreover, PM2.5 microbial diversity was affected by organic compounds within PM2.5, with fatty acids exhibited a positive correlation with fungal diversity, while dicarboxylic acids exhibited a negative correlation with it. Furthermore, network analysis revealed direct links between air pollutants and dominant bacterial and fungal genera. The air pollutants exhibited a strong correlation with bacterial genera, such as Arthrospira and Clostridium, and fungal genera, including Aureobasidium and Cladosporium. These results will contribute to our understanding of the ecological dynamics of airborne microorganisms and provide insights into the potential risks associated with PM2.5 exposure.


Air Pollutants , Microbiota , Seasons , Seoul , Environmental Monitoring , Air Microbiology , Particulate Matter/analysis , Air Pollutants/analysis , Bacteria , Ions/analysis , Water/analysis
3.
Article En | MEDLINE | ID: mdl-34304695

Control over particulate matter (PM) emission from grilling is required for improving public health and air quality. The performance of mirror-symmetrical multi-compartment scrubbers with an upflow (U-type) and downflow baffle (D-type) configuration was evaluated for PM emission control from grilling at a flow rate of 30 m3 min-1. The PM removal efficiency of the U-type scrubber was the highest when the water level was 8 cm (95.6%), and the pressure drops recorded at the water levels of 6, 8 and 10 cm were 103, 122 and 153 mmH2O, respectively. Although PM removal efficiency of the D-type scrubber was over 91.0% at the water levels of 8, 10 and 12 cm, the pressure drops were 124, 142 and 185 mmH2O, respectively. A comprehensive evaluation of the water volume, pressure drop and PM removal performance, as well as device size, revealed that the U-type scrubber with a PM removal efficiency of 92% or higher and a pressure drop of 122 mmH2O or lower at the water levels of 6-8 cm was more economical for removing PM from grilling gas than the D-type scrubber.


Air Pollutants , Air Pollution , Air Pollutants/analysis , Particulate Matter/analysis
5.
J Perianesth Nurs ; 35(3): 298-306, 2020 Jun.
Article En | MEDLINE | ID: mdl-32005604

PURPOSE: To evaluate the effect of using the evidence-based hypothermia guideline developed by the American Society of PeriAnesthesia Nurses on body temperature, shivering, thermal discomfort and comfort, and incidence of hypothermia. DESIGN: Randomized controlled trial with 54 patients undergoing upper arm surgery with general anesthesia in the Republic of Korea. METHODS: Participants in the experimental group received a head turban, sleeping socks, a heated blanket, a Bair Hugger for forced-air warming, and a Mega Acer kit (ACE Medical Co, Seoul, Korea) for warming intravenous fluid. Participants in the control group received a typical hospital cotton blanket. FINDINGS: Body temperature, shivering, thermal discomfort, and thermal comfort showed significant improvements in the experimental group compared with the control group. CONCLUSIONS: The American Society of PeriAnesthesia Nurses guideline is applicable for preventing hypothermia under general anesthesia, which, in turn, aids in patient recovery through the suppression of various hypothermia-related complications.


Anesthesia, General , Hypothermia , Humans , Hypothermia/prevention & control , Republic of Korea , Silicate Cement , Upper Extremity
6.
Oral Dis ; 24(8): 1591-1598, 2018 Nov.
Article En | MEDLINE | ID: mdl-29920852

OBJECTIVES: This placebo-controlled randomized double-blinded clinical study assessed the analgesic efficacy of intramuscular morphine in TMD patients with myofascial pain and sex-dependent responses of the morphine treatment. SUBJECTS AND METHODS: Men and women with TMD were treated with morphine (1.5 or 5 mg), lidocaine, or saline in the masseter muscle. VAS of pain intensity, PPT, and PPtol were compared between treatment groups and gender. An additional group was treated with morphine in the trapezius muscle to evaluate the systemic effect of morphine that may reduce pain in the masseter muscle. RESULTS: There was a significant difference in VAS scores between the morphine 5 mg group and the saline group favoring morphine, but not between the morphine 5 mg and lidocaine. Morphine 1.5 and 5 mg treatments led to consistently and significantly elevated PPT and PPtol measures in men, but not in women. Morphine administered in the trapezius muscle did not affect the outcome measures. CONCLUSION: A single dose intramuscular morphine produced analgesic effects up to 48 hr in patients with myofascial pain. Intramuscular morphine elevated mechanical pain threshold and tolerance in the masseter only in male patients, suggesting sex differences in local morphine effects. No systemic effect of intramuscular morphine was detected.


Analgesics, Opioid/therapeutic use , Facial Pain/drug therapy , Morphine/therapeutic use , Pain Threshold/drug effects , Temporomandibular Joint Disorders/drug therapy , Adult , Analgesics, Opioid/administration & dosage , Anesthetics, Local/therapeutic use , Double-Blind Method , Facial Pain/etiology , Female , Humans , Injections, Intramuscular , Lidocaine/therapeutic use , Male , Masseter Muscle , Morphine/administration & dosage , Pain Measurement , Pilot Projects , Pressure/adverse effects , Sex Factors , Temporomandibular Joint Disorders/complications , Young Adult
7.
Int J Mol Sci ; 19(6)2018 06 12.
Article En | MEDLINE | ID: mdl-29895782

The present study evaluated the protective effects of melatonin in ethanol (EtOH)-induced senescence and osteoclastic differentiation in human periodontal ligament cells (HPDLCs) and cementoblasts and the underlying mechanism. EtOH increased senescence activity, levels of reactive oxygen species (ROS) and the expression of cell cycle regulators (p53, p21 and p16) and senescence-associated secretory phenotype (SASP) genes (interleukin [IL]-1ß, IL-6, IL-8 and tumor necrosis factor-α) in HPDLCs and cementoblasts. Melatonin inhibited EtOH-induced senescence and the production of ROS as well as the increased expression of cell cycle regulators and SASP genes. However, it recovered EtOH-suppressed osteoblastic/cementoblastic differentiation, as evidenced by alkaline phosphatase activity, alizarin staining and mRNA expression levels of Runt-related transcription factor 2 (Runx2) and osteoblastic and cementoblastic markers (glucose transporter 1 and cementum-derived protein-32) in HPDLCs and cementoblasts. Moreover, it inhibited EtOH-induced osteoclastic differentiation in mouse bone marrow⁻derived macrophages (BMMs). Inhibition of protein never in mitosis gene A interacting-1 (PIN1) by juglone or small interfering RNA reversed the effects of melatonin on EtOH-mediated senescence as well as osteoblastic and osteoclastic differentiation. Melatonin blocked EtOH-induced activation of mammalian target of rapamycin (mTOR), AMP-activated protein kinase (AMPK), mitogen-activated protein kinase (MAPK) and Nuclear factor of activated T-cells (NFAT) c-1 pathways, which was reversed by inhibition of PIN1. This is the first study to show the protective effects of melatonin on senescence-like phenotypes and osteoclastic differentiation induced by oxidative stress in HPDLCs and cementoblasts through the PIN1 pathway.


Dental Cementum/cytology , Ethanol/pharmacology , Melatonin/pharmacology , Osteoclasts/drug effects , Osteoclasts/metabolism , Periodontal Ligament/cytology , Cell Differentiation/drug effects , Cells, Cultured , Cellular Senescence/drug effects , Dental Cementum/metabolism , Humans , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoclasts/cytology , Periodontal Ligament/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
8.
Oral Surg Oral Med Oral Pathol Oral Radiol ; 124(2): e164-e170, 2017 Aug.
Article En | MEDLINE | ID: mdl-28522185

Complex regional pain syndrome (CRPS) is one of the most challenging chronic pain conditions and is characterized by burning pain, allodynia, hyperalgesia, autonomic changes, trophic changes, edema, and functional loss involving mainly the extremities. Until recently, very few reports have been published concerning CRPS involving the orofacial area. We report on a 50-year-old female patient who presented with unbearable pain in all of her teeth and hypersensitivity of the facial skin. She also reported intractable pain in both extremities accompanied by temperature changes and orofacial pain that increased when the other pains were aggravated. In the case of CRPS with trigeminal neuropathic pain, protocols for proper diagnosis and prompt treatment have yet to be established in academia or in the clinical field. We performed functional magnetic resonance imaging for a thorough analysis of the cortical representation of the affected orofacial area immediately before and immediately after isolated light stimulus of the affected hand and foot and concluded that CRPS can be correlated with trigeminal neuropathy in the orofacial area. Furthermore, the patient was treated with carbamazepine administration and stellate ganglion block, which can result in a rapid improvement of pain in the trigeminal region.


Complex Regional Pain Syndromes/diagnostic imaging , Complex Regional Pain Syndromes/physiopathology , Facial Pain/diagnostic imaging , Facial Pain/physiopathology , Magnetic Resonance Imaging , Female , Humans , Middle Aged , Pain Measurement , Radiography, Panoramic
9.
Muscle Nerve ; 56(3): 510-518, 2017 09.
Article En | MEDLINE | ID: mdl-27977864

INTRODUCTION: N-methyl-d-aspartate (NMDA) is expressed in sensory neurons and plays important roles in peripheral pain mechanisms. The aim of this study was to examine the effects and molecular mechanisms of NMDA on C2C12 myoblast proliferation and differentiation. METHODS: Cytotoxicity and differentiation were examined by the MTT assay, reverse transcription-polymerase chain reaction, and immunofluorescence. RESULTS: NMDA had no cytotoxicity (10-500 µM) and inhibited myoblastic differentiation of C2C12 cells, as assessed by F-actin immunofluorescence and levels of mRNAs encoding myogenic markers such as myogenin and myosin heavy-chain 2. It inhibited phosphorylation of mammalian target of rapamycin (mTOR) by inactivating mitogen-activated protein kinases (MAPKs), including extracellular signal-regulated kinase, c-Jun N-terminal kinase, and p38. It induced reactive oxygen species production. Furthermore, NMDA-suppressed expression of F-actin was reversed by adding the antioxidant N-acetylcysteine. CONCLUSIONS: Collectively, these results indicate that NMDA impairs myogenesis or myogenic differentiation in C2C12 cells through the mTOR/MAPK signaling pathways and may lead to skeletal muscle degeneration. Muscle Nerve 56: 510-518, 2017.


Cell Differentiation/drug effects , Cell Proliferation/drug effects , Muscle Development/drug effects , Myoblasts/drug effects , N-Methylaspartate/toxicity , Animals , Cell Differentiation/physiology , Cell Line , Cell Proliferation/physiology , Mice , Muscle Development/physiology , Myoblasts/metabolism , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism
10.
Dent Mater ; 32(11): 1301-1311, 2016 11.
Article En | MEDLINE | ID: mdl-27634479

OBJECTIVE: Magnetic biomaterials have recently gained great attention due to their some intriguing cell and tissue responses. However, little attention has been given to the fields of dental tissue regeneration. In this sense, we aim to investigate the effects of magnetic nanofiber scaffolds on the human dental pulp cell (HDPC) behaviors and to elucidate the underlying signaling mechanisms in the events. METHODS: Magnetic nanofiber scaffolds incorporating magnetic nanoparticles at varying contents were prepared into nanofibrous matrices to cultivate cells. Cell growth by MTS assay, odontoblastic differentiation by alkaline phosphatase (ALP) activity, mineralization, and the mRNA expression of differentiation-related genes of HDPCs, in vitro angiogenesis by migration and capillary tube formation in endothelial cells on the conditioned medium obtained from HDPSCs in the presence or absence of scaffolds. Western blot analysis and confocal immunofluorescene were used to asses signaling pathways. RESULTS: The growth of HDPCs was significantly enhanced on the magnetic scaffolds with respect to the non-magnetic counterpart. The odontogenic differentiation of cells was significantly up-regulated by the culture with magnetic scaffolds. Furthermore, the magnetic scaffolds promoted the HDPC-induced angiogenesis of endothelial cells. The expression of signaling molecules, Wnt3a, phosphorylated GSK-3ß and nuclear ß-catenin, was substantially stimulated by the magnetic scaffolds; in parallel, the MAPK and NF-κB were highly activated when cultured on the magnetic nanofiber scaffolds. SIGNIFICANCE: This study is the first to demonstrate that magnetic nanofiber scaffolds stimulate HDPCs in the events of growth, odontogenic differentiation, and pro-angiogenesis, and the findings imply the novel scaffolds can be potentially useful as dentin-pulp regenerative matrices.


Dental Pulp/metabolism , NF-kappa B/metabolism , Nanofibers , Neovascularization, Physiologic , Odontogenesis , Cell Differentiation , Humans , Mitogen-Activated Protein Kinase Kinases/metabolism , Wnt Proteins/metabolism
11.
Am J Chin Med ; 43(7): 1439-52, 2015.
Article En | MEDLINE | ID: mdl-26477796

Cudraxanthone H (CH) is a natural compound isolated from a methanol extract of the root bark of Cudrania tricuspidata, a herbal plant also known as Moraceae. However, the effect of CH on human cancer cells has not been reported previously. The aim of this study was to investigate the anticancer effects and mechanism of action of CH on oral squamous cell carcinoma (OSCC) cells. CH exerted significant antiproliferative effects on OSCC cells in dose- and time-dependent manners. CH also induced apoptosis in OSCC cells, as evidenced by an increased percentage of cells in the sub-G1 phase of the cell cycle, annexin V-positive/propidium iodide-negative cells, and nuclear morphology. This antiproliferative effect of CH was associated with a marked reduction in the expression of cyclin D1 and cyclin E, with a concomitant induction of cyclin-dependent kinase inhibitor (CDKI) expression (p21 and p27). CH inhibited the phosphorylation and degradation of IκB-α and the nuclear translocation of NF-κB p65. Furthermore, CH treatment down-regulated PIN1 mRNA and protein expression in a dose-dependent manner. PIN1 overexpression by infection with adenovirus-PIN1 (Ad-PIN1) attenuated the CH-induced growth-inhibiting and apoptosis-inducing effects, blocked CH-enhanced CDKI expression and restored cyclin levels. In contrast, inhibiting PIN1 expression via juglone exerted the opposite effects. The present study is the first to demonstrate antiproliferative and apoptosis-inducing effects of CH, which exerts its effects by inhibiting NF-κB and PIN1. These data suggest that it might be a novel alternative chemotherapeutic agent for use in the treatment of oral cancer.


Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/therapeutic use , Apoptosis/drug effects , Apoptosis/genetics , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , NF-kappa B/physiology , Peptidylprolyl Isomerase/physiology , Phytotherapy , Signal Transduction/drug effects , Signal Transduction/genetics , Xanthones/pharmacology , Antineoplastic Agents, Phytogenic/isolation & purification , Carcinoma, Squamous Cell/drug therapy , Cell Line, Tumor , Humans , Moraceae/chemistry , Mouth Neoplasms/drug therapy , NIMA-Interacting Peptidylprolyl Isomerase , Xanthones/isolation & purification
12.
J Cell Biochem ; 116(7): 1241-55, 2015 Jul.
Article En | MEDLINE | ID: mdl-25565665

Recent reports suggest that hypoxia inducible factor-2α (HIF-2α) is a key regulator of osteoarthritis cartilage destruction. However, the precise role of HIF-2α in the inflammatory response and osteoclast differentiation remains unclear. The purpose of this study was to investigate the effect of HIF-2α on inflammatory cytokines, extracellular matrix (ECM) destruction enzymes, and osteoclastic differentiation in nicotine and lipopolysaccharide (LPS)-stimulated human periodontal ligament cells (PDLCs). HIF-2α was upregulated in chronically inflamed PDLCs of periodontitis patients, and in nicotine- and LPS-exposed PDLC in dose- and time-dependent manners. HIF-2α inhibitor and HIF-2α siRNA attenuated the nicotine- and LPS- induced production of NO and PGE2 , upregulation of iNOS, COX-2, pro-inflammatory cytokines (IL-1ß, TNF-α, IL-1ß, IL-6, IL-8, IL-10, IL-11, and IL-17), and matrix metalloproteinases (MMPs; MMP-1, -8, -13, -2 and -9), and reversed the effect on TIMPs (TIMP-1 and -2) in PDLCs. The conditioned medium produced by nicotine and LPS-treated PDLCs increased the number of TRAP-stained osteoclasts, TRAP activity and osteoclast-specific genes, which has been blocked by HIF-2α inhibition and silencing. HIF-2α inhibitor and HIF-2α siRNA inhibited the effects of nicotine and LPS on the activation of Akt, JAK2 and STAT3, ERK and JNK MAPK, nuclear factor-κB, c-Jun, and c-Fos. Taken together, this study is the first to demonstrate that HIF-2α inhibition exhibits anti-inflammatory activity through the inhibition of inflammatory cytokines and impairment of ECM destruction, as well as blocking of osteoclastic differentiation in a nicotine- and periodontopathogen-stimulated PDLCs model. Thus, HIF-2α inhibition may be a novel molecular target for therapeutic approaches in periodontitis.


Basic Helix-Loop-Helix Transcription Factors/metabolism , Osteoclasts/cytology , Periodontal Ligament/pathology , Periodontitis/metabolism , Transcription Factors/metabolism , Animals , Cell Differentiation/drug effects , Cells, Cultured , Culture Media, Conditioned/pharmacology , Gene Expression Regulation/drug effects , Humans , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred ICR , Nicotine/pharmacology , Osteoclasts/drug effects , Osteoclasts/metabolism , Periodontal Ligament/cytology , Periodontitis/chemically induced , Periodontitis/pathology , Up-Regulation
13.
Clin Oral Investig ; 19(6): 1419-28, 2015 Jul.
Article En | MEDLINE | ID: mdl-25467233

PURPOSE: The expression levels of intracellular pyrin domain-containing 3 (NLRP3) and microbial pattern-recognition receptors, such as nucleotide-binding oligomerization domain 2 (NOD2), have been reported in human dental pulp cells (HDPCs) and inflamed dental pulp tissue, but the role of NLRP3 and Toll-like receptors (TLRs) in the production of human beta defensin 2 (hBD2) and inflammatory cytokines against invading pathogens remains poorly defined. The aim of this study was to determine whether the NOD2 ligand muramyl dipeptide (MDP) upregulates hBD2 and inflammatory cytokines and whether this response is dependent on TLRs and NLRP inflammasomes in HDPCs. METHODOLOGY: The effects of MDP on the expression of hBD2, TLRs, inflammasomes, and pro-inflammatory mediators in HDPCs were examined using Western blotting and reverse transcription-polymerase chain reaction. Levels of pro-inflammatory cytokines, such as nitric oxide (NO) and prostaglandin E2 (PGE2), were determined by enzyme-linked immunosorbent assay. RESULTS: MDP upregulated hBD2, TLR2, and TLR4 mRNAs and protein levels in a dose- and time-dependent manner. TLR2 and TLR4 neutralizing blocking antibodies and NOD2- and hBD2-specific small interfering RNAs (siRNAs) attenuated the MDP-induced production of NO, PGE2, tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and IL-8 and upregulated inducible nitric oxide synthase (iNOS) and cyclooxygenase 2 (COX2) in HDPCs. Additionally, MDP activated inflammasome-related genes, such as NLRP3, caspase 1, apoptotic speck protein containing a caspase recruitment domain, and IL-1ß. Furthermore, silencing of the NLRP3 gene using a siRNA significantly decreased the MDP-induced expression of hBD2 and cytokines, such as iNOS-derived NO, COX2, PGE2, TNF-α, IL-6, and IL-8. CONCLUSION: These results suggest that NOD2 activates the TLR2, TLR4, and NLRP3 inflammasome-signaling pathways in HDPCs to induce the production of multiple inflammatory mediators and antimicrobial peptides, which in turn promote pulp immune defense against microbial challenge. CLINICAL RELEVANCE: The TLR and NLRP3 inflammasome pathways may represent an important modulatory mechanism of immune defense responses during the progression of pulpitis. Our results suggest that local inhibition of NLRP3 and TLRs may reduce the impact of cytokine-mediated host destructive processes in pulpitis.


Acetylmuramyl-Alanyl-Isoglutamine/pharmacology , Dental Pulp/cytology , Dental Pulp/metabolism , Inflammasomes/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Toll-Like Receptors/metabolism , beta-Defensins/metabolism , Blotting, Western , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
14.
J Endod ; 40(10): 1642-8, 2014 Oct.
Article En | MEDLINE | ID: mdl-25260738

INTRODUCTION: The objective of this study was to compare the cytotoxicity, inflammatory response, osteogenic effect, and the signaling mechanism of these biologic activities of 4 calcium compound-based root canal sealers (ie, Sealapex [Sybron Kerr, WA], apatite root sealer [ARS; Dentsply Sankin, Tokyo, Japan], MTA Fillapex [Angelus Indústria de Produtos Odontológicos S/A, Londrina, PR, Brazil], and iRoot SP [Innovative BioCreamix Inc, Vancouver, Canada]) in human periodontal ligament cells. METHODS: Cytotoxicity was assessed using the 3-(4,5-dimethylthiazolyl-2-yl)-2,5-diphenyltetrazolium bromide assay. Levels of inflammatory mediators were measured by enzyme-linked immunosorbent assay, reverse-transcription polymerase chain reaction, and Western blot analysis. Osteogenic potential was evaluated by alkaline phosphatase activity, alizarin red staining, and marker genes by reverse-transcription polymerase chain reaction. The signal transduction pathways were examined by Western blotting. RESULTS: None of the sealers were cytotoxic. ARS, MTA Fillapex, and iRoot SP induced a lower expression of proinflammatory mediators than Sealapex. All sealers increased ALP activity and the formation of mineralized nodules and up-regulated the expression of osteoblastic marker messenger RNA. ARS, MTA Fillapex, and iRoot SP showed superior osteogenic potential compared with Sealapex. The expression and/or activation of integrin receptors and downstream signaling molecules, including focal adhesion kinase, paxillin, Akt, mitogen-activated protein kinase, and nuclear factor κB, was induced by ARS, MTA Fillapex, and iRoot SP treatment but not by Sealapex treatment. CONCLUSIONS: We show for the first time that ARS, MTA Fillapex, and iRoot SP induce a lower expression of inflammatory mediators and enhance osteoblastic differentiation of PDLCs via the integrin-mediated signaling pathway compared with Sealapex.


Biocompatible Materials/pharmacology , Inflammation Mediators/pharmacology , Osteogenesis/drug effects , Root Canal Filling Materials/pharmacology , Alkaline Phosphatase/analysis , Alkaline Phosphatase/drug effects , Aluminum Compounds/pharmacology , Aluminum Compounds/toxicity , Anthraquinones , Biocompatible Materials/toxicity , Calcium Compounds/pharmacology , Calcium Compounds/toxicity , Calcium Hydroxide/pharmacology , Calcium Hydroxide/toxicity , Cell Differentiation/drug effects , Cell Line , Cell Survival/drug effects , Coloring Agents , Drug Combinations , Durapatite/pharmacology , Durapatite/toxicity , Focal Adhesion Kinase 1/drug effects , Humans , Inflammation Mediators/toxicity , Materials Testing , Mitogen-Activated Protein Kinases/drug effects , NF-kappa B/drug effects , Nanoparticles , Osteoblasts/drug effects , Oxides/pharmacology , Oxides/toxicity , Paxillin/drug effects , Periodontal Ligament/cytology , Periodontal Ligament/drug effects , Proto-Oncogene Proteins c-akt/drug effects , Root Canal Filling Materials/toxicity , Salicylates/pharmacology , Salicylates/toxicity , Signal Transduction/drug effects , Silicates/pharmacology , Silicates/toxicity , Tetrazolium Salts , Thiazoles
15.
Biomed Res Int ; 2014: 934691, 2014.
Article En | MEDLINE | ID: mdl-25105148

Isocudraxanthone K (IK) is a novel, natural compound from a methanol extract of the root bark of Cudrania tricuspidata. It has not been shown previously that IK possessed antitumor activity. We investigated the antitumor effects and molecular mechanism of IK and related signal transduction pathway(s) in oral squamous cell carcinoma cells (OSCCCs). The MTT assay revealed that IK had an antiproliferative effect on OSCCCs, in a dose- and time-dependent manner. IK induced apoptosis in OSCCCs, as identified by a cell-cycle analysis, annexin V-FITC and propidium iodide staining, and the nuclear morphology in cell death. IK caused time-dependent phosphorylation of Akt, p38, and ERK (extracellular signal-regulated kinase). In addition, IK increased the cytosolic to nuclear translocation of nuclear factor-κB (NF-κB) p65 and the degradation and phosphorylation of IκB-α in HN4 and HN12 cells. Furthermore, IK treatment downregulated hypoxia-inducible factor 1α (HIF-1α) and its target gene, vascular endothelial growth factor (VEGF). Cobalt chloride (CoCl2), a HIF-1α activator, attenuated the IK-induced growth-inhibiting and apoptosis-inducing effects, and blocked IK-induced expression of apoptosis regulatory proteins, such as Bax, Bcl-2, caspase-3, caspase-8, and caspase-9, and cytochrome c. Collectively, these data provide the first evidence of antiproliferative and apoptosis-inducing effects of IK as a HIF-1α inhibitor and suggest it may be a drug candidate for chemotherapy against oral cancer.


Apoptosis/drug effects , Cell Proliferation/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Moraceae/chemistry , Mouth Neoplasms , Plant Bark/chemistry , Plant Extracts/pharmacology , Plant Roots/chemistry , Xanthones/pharmacology , Caspases/genetics , Caspases/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Cytochromes c/genetics , Cytochromes c/metabolism , Drug Screening Assays, Antitumor , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , I-kappa B Proteins/genetics , I-kappa B Proteins/metabolism , Mouth Neoplasms/drug therapy , Mouth Neoplasms/genetics , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Plant Extracts/chemistry , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , Xanthones/chemistry , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
16.
Cell Tissue Res ; 357(1): 119-32, 2014 Jul.
Article En | MEDLINE | ID: mdl-24817031

The aim of this study is to determine the effects of the combination of recombinant human BMP-2 (rh-BMP-2) and dentin sialoprotein (rh-DSP) on growth and differentiation in human cementoblasts and determine the underlying signal transduction mechanism. Compared to treatment of cementoblasts with either rh-BMP-2 or rh-DSP alone, the combination of rh-BMP-2 and rh-DSP synergistically increased cell growth, ALP activity, nodule formation and expression of differentiation markers. The differentiation-promoting effect was also observed in periodontal ligament cells and an osteoblastic cell line. Likewise, combination of rh-DSP and rh-BMP-2 increased BMP-2 mRNA expression and Smad1/5/8 phosphorylation, which was blocked by the BMP antagonist noggin. The expression levels of α2ß1 integrin and RhoA, as well as the phosphorylation status of FAK and Akt, were increased by the combination of rh-BMP-2 and rh-DSP in a time-dependent manner. In addition, rh-BMP-2 and rh-DSP enhanced expression of Wnt ligands, ß-catenin activation and GSK-3ß phosphorylation, all of which were inhibited by the Wnt receptor antagonist DKK1. Furthermore, treatment with rh-DSP plus rh-BMP-2 resulted in phosphorylation of extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK) and p38 and also induced the nuclear translocation of the NF-κB p65 subunit, which was blocked by noggin. This study demonstrates for the first time that rh-DSP and rh-BMP-2 act synergistically, enhancing each other's ability to stimulate cementoblastic cell growth and differentiation in vitro via autocrine BMP, integrin, Wnt/ß-catenin, MAP kinase and NF-κB pathways. These results support the therapeutic potential of a combination strategy for aiding periodontal regeneration.


Bone Morphogenetic Protein 2/pharmacology , Dental Cementum/cytology , Dental Cementum/drug effects , Extracellular Matrix Proteins/pharmacology , Phosphoproteins/pharmacology , Sialoglycoproteins/pharmacology , Transforming Growth Factor beta/pharmacology , Cell Differentiation/drug effects , Cells, Cultured , Dental Cementum/metabolism , Humans , Periodontal Ligament/cytology , Periodontal Ligament/drug effects , Periodontal Ligament/metabolism , Recombinant Proteins/pharmacology , Signal Transduction/drug effects
17.
Planta Med ; 79(14): 1298-306, 2013 Sep.
Article En | MEDLINE | ID: mdl-23881456

The goal of this study was to investigate the effect and molecular mechanism of cudraflavone B, a prenylated flavonoid isolated from the root bark of Cudrania tricuspidata, against oral squamous cell carcinoma cells. We observed that cudraflavone B inhibited proliferation of these cells in a time- and dose-dependent manner. At 15 µM, cudraflavone B induced cell death via apoptosis (characterized by the appearance of nuclear morphology) and increased the accumulation of the sub-G1 peak (portion of apoptotic annexin V positive cells). Treatment with cudraflavone B triggered the mitochondrial apoptotic pathway (indicated by induction of the proapoptotic protein p53 and the p21 and p27 effector proteins), downregulation of cell cycle regulatory proteins (e.g., p-Rb, changing Bax/Bcl-2 ratios, cytochrome-c release), and caspase-3 activation. Cudraflavone B time-dependently activated NF-κB, the MAP kinases p38, and ERK, and induced the expression of SIRT1. SIRT1 activator, resveratrol, dose-dependently attenuated the growth-inhibitory and apoptosis-inducing effect of cudraflavone B and blocked cudraflavone B-induced regulatory protein expressions in the mitochondrial pathway such as p53, p21, p27, Bax, caspase-3, and cytochrome-c. Conversely, treatment with SIRT1 inhibitor sirtinol caused opposite effects. These results demonstrate for the first time that the molecular mechanism underlying the antitumor effect in oral squamous cell carcinoma cells is related to the activation of MAPK/and NF-κB as well as of the SIRT1 pathway. Therefore, cudraflavone B may be a lead for the development of a potential candidate for human oral squamous cell carcinoma cells.


Carcinoma, Squamous Cell/drug therapy , Flavonoids/therapeutic use , Mitogen-Activated Protein Kinases/metabolism , Moraceae/chemistry , Mouth Neoplasms/drug therapy , NF-kappa B/metabolism , Sirtuin 1/metabolism , Antineoplastic Agents, Phytogenic/isolation & purification , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/therapeutic use , Apoptosis/drug effects , Carcinoma, Squamous Cell/metabolism , Caspase 3/metabolism , Cell Cycle/drug effects , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Flavonoids/isolation & purification , Flavonoids/pharmacology , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Mouth Neoplasms/metabolism , Phytotherapy , Plant Bark , Plant Extracts/chemistry , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Plant Roots , Protein Kinases/metabolism , Signal Transduction
18.
Article En | MEDLINE | ID: mdl-22384812

Abstract Aims: We explore the region-specific impact of nitric oxide (NO) on adult neural stem cell (aNSC) niches with regard to neurogenesis and NSC damage and investigate the underlying mechanisms in Niemann-Pick disease type C (NPC) mice. Results: Among the two anatomical stem-cell niches of the brain, subventricular zone (SVZ)-derived aNSCs enhanced c-Jun N-terminal kinase (JNK) activity because of excessive NO production by the cholesterol accumulation. Activated JNK interacts with γH2AX, a marker for DNA damage; however, almost none of the aNSCs in the dentate gyrus (DG) showed either JNK signaling activation or abundant DNA damage. SVZ-derived aNSCs were protected from DNA damage by the treatment of Nω-nitro-L-arginine methyl ester (L-NAME), a NO synthase (NOS) inhibitor, both in vitro and in vivo. We also observed that U18666A, an inducer of cholesterol accumulation, increased inducible NOS expression, JNK activation, and DNA damage in the wild type (WT)-aNSCs. Interestingly, we found that endogenous cholesterol efflux transporters and their regulator were less activated in the SVZ than in the DG, in both WT and NPC mice. This result explains the high vulnerability of SVZ-derived aNSCs to the cholesterol imbalance as observed in NPC mice. Innovation and Conclusion: In this study, we demonstrated that the SVZ-derived aNSCs might be major targets of NPC. Significantly, aNSCs showed different responses depending on their anatomical origins due to dissimilarities in their cholesterol transporting system and NO-dependent JNK activation. These findings can contribute to the understanding of the region-specific nature of the two SVZ and DG neurogenic niches. Antioxid. Redox Signal. 00, 000-000.

19.
Article En | MEDLINE | ID: mdl-22364277

UNLABELLED: Abstract Aims: The fate decision of adult stem cells is determined by the activation of specific intracellular signaling pathways after exposure to specific stimuli. In this study, we demonstrated specific functions of a novel small molecule, CBM-1078, that induced cell self-renewal via Oct4- and canonical Wnt/ß-catenin-mediated deaging in cultured human adipose tissue-derived stem cells (hATSCs). RESULTS: As a potential glycogen synthase kinase-3ß (GSK-3ß) inhibitor, CBM-1078 primarily activated ß-catenin and Oct4 expression after inhibition of GSK-3ß. Treatment of hATSCs with CBM-1078 led to transdifferentiation toward a neural precursor cell fate after transient self-renewal, and the cells were capable of differentiation into gamma-Aminobutyric acid (GABA)-secreting neuronal cells with pain-modulating functions in an animal model of neuropathic pain. During cell self-renewal, CBM-1078 directs the translocalization of ß-catenin and Oct4 into the nucleus, an event that is crucial for the cooperative activation of hATSC neurogenesis via Oct4 and Wnt/ß-catenin. Nuclear-localized ß-catenin and Oct4 act together to regulate the expression of Oct4, Nanog, Sox2, ß-catenin, c-Myc, and STAT3 after binding to the regulatory regions of these genes. Nuclear Oct4 and Wnt3a/ß-catenin also control cell growth by binding to the promoters of STAT3, Gli3, and c-Myc after complex formation and direct interaction. CBM-1078 actively enhanced the DNA-binding affinity of Oct4 and ß-catenin to functional genes and activated the Wnt/ß-catenin pathway to promote hATSC reprogramming. INNOVATION AND CONCLUSION: This study revealed the value of a single small molecule, CBM-1078, showing a definitive cell reprogramming mechanism. Finally, we confirmed the therapeutic potential of GABA-hATSCs for treatment of neuropathic pain, which could be used for therapeutic purposes in humans. Antioxid. Redox Signal. 00, 000-000.

20.
Stem Cells ; 30(5): 876-87, 2012 May.
Article En | MEDLINE | ID: mdl-22311737

CD49f (integrin subunit α6) regulates signaling pathways in a variety of cellular activities. However, the role of CD49f in regulating the differentiation and pluripotency of stem cells has not been fully investigated. Therefore, in this study, human mesenchymal stem cells (hMSCs) were induced to form spheres under nonadherent culture conditions, and we found that the CD49f-positive population was enriched in MSC spheres compared with MSCs in a monolayer. The expression of CD49f regulated the ability of hMSCs to form spheres and was associated with an activation of the phosphatidylinositol 3-kinase (PI3K)/AKT signaling pathway. Furthermore, the forced expression of CD49f modulated the proliferation and differentiation potentials of hMSCs through prolonged activation of PI3K/AKT and suppressed the level of p53. We showed that the pluripotency factors OCT4 and SOX2 were recruited to the putative promoter region of CD49f, indicating that OCT4 and SOX2 play positive roles in the expression of CD49f. Indeed, CD49f expression was upregulated in human embryonic stem cells (hESCs) compared with hMSCs. The elevated level of CD49f expression was significantly decreased upon embryoid body formation in hESCs. In hESCs, the knockdown of CD49f downregulated PI3K/AKT signaling and upregulated the level of p53, inducing differentiation into three germ layers. Taken together, our data suggest that the cell-surface protein CD49f has novel and dynamic roles in regulating the differentiation potential of hMSCs and maintaining pluripotency.


Embryonic Stem Cells/metabolism , Integrin alpha6/biosynthesis , Mesenchymal Stem Cells/metabolism , Multipotent Stem Cells/metabolism , Octamer Transcription Factor-3/biosynthesis , SOXB1 Transcription Factors/biosynthesis , Cell Differentiation/physiology , Cells, Cultured , Embryonic Stem Cells/cytology , Gene Expression Regulation/physiology , Humans , Mesenchymal Stem Cells/cytology , Multipotent Stem Cells/cytology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , Tumor Suppressor Protein p53/metabolism
...