Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters

Complementary Medicines
Country/Region as subject
Publication year range
1.
Int Marit Health ; 74(4): 243-252, 2023.
Article in English | MEDLINE | ID: mdl-38111244

ABSTRACT

BACKGROUND: Seafarers, confronted with unique health challenges, occasionally necessitate medical repatriation. This study examines the trends in medical repatriation cases among Filipino seafarers employed by OSM Maritime shipping company over a 10-year period from 2013 to 2022. MATERIALS AND METHODS: Medical records of OSM Maritime seafarers were reviewed, obtaining causes for and dates of medical repatriation. International Classification of Diseases (ICD-11) was utilised to classify repatriation cases. Proportion of repatriation cases were calculated and their annual trends were analysed. RESULTS: Our findings reveal that the majority of repatriation cases are attributed to injury/trauma (19.91%), musculoskeletal (18.40%), gastrointestinal (16.56%), cardiovascular (8.77%), infectious (6.82%), and genitourinary conditions (5.30%). Significantly, the study identifies a declining trend in the proportion of cardiovascular, gastrointestinal, and genitourinary conditions in annual repatriation cases, particularly in ischaemic heart conditions, cholelithiasis, cholecystitis, and urinary calculus. CONCLUSIONS: These results emphasize the critical need for multisectoral collaboration to enhance seafarers' health and well-being. Prioritizing comprehensive care programmes, ensuring safe working conditions, and exploring holistic healthcare initiatives are essential steps to enhance seafarers' occupational health.


Subject(s)
Naval Medicine , Occupational Health , Humans , Philippines , Ships , Oncostatin M
2.
Phytomedicine ; 80: 153372, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33113505

ABSTRACT

BACKGROUND: Feiyangchangweiyan capsule (FYC) is a traditional Chinese medicine formulation used in the clinical treatment of acute and chronic gastroenteritis and bacterial dysentery. However, the effect of FYC on ulcerative colitis (UC) and the mechanism thereof remains unknown. PURPOSE: To investigate the protective effect of FYC on UC mice induced by dextran sulfate sodium and illustrate the potential mechanism of this effect. METHODS: Here, we established a model of UC mice by dextran sulfate sodium and administered with FYC. The disease activity index (DAI), colon length, myeloperoxidase (MPO) content in serum, pathological structure and ultrastructural changes, and inflammatory cell infiltration of colon tissue were evaluated. Transcriptome and 16S rDNA sequencing were employed to illuminate the mechanism of FYC in the protection of UC mice. RESULTS: FYC significantly alleviates the pathological damage and the infiltration of inflammatory cells in colon tissue of dextran sulfate sodium induced UC mice, rescues shortened colon length, reduces DAI score, MPO content in serum, and pro-inflammatory factors including IL-1ß, IL-6, CCL11, MCP-1 and MIP-2, and increases anti-inflammatory factors such as IL-10. Transcriptomics revealed that Oncostatin M (OSM) and its receptor (OSMR) are the critical pathway for UC treatment by FYC. OSM and OSMR increased in UC mice compared to control mice, and decreased with FYC, which was verified via measurement of OSM and OSMR mRNA and protein levels. Furthermore, we observed that FYC modulates intestinal microbiome composition (e.g., the proportion of Barnesiella/Proteobacteria) by affecting the inflammatory factors. CONCLUSION: FYC exerts an effect on UC by inhibiting the OSM/OSMR pathway and regulating inflammatory factors to improve the intestinal flora.


Subject(s)
Colitis, Ulcerative/drug therapy , Drugs, Chinese Herbal/pharmacology , Gastrointestinal Microbiome/drug effects , Oncostatin M Receptor beta Subunit/metabolism , Oncostatin M/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Capsules , Chemokines/blood , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/microbiology , Colon/drug effects , Colon/metabolism , Colon/pathology , Colon/ultrastructure , Cytokines/blood , Dextran Sulfate/toxicity , Disease Models, Animal , Gastrointestinal Microbiome/genetics , Male , Mice, Inbred C57BL , Oncostatin M/genetics , Oncostatin M Receptor beta Subunit/genetics , Protective Agents/pharmacology
3.
Differentiation ; 114: 36-48, 2020.
Article in English | MEDLINE | ID: mdl-32563741

ABSTRACT

The capability to produce and maintain functional human adult hepatocytes remains one of the major challenges for the use of in-vitro models toward liver cell therapy and industrial drug-screening applications. Among the suggested strategies to solve this issue, the use of human-induced pluripotent stem cells (hiPSCs), differentiated toward hepatocyte-like cells (HLCs) is promising. In this work, we propose a 31-day long protocol, that includes a final 14-day long phase of oncostatin treatment, as opposed to a 7-day treatment which led to the formation of a hepatic tissue functional for CYP1A2, CYP2B6, CYP2C8, CYP2D6, and CYP3A4. The production of albumin, as well as bile acid metabolism and transport, were also detected. Transcriptome profile comparisons and liver transcription factors (TFs) motif dynamics revealed increased expression of typical hepatic markers such as HNF1A and of important metabolic markers like PPARA. The performed analysis has allowed for the extraction of potential targets and pathways which would allow enhanced hepatic maturation in-vitro. From this investigation, NRF1 and SP3 appeared as transcription factors of importance. Complex epithelial-mesenchymal transition (EMT) and mesenchymal-epithelial transition (MET) patterns were also observed during the differentiation process. Moreover, whole transcriptome analysis highlighted a response typical of the one observed in liver regeneration and hepatocyte proliferation. While a complete maturation of hepatocytes was yet to be obtained, the results presented in this work provide new insights into the process of liver development and highlight potential targets aimed to improve in-vitro liver regeneration.


Subject(s)
Cell Differentiation/genetics , Hepatocytes/cytology , Induced Pluripotent Stem Cells/cytology , Liver Regeneration , Liver/growth & development , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cytochrome P-450 CYP1A2/genetics , Cytochrome P-450 CYP2B6/genetics , Cytochrome P-450 CYP2C8/genetics , Cytochrome P-450 CYP2D6/genetics , Cytochrome P-450 CYP3A/genetics , Drug Evaluation, Preclinical , Epithelial-Mesenchymal Transition/drug effects , Hepatocytes/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Liver/cytology , Liver/drug effects , Nuclear Respiratory Factor 1/genetics , Oncostatin M/pharmacology , Sp3 Transcription Factor/genetics , Transcriptome/drug effects
4.
Cell Death Dis ; 11(4): 271, 2020 04 24.
Article in English | MEDLINE | ID: mdl-32332711

ABSTRACT

Ulcerative colitis (UC) is a chronic and etiologically refractory inflammatory gut disorder. Although berberine, an isoquinoline alkaloid, has been revealed to exert protective effects on experimental colitis, the underlying molecular mechanism in chronic intestinal inflammation remains ill-defined. This study was designed to uncover the therapeutic efficacy and immunomodulatory role of berberine in chronic UC. Therapeutic effects of oral administration of berberine were investigated in dextran sodium sulfate (DSS)-induced murine chronic UC and the underlying mechanisms were further identified by si-OSMR transfection in human intestinal stromal cells. Berberine significantly attenuated the experimental symptoms and gut inflammation of chronic UC. Berberine treatment could also maintain the intestinal barrier function and rectify tissue fibrosis. In accordance with infiltrations of antigen-presenting cells (APCs), innate lymphoid cells (ILCs), and activated NK cells in colonic lamina propria, increased expression of OSM and OSMR were observed in the inflamed tissue of chronic UC, which were decreased following berberine treatment. Moreover, berberine inhibited the overactivation of human intestinal stromal cells through OSM-mediated JAK-STAT pathway, which was obviously blocked upon siRNA targeting OSMR. The research provided an infusive mechanism of berberine and illustrated that OSM and OSMR intervention might function as the potential target in chronic UC.


Subject(s)
Berberine/therapeutic use , Colitis, Ulcerative/drug therapy , Inflammation/chemically induced , Intestinal Mucosa/drug effects , Oncostatin M/adverse effects , Animals , Berberine/pharmacology , Chronic Disease , Humans , Male , Mice , Transfection
5.
J Int Med Res ; 48(3): 300060519872898, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31813325

ABSTRACT

OBJECTIVE: This study aimed to explore whether initial hyperbaric oxygen treatment affects the stemness of glioma stem cells using an in vivo basal ganglia glioma model. METHODS: A basal ganglia glioma rat model was established. Rats were exposed to normal oxygen or hyperbaric oxygen on days 2, 4, 6, 8, 10, and 12. After 16 days of glioma cell inoculation, western blot, ELISA, and flow cytometry were performed to examine stemness-associated properties by examining the expression of CD133, A2B5, Nanog, oncostatin M, ß-catenin, Oct-3/4, Sox2, and Nestin. RESULTS: Initial hyperbaric oxygen treatment began to affect glioma stemness-associated properties. The proportion of CD133+A2B5+ cells was significantly reduced after initial hyperbaric oxygen treatment. Additionally, the expression of stemness-related genes such as Nanog and oncostatin M was reduced, while TGF-ß and ß-catenin were increased. CONCLUSIONS: Initial hyperbaric oxygen treatment not only alters the hypoxic microenvironment but also affects the stemness-associated properties of cancer stem cells.


Subject(s)
Glioma , Hyperbaric Oxygenation , Animals , Cell Line, Tumor , Glioma/therapy , Neoplastic Stem Cells , Oncostatin M , Oxygen , Rats , Tumor Microenvironment , beta Catenin/genetics
6.
Arthritis Rheumatol ; 70(12): 1959-1970, 2018 12.
Article in English | MEDLINE | ID: mdl-29790294

ABSTRACT

OBJECTIVE: To examine the effects of tofacitinib on metabolic activity, mitochondrial function, and proinflammatory mechanisms in rheumatoid arthritis (RA). METHODS: Ex vivo RA synovial explants and primary RA synovial fibroblasts (RASFs) were cultured with 1 µM tofacitinib. RASF bioenergetics were assessed using an XF24 analyzer, and key metabolic genes were assessed by reverse transcription-polymerase chain reaction (RT-PCR) analysis. Mitochondrial function was assessed using specific cell fluorescent probes and by mitochondrial gene arrays. Mitochondrial mutagenesis was quantified using a mitochondrial random mutation capture assay, and lipid peroxidation was quantified by enzyme-linked immunosorbent assay (ELISA). The effect of tofacitinib on spontaneous release of proinflammatory mediators from RA whole tissue synovial explants was quantified by ELISAs/MSD multiplex assays, and metabolic markers were quantified by RT-PCR. Finally, RASF invasion, matrix degradation, and synovial outgrowths were assessed by transwell invasion/Matrigel outgrowth assays and ELISA. RESULTS: Tofacitinib significantly decreased mitochondrial membrane potential, mitochondrial mass, and reactive oxygen species production by RASFs and differentially regulated key mitochondrial genes. Tofacitinib significantly increased oxidative phosphorylation, ATP production, and the maximal respiratory capacity and the respiratory reserve in RASFs, an effect paralleled by a decrease in glycolysis and the genes for the key glycolytic enzymes hexokinase 2 (HK2), glycogen synthase kinase 3α (GSK-3α), lactate dehydrogenase A, and hypoxia-inducible factor 1α. Tofacitinib inhibited the effect of oncostatin M (OSM) on interleukin-6 (IL-6) and monocyte chemotactic protein 1 and reversed the effects of OSM on RASF cellular metabolism. Using RA whole tissue synovial explants, we found that tofacitinib inhibited the key metabolic genes for glucose transporter 1, 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3, 3'-phosphoinositide-dependent protein kinase 1, HK2, and GSK-3α, the proinflammatory mediators IL-6, IL-8, IL-1ß, intercellular adhesion molecule 1, vascular endothelial growth factor, and TIE-2, and RASF outgrowth from synovial explants, RASF invasion, and matrix metalloproteinase 1 activity. CONCLUSION: This study demonstrates that JAK/STAT signaling mediates the complex interplay between inflammation and cellular metabolism in RA pathogenesis.


Subject(s)
Arthritis, Rheumatoid/metabolism , Inflammation Mediators/metabolism , Mitochondria/metabolism , Signal Transduction/physiology , Synovial Membrane/metabolism , Arthritis, Rheumatoid/drug therapy , Cells, Cultured , Energy Metabolism , Fibroblasts/metabolism , Humans , Janus Kinases/physiology , Oncostatin M/metabolism , Phosphorylation/drug effects , Piperidines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Pyrroles/pharmacology , STAT Transcription Factors/physiology
7.
Sci Rep ; 7(1): 16734, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29196668

ABSTRACT

Current induction methods of hepatocytes from human induced pluripotent stem cells (hiPSCs) are neither low cost nor stable. By screening a chemical library of 1,120 bioactive compounds and known drugs, we identified the α1-adrenergic receptor agonist methoxamine hydrochloride as a small molecule that promotes the differentiation of hiPSC-derived hepatoblasts into ALBUMIN+ hepatocyte-like cells. Other α1-adrenergic receptor agonists also induced the differentiation of hepatocyte-like cells, and an α1-receptor antagonist blocked the hepatic-inducing activity of methoxamine hydrochloride and that of the combination of hepatocyte growth factor (HGF) and Oncostatin M (OsM), two growth factors often used for the induction of hepatoblasts into hepatocyte-like cells. We also confirmed that treatment with methoxamine hydrochloride activates the signal transducer and activator of transcription 3 (STAT3) pathway downstream of IL-6 family cytokines including OsM. These findings allowed us to establish hepatic differentiation protocols for both mouse embryonic stem cells (mESCs) and hiPSCs using small molecules at the step from hepatoblasts into hepatocyte-like cells. The results of the present study suggest that α1-adrenergic agonists induce hepatocyte-like cells by working downstream of HGF and OsM to activate STAT3.


Subject(s)
Adrenergic alpha-1 Receptor Agonists/pharmacology , Hepatocytes/cytology , Human Embryonic Stem Cells/cytology , Induced Pluripotent Stem Cells/cytology , Methoxamine/pharmacology , Adrenergic alpha-1 Receptor Antagonists/pharmacology , Cell Differentiation/drug effects , Cell Line , Drug Evaluation, Preclinical , Hepatocyte Growth Factor/pharmacology , Hepatocytes/drug effects , Hepatocytes/metabolism , Human Embryonic Stem Cells/drug effects , Humans , Induced Pluripotent Stem Cells/drug effects , Oncostatin M/pharmacology , STAT3 Transcription Factor/metabolism , Serum Albumin, Human/metabolism , Signal Transduction/drug effects
8.
Mol Med Rep ; 15(5): 3088-3092, 2017 May.
Article in English | MEDLINE | ID: mdl-28358419

ABSTRACT

William's E (WE) is a suitable medium for the differentiation of human induced pluripotent stem (iPS) cells to the hepatocyte lineage. The aim of the present study was to investigate various growth factors in their ability to promote hepatocyte differentiation of iPS cells in WE medium. Human iPS 201B7 cells were cultured in WE medium supplemented with growth factors, and mRNA expression levels and promoter activities of α­fetoprotein (AFP) and albumin were examined by reverse transcription­quantitative polymerase chain reaction and luciferase assay, respectively. In addition, time course analysis of AFP mRNA expression was performed in 201B7 cells cultured in WE medium supplemented with oncostatin M. The results demonstrated that mRNA expression levels of AFP were significantly elevated by most growth factors tested as supplements in WE medium, except all­trans retinoic acid, compared with cells cultured in ReproFF (a medium that maintains pluripotency). The highest increase in AFP mRNA expression levels was observed by oncostatin M stimulation. Albumin mRNA expression levels were increased by all­trans retinoic acid and insulin­transferrin­selenium supplementation in WE medium compared with cells cultured in ReproFF. Oncostatin M supplementation significantly stimulated the promoter activity of the AFP gene, but no growth factor tested significantly stimulated the promoter activity of the albumin gene. By time course analysis, significant increase of AFP mRNA expression was observed on the sixth day post­stimulation, compared with cells cultured in WE medium alone. In conclusion, the present study demonstrated that oncostatin M supplementation in WE medium was sufficient to initiate hepatocyte differentiation in iPS cells.


Subject(s)
Cell Differentiation/drug effects , Hepatocytes/cytology , Hepatocytes/metabolism , Induced Pluripotent Stem Cells/drug effects , Oncostatin M/pharmacology , Organ Preservation Solutions/chemistry , Albumins/drug effects , Albumins/genetics , Albumins/metabolism , Cell Line , Cells, Cultured , Culture Media , Hepatocyte Growth Factor/pharmacology , Humans , Induced Pluripotent Stem Cells/cytology , RNA, Messenger/biosynthesis , Tretinoin/pharmacology , alpha-Fetoproteins/drug effects , alpha-Fetoproteins/genetics , alpha-Fetoproteins/metabolism
9.
BMC Complement Altern Med ; 16: 205, 2016 Jul 11.
Article in English | MEDLINE | ID: mdl-27400672

ABSTRACT

BACKGROUND: Rheumatoid arthritis (RA) is an autoimmune disease associated with chronic inflammatory arthritis. TNF-α and OSM are pro-inflammatory cytokines that play a key role in RA progression. Thus, reducing the effects of both cytokines is practical in order to relieve the progression of the disease. This current study is interested in sesamin, an active compound in sesame seeds. Sesamin has been shown to be a chondroprotective agent in osteoarthritis models. Here, we have evaluated a porcine cartilage explant as a cartilage degradation model related to RA induced by TNF-α and/or OSM in order to investigate the effects of sesamin on TNF-α and OSM in the cartilage degradation model. METHODS: A porcine cartilage explant was induced with a combination of TNF-α and OSM (test group) or IL-1ß and OSM (control group) followed by a co-treatment of sesamin over a long-term period (35 days). After which, the tested explants were analyzed for indications of both the remaining and the degradation aspects using glycosaminoglycan and collagen as an indicator. RESULTS: The combination of TNF-α and OSM promoted cartilage degradation more than either TNF-α or OSM alone and was comparable with the combination of IL-1ß and OSM. Sesamin could be offering protection against cartilage degradation by reducing GAGs and collagen turnover in the generated model. CONCLUSIONS: Sesamin might be a promising agent as an alternative treatment for RA patients. Furthermore, the generated model revealed itself to be an impressive test model for the analysis of phytochemical substances against the cartilage degradation model for RA. The model could be used to test for the prevention of cartilage degradation in other biological agents induced with TNF-α and OSM as well.


Subject(s)
Cartilage/drug effects , Dioxoles/pharmacology , Lignans/pharmacology , Oncostatin M/metabolism , Protective Agents/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Animals , Arthritis, Rheumatoid , Cartilage/metabolism , Dioxoles/chemistry , Immunohistochemistry , Lignans/chemistry , Models, Biological , Protective Agents/chemistry , Swine
10.
J Nutr ; 145(11): 2448-55, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26423732

ABSTRACT

BACKGROUND: Preclinical and epidemiologic studies suggest that garlic intake is inversely associated with the progression of cancer and cardiovascular disease. OBJECTIVE: We designed a study to probe the mechanisms of garlic action in humans. METHODS: We conducted a randomized crossover feeding trial in which 17 volunteers consumed a garlic-containing meal (100 g white bread, 15 g butter, and 5 g raw, crushed garlic) or a garlic-free control meal (100 g white bread and 15 g butter) after 10 d of consuming a controlled, garlic-free diet. Blood was collected before and 3 h after test meal consumption for gene expression analysis in whole blood. Illumina BeadArray was used to screen for genes of interest, followed by real-time quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR) on selected genes. To augment human study findings, Mono Mac 6 cells were treated with a purified garlic extract (0.5 µL/mL), and mRNA was measured by qRT-PCR at 0, 3, 6, and 24 h. RESULTS: The following 7 genes were found to be upregulated by garlic intake: aryl hydrocarbon receptor (AHR), aryl hydrocarbon receptor nuclear translocator (ARNT), hypoxia-inducible factor 1α (HIF1A), proto-oncogene c-Jun (JUN), nuclear factor of activated T cells (NFAT) activating protein with immunoreceptor tyrosine-based activation motif 1 (NFAM1), oncostatin M (OSM), and V-rel avian reticuloendotheliosis viral oncogene homolog (REL). Fold-increases in mRNA transcripts ranged from 1.6 (HIF1A) to 3.0 (NFAM1) (P < 0.05). The mRNA levels of 5 of the 7 genes that were upregulated in the human trial were also upregulated in cell culture at 3 and 6 h: AHR, HIF1A, JUN, OSM, and REL. Fold-increases in mRNA transcripts in cell culture ranged from 1.7 (HIF1A) to 12.1 (JUN) (P < 0.01). OSM protein was measured by ELISA and was significantly higher than the control at 3, 6, and 24 h (24 h: 19.5 ± 1.4 and 74.8 ± 1.4 pg/mL for control and garlic, respectively). OSM is a pleiotropic cytokine that inhibits several tumor cell lines in culture. CONCLUSION: These data indicate that the bioactivity of garlic is multifaceted and includes activation of genes related to immunity, apoptosis, and xenobiotic metabolism in humans and Mono Mac 6 cells. This trial is registered at clinicaltrials.gov as NCT01293591.


Subject(s)
Administration, Oral , B-Lymphocytes/immunology , Garlic , T-Lymphocytes/immunology , Aryl Hydrocarbon Receptor Nuclear Translocator/blood , Aryl Hydrocarbon Receptor Nuclear Translocator/genetics , Basic Helix-Loop-Helix Transcription Factors/blood , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Line , Cross-Over Studies , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/blood , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Male , Membrane Proteins/blood , Membrane Proteins/genetics , Middle Aged , Oncostatin M/blood , Oncostatin M/genetics , Proto-Oncogene Mas , Proto-Oncogene Proteins c-jun/blood , Proto-Oncogene Proteins c-jun/genetics , RNA, Messenger/blood , RNA, Messenger/metabolism , Receptors, Aryl Hydrocarbon/blood , Receptors, Aryl Hydrocarbon/genetics , Transcription Factor RelA/blood , Transcription Factor RelA/genetics , Up-Regulation
11.
Biochim Biophys Acta ; 1853(10 Pt A): 2306-25, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25986861

ABSTRACT

Oncostatin M (OSM), a cytokine in the interleukin-6 (IL-6) family, has been proposed to play a protective role in the central nervous system, such as attenuation of excitotoxicity induced by N-methyl-D-aspartate (NMDA) and glutamate. However, the potential neuroprotective effects of OSM against mitochondrial dysfunction have never been reported. In the present study, we tested the hypothesis that OSM may confer neuronal resistance against 3-nitropropionic acid (3-NP), a plant toxin that irreversibly inhibits the complex II of the mitochondrial electron transport chain, and characterized the underlying molecular mechanisms. We found that OSM preconditioning dose- and time-dependently protected cortical neurons against 3-NP toxicity. OSM stimulated expression of myeloid cell leukemia-1 (Mcl-1), an anti-apoptotic Bcl-2 family member expressed in differentiating myeloid cells, that required prior phosphorylation of Janus kinase-1 (JAK1), JAK2, extracellular signal-regulated kinase-1/2 (ERK1/2), signal transducer and activator of transcription-3 (STAT3), STAT1, and cAMP-response element-binding protein (CREB). Pharmacological inhibitors of JAK1, JAK2, ERK1/2, STAT3, STAT1, and CREB as well as the siRNA targeting at STAT3 and Mcl-1 all abolished OSM-dependent 3-NP resistance. Finally, OSM-dependent Mcl-1 induction contributed to the enhancements of mitochondrial bioenergetics including increases in spare respiratory capacity and ATP production. In conclusion, our findings indicated that OSM induces Mcl-1 expression via activation of ERK1/2, JAK1/2, STAT1/3, and CREB; furthermore, OSM-mediated Mcl-1 induction contributes to bioenergetic improvements and neuroprotective effects against 3-NP toxicity in cortical neurons. OSM may thus serve as a novel neuroprotective agent against mitochondrial dysfunction commonly associated with pathogenic mechanisms underlying neurodegeneration.


Subject(s)
Cerebral Cortex/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Energy Metabolism/physiology , Janus Kinase 1/metabolism , Janus Kinase 2/metabolism , Mitochondria/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Neurons/metabolism , Oncostatin M/metabolism , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism , Animals , Antihypertensive Agents/adverse effects , Antihypertensive Agents/pharmacology , Cerebral Cortex/cytology , Energy Metabolism/drug effects , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Neurons/cytology , Nitro Compounds/adverse effects , Nitro Compounds/pharmacology , Propionates/adverse effects , Propionates/pharmacology , Rats
12.
J Cell Biochem ; 116(8): 1479-89, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25683148

ABSTRACT

Human induced pluripotent stem (hiPS) cells are an ideal source for hepatocytes. Glucose and arginine are necessary for cells to survive. Hepatocytes have galactokinase (GALK), which metabolizes galactose for gluconeogenesis, and ornithine transcarbamylase (OTC), which converts ornithine to arginine in the urea cycle. Hepatocyte selection medium (HSM) lacks both glucose and arginine, but contains galactose and ornithine. Although human primary hepatocytes survive in HSM, all the hiPS cells die in 3 days. The aim of this study was to modify HSM so as to initiate hepatocyte differentiation in hiPS cells within 2 days. Hepatocyte differentiation initiating medium (HDI) was prepared by adding oncostatin M (10 ng/ml), hepatocyte functional proliferation inducer (10 nM), 2,2'-methylenebis (1,3-cyclohexanedione) (M50054) (100 µg/ml), 1× non-essential amino acid, 1× sodium pyruvate, nicotinamide (1.2 mg/ml), L-proline (30 ng/ml), and L-glutamine (0.3 mg/ml) to HSM. HiPS cells (201B7 cells) were cultured in HDI for 2 days. RNA was isolated, used as template for cDNA, and subjected to real-time quantitative polymerase chain reaction. Alpha-fetoprotein, γ-glutamyl transpeptidase, and delta-like 1 were upregulated. Expression of albumin was not observed. Expression of transcription factors specific to hepatocytes was upregulated. The expression of GALK2, OTC, and CYP3A4 were increased. In conclusion, differentiation of 201B7 cells to hepatoblast-like cells was initiated in HDI. Limitations were small number of cells were obtained, and the cells with HDI were not mature hepatocytes.


Subject(s)
Culture Media/chemistry , Culture Media/pharmacology , Gene Expression Regulation/drug effects , Hepatocytes/cytology , Induced Pluripotent Stem Cells/drug effects , Cell Differentiation , Cell Line , Cell Survival/drug effects , Cytochrome P-450 CYP3A/genetics , Galactokinase/genetics , Galactokinase/pharmacology , Glutamine/pharmacology , Hepatocytes/drug effects , Humans , Oncostatin M/pharmacology , Ornithine Carbamoyltransferase/genetics , Proline/pharmacology
13.
Nutrients ; 7(2): 1333-51, 2015 Feb 16.
Article in English | MEDLINE | ID: mdl-25690421

ABSTRACT

The aim of this study was to evaluate the osteoprotective effect of aqueous Rhizoma Dioscoreae extract (RDE) on the alveolar bone of rats with ovariectomy-induced bone loss. Female Wistar rats underwent either ovariectomy or sham operation (SHAM). The ovariectomized (OVX) rats were treated with vehicle (OVX), estradiol valerate (EV), or RDE. After treatments, the bone mineral density (BMD) and the three-dimensional microarchitecture of the alveolar bone were analyzed to assess bone mass. Microarrays were used to evaluate microRNA expression profiles in alveolar bone from RDE-treated and OVX rats. The differential expression of microRNAs was validated using real-time quantitative RT-PCR (qRT-PCR), and the target genes of validated microRNAs were predicted and further analyzed using Ingenuity Pathway Analysis (IPA). The key findings were verified using qRT-PCR. Our results show that RDE inhibits alveolar bone loss in OVX rats. Compared to the OVX rats, the RDE-treated rats showed upregulated expression levels of 8 microRNAs and downregulated expression levels of 8 microRNAs in the alveolar bone in the microarray analysis. qRT-PCR helped validate 13 of 16 differentially expressed microRNAs, and 114 putative target genes of the validated microRNAs were retrieved. The IPA showed that these putative target genes had the potential to code for proteins that were involved in the transforming growth factor (TGF)-ß/bone morphogenetic proteins (BMPs)/Smad signaling pathway (Tgfbr2/Bmpr2, Smad3/4/5, and Bcl-2) and interleukin (IL)-6/oncostatin M (OSM)/Jak1/STAT3 signaling pathway (Jak1, STAT3, and Il6r). These experiments revealed that RDE could inhibit ovariectomy-induced alveolar bone loss in rats. The mechanism of this anti-osteopenic effect in alveolar bone may involve the simultaneous inhibition of bone formation and bone resorption, which is associated with modulation of the TGF-ß/BMPs/Smad and the IL-6/OSM/Jak1/STAT3 signaling pathways via microRNA regulation.


Subject(s)
Alveolar Bone Loss/diet therapy , Bone Density/drug effects , Dioscorea , MicroRNAs/drug effects , Phytotherapy/methods , Plant Preparations/pharmacology , Alveolar Bone Loss/metabolism , Animals , Bone Morphogenetic Proteins/metabolism , Estradiol/administration & dosage , Estradiol/analogs & derivatives , Estradiol/pharmacology , Female , Interleukin-6/metabolism , Janus Kinase 1/metabolism , MicroRNAs/metabolism , Oncostatin M/metabolism , Ovariectomy/adverse effects , Plant Preparations/administration & dosage , Rats , Rats, Wistar , STAT3 Transcription Factor/metabolism , Transforming Growth Factor beta/metabolism
14.
Korean J Intern Med ; 29(5): 647-55, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25228841

ABSTRACT

BACKGROUND/AIMS: SKI306X, a mixed extract of three herbs, Clematis mandshurica (CM), Prunella vulgaris (PV), and Trichosanthes kirilowii (TK), is chondroprotective in animal models of osteoarthritis (OA). The objectives of this study were to investigate its effect on interleukin (IL)-1ß-induced degradation of glycosaminoglycan (GAG) and the basis of its action in human OA cartilage, as well as to screen for the presence of inhibitors of matrix metalloproteinase (MMP)-13 and a disintegrin and metalloprotease with thrombospondin motifs (ADAMTS)-4 in SKI306X and its component herbs, as well as in fractions from SKI306X. METHODS: Human OA chondrocytes and cartilage explants were obtained during total knee replacements and incubated with IL-1ß ± oncostatin M with or without SKI306X or its component herb extracts. GAG degradation was assayed in cartilage explants using a commercial kit. Expression of genes involved in cartilage destruction was measured by real-time polymerase chain reaction using chondrocyte RNA. SKI306X was fractionated by preparative liquid chromatography to test for the presence of inhibitors of MMP-13 and ADAMTS-4. RESULTS: SKI306X and PV inhibited IL-1ß-induced GAG release from cartilage explants, and SKI306X, CM, PV, and TK inhibited IL-1ß-induced MMP gene expression. Unexpectedly, SKI306X greatly stimulated IL-1ß + oncostatin M-induced ADAMTS-4 gene expression, probably due to its TK component. Some fractions of SKI306X also inhibited ADAMTS-4 activity. CONCLUSIONS: SKI306X and its herbal components inhibit GAG degradation and catabolic gene expression in human OA chondrocytes and cartilage explants. SKI306X likely also contains one or more ADAMTS-4 inhibitor.


Subject(s)
Cartilage, Articular/drug effects , Cartilage, Articular/metabolism , Drugs, Chinese Herbal/pharmacology , Glycosaminoglycans/metabolism , ADAM Proteins/antagonists & inhibitors , ADAMTS4 Protein , Cells, Cultured , Chondrocytes/drug effects , Chondrocytes/metabolism , Down-Regulation/drug effects , Humans , Interleukin-1beta/metabolism , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase Inhibitors/pharmacology , Oncostatin M/metabolism , Osteoarthritis, Knee/drug therapy , Osteoarthritis, Knee/genetics , Osteoarthritis, Knee/metabolism , Procollagen N-Endopeptidase/antagonists & inhibitors
15.
Hepatology ; 60(5): 1727-40, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24585441

ABSTRACT

UNLABELLED: In severe liver injury, ductular reactions (DRs) containing bipotential hepatic progenitor cells (HPCs) branch from the portal tract. Neural cell adhesion molecule (NCAM) marks bile ducts and DRs, but not mature hepatocytes. NCAM mediates interactions between cells and surrounding matrix; however, its role in liver development and regeneration is undefined. Polysialic acid (polySia), a unique posttranslational modifier of NCAM, is produced by the enzymes, ST8SiaII and ST8SiaIV, and weakens NCAM interactions. The role of polySia with NCAM synthesizing enzymes ST8SiaII and ST8SiaIV were examined in HPCs in vivo using the choline-deficient ethionine-supplemented and 3,5-diethoxycarbonyl-1,4-dihydrocollidine diet models of liver injury and regeneration, in vitro using models of proliferation, differentiation, and migration, and by use of mouse models with gene defects in the polysialyltransferases (St8sia 2+/-4+/-, and St8sia2-/-4-/-). We show that, during liver development, polySia is required for the correct formation of bile ducts because gene defects in both the polysialyltransferases (St8sia2+/-4+/- and St8sia2-/-4-/- mice) caused abnormal bile duct development. In normal liver, there is minimal polySia production and few ductular NCAM+ cells. Subsequent to injury, NCAM+ cells expand and polySia is produced by DRs/HPCs through ST8SiaIV. PolySia weakens cell-cell and cell-matrix interactions, facilitating HGF-induced migration. Differentiation of HPCs to hepatocytes in vitro results in both transcriptional down-regulation of polySia and cleavage of polySia-NCAM. Cleavage of polySia by endosialidase (endoN) during liver regeneration reduces migration of DRs into parenchyma. CONCLUSION: PolySia modification of NCAM+ ductules weakens cell-cell and cell-matrix interactions, allowing DRs/HPCs to migrate for normal development and regeneration. Modulation of polySia levels may provide a therapeutic option in liver regeneration.


Subject(s)
Liver Regeneration , Neural Cell Adhesion Molecules/metabolism , Sialic Acids/metabolism , Animals , Bile Ducts, Intrahepatic/growth & development , Cell Differentiation , Cell Movement , Coculture Techniques , Hepatocytes/cytology , Male , Mice , Mice, Inbred C57BL , Myofibroblasts/metabolism , Neuraminidase , Oncostatin M , Stem Cells/physiology
16.
Article in English | WPRIM | ID: wpr-108336

ABSTRACT

BACKGROUND/AIMS: SKI306X, a mixed extract of three herbs, Clematis mandshurica (CM), Prunella vulgaris (PV), and Trichosanthes kirilowii (TK), is chondroprotective in animal models of osteoarthritis (OA). The objectives of this study were to investigate its effect on interleukin (IL)-1beta-induced degradation of glycosaminoglycan (GAG) and the basis of its action in human OA cartilage, as well as to screen for the presence of inhibitors of matrix metalloproteinase (MMP)-13 and a disintegrin and metalloprotease with thrombospondin motifs (ADAMTS)-4 in SKI306X and its component herbs, as well as in fractions from SKI306X. METHODS: Human OA chondrocytes and cartilage explants were obtained during total knee replacements and incubated with IL-1beta +/- oncostatin M with or without SKI306X or its component herb extracts. GAG degradation was assayed in cartilage explants using a commercial kit. Expression of genes involved in cartilage destruction was measured by real-time polymerase chain reaction using chondrocyte RNA. SKI306X was fractionated by preparative liquid chromatography to test for the presence of inhibitors of MMP-13 and ADAMTS-4. RESULTS: SKI306X and PV inhibited IL-1beta-induced GAG release from cartilage explants, and SKI306X, CM, PV, and TK inhibited IL-1beta-induced MMP gene expression. Unexpectedly, SKI306X greatly stimulated IL-1beta + oncostatin M-induced ADAMTS-4 gene expression, probably due to its TK component. Some fractions of SKI306X also inhibited ADAMTS-4 activity. CONCLUSIONS: SKI306X and its herbal components inhibit GAG degradation and catabolic gene expression in human OA chondrocytes and cartilage explants. SKI306X likely also contains one or more ADAMTS-4 inhibitor.


Subject(s)
Humans , ADAM Proteins/antagonists & inhibitors , Cartilage, Articular/drug effects , Cells, Cultured , Chondrocytes/drug effects , Down-Regulation/drug effects , Drugs, Chinese Herbal/pharmacology , Glycosaminoglycans/metabolism , Interleukin-1beta/metabolism , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase Inhibitors/pharmacology , Oncostatin M/metabolism , Osteoarthritis, Knee/drug therapy , Procollagen N-Endopeptidase/antagonists & inhibitors
17.
Differentiation ; 86(1-2): 65-74, 2013.
Article in English | MEDLINE | ID: mdl-23995451

ABSTRACT

Oncostatin M (OSM), one of the IL-6 family cytokines, inhibits adipogenic differentiation and stimulates osteoblastogenic differentiation from human bone marrow mesenchymal stem cells (hBMSCs). This functional study of OSM enabled us to develop a two-dimensional small-molecule screen that shifts hBMSC differentiation from adipocyte to osteoblast. Several structurally related compounds (isoxazoles) inhibited the accumulation of intracellular lipid droplets, whereas they promoted alkaline phosphatase activity and extracellular matrix calcification. Isoxazoles also reduced the expression of adipogenic transcription factor PPARγ and increased the levels of osteogenic transcription factors Runx2 and Osterix. They also induced the expression of the Wnt/ß-catenin downstream gene and TOPflash reporter; however, the dephosphorylated ß-catenin-active form was not significantly increased. Interestingly, the slight modification of the active compound led to a complete reversion of the dual differentiation activities. In summary, we have identified isoxazoles with anti-adipogenic and pro-osteogenic activities that provide a potential new tool for exploring the lineage commitment of mesenchymal stem cells and a possible lead for therapeutic intervention in osteopenia and osteoporosis.


Subject(s)
Adipogenesis/drug effects , Isoxazoles/pharmacology , Osteogenesis/drug effects , Small Molecule Libraries/pharmacology , Alkaline Phosphatase/metabolism , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Drug Evaluation, Preclinical , HEK293 Cells , High-Throughput Screening Assays , Humans , Isoxazoles/isolation & purification , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Oncostatin M/pharmacology , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , PPAR gamma/genetics , PPAR gamma/metabolism , Sp7 Transcription Factor , Transcription Factors/genetics , Transcription Factors/metabolism , beta Catenin/genetics , beta Catenin/metabolism
18.
J Endod ; 38(4): 475-80, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22414832

ABSTRACT

INTRODUCTION: We have previously differentiated hepatocyte like cells from deciduous tooth pulp stem and extracted third molar pulp stem cells with a protocol that used fetal bovine serum, but it showed high contaminations of nondifferentiated cells. Both the lower purity of hepatically differentiated cells and usage of serum are obstacles for application of cell therapy or regenerative medicine. Objective of this study was to investigate the capacity for and purity of hepatocyte-like differentiation of CD117-positive dental pulp stem cells without serum. METHODS: Mesenchymal cells from human deciduous and extracted third molar pulp were isolated and expanded in vitro. We separated CD117-positive cells by using a magnetic-activated cell sorter. The cells were characterized immunofluorescently by using known stem cell markers. For hepatic differentiation, the media were supplemented with hepatic growth factor, insulin-transferrin-selenium-x, dexamethasone, and oncostatin M. Expression of hepatic markers alpha fetoprotein, albumin, hepatic nuclear factor-4 alpha, insulin-like growth factor-1, and carbamoyl phosphate synthetase was examined immunofluorescently after differentiation. The amount of differentiated cells was assessed by using flow cytometry. Glycogen storage and urea concentration in the medium were defined. RESULTS: Both cell cultures demonstrated a number of cells positive for all tested hepatic markers after differentiation, ie, albumin-positive cells were almost 90% of differentiated deciduous pulp cells. The concentration of urea in the media increased significantly after differentiation. Significant amount of cytoplasmic glycogen storage was found in the cells. CONCLUSIONS: Without serum both cell types differentiated into high-purity hepatocyte-like cells. These cells offer a source for hepatocyte lineage differentiation for transplantation in the future.


Subject(s)
Cell Culture Techniques/methods , Dental Pulp/cytology , Hepatocytes/physiology , Stem Cells/physiology , Biomarkers/analysis , Carbamoyl-Phosphate Synthase (Ammonia)/analysis , Cell Differentiation , Cell Lineage , Culture Media, Serum-Free , Dexamethasone/pharmacology , Flow Cytometry , Fluorescent Antibody Technique , Glucocorticoids/pharmacology , Glycogen/analysis , Growth Inhibitors/pharmacology , Hepatocyte Growth Factor/pharmacology , Hepatocyte Nuclear Factor 4/analysis , Humans , Insulin/pharmacology , Insulin-Like Growth Factor I/analysis , Oncostatin M/pharmacology , Proto-Oncogene Proteins c-kit/analysis , Selenium/pharmacology , Serum Albumin/analysis , Transferrin/pharmacology , Urea/analysis , alpha-Fetoproteins/analysis
19.
Int Immunopharmacol ; 11(6): 670-4, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21255696

ABSTRACT

CXC chemokine ligand 10 (CXCL10) plays an important role in the infiltration of Th1 cells and thus in the exacerbation of periodontal disease. Theaflavin-3,3'-digallate (TFDG), polyphenol in black tea, has some beneficial effects but the effect of TFDG on CXCL10 production from human gingival fibroblasts (HGFs) is uncertain. In this study, we investigated the mechanisms by which TFDG may inhibit oncostatin M (OSM)-induced CXCL10 production in human gingival fibroblasts. TFDG prevented OSM-mediated CXCL10 production by HGFs in a dose dependent manner. TFDG significantly inhibited OSM-induced phosphorylation of c-Jun N terminal kinase (JNK), protein kinase B (Akt) (Ser473) that are related to CXCL10 production from OSM-stimulated HGFs. In addition, TFDG suppressed OSM receptor (OSMR) ß expression on HGFs. These data provide a novel mechanism where the black tea flavonoid, theaflavin, could provide direct benefits in periodontal disease.


Subject(s)
Biflavonoids/pharmacology , Catechin/pharmacology , Fibroblasts/metabolism , Flavonoids/pharmacology , Gallic Acid/analogs & derivatives , Gingiva/pathology , Periodontal Diseases/drug therapy , Phenols/pharmacology , Cells, Cultured , Chemokine CXCL10/genetics , Chemokine CXCL10/metabolism , Fibroblasts/drug effects , Fibroblasts/pathology , Gallic Acid/pharmacology , Humans , MAP Kinase Kinase 4/metabolism , Oncostatin M/immunology , Oncostatin M/metabolism , Oncostatin M Receptor beta Subunit/genetics , Oncostatin M Receptor beta Subunit/metabolism , Periodontal Diseases/immunology , Phosphorylation/drug effects , Polyphenols , Proto-Oncogene Proteins c-akt/metabolism , STAT3 Transcription Factor/metabolism , Tea
20.
Mol Endocrinol ; 24(4): 745-53, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20197307

ABSTRACT

Hyperbilirubinemia remains a common condition in neonates. The constitutive androstane receptor (CAR) is an orphan nuclear receptor that has been shown to participate in the activation of the uridine diphosphate-5'-glucuronosyltransferase 1A1 (UGT1A1) gene, which plays an important role in bilirubin clearance. Oncostatin M (OSM), a member of the IL-6 family, is involved in the maturation of fetal hepatocytes. We have demonstrated that low OSM levels are a potential indicator of neonatal jaundice and the need for phototherapy. In this study we examined the effects of OSM on CAR-mediated signaling to investigate its potential role in neonatal jaundice via the CAR-UGT1A1 pathway. We observed that OSM positively augmented the CAR and UGT1A1 expressions and CAR-mediated signaling in vivo and in vitro, through cross talk between the nuclear CAR receptor and the plasma membrane OSM receptor, via the MAPK cascade. These data suggest that OSM might play a role in bilirubin metabolism via the CAR-UGT1A1 pathway.


Subject(s)
Glucuronosyltransferase/metabolism , Oncostatin M/pharmacology , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Animals, Newborn , Blotting, Western , Constitutive Androstane Receptor , Cytochrome P-450 CYP3A/genetics , Cytochrome P-450 CYP3A/metabolism , Flavonoids/pharmacology , Glucuronosyltransferase/genetics , Hep G2 Cells , Humans , Infant, Newborn , Jaundice, Neonatal/metabolism , Mice , Mice, Inbred ICR , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Oximes/pharmacology , Phenobarbital/pharmacology , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics , Thiazoles/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL