Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Biochim Biophys Acta Mol Basis Dis ; 1870(6): 167250, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38763409

ABSTRACT

Despite considerable therapeutic advancements, the global survival rate for lung cancer patients remains poor, posing challenges in developing an effective treatment strategy. In many cases, microRNAs (miRNAs) exhibit abnormal expression levels in cancers, including lung cancer. Dysregulated miRNAs often play a crucial role in the development and progression of cancer. Therefore, understanding the mechanisms underlying aberrant miRNA expression during carcinogenesis may provide crucial clues to develop novel therapeutics. In this study, we identified and cloned a novel miRNA, hsa-miR-CHA2, which is abnormally downregulated in non-small cell lung cancer (NSCLC)-derived cell lines and tissues of patients with NSCLC. Furthermore, we found that hsa-miR-CHA2 regulates the post-transcriptional levels of Cyclin E1 (CCNE1) by binding to the 3'-UTR of CCNE1 mRNA. CCNE1, a cell cycle regulator involved in the G1/S transition, is often amplified in various cancers. Notably, hsa-miR-CHA2 overexpression led to the alteration of the Rb-E2F pathway, a significant signaling pathway in the cell cycle, by targeting CCNE1 in A549 and SK-LU-1 cells. Subsequently, we confirmed that hsa-miR-CHA2 induced G1-phase arrest and exhibited an anti-proliferative effect by targeting CCNE1. Moreover, in subcutaneous xenograft mouse models, intra-tumoral injection of polyplexed hsa-miR-CHA2 mimic suppressed tumor growth and development. In conclusion, hsa-miR-CHA2 exhibited an anticancer effect by targeting CCNE1 both in vitro and in vivo. These findings suggest the potential role of hsa-miR-CHA2 as an important regulator of cell proliferation in molecular-targeted therapy for NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Cyclin E , Gene Expression Regulation, Neoplastic , Lung Neoplasms , MicroRNAs , Oncogene Proteins , Humans , Cyclin E/genetics , Cyclin E/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Animals , Mice , Cell Proliferation/genetics , Cell Line, Tumor , A549 Cells , Mice, Nude , Xenograft Model Antitumor Assays , 3' Untranslated Regions/genetics , Mice, Inbred BALB C , Signal Transduction
2.
Int J Mol Sci ; 25(5)2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38474002

ABSTRACT

Psoriasis is a chronic inflammatory skin disease characterized by the rapid abnormal growth of skin cells in the epidermis, driven by an overactive immune system. Consequently, a complex interplay among epidermal cells, immune cells, and sensory neurons contributes to the development and progression of psoriasis. In these cellular contexts, various ion channels, such as acetylcholine receptors, TRP channels, Ca2+ release-activated channels, chloride channels, and potassium channels, each serve specific functions to maintain the homeostasis of the skin. The dysregulation of ion channels plays a major role in the pathophysiology of psoriasis, affecting various aspects of epidermal cells, immune responses, and sensory neuron signaling. Impaired function of ion channels can lead to altered calcium signaling, inflammation, proliferation, and sensory signaling, all of which are central features of psoriasis. This overview summarizes the pathophysiological roles of ion channels in epidermal cells, immune cells, and sensory neurons during early and late psoriatic processes, thereby contributing to a deeper understanding of ion channel involvement in the interplay of psoriasis and making a crucial advance toward more precise and personalized approaches for psoriasis treatment.


Subject(s)
Keratinocytes , Psoriasis , Humans , Keratinocytes/physiology , Epidermis , Epidermal Cells , Sensory Receptor Cells , Ion Channels
3.
Int J Mol Sci ; 23(13)2022 Jun 30.
Article in English | MEDLINE | ID: mdl-35806296

ABSTRACT

Excessive use of alcohol can induce neurobiological and neuropathological alterations in the brain, including the hippocampus and forebrain, through changes in neurotransmitter systems, hormonal systems, and neuroimmune processes. We aimed to investigate the effects of ethanol on the expression of coding and noncoding RNAs in a brain-derived cell line exposed to ethanol. After exposing Neuro2a cells, a neuroblastoma cell line, to ethanol for 24 and 72 h, we observed cell proliferation and analyzed up- and downregulated mRNAs and long noncoding RNAs (lncRNAs) using total RNA-Seq technology. We validated the differential expression of some mRNAs and lncRNAs by RT-qPCR and analyzed the expression of Cebpd and Rnu3a through knock-down of Cebpd. Cell proliferation was significantly reduced in cells exposed to 100 mM ethanol for 72 h, with 1773 transcripts up- or downregulated by greater than three-fold in ethanol-treated cells compared to controls. Of these, 514 were identified as lncRNAs. Differentially expressed mRNAs and lncRNAs were mainly observed in cells exposed to ethanol for 72 h, in which Atm and Cnr1 decreased, but Trib3, Cebpd, and Spdef increased. On the other hand, lncRNAs Kcnq1ot1, Tug1, and Xist were changed by ethanol, and Rnu3a in particular was greatly increased by chronic ethanol treatment through inhibition of Cebpd. Our results increase the understanding of cellular and molecular mechanisms related to coding and noncoding RNAs in an in vitro model of acute and chronic exposure to ethanol.


Subject(s)
Neuroblastoma , RNA, Long Noncoding , Animals , Cell Proliferation , Ethanol/pharmacology , Gene Expression Profiling/methods , Mice , Neuroblastoma/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
4.
Int J Mol Sci ; 22(13)2021 Jul 01.
Article in English | MEDLINE | ID: mdl-34281197

ABSTRACT

Psoriasis, a long-lasting and multifactorial skin disease, is related to comorbidities such as metabolic disease, depression, and psoriatic arthritis. Psoriasis occurs due to a variety of factors including keratinocyte hyperproliferation, inflammation, and abnormal differentiation. Proinflammatory cytokines upregulated by increased activation of keratinocytes and immune cells in the skin trigger progression of psoriasis. This study aimed to investigate the effects of anoctamin1 (ANO1) on psoriasis development in vitro and in vivo. We analyzed the proliferation of HaCaT keratinocytes and ANO1-related ERK and AKT signaling pathways after ANO1 inhibitor (T16Ainh-A01 and Ani9) treatment and knock-down of ANO1. Furthermore, after applying imiquimod (IMQ) cream or coapplying IMQ cream and T16Ainh-A01 on mouse ears, we not only observed psoriatic symptoms, including ear thickening, but also quantified the effects of treatment on ERK and AKT signaling-involved proteins and proinflammatory cytokines. Inhibition of ANO1 attenuated the proliferation of HaCaT cells and induced reduction of pERK1/2. Coapplication of IMQ and T16Ainh-A01 on ears of mice reduced not only symptoms of IMQ-induced psoriasis such as thickening and erythema, but also expression of ANO1 and pERK1/2 compared to that of application of IMQ alone. In addition, the expression levels of IL-17A, IL-17F, IL-22, IL-23, IL-6, IL-1ß, and TNF-α increased after applying IMQ and were significantly reduced by coapplying IMQ and T16Ainh-A01. These results aid in understanding the underlying mechanisms of ANO1 in epidermal layer keratinocyte hyperproliferation and suggest the potential of ANO1 as a target to treat psoriasis.


Subject(s)
Anoctamin-1/pharmacology , Keratinocytes/drug effects , Psoriasis/chemically induced , Acetamides/pharmacology , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cytokines/metabolism , Disease Models, Animal , Female , HaCaT Cells , Humans , Hydrazones/pharmacology , Imiquimod/adverse effects , Imiquimod/pharmacology , Inflammation/metabolism , Inflammation/pathology , Interleukins/metabolism , Keratinocytes/metabolism , Keratinocytes/pathology , MAP Kinase Signaling System , Mice , Mice, Inbred BALB C , Psoriasis/metabolism , Psoriasis/pathology , Pyrimidines/pharmacology , Thiazoles/pharmacology
5.
Molecules ; 26(3)2021 Jan 26.
Article in English | MEDLINE | ID: mdl-33530645

ABSTRACT

The research on resveratrol (1) has been conducted intensively over a long time due to its proven antioxidant activity and disease-fighting capabilities. Many efforts have also been made to increase these biological effects. In the present study, six new extended aromatic resveratrol analogues containing naphthalene (2) and its bioisosteres quinoline (3 and 4), isoquinoline (5) quinoxaline (6) and quinazoline (7) scaffolds were designed and synthesized using an annulation strategy. The antioxidant and anti-inflammatory activities of these compounds were investigated. All compounds showed better antioxidant activity than resveratrol in ABTS assay. As for the anti-inflammatory test, 5 and 7 exhibited better activity than resveratrol. It is worth noting that nitrogen substitution on the extended aromatic resveratrol analogues has a significant impact on cell viability. Taking the antioxidant activities and NO inhibition activities into consideration, we conclude that isoquinoline analogue 5 may qualify for the further investigation of antioxidant and anti-inflammatory therapy. Furthermore, our study results suggest that in order to improve the biological activity of polyphenolic compounds, extended aromaticity and nitrogen substitution strategy could be a viable method for the design of future drug candidates.


Subject(s)
Antioxidants/chemical synthesis , Interleukin-1beta/genetics , Interleukin-6/genetics , Lipopolysaccharides/adverse effects , Resveratrol/analogs & derivatives , Stilbestrols/chemical synthesis , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Antioxidants/chemistry , Antioxidants/pharmacology , Cell Survival/drug effects , Gene Expression Regulation/drug effects , Isoquinolines/chemistry , Mice , Naphthalenes/chemistry , Quinazolines/chemistry , Quinolines/chemistry , Quinoxalines/chemistry , RAW 264.7 Cells , Resveratrol/chemistry , Stilbestrols/chemistry , Stilbestrols/pharmacology , Structure-Activity Relationship
6.
Int Immunopharmacol ; 86: 106553, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32563057

ABSTRACT

Although tacrolimus (FK-506) has been shown to be an effective monotherapy for psoriasis, it does not always work well. Currently, combination therapy is frequently used to manage psoriasis because clinical trials have shown it may provide additive or synergistic benefits and reduce risks of adverse effects. Myeloid-derived suppressor cells (MDSCs) have potent immunomodulatory and anti-inflammatory properties in autoimmune diseases. We previously reported that MDSCs had protective effects in a murine model of imiquimod (IMQ)-induced psoriasis. The present study was undertaken to investigate the systemic immunomodulatory and therapeutic efficacy effects of MDSC plus FK-506 in an IMQ-induced mouse model of psoriasis and to investigate the immunomodulatory mechanisms involved. Systemic MDSC plus FK-506 therapy was found to have a significant anti-psoriatic effect in the murine model, to reduce levels of pro-inflammatory cytokines Th1 cytokines (TNF-α and IFN-γ) and Th17 cytokines (IL-17A and IL-23) in serum and skin. However, treatment with MDSCs or FK-506 alone had little impact. Furthermore, the anti-psoriatic effects of MDSC plus FK-506 were associated with histopathological reductions in inflammatory infiltration, epidermal hyperplasia, and hyperkeratosis. In addition, this combined treatment also attenuated IMQ-induced splenomegaly, and increased the proportion of CD4+CD25+FoxP3+ regulatory T (Treg) cells and decreased the proportions of CD4+IFN-γ+ Th1 cells and CD4+IL-17+ Th17 cells in spleen. Taken together, our results show systemic combination therapy with MDSCs and FK-506 had a better therapeutic effect in our IMQ-induced psoriasis model than either agent alone, and suggest that this combinatorial therapy might be useful for the management of autoimmune skin diseases like psoriasis.


Subject(s)
Immunosuppressive Agents/pharmacology , Myeloid-Derived Suppressor Cells/immunology , Psoriasis/drug therapy , Tacrolimus/pharmacology , Animals , Cytokines/metabolism , Disease Models, Animal , Drug Therapy, Combination , Female , Imiquimod/toxicity , Immunomodulation/drug effects , Immunosuppressive Agents/therapeutic use , Mice, Inbred C57BL , Psoriasis/chemically induced , Psoriasis/pathology , Skin/metabolism , Skin/pathology , Spleen/drug effects , T-Lymphocytes, Regulatory/drug effects , Tacrolimus/therapeutic use , Th1 Cells/drug effects , Th17 Cells/drug effects
7.
Lung Cancer ; 132: 99-106, 2019 06.
Article in English | MEDLINE | ID: mdl-31097102

ABSTRACT

OBJECTIVES: MicroRNAs have critical roles in cancer development by regulating the expression of oncogenes or tumor suppressor genes. We identified and characterized a novel miRNA, miR-CHA1, in human lung cancer cells. The aim of this study was to investigate its novel function in human lung cancer by targeting XIAP. MATERIAL AND METHODS: Novel miRNA cloning, Real-time qRT-PCR, western blotting, dual luciferase assay, miRNA transfection, proliferation and apoptosis assay were carried on human lung cancer cell line A549. Fifteen paired NSCLC tissues and noncancerous lung tissues were collected. In vivo xenograft assay was performed. RESULTS: Expression of miR-CHA1 was downregulated in human lung cancer cell lines and tissues compared with normal cells and tissues. We identified a putative target gene, XIAP, whose expression was regulated by miR-CHA1 overexpression. XIAP is an inhibitor of apoptosis that represses the activation of caspase 3 and 9. XIAP mRNA and protein levels were directly suppressed by miR-CHA1. XIAP has an important role in carcinogenesis, and previous studies suggest that it may regulate cell survival and proliferation by its anti-apoptotic ability. CONCLUSION: Taken together, miR-CHA1 inhibited cell proliferation and induced apoptosis in vitro and in vivo by targeting XIAP. These data can be applied to identify novel therapeutic targets for lung cancer therapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/genetics , MicroRNAs/genetics , X-Linked Inhibitor of Apoptosis Protein/metabolism , A549 Cells , Animals , Apoptosis , Carcinogenesis/genetics , Caspase 3/metabolism , Caspase 9/metabolism , Cell Proliferation , Cloning, Molecular , Gene Expression Regulation, Neoplastic , Humans , Male , Mice, Nude , X-Linked Inhibitor of Apoptosis Protein/genetics , Xenograft Model Antitumor Assays
8.
Scand J Immunol ; 89(3): e12742, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30548969

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) play an important role in controlling the immune response against cancer and in suppression of autoimmunity and allergic inflammation. However, the beneficial effects of MDSCs on the experimental mouse model of psoriasis have not been reported. Therefore, we investigated the anti-psoriatic effect of MDSCs on IMQ-induced skin inflammation in mice and explored the mechanisms involved. Our results showed that administration of MDSCs (1 × 106 or 2 × 106  cells) suppressed the development of IMQ-induced skin inflammation in mice as exemplified by a significant reduction in clinical severity scores and was associated with a reduction of histopathological changes, including inflammatory infiltration, epidermal hyperplasia and hyperkeratosis. The immunosuppressive effect of MDSCs (1 × 106 or 2 × 106  cells) corresponded to the production of Th1 cytokines (TNF-α, IFN-γ) and Th17 cytokines (IL-17A and IL-23) in the serum and dorsal skin. Administration of MDSCs (1 × 106 or 2 × 106  cells) also inhibited splenomegaly. Moreover, an increased percentage of CD4+ CD25+ FoxP3+ regulatory T (Treg) cells and decreased percentage of Th1 and Th17 cells were found in mice treated with MDSCs. Taken together, these results imply that MDSCs have immunomodulatory and immunosuppressive effects on disease progression in a murine model of psoriasis and that MDSCs could be used in preventive or therapeutic strategies for the management of autoimmune inflammatory skin disorders, such as psoriasis.


Subject(s)
Imiquimod/pharmacology , Myeloid-Derived Suppressor Cells/physiology , Psoriasis/therapy , Animals , Cytokines/biosynthesis , Dermatitis/immunology , Dermatitis/pathology , Female , Mice , Mice, Inbred C57BL , Psoriasis/immunology , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Th17 Cells/immunology
9.
Cell Death Dis ; 9(5): 493, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29712908

ABSTRACT

Lung cancer cases are increasing yearly; however, few novel therapeutic strategies for treating this disease have been developed. Here the dysregulation between microRNAs and oncogenes or tumour-suppressor genes forms a close connection-loop to the development or progression in human lung carcinogenesis. That is, the relationship between microRNAs and carcinogenic mechanism may find the critical clue to improve the treatment efficacy. Accordingly, we identified and characterised a novel microRNA, hsa-miR-12528, in A549 cells. The miR-12528 expression was aberrantly downregulated in cancer cell lines and in the patient tissues derived from human non-small cell lung cancer. In addition, we found that miR-12528 post-transcriptionally controls the translation of the insulin-like growth factor 1 receptor (IGF-1R) gene by directly targeting the 3'-untranslated region of IGF-1R mRNA. Notably, the IGF-1R gene is elevated in the majority of cancers and may be an attractive therapeutic target for anticancer therapy because elevated IGF-1R mediates the signalling amplification of a major oncogenic pathway in neoplasia. In A549 cells, miR-12528 overexpression epigenetically altered the downstream phosphorylation of the primary IGF-1R networks, negatively regulated proliferation, apoptosis and migratory activity, and consequently inhibited tumourigenesis and metastasis in vivo. Therefore, our discovery of hsa-miR-12528 may be able to be applied to the development of molecular-target therapeutic strategies and diagnosis-specific biomarkers for human lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung/enzymology , Cell Movement , Cell Proliferation , Lung Neoplasms/enzymology , MicroRNAs/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, IGF Type 1/metabolism , 3' Untranslated Regions , A549 Cells , Animals , Apoptosis , Binding Sites , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Cycle , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Neoplasm Invasiveness , Neoplasm Metastasis , Phosphorylation , Receptor, IGF Type 1/genetics , Signal Transduction
10.
J Immunol ; 185(11): 7037-46, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-20980634

ABSTRACT

Galectin-3, a ß-galactoside-binding lectin, has been proposed to have multifaceted functions in various pathophysiological conditions. However, the characteristics of galectin-3 and its molecular mechanisms of action are still largely unknown. In this study, we show that galectin-3 exerts cytokine-like regulatory actions in rat and mouse brain-resident immune cells. Both the expression of galectin-3 and its secretion into the extracellular compartment were significantly enhanced in glia under IFN-γ-stimulated, inflamed conditions. After exposure to galectin-3, glial cells produced high levels of proinflammatory mediators and exhibited activated properties. Notably, within minutes after exposure to galectin-3, JAK2 and STAT1, STAT3, and STAT5 showed considerable enhancement of tyrosine phosphorylation; thereafter, downstream events of STAT signaling were also significantly enhanced. Treatment of the cells with pharmacological inhibitors of JAK2 reduced the galectin-3-stimulated increases of inflammatory mediators. Using IFN-γ receptor 1-deficient mice, we further found that IFN-γR 1 might be required for galectin-3-dependent activation of the JAK-STAT cascade. However, galectin-3 significantly induced phosphorylation of STATs in glial cells from IFN-γ-deficient mice, suggesting that IFN-γ does not mediate activation of STATs. Collectively, our findings suggest that galectin-3 acts as an endogenous danger signaling molecule under pathological conditions in the brain, providing a potential explanation for the molecular basis of galectin-3-associated pathological events.


Subject(s)
Cytokines/physiology , Galectin 3/physiology , Janus Kinase 2/physiology , STAT1 Transcription Factor/physiology , STAT3 Transcription Factor/physiology , Signal Transduction/immunology , Animals , Astrocytes/immunology , Astrocytes/metabolism , Astrocytes/pathology , Brain Diseases/immunology , Brain Diseases/metabolism , Brain Diseases/pathology , Brain Neoplasms/immunology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cells, Cultured , Cytokines/biosynthesis , Cytokines/metabolism , Galectin 3/biosynthesis , Galectin 3/metabolism , Inflammation Mediators/metabolism , Inflammation Mediators/physiology , Interferon-gamma/physiology , Janus Kinase 2/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microglia/immunology , Microglia/metabolism , Microglia/pathology , Phosphorylation/immunology , Rats , Rats, Sprague-Dawley , Receptors, Interferon/deficiency , Receptors, Interferon/genetics , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism , Interferon gamma Receptor
11.
Korean J Physiol Pharmacol ; 13(4): 265-71, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19885009

ABSTRACT

Nitric oxide (NO) has both neuroprotective and neurotoxic effects depending on its concentration and the experimental model. We tested the effects of NG-nitro-L-arginine methyl ester (L-NAME), a nonselective nitric oxide synthase (NOS) inhibitor, and aminoguanidine, a selective inducible NOS (iNOS) inhibitor, on kainic acid (KA)-induced seizures and hippocampal CA3 neuronal death. L-NAME (50 mg/kg, i.p.) and/or aminoguanidine (200 mg/kg, i.p.) were administered 1 h prior to the intracerebroventricular (i.c.v.) injection of KA. Pretreatment with L-NAME significantly increased KA-induced CA3 neuronal death, iNOS expression, and activation of microglia. However, pretreatment with aminoguanidine significantly suppressed both the KA-induced and L-NAME-aggravated hippocampal CA3 neuronal death with concomitant decreases in iNOS expression and microglial activation. The protective effect of aminoguanidine was maintained for up to 2 weeks. Furthermore, iNOS knockout mice (iNOS(-/-)) were resistant to KA-induced neuronal death. The present study demonstrates that aminoguanidine attenuates KA-induced neuronal death, whereas L-NAME aggravates neuronal death, in the CA3 region of the hippocampus, suggesting that NOS isoforms play different roles in KA-induced excitotoxicity.

12.
Biomed Mater ; 4(4): 044106, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19671952

ABSTRACT

In this study, we prepared a blended nanofiber scaffold using synthetic and natural polymers, polyurethane (PU) and gelatin respectively, using the electrospinning method to prepare a material for wound dressing. In order to confirm the properties of this gelatin/PU blended nanofiber scaffold, we performed scanning electron microscopy, atomic force microscopy, attenuated total reflectance Fourier-transform infrared spectroscopy, thermal gravimetric analysis, contact angle, water uptake, mechanical property, recovery, and degradation tests, and cellular response. The results obtained indicate that the mean diameter of these nanofibers was uniformly electrospun and ranged from 0.4 to 2.1 microm. According to the results, when the amount of gelatin in the blended solution decreased, the contact angle increased and water uptake of the scaffold decreased concurrently. In the mechanical tests, the blended nanofibrous scaffolds were elastic, and elasticity increased as the total amount of PU increased. Moreover, as the total amount of gelatin increased, the cell proliferation increased with the same amount of culture time. Therefore, this gelatin/PU blended nanofiber scaffold has potential application for use as a wound dressing.


Subject(s)
Gelatin/chemistry , Nanofibers/chemistry , Polymers/chemistry , Polyurethanes/chemistry , Wound Healing/physiology , Animals , Cell Proliferation , Gelatin/analysis , Mice , Microscopy, Atomic Force , Microscopy, Electron, Scanning , NIH 3T3 Cells , Spectroscopy, Fourier Transform Infrared
13.
Mol Cells ; 28(2): 119-24, 2009 Aug 31.
Article in English | MEDLINE | ID: mdl-19714313

ABSTRACT

Anti cancer agent 5-FU (Fluoro Uracil) is a prodrug that can be metabolized and then activated to interfere with RNA and DNA homeostasis. However, the majority of administered 5-FU is known to be catabolized in vivo in the liver where Dihydropyrimidine dehydrogenase (DPD) is abundantly expressed to degrade 5-FU. The biological factors that correlate with the response to 5-FU-based chemotherapy have been proposed to include uridine phosphorylase (UPP), thymidine phosphorylase (TPP), p53 and microsatellite instability. Among these, the expression of UPP is known to be controlled by cytokines such as TNF-alpha, IL1 and IFN-gamma. Our preliminary study using a DNA microarray technique showed that basic fibroblast growth factor (bFGF) markedly induced the expression of UPP1 at the transcription level. In the present study, we investigated whether bFGF could modulate the expression of UPP1 in osteo-lineage cells and examined the sensitivity of these cells to 5-FU mediated apoptosis.


Subject(s)
Apoptosis/drug effects , Fibroblast Growth Factor 2/pharmacology , Fluorouracil/pharmacology , Up-Regulation/drug effects , Uridine Phosphorylase/genetics , Animals , Antimetabolites, Antineoplastic/pharmacology , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Synergism , Flow Cytometry , Gene Expression Regulation, Enzymologic/drug effects , Humans , Myoblasts/cytology , Myoblasts/drug effects , Myoblasts/metabolism , Osteosarcoma/genetics , Osteosarcoma/pathology , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction
14.
Biologicals ; 37(4): 203-9, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19264508

ABSTRACT

Genetic modification of hematopoietic stem cells holds great promise in the treatment of hematopoietic disorders. However, clinical application of gene delivery has been limited, in part, by low gene transfer efficiency. To overcome this problem, we investigated the effect of retronectin (RN) on lentiviral-mediated gene delivery into hematopoietic progenitor cells (HPCs) derived from bone marrow both in vitro and in vivo. RN has been shown to enhance transduction by promoting colocalization of lentivirus and target cells. We found that RN enhanced lentiviral transfer of the VENUS transgene into cultured c-Kit(+) Lin(-) HPCs. As a complementary approach, in vivo gene delivery was performed by subjecting mice to intra-bone marrow injection of lentivirus or a mixture of RN and lentivirus. We found that co-injection with RN increased the number of VENUS-expressing c-Kit(+) Lin(-) HPCs in bone marrow by 2-fold. Further analysis of VENUS expression in colony-forming cells from the bone marrow of these animals revealed that RN increased gene delivery among these cells by 4-fold. In conclusion, RN is effective in enhancing lentivirus-mediated gene delivery into HPCs.


Subject(s)
Fibronectins/pharmacology , Gene Transfer Techniques , Hematopoietic Stem Cells/drug effects , Lentivirus/genetics , Recombinant Proteins/pharmacology , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cells, Cultured , Drug Evaluation, Preclinical , Fibronectins/chemistry , Hematopoietic Stem Cells/metabolism , Humans , Lentivirus/physiology , Mice , Mice, Inbred C57BL , Multipotent Stem Cells/drug effects , Multipotent Stem Cells/metabolism , Peptide Fragments/pharmacology , Protein Structure, Tertiary , Up-Regulation
15.
Arch Pharm Res ; 31(8): 978-82, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18787784

ABSTRACT

Eight compounds were isolated from the bark of Salix hulteni. Based on spectral data, the isolated compounds were identified as 4-hydroxyacetophenone (1), naringenin (2), aromadendrin (3), catechin (4), picein (5), sachaliside 1 (6), 1-p-coumaroyl-beta-D-glucoside (7), and dihydromyricetin (8). Their cytotoxic activities against brine shrimp and a human lung cancer cell line (H1299) were evaluated.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Salix/chemistry , Animals , Antineoplastic Agents, Phytogenic/isolation & purification , Artemia , Cell Line, Tumor , Cell Survival/drug effects , Chromatography, High Pressure Liquid , Chromatography, Ion Exchange , Chromatography, Thin Layer , Drug Screening Assays, Antitumor , Humans , Hydrolysis , Lung Neoplasms/drug therapy , Mass Spectrometry , Plant Bark/chemistry , Spectrophotometry, Infrared , Spectrophotometry, Ultraviolet , Spectroscopy, Fourier Transform Infrared , Tetrazolium Salts , Thiazoles
16.
J Korean Med Sci ; 22(2): 380-2, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17449956

ABSTRACT

Cervical spinal epidural abscess, caused by fish bone injury and a secondary infection by Eikenella corrodens which is part of the normal flora, has not been reported. A 72-yr-old man came to the hospital with pain in his posterior neck and both shoulders for 2 months. He also was experiencing weakness on his right side for 3 days. A fish bone had been stuck in his throat for about 2 months. Neurological examination revealed right hemiparesis, hypesthesia on the left extremities and neck stiffness. Laboratory findings showed an elevated ESR/CRP and leukocytosis, and magnetic resonance imaging revealed a retropharyngeal abscess and cervical myelitis. The patient was treated with emergency surgical decompression and antibiotics. A fish bone was removed from the C3-C4 intervertebral disc space. In the culture of chocolate blood agar and 5% sheep blood agar plate, E. corrodens was detected as a causative organism.


Subject(s)
Eikenella corrodens/isolation & purification , Epidural Abscess/etiology , Epidural Abscess/surgery , Food/adverse effects , Foreign Bodies/complications , Foreign Bodies/surgery , Gram-Negative Bacterial Infections/etiology , Aged , Animals , Anti-Bacterial Agents/administration & dosage , Bone and Bones , Decompression, Surgical , Epidural Abscess/diagnosis , Fishes , Gram-Negative Bacterial Infections/diagnosis , Gram-Negative Bacterial Infections/prevention & control , Humans , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...