• 제목/요약/키워드: Signal transducer and activator of transcription 1

검색결과 74건 처리시간 0.029초

Effects of β-Glucan on the Release of Nitric Oxide by Macrophages Stimulated with Lipopolysaccharide

  • Choi, E.Y.;Lee, S.S.;Hyeon, J.Y.;Choe, S.H.;Keum, B.R.;Lim, J.M.;Park, D.C.;Choi, I.S.;Cho, K.K.
    • Asian-Australasian Journal of Animal Sciences
    • /
    • 제29권11호
    • /
    • pp.1664-1674
    • /
    • 2016
  • This research analyzed the effect of ${\beta}$-glucan that is expected to alleviate the production of the inflammatory mediator in macrophagocytes, which are processed by the lipopolysaccharide (LPS) of Escherichia. The incubated layer was used for a nitric oxide (NO) analysis. The DNA-binding activation of the small unit of nuclear factor-${\kappa}B$ was measured using the enzyme-linked immunosorbent assay-based kit. In the RAW264.7 cells that were vitalized by Escherichia coli (E. coli) LPS, the ${\beta}$-glucan inhibited both the combatant and rendering phases of the inducible NO synthase (iNOS)-derived NO. ${\beta}$-Glucan increased the expression of the heme oxygenase-1 (HO-1) in the cells that were stimulated by E. coli LPS, and the HO-1 activation was inhibited by the tin protoporphyrin IX (SnPP). This shows that the NO production induced by LPS is related to the inhibition effect of ${\beta}$-glucan. The phosphorylation of c-Jun N-terminal kinases (JNK) and the p38 induced by the LPS were not influenced by the ${\beta}$-glucan, and the inhibitory ${\kappa}B-{\alpha}$ ($I{\kappa}B-{\alpha}$) decomposition was not influenced either. Instead, ${\beta}$-glucan remarkably inhibited the phosphorylation of the signal transducer and activator of transcription-1 (STAT1) that was induced by the E. coli LPS. Overall, the ${\beta}$-glucan inhibited the production of NO in macrophagocytes that was vitalized by the E. coli LPS through the HO-1 induction and the STAT1 pathways inhibition in this research. As the host immune response control by ${\beta}$-glucan weakens the progress of the inflammatory disease, ${\beta}$-glucan can be used as an effective immunomodulator.

Anti-Inflammatory Activities of (+)-Afzelechin against Lipopolysaccharide-Induced Inflammation

  • In-Chul Lee;Jong-Sup Bae
    • Biomolecules & Therapeutics
    • /
    • 제32권4호
    • /
    • pp.467-473
    • /
    • 2024
  • In this study, we investigated the potential protective effects of (+)-afzelechin (AZC), a natural compound that is derived from Bergenia ligulata, on lipopolysaccharide (LPS)-induced inflammatory responses. AZC is known to have antioxidant, anticancer, antimicrobial, and cardiovascular protective properties. However, knowledge regarding the therapeutic potential of AZC against LPS-induced inflammatory responses is limited. Thus, we investigated the protective attributes of AZC against inflammatory damage caused by LPS exposure. We examined the effects of AZC on heme oxygenase (HO)-1, cyclooxygenase (COX)-2, and inducible nitric oxide synthase (iNOS) in LPS-activated human umbilical vein endothelial cells (HUVECs). In addition, the effects of AZC on the expression of iNOS, tumor necrosis factor (TNF)-α, and interleukin (IL)-1β were analyzed in the lung tissues of LPS-injected mice. Data revealed that AZC promoted the production of HO-1, inhibited the interaction between luciferase and nuclear factor (NF)-κB, and reduced the levels of COX-2/PGE2 and iNOS/NO, thereby leading to a decrease in the signal transducer and activator of transcription (STAT)-1 phosphorylation. Moreover, AZC facilitated the nuclear translocation of Nrf2, increased the binding activity between Nrf2 and the antioxidant response elements (AREs), and lowered the expression of IL-1β in the LPS-treated HUVECs. In the animal model, AZC significantly reduced the expression of iNOS in the lung tissue structure and the TNF-α level in the bronchoalveolar lavage fluid. These findings demonstrate that AZC possesses anti-inflammatory properties that regulate iNOS through the inhibition of both NF-κB expression and p-STAT-1. Consequently, AZC has potential as a future candidate for the development of new clinical substances for the treatment of pathological inflammation.

TNF-α/IFN-γ 유도된 각질형성세포 염증에서 가자 추출물의 케모카인 저해 효과 (Inhibitory activity of Terminalia chebula extract against TNF-α/IFN-γ-induced chemokine increase on human keratinocyte, HaCaT cells)

  • 조일주
    • 대한본초학회지
    • /
    • 제37권3호
    • /
    • pp.41-47
    • /
    • 2022
  • Objectives : Terminalia chebula (TC) has been used as a traditional remedy to treat gastrointestinal infectious and inflammatory diseases. However, its protective effects and mechanisms against skin inflammation have not been well-elucidated. Thus, the aim of this study is to evaluate the protective effects of the TC water extract and also to suggest a putative mechanism of TC against skin injury on human keratinocytes, HaCaT cells. Methods : HaCaT cells were pre-treated with TC for 1 h and then stimulated with tumor necrosis factor-alpha (TNF-α) and interferon-gamma (IFN-γ) (10 ng/mL each) to induce skin inflammation and injury. After 24 h, the cells were harvested to evaluate the expression of Th2 chemokines, such as C-C motif chemokine ligand 5 (CCL5, also known as RANTES), C-C chemokine ligand 17 (CCL17, also known as TARC) and C-C chemokine ligand 22 (CCL22, also known as MDC). To investigate the regulatory mechanisms of TC, we also assessed the phosphorylation of signal transducer and activator of transcription 1 (STAT1) signaling pathways in HaCaT cells. Results : Treatment of TC decreased the mRNA levels of RANTES, TARC and MDC with a concentration dependent manner against co-stimulation of TNF-α and IFN-γ. In addition, TC significantly reduced TNF-α and IFN-γ induced phosphorylation of STAT1. Conclusions : In summary, we propose that TC may be a promising candidate for anti-inflammatory skin protector through the inhibition of chemokines via STAT1 deactivation.

Eupatilin Inhibits Gastric Cancer Cell Growth by Blocking STAT3-Mediated VEGF Expression

  • Cheong, Jae-Ho;Hong, Sung-Yi;Zheng, Yanjun;Noh, Sung-Hoon
    • Journal of Gastric Cancer
    • /
    • 제11권1호
    • /
    • pp.16-22
    • /
    • 2011
  • Purpose: Eupatilin is an antioxidative flavone and a phytopharmaceutical derived from Artemisia asiatica. It has been reported to possess anti-tumor activity in some types of cancer including gastric cancer. Eupatilin may modulate the angiogenesis pathway which is part of anti-inflammatory effect demonstrated in gastric mucosal injury models. Here we investigated the anti-tumor effects of eupatilin on gastric cancer cells and elucidated the potential underlying mechanism whereby eupatilin suppresses angiogenesis and tumor growth. Materials and Methods: The impact of eupatilin on the expression of angiogenesis pathway proteins was assessed using western blots in MKN45 cells. Using a chromatin immunoprecipitation assay, we tested whether eupatilin affects the recruitment of signal transducer and activator of transcription 3 (STAT3), aryl hydrocarbon receptor nuclear translocator (ARNT) and hypoxia-inducible factor-$1{\alpha}$ (HIF-$1{\alpha}$) to the human VEGF promoter. To investigate the effect of eupatilin on vasculogenesis, tube formation assays were conducted using human umbilical vein endothelial cells (HUVECs). The effect of eupatilin on tumor suppression in mouse xenografts was assessed. Results: Eupatilin significantly reduced VEGF, ARNT and STAT3 expression prominently under hypoxic conditions. The recruitment of STAT3, ARNT and HIF-$1{\alpha}$ to the VEGF promoter was inhibited by eupatilin treatment. HUVECs produced much foreshortened and severely broken tubes with eupatilin treatment. In addition, eupatilin effectively reduced tumor growth in a mouse xenograft model. Conclusions: Our results indicate that eupatilin inhibits angiogenesis in gastric cancer cells by blocking STAT3 and VEGF expression, suggesting its therapeutic potential in the treatment of gastric cancer.

대식세포 분화 조절을 통한 대시호탕의 암세포 전이 억제 효과 (Anti-migration Effects of the Daesiho-tang (Da Chai Hu-Tang) Water Extract in Cancer Cells by Regulating Macrophage Polarization)

  • 정재훈;박신형
    • 동의생리병리학회지
    • /
    • 제38권1호
    • /
    • pp.32-37
    • /
    • 2024
  • The aim of this study was to investigate the role of Daesiho-tang (Da Chai Hu-Tang) water extract (DSTE) in regulating chronic stress-induced cancer progression, focusing on its activity in modulating tumor-associated macrophages (TAMs). Different stimuli can polarize TAMs into immune-stimulating M1 macrophages or immunosuppressive M2 macrophages. During cancer progression, M2 phenotype increases and supports tumor growth, angiogenesis and metastasis. Notably, chronic stress-induced catecholamines promote M2 macrophage polarization. In this study, we investigated whether DSTE regulates norepinephrine (NE)-induced M2 macrophage polarization in RAW 264.7 mouse macrophage cells. Even though NE itself did not increase the expression of M2 markers, the conditioned media of NE-treated 4T1 mouse breast cancer cells (NE CM) significantly up-regulated M2 markers in RAW 264.7 cells, suggesting that NE-regulated cancer cell secretome stimulated M2 polarization. However, such increase was abrogated by DSTE. NE CM also induced phosphorylation of signal transducer and activator of transcription 6 (STAT6) in RAW 264.7 cells, which was clearly reversed by pretreatment with DSTE, demonstrating that DSTE inhibited M2 polarization by inactivating STAT6. Finally, M2-polarized RAW264.7 cells by NE CM markedly increased the migration of 4T1 cells. However, such increase was completely reversed by co-treating RAW264.7 cells with NE CM and DSTE, indicating that DSTE attenuated cancer cell migration by blocking M2 polarization. Taken together, our results suggest a probable use of DSTE for cancer patients under chronic stress by regulating M2 macrophage polarization.

도두(刀豆)의 조혈증진 작용에 관한 연구 (Enhancing effect of Canavalia gladiata DC semen on the hematopoietic expansion and function of stem cells)

  • 김승형;김근회;지경엽;조인식;김한영;이영철
    • 대한본초학회지
    • /
    • 제27권4호
    • /
    • pp.9-16
    • /
    • 2012
  • Objective : Canavalia gladiata DC semen (CGS) have been used to improve hematopoietic activity. In the current study, we investigated whether CGS regulate hemato-potentiating function using hematopoietic stem cells (HSCs) as a testing system. Methods : HSCs isolated from femur in mice with leukopenia and thrombocytopenia induced induced by CTX. Then, Real-time PCR was performed to measure the mRNA expression and hematopoietic related gene (EPO, IL-3, SCF, c-kit, GM-CSF), the phoaphorylation of GATA-1 and STAT-5a/b were observed by ELISA method, and the number of granulocyte erythrocyte monocyte macrophage colony-forming units (CFU-GEMM) and erythroid burst forming units (BFU-E), semisolid clonogenic assay was performed. Result : When HSCs were treated with CGS, the expression of hematopoietic related genes (EPO, IL-3, SCF, c-kit, and GM-CSF) were significantly increased at the levels of mRNA as well as production in HSCs. Additionally, CGS enhanced phosphorylation of STAT-1 and signal transducer and activator of transcription-5a/b (STAT-5a/b) in HSCs. Furthermore, CGS significantly enhanced the growth rate of granulocyte erythrocyte monocyte macrophage colony-forming units (CFU-GEMM) and erythroid burst forming units (BFU-E) in vitro. Conclusion : These result suggest that CGS has hematopoietic enhancement via hematopoietic cytokine-mediated GATA-1/STAT-5a/b pathway.

Sanghuangporus sanghuang extract inhibits the proliferation and invasion of lung cancer cells in vitro and in vivo

  • Weike Wang;Jiling Song;Na Lu;Jing Yan;Guanping Chen
    • Nutrition Research and Practice
    • /
    • 제17권6호
    • /
    • pp.1070-1083
    • /
    • 2023
  • BACKGROUND/OBJECTIVES: Sanghuangporus sanghuang (SS) has various medicinal effects, including anti-inflammation and anticancer activities. Despite the extensive research on SS, its molecular mechanisms of action on lung cancer are unclear. This study examined the impact of an SS alcohol extract (SAE) on lung cancer using in vitro and in vivo models. MATERIALS/METHODS: Different concentrations of SAE were used to culture lung cancer cells (A549 and H1650). A cell counting kit-8 assay was used to detect the survival ability of A549 and H1650 cells. A scratch assay and transwell cell invasion assay were used to detect the migration rate and invasive ability of SAE. Western blot analysis was used to detect the expression of B-cell lymphoma-2 (Bcl-2), Bcl2-associated X (Bax), cyclin D1, cyclin-dependent kinases 4 (CDK4), signal transducer and activator of transcription 3 (STAT3), and phosphorylated STAT3 (p-STAT3). Lung cancer xenograft mice were used to detect the inhibiting ability of SAE in vivo. Hematoxylin and eosin staining and immunohistochemistry were used to detect the effect of SAE on the structural changes to the tumor and the expression of Bcl-2, Bax, cyclin D1, CDK4, STAT3, and p-STAT3 in lung cancer xenograft mice. RESULTS: SAE could inhibit lung cancer proliferation significantly in vitro and in vivo without cytotoxicity. SAE suppressed the viability, migration, and invasion of lung cancer cells in a dose and time-dependent manner. The SAE treatment significantly decreased the proapoptotic Bcl-2/Bax ratio and the expression of pro-proliferative proteins Cyclin D1 and CDK4 in vitro and in vivo. Furthermore, SAE also inhibited STAT3 expression. CONCLUSIONS: SAE reduced the cell viability and suppressed cell migration and invasion in human lung cancer cells. Moreover, SAE also exhibited anti-proliferation effects in vivo. Therefore, SAE may have benefits in cancer therapy.

Atorvastatin inhibits the proliferation of MKN45-derived gastric cancer stem cells in a mevalonate pathway-independent manner

  • Choi, Ye Seul;Cho, Hee Jeong;Jung, Hye Jin
    • The Korean Journal of Physiology and Pharmacology
    • /
    • 제26권5호
    • /
    • pp.367-375
    • /
    • 2022
  • Gastric cancer stem cells (GCSCs) are a major cause of radioresistance and chemoresistance in gastric cancer (GC). Therefore, targeting GCSCs is regarded as a powerful strategy for the effective treatment of GC. Atorvastatin is a widely prescribed cholesterol-lowering drug that inhibits 3-hydroxy-3-methylglutaryl-coenzyme A reductase, a rate-limiting enzyme in the mevalonate pathway. The anticancer activity of atorvastatin, a repurposed drug, is being investigated; however, its therapeutic effect and molecular mechanism of action against GCSCs remain unknown. In this study, we evaluated the anticancer effects of atorvastatin on MKN45-derived GCSCs. Atorvastatin significantly inhibited the proliferative and tumorsphere-forming abilities of MKN45 GCSCs in a mevalonate pathway-independent manner. Atorvastatin induced cell cycle arrest at the G0/G1 phase and promoted apoptosis by activating the caspase cascade. Furthermore, atorvastatin exerted an antiproliferative effect against MKN45 GCSCs by inhibiting the expression of cancer stemness markers, such as CD133, CD44, integrin α6, aldehyde dehydrogenase 1A1, Oct4, Sox2, and Nanog, through the downregulation of β-catenin, signal transducer and activator of transcription 3, and protein kinase B activities. Additionally, the combined treatment of atorvastatin and sorafenib, a multi-kinase targeted anticancer drug, synergistically suppressed not only the proliferation and tumorsphere formation of MKN45 GCSCs but also the in vivo tumor growth in a chick chorioallantoic membrane model implanted with MKN45 GCSCs. These findings suggest that atorvastatin can therapeutically eliminate GCSCs.

Preparation of Branched Dextran Microspheres of Soluble Interferon-alpha and its Activity In Vitro and In Vivo

  • Hong, Hua;Jo, Jeong-Rang;Yeon, Ji-Hyeon;Hong, Jun-Tack;Jung, Kyung-Hwan;Yoo, Sun-Kyun;Jang, Byeong-Churl
    • Journal of Microbiology and Biotechnology
    • /
    • 제21권2호
    • /
    • pp.176-182
    • /
    • 2011
  • The study objective was to prepare biodegradable branched dextran microspheres encapsulated with His-tagged interferon-alpha (BDM-hIFN-${\alpha}$) and evaluate its activity in vitro and in vivo. The glycidyl methacrylate derivatized dextrans (Dex-GMA) as precursor was primarily synthesized by substituting hydroxyl groups of either the branched or linear type of dextran with GMA. Dex-GMA microspheres loaded with hIFN-${\alpha}$ was then prepared by the water-in-water emulsion technique. In vitro release and Western blotting experiments demonstrated the retained activity of hIFN-${\alpha}$ released from branched dextran microspheres at 24 h by inducing phosphorylation of signal transducer and activator transcription-1 (STAT-1), a down-stream effector of IFN-${\alpha}$, in HepG2 cells. Animal data further revealed a peak of plasma levels of IFN-${\alpha}$ in rats injected intravenously with BDM-hIFN-${\alpha}$ at 10 min post-injection, but a sharp decline at 2 h. High plasma levels of neopterin, a plasma protein induced by IFN-${\alpha}$, were also detected in rats injected with BDM-hIFN-${\alpha}$ at 10 min post-injection. Notably, plasma levels of neopterin remained high at 4 h, but largely declined thereafter.

Cripto Enhances Proliferation and Survival of Mesenchymal Stem Cells by Up-Regulating JAK2/STAT3 Pathway in a GRP78-Dependent Manner

  • Yun, SeungPil;Yun, Chul Won;Lee, Jun Hee;Kim, SangMin;Lee, Sang Hun
    • Biomolecules & Therapeutics
    • /
    • 제26권5호
    • /
    • pp.464-473
    • /
    • 2018
  • Cripto is a small glycosylphosphatidylinositol-anchored signaling protein that can detach from the anchored membrane and stimulate proliferation, migration, differentiation, vascularization, and angiogenesis. In the present study, we demonstrated that Cripto positively affected proliferation and survival of mesenchymal stem cells (MSCs) without affecting multipotency. Cripto also increased expression of phosphorylated janus kinase 2 (p-JAK2), phosphorylated signal transducer and activator of transcription 3 (p-STAT3), 78 kDa glucose-regulated protein (GRP78), c-Myc, and cyclin D1. Notably, treatment with an anti-GRP78 antibody blocked these effects. In addition, pretreatment with STAT3 short interfering RNA (siRNA) inhibited the increase in p-JAK2, c-Myc, cyclin D1, and BCL3 levels caused by Cripto and attenuated the pro-survival action of Cripto on MSCs. We also found that incubation with Cripto protected MSCs from apoptosis caused by hypoxia or $H_2O_2$ exposure, and the level of caspase-3 decreased by the Cripto-induced expression of B-cell lymphoma 3-encoded protein (BCL3). These effects were sensitive to down-regulation of BCL3 expression by BCL3 siRNA. Finally, we showed that Cripto enhanced expression levels of vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF), and hepatocyte growth factor (HGF). In summary, our results demonstrated that Cripto activated a novel biochemical cascade that potentiated MSC proliferation and survival. This cascade relied on phosphorylation of JAK2 and STAT3 and was regulated by GRP78. Our findings may facilitate clinical applications of MSCs, as these cells may benefit from positive effects of Cripto on their survival and biological properties.