Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(2)2024 Jan 22.
Article in English | MEDLINE | ID: mdl-38279326

ABSTRACT

Liver tumor organoids derived from liver tumor tissues and pluripotent stem cells are used for liver tumor research but have several challenges in primary cell isolation and stem cell differentiation. Here, we investigated the potential of HepG2-based liver tumor organoids for screening anticancer drugs by evaluating their responsiveness to IFN-ß produced by mesenchymal stem cells (MSCs). Liver tumor organoids were prepared in three days on Matrigel using HepG2, primary liver sinusoidal epithelial cells (LSECs), LX-2 human hepatic stellate cells, and THP-1-derived macrophages at a ratio of 4:4:1:1, with 105 total cells. Hepatocyte-related and M2 macrophage-associated genes increased in liver tumor organoids. IFN-ß treatment decreased the viability of liver tumor organoids and increased M1 macrophage marker expression (i.e., TNF-α and iNOS) and TRAIL. TRAIL expression was increased in all four cell types exposed to IFN-ß, but cell death was only observed in HepG2 cells and macrophages. Further, MSCs overexpressing IFN-ß (ASC-IFN-ß) also expressed TRAIL, contributing to the reduced viability of liver tumor organoids. In summary, IFN-ß or ASC-IFN-ß can induce TRAIL-dependent HepG2 and macrophage cell death in HepG2-based liver tumor organoids, highlighting these liver tumor organoids as suitable for anticancer drug screening and mechanistic studies.


Subject(s)
Interferon-beta , Liver Neoplasms , Humans , Apoptosis , Cell Death , Interferon-beta/pharmacology , Liver Neoplasms/metabolism , Macrophages/metabolism , Organoids/metabolism , Stem Cells/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Tumor Necrosis Factor-alpha/physiology
2.
Cells ; 12(21)2023 10 24.
Article in English | MEDLINE | ID: mdl-37947592

ABSTRACT

Liver organoids generated with single or multiple cell types have been used to investigate liver fibrosis development, toxicity, pathogenesis, and drug screening. However, organoid generation is limited by the availability of cells isolated from primary tissues or differentiated from various stem cells. To ensure cell availability for organoid formation, we investigated whether liver organoids could be generated with cell-line-based Huh-7 hepatocellular carcinoma cells, macrophages differentiated from THP-1 monocytes, and LX-2 hepatic stellate cells (HSCs) and primary liver sinusoidal endothelial cells (LSECs). In liver organoids, hepatocyte-, LSEC-, macrophage-, and HSC-related gene expression increased relative to that in two-dimensional (2D)-cultured Huh-7/LSEC/THP-1/LX-2 cells without Matrigel. Thioacetamide (TAA) increased α-smooth muscle actin expression in liver organoids but not in 2D-cultured cells, whereas in TAA-treated organoids, the expression of hepatic and LSEC markers decreased and that of macrophage and HSC markers increased. TAA-induced fibrosis was suppressed by treatment with N-acetyl-L-cysteine or tumor-necrosis-factor-stimulated gene 6 protein. The results showed that liver toxicants could induce fibrotic and inflammatory responses in liver organoids comprising Huh-7/LSEC/macrophages/LX-2 cells, resulting in fibrotic liver organoids. We propose that cell-line-based organoids can be used for disease modeling and drug screening to improve liver fibrosis treatment.


Subject(s)
Endothelial Cells , Hepatic Stellate Cells , Humans , Hepatic Stellate Cells/metabolism , Endothelial Cells/metabolism , Liver Cirrhosis/metabolism , Hepatocytes/metabolism , Macrophages/metabolism , Organoids/metabolism
3.
Biochim Biophys Acta Mol Cell Res ; 1870(8): 119556, 2023 12.
Article in English | MEDLINE | ID: mdl-37544381

ABSTRACT

Several diseases are associated with improper regulation of the Hippo pathway, which plays an important role in cell proliferation and cancer metastasis. Overactivation of the YAP and TAZ proteins accelerates cell proliferation, invasion, and migration during tumorigenesis. Tolfenamic acid (TA) is a non-steroidal anti-inflammatory drug (NSAID) that exhibits activity against various types of cancer. In this study, we observed that TA decreased YAP and TAZ protein levels in cancer cells. TA increased the phosphorylation of YAP and TAZ, leading to the degradation of YAP and TAZ in the cytoplasm and nucleus. TA predominantly affected multiple phosphodegron sites in the YAP and TAZ and lowered 14-3-3ß protein expression, causing YAP and TAZ to enter the ubiquitination pathway. Proteins that affect YAP and TAZ regulation, such as NAG-1 and several YAP/TAZ E3 ligases, were not involved in TA-mediated YAP/TAZ degradation. In summary, our results indicate that TA affects phosphodegron sites on YAP/TAZ, demonstrating a novel effect of TA in tumorigenesis.


Subject(s)
Adaptor Proteins, Signal Transducing , Transcription Factors , Humans , Transcription Factors/genetics , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , YAP-Signaling Proteins , Carcinogenesis , Cell Transformation, Neoplastic
4.
Int J Mol Sci ; 25(1)2023 Dec 29.
Article in English | MEDLINE | ID: mdl-38203646

ABSTRACT

Mesenchymal stem cells (MSCs) regulate immune cell activity by expressing tumor necrosis factor-α (TNF-α)-stimulated gene 6 (TSG-6) in inflammatory environments; however, whether anti-inflammatory responses affect TSG-6 expression in MSCs is not well understood. Therefore, we investigated whether transforming growth factor-ß (TGF-ß) regulates TSG-6 expression in adipose tissue-derived stem cells (ASCs) and whether effective immunosuppression can be achieved using ASCs and TGF-ß signaling inhibitor A83-01. TGF-ß significantly decreased TSG-6 expression in ASCs, but A83-01 and the p38 inhibitor SB202190 significantly increased it. However, in septic C57BL/6 mice, A83-01 further reduced the survival rate of the lipopolysaccharide (LPS)-treated group and ASC transplantation did not improve the severity induced by LPS. ASC transplantation alleviated the severity of sepsis induced by LPS+A83-01. In co-culture of macrophages and ASCs, A83-01 decreased TSG-6 expression whereas A83-01 and SB202190 reduced Cox-2 and IDO-2 expression in ASCs. These results suggest that TSG-6 expression in ASCs can be regulated by high concentrations of pro-inflammatory cytokines in vitro and in vivo, and that A83-01 and SB202190 can reduce the expression of immunomodulators in ASCs. Therefore, our data suggest that co-treatment of ASCs with TGF-ß or p38 inhibitors is not adequate to modulate inflammation.


Subject(s)
Pyrazoles , Thiosemicarbazones , Transforming Growth Factor beta , p38 Mitogen-Activated Protein Kinases , Mice , Animals , Mice, Inbred C57BL , Lipopolysaccharides/pharmacology , Stem Cells , Adipose Tissue
5.
Int J Mol Sci ; 23(21)2022 Oct 28.
Article in English | MEDLINE | ID: mdl-36361907

ABSTRACT

The pro-inflammatory cytokines tumor necrosis factor-alpha (TNF-α) and interleukin (IL)-1ß upregulate TNF-α-stimulated gene 6 (TSG-6); however, current knowledge about the optimal conditions for TSG-6 expression in mesenchymal stem cells (MSCs) is limited. Here, we investigated whether TSG-6 expression varies depending on the polarization state of macrophages co-cultured with adipose tissue-derived stem cells (ASCs) and analyzed the optimal conditions for TSG-6 expression in ASCs. TSG-6 expression increased in ASCs co-cultured with M0, M1, and M2 macrophages indirectly; among them, M1 macrophages resulted in the highest increase in TSG-6 expression in ASCs. TSG-6 expression in ASCs dramatically increased by combination (but not single) treatment of TNF-α, IL-1ß, interferon-gamma (IFN-γ), and lipopolysaccharide (LPS). In addition, phosphorylation of signal transducer and activator of transcription (STAT) 1/3 was observed in response to IFN-γ and LPS treatment but not TNF-α and/or IL-1ß. STAT1/3 activation synergistically increased TNF-α/IL-1ß-dependent TSG-6 expression, and JAK inhibitors suppressed TSG-6 expression both in ASCs and macrophages. In LX-2 hepatic stellate cells, TSG-6 inhibited TGF-ß-induced Smad3 phosphorylation, resulting in decreased α-smooth muscle actin (SMA) expression. Moreover, fibrotic activities of LX-2 cells induced by TGF-ß were dramatically decreased after indirect co-culture with ASCs and M1 macrophages. These results suggest that a comprehensive inflammatory microenvironment may play an important role in determining the therapeutic properties of ASCs by increasing TSG-6 expression through STAT1/3 activation.


Subject(s)
Lipopolysaccharides , Mesenchymal Stem Cells , Coculture Techniques , Lipopolysaccharides/pharmacology , Lipopolysaccharides/metabolism , Macrophages/metabolism , Mesenchymal Stem Cells/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Interferon-gamma/metabolism , Transforming Growth Factor beta/metabolism
6.
Cells ; 12(1)2022 12 26.
Article in English | MEDLINE | ID: mdl-36611885

ABSTRACT

Although protein kinase C (PKC) regulates various biological activities, including cell proliferation, differentiation, migration, tissue remodeling, gene expression, and cell death, the antifibrotic effect of PKC in myofibroblasts is not fully understood. We investigated whether 12-O-tetradecanoylphorbol-13-acetate (TPA), a PKC activator, reduced the activation of hepatic stellate cells (HSCs) and explored the involvement of the Hippo pathway transcriptional coactivator YAP. We analyzed the effect of TPA on the proliferation and expression of α-smooth muscle actin (SMA) in the LX-2 HSC line. We also analyzed the phosphorylation of the Hippo pathway molecules YAP and LATS1 and investigated YAP nuclear translocation. We examined whether Gö 6983, a pan-PKC inhibitor, restored the TPA-inhibited activities of HSCs. Administration of TPA decreased the growth rate of LX-2 cells and inhibited the expression of α-SMA and collagen type I alpha 1 (COL1A1). In addition, TPA induced phosphorylation of PKCδ, LATS1, and YAP and inhibited the nuclear translocation of YAP compared with the control. These TPA-induced phenomena were mostly ameliorated by Gö 6983. Our results indicate that PKCδ exerts an antifibrotic effect by inhibiting the Hippo pathway in HSCs. Therefore, PKCδ and YAP can be used as therapeutic targets for the treatment of fibrotic diseases.


Subject(s)
Adaptor Proteins, Signal Transducing , Hippo Signaling Pathway , Adaptor Proteins, Signal Transducing/metabolism , Signal Transduction , Hepatic Stellate Cells/metabolism , YAP-Signaling Proteins , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Acetates/metabolism
7.
Cell Biol Int ; 45(12): 2443-2451, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34374483

ABSTRACT

Skeletal muscle satellite cells (SkMSCs) play crucial roles in muscle fiber maintenance, repair, and remodeling; however, it remains unknown if these properties are preserved in cultured SkMSCs. In this study, we investigated the characteristics of cultured SkMSCs and their ability to regulate the activity of M1 macrophages. SkMSCs grew well with an average population doubling time of 26.26 ± 6.85 h during 10 passages (P). At P5, Pax7, MyoD, cluster of differentiation (CD)34, and CD56 were not expressed in SkMSCs, but the MSC markers CD73, CD105, and CD90 were expressed and the cells were differentiated into adipocytes and osteoblasts. When SkMSCs were cocultured with macrophages, interleukin (IL)-1ß secretion was decreased, prostaglandin (PG)E2 was produced in coculture, and cyclooxygenase-2 protein was induced in an SkMSC-dependent manner. Hepatocyte growth factor (HGF) was highly secreted by monocultured SkMSCs; interferon-γ and lipopolysaccharide reduced its expression level. However, HGF expression recovered when SkMSCs and macrophages were cocultured. Although exogenous PGE2 upregulated macrophage pro-IL-1ß expression, it suppressed the secretion of cleaved IL-1ß. In contrast, HGF decreased active IL-1ß secretion without affecting pro-IL-1ß expression. Co-treatment of macrophages with HGF and PGE2 reduced pro-IL-1ß expression level and active IL-1ß secretion. Our results suggest that SkMSCs lose their satellite cell properties during serial passaging but acquire mesenchymal stem cell properties including the ability to exert an anti-inflammatory response for macrophages through PGE2 and HGF.


Subject(s)
Anti-Inflammatory Agents/metabolism , Dinoprostone/metabolism , Hepatocyte Growth Factor/metabolism , Mesenchymal Stem Cells/metabolism , Satellite Cells, Skeletal Muscle/metabolism , Adipose Tissue/metabolism , Biomarkers/metabolism , Cell Differentiation/physiology , Cells, Cultured , Cyclooxygenase 2/metabolism , Hepatocytes/metabolism , Humans , Interleukin-1beta/metabolism , Macrophages/metabolism , THP-1 Cells/metabolism
8.
Anticancer Res ; 41(6): 2859-2866, 2021 Jun.
Article in English | MEDLINE | ID: mdl-34083276

ABSTRACT

BACKGROUND/AIM: Genetic manipulation of stem cells using non-viral vectors is still limited due to low transfection efficiency. We investigated whether the DNA-binding cell-permeation peptides (CPP) can enhance the transfection efficiency of non-viral vectors in adipose tissue-derived mesenchymal stem cells (ASCs) and whether ASCs over-expressing TRAIL through CPP can inhibit the growth of glioma U251MG cells in vitro and in vivo. MATERIALS AND METHODS: ASCs were genetically engineered to over-express TRAIL by using CPP, pCMV3-TRAIL and lipid-based transfection reagents (X-tremeGENE). RESULTS: The transfection efficiency of ASCs increased by approximately 7% using CPP; 53.9% of ASCs were transfected and TRAIL expression in ASCs increased by approximately 3 times compared to X-tremeGENE alone. ASCs over-expressing TRAIL using CPP inhibited growth of glioma U251MG cells both in vitro and in the U251MG xenograft model. CONCLUSION: CPP can be used as an enhancer for genetically manipulating ASCs and tumor treatment.


Subject(s)
Adipose Tissue/cytology , Brain Neoplasms/pathology , Cell-Penetrating Peptides/metabolism , DNA/metabolism , Glioma/pathology , Mesenchymal Stem Cells/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Humans , Mesenchymal Stem Cells/cytology , Mice , Mice, Nude , Protein Binding , Xenograft Model Antitumor Assays
9.
Cancer Genomics Proteomics ; 18(4): 569-578, 2021.
Article in English | MEDLINE | ID: mdl-34183389

ABSTRACT

BACKGROUND/AIM: Mesenchymal stem cell-based tumor therapy is still limited due to the insufficient secretion of effectors and discrepancies between their in vitro and in vivo efficacy. We investigated whether genetically engineered adipose tissue-derived mesenchymal stem cells (ASCs) overexpressing tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) had inhibitory effects on H460 tumor growth both in vitro and in an H460 xenograft model. MATERIALS AND METHODS: Genetically engineered adipose tissue-derived mesenchymal stem cells (ASCs) overexpressing tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) were obtained from plasmid transfection with pCMV3-TRAIL and -interferon (IFN)-ß (producing ASC-TRAIL and ASC-IFN-ß, respectively). Death of H460 cells co-cultured with ASCs, ASC-TRAIL, and ASC-IFN-ß or exposed to their conditioned medium was evaluated via apoptosis and cytotoxicity assays. In addition, in an H460 xenograft model (n=10 per group), the antitumor potential of TRAIL-overexpressing, and IFN-ß-overexpressing ASCs was investigated. RESULTS: Conditioned medium obtained from ASC-IFN-ß increased apoptosis of H460 cells more than did ASC-TRAIL. Additionally, in H460 xenograft models, while native ASCs promoted tumor growth, ASC-TRAIL and ASC-IFN-ß both dramatically suppressed tumor growth. Interestingly, in the context of ASC-IFN-ß, tumors were detected only in 20% of nude mice, with smaller sizes and lower weights than those of the control group. CONCLUSION: TRAIL-overexpressing ASCs can be used to treat tumors; ASC-IFN-ß in particular secrete a higher level of TRAIL.


Subject(s)
Adipose Tissue/metabolism , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Adipose Tissue/cytology , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Culture Media, Conditioned/metabolism , Culture Media, Conditioned/pharmacology , Humans , Interferon-beta/genetics , Interferon-beta/metabolism , Mice , Mice, Nude , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , TNF-Related Apoptosis-Inducing Ligand/genetics , Xenograft Model Antitumor Assays
10.
Curr Opin Gastroenterol ; 37(3): 216-223, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33769377

ABSTRACT

PURPOSE OF REVIEW: Liver transplantation is the gold standard for the treatment of end-stage liver disease. However, a shortage of donor organs, high cost, and surgical complications limit the use of this treatment. Cellular therapies using hepatocytes, hematopoietic stem cells, bone marrow mononuclear cells, and mesenchymal stem cells (MSCs) are being investigated as alternative treatments to liver transplantation. The purpose of this review is to describe studies using MSC transplantation for liver diseases based on the reported literature and to discuss prospective research designed to improve the efficacy of MSC therapy. RECENT FINDINGS: MSCs have several properties that show potential to regenerate injured tissues or organs, such as homing, transdifferentiation, immunosuppression, and cellular protective capacity. Additionally, MSCs can be noninvasively isolated from various tissues and expanded ex vivo in sufficient numbers for clinical evaluation. SUMMARY: Currently, there is no approved MSC therapy for the treatment of liver disease. However, MSC therapy is considered a promising alternative treatment for end-stage liver diseases and is reported to improve liver function safely with no side effects. Further robust preclinical and clinical studies will be needed to improve the therapeutic efficacy of MSC transplantation.


Subject(s)
Liver Diseases , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Cell- and Tissue-Based Therapy , Humans , Liver Diseases/therapy , Prospective Studies
11.
Anticancer Res ; 39(5): 2405-2413, 2019 May.
Article in English | MEDLINE | ID: mdl-31092433

ABSTRACT

BACKGROUND/AIM: Plasma medicine is a new field that provides great potential for the treatment of human diseases including cancer in addition to sterilizing the surface of skin and facilitating wound healing. Recently, non-thermal atmospheric plasma (or cold atmospheric plasma, CAP) was introduced, not only for denaturing cells and tissues, but also for operating under the threshold of thermal damage and for chemically inducing a specific response or modification. MATERIALS AND METHODS: Microwave-mediated CAP was used in this study. RESULTS: CAP increased high-mobility group box 1 protein (HMGB1) expression, thereby increasing HMGB-1 secretion. In addition, we observed that the calreticulin (CRT) protein was concentrated at the cellular membrane when plasma was treated, representing immunogenic cell death. CONCLUSION: Overall, plasma treatment induces apoptosis via immunogenic cell death in cancer cells, implying a potential application to human cancer therapy and for the treatment of other human diseases.


Subject(s)
Calreticulin/genetics , HMGB1 Protein/genetics , Neoplasms/therapy , Plasma Gases/therapeutic use , Apoptosis/radiation effects , Gene Expression Regulation, Neoplastic/radiation effects , HCT116 Cells , Humans , Immunogenetic Phenomena/radiation effects , Microwaves/therapeutic use , Neoplasms/genetics , Neoplasms/pathology
12.
Oncol Lett ; 16(6): 7029-7034, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30546436

ABSTRACT

Cancer is one of the leading causes of mortality worldwide. Phytochemicals may be promising anticancer agents given their various chemical structures and diverse biological activities. Damnacanthal (DAM) is a major bioactive component of Noni, which has been investigated previously as a cancer-preventive or chemotherapeutic agent. DAM has also been reported to exhibit anti-proliferative activity in several cancer types. In the present study, it was identified that DAM downregulates chromosome maintenance protein 1 (CRM1) expression in human cancer cells. The application of chitosan-based nanoparticles (NPs) with DAM also induced CRM1 downregulation, which suggests that chitosan-based NPs may be effective vehicles for delivery of phytochemicals such as DAM. It was also identified that DAM increased the levels of the tumor suppressor non-steroidal anti-inflammatory drugs-activated gene 1 in the nucleus, thereby leading to enhanced anticancer effects. The results of the present study indicate that DAM and its nanoformulation may be a candidate anticancer drug.

13.
PLoS One ; 13(8): e0202263, 2018.
Article in English | MEDLINE | ID: mdl-30118500

ABSTRACT

Naturally-occurring chalcones and synthetic chalcone analogues have been demonstrated to have many biological effects, including anti-inflammatory, anti-malarial, anti-fungal, and anti-oxidant/anti-cancerous activities. Compared to other chalcones, trans-chalcone exhibits superior inhibitory activity in cancer cell growth as shown via in vitro assays, and exerts anti-cancerous effects via the activation of the p53 tumor suppressor protein. Thus, characterization of the specific mechanisms, by which trans-chalcone activates p53, can aid development of new chemotherapeutic drugs that can be used individually or synergistically with other drugs. In this report, we found that trans-chalcone modulates many p53 target genes, HSP40 being the most induced gene in the RNA-Seq data using trans-chalcone-treated cells. CRM1 is also inhibited by trans-chalcone, resulting in the accumulation of p53 and other tumor suppressor proteins in the nucleus. Similar effects were seen using trans-chalcone derivatives. Overall, trans-chalcone could provide a strong foundation for the development of chalcone-based anti-cancer drugs.


Subject(s)
Antineoplastic Agents/pharmacology , Chalcone/pharmacology , HSP40 Heat-Shock Proteins/metabolism , Karyopherins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Tumor Suppressor Protein p53/metabolism , Activating Transcription Factor 3/metabolism , Antineoplastic Agents/chemistry , Caspase 3/metabolism , Caspase 7/metabolism , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Chalcone/chemistry , Growth Differentiation Factor 15/metabolism , Humans , Karyopherins/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Signal Transduction/drug effects , Transcription, Genetic/drug effects , Exportin 1 Protein
14.
Mol Immunol ; 87: 60-66, 2017 07.
Article in English | MEDLINE | ID: mdl-28407559

ABSTRACT

Prostaglandins (PGs) are bioactive lipid mediators generated from the phospholipids of cell membrane in response to various inflammatory signals. To understand the potential role of PGs in PG production itself during immune inflammatory responses, we examined the effect of PGE2, PGF2α, and beraprost on COX-2 expression using follicular dendritic cell (FDC)-like HK cells isolated from human tonsils. Those three PGs specifically augmented COX-2 protein expression in a dose-dependent manner after 4 or 8h of treatment. The enhancing effect was also reflected in the actual production of PGs and the viable cell recovery of germinal center B-cells. To investigate the underlying molecular mechanism, we examined the impact of PI3K inhibitors on PG-induced COX-2 expression. Interestingly, COX-2 induction by PGE2 and beraprost, but not PGF2α, was enhanced by wortmannin and LY294002. In line with this result, Akt phosphorylation was inhibited by PGE2 and beraprost but not by PGF2α. The distinct effect of PGE2 and beraprost from PGF2α was reproduced in Akt-knockdowned HK cells. Our current findings imply that PGE2 and PGI2 stimulate COX-2 expression in FDC by inhibiting Akt phosphorylation. Additional studies are warranted to determine the potential role of Akt as a therapeutic target in patients with inflammatory disorders.


Subject(s)
Cyclooxygenase 2/metabolism , Dendritic Cells, Follicular/metabolism , Dinoprostone/metabolism , Phosphorylation/physiology , Proto-Oncogene Proteins c-akt/metabolism , B-Lymphocytes/metabolism , Cell Line , Epoprostenol/analogs & derivatives , Epoprostenol/metabolism , Germinal Center/metabolism , Humans , Phosphatidylinositol 3-Kinases/metabolism
15.
Mol Immunol ; 66(2): 189-96, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25818476

ABSTRACT

IFN-γ plays a critical role in the regulation of innate and adaptive immunity. Paying attention to the emerging role of prostaglandins (PGs) as immune regulators, we attempted to establish the effect of IFN-γ on PG production in human follicular dendritic cell-like HK cells and the underlying signaling pathway by using RNA interference technology. IFN-γ induced COX-2 protein expression in HK cells in a time- and dose-dependent manner, which was not observed in peripheral blood monocytes. Although IFN-γ induced phosphorylation of STAT1, STAT3, and STAT5, only STAT1 was essential for the COX-2 augmentation. The JAK kinases responsible for IFN-γ-triggered STAT1 phosphorylation were JAK1 and JAK2, which were also required for the COX-2 induction. The essential requirement of JAK1 and JAK2 was verified by confocal microscopic analysis, since STAT1 phosphorylation and nuclear translocation were impaired in HK cells with these two kinases knocked down. Finally, we demonstrated that JAK1, JAK2, and STAT1 were indispensable for the actual enhancement of PG production in response to IFN-γ stimulation. These results provide a novel insight into our understanding of IFN-γ under inflammatory conditions and support the emerging concept of PGs as important immune regulators.


Subject(s)
Dendritic Cells/drug effects , Interferon-gamma/pharmacology , Janus Kinase 1/immunology , Janus Kinase 2/immunology , Monocytes/drug effects , Prostaglandins/agonists , STAT1 Transcription Factor/immunology , Cell Nucleus/drug effects , Cyclooxygenase 2/genetics , Cyclooxygenase 2/immunology , Cytoplasm/drug effects , Dendritic Cells/cytology , Dendritic Cells/immunology , Dose-Response Relationship, Drug , Gene Expression Regulation , Humans , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 1/genetics , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/genetics , Monocytes/cytology , Monocytes/immunology , Phosphorylation , Primary Cell Culture , Prostaglandins/biosynthesis , Protein Transport/drug effects , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , STAT1 Transcription Factor/antagonists & inhibitors , STAT1 Transcription Factor/genetics , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/immunology , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/immunology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...