• 제목/요약/키워드: Cell hypoxia

검색결과 307건 처리시간 0.034초

저산소증에 의한 활막 섬유모세포의 ICAM-1 발현에 대한 항산화제의 영향 (Effects of Antioxidant on the Hypoxia-induced Expression of ICAM-1 in Cultured Human Synovial Fibroblasts)

  • 김정렬;류완희
    • IMMUNE NETWORK
    • /
    • 제2권1호
    • /
    • pp.25-34
    • /
    • 2002
  • Background: Rheumatoid arthritis (RA) is a chronic inflammatory disease characterized by synovial hyperplasia and joint destruction. The synovial fibroblasts express cell adhesion molecules and have a role in adhesive interation with inflammatory cells in synovial tissue. It has been suggested that hypoxic conditioins are thought to exist in arthritic joints, and several studies indicate that reactive oxygen species (ROS) produced in hypoxic condition can initiate events that lead to pro-adhesive changes via increased expression of adhesion molecules. So, this study wsa designed to examine whether antioxidant can inhibit hypoxia-induced expression of ICAM-1 in cultured human synovial fibroblasts. Methods: Synovial fibroblasts were isolated from synovial tissue in patients with RA and cultured at hypoxic condition. Antioxidant, PDTC (pyrrolidine dithiocarbamate) were pre-treated for an hour before the hypoxic culture and synovial fibroblasts were harvested at 0, 6, 12, 24, 48 hours time points. Cell surface ICAM-1 expression in synovial fibroblasts was examined by the flow cytometric analysis. To analyse the expression of ICAM-1 mRNA, reverse-transcriptase polymerase chain reaction (RT-PCR) was performed. The levels of cytokines in culture supernatants were measured by ELISA, and activation of NF-${\kappa}B$ was analysed by electrophoretic mobility shift assay. The adhesive reaction between synovial fibroblasts and lymphocytes was assayed by measurement of fluorescent intensity of BCECF-AM in lymphocytes. Results: Hypoxic stimuli up-regulated the ICAM-1 expression as well as the adhesive interaction of human synvial fibroblasts to lymphocytes in a time-dependent manner, and PDTC inhibited hpyoxia-induced ICAM-1 expression and cell-cell interaction. PDTC also inhibited the hypoxia-induced activation of intracellular transcription factor, NF-${\kappa}B$. PDTC decreased the amount of hypoxia-induced production of IL-$1{\beta}$ and TNF-${\alpha}$. Conclusion: These studies demonstrate that PDTC inhibit the hypoxia-induced expression of the adhesion molecule, ICAM-1 and activation of NF-${\kappa}B$ in cultured human synovial fibroblasts.

Differential Changes of ATP-sensitive Potassium Channel Current after Hypoxia-reperfusion Treatment in Mouse Neuroblastoma 2a (N2a) Cell

  • Park, Ji-Ho
    • The Korean Journal of Physiology and Pharmacology
    • /
    • 제6권4호
    • /
    • pp.183-186
    • /
    • 2002
  • Ischemic damage is one of the most serious problems. The openers of KATP channel have been suggested to have an effect to limit the ischemic damage. However, it is not yet clear how KATP channels of a cell correspond to hypoxic damage. To address the question, N2a cells were exposed to two different hypoxic conditions as follows: 6 hours hypoxia followed by 3 hours reperfusion and 12 hours hypoxia followed by 3 hours reperfusion. As the results, 6 hours hypoxic treatment increased glibenclamide- sensitive basal $K_{ATP}$ current activity (approximately 6.5-fold at 0 mV test potential) when compared with nomoxic condition. In contrast, 12 hours hypoxic treatment induced a relatively smaller change in the $K_{ATP}$ current density (2.5-fold at 0 mV test potential). Additionally, in experiments where $K_{ATP}$ channels were opened using diazoxide, the hypoxia for 6 hours significantly increased the current density in comparison to control condition (p<0.001). Interestingly, the augmentation in the $K_{ATP}$ current density reduced after exposure to the 12 hours hypoxic condition (p<0.001). Taken together, these results suggest that $K_{ATP}$ channels appear to be recruited more in cells exposed to the 6 hours hypoxic condition and they may play a protective role against hypoxia-reperfusion damage within the time range.

Hypoxia Upregulates Mitotic Cyclins Which Contribute to the Multipotency of Human Mesenchymal Stem Cells by Expanding Proliferation Lifespan

  • Lee, Janet;Kim, Hyun-Soo;Kim, Su-Min;Kim, Dong-Ik;Lee, Chang-Woo
    • Molecules and Cells
    • /
    • 제41권3호
    • /
    • pp.207-213
    • /
    • 2018
  • Hypoxic culture is widely recognized as a method to efficiently expand human mesenchymal stem cells (MSCs) without loss of stem cell properties. However, the molecular basis of how hypoxia priming benefits MSC expansion remains unclear. In this report, our systemic quantitative proteomic and RT-PCR analyses revealed the involvement of hypoxic conditioning activated genes in the signaling process of the mitotic cell cycle. Introduction of screened two mitotic cyclins, CCNA2 and CCNB1, significantly extended the proliferation lifespan of MSCs in normoxic condition. Our results provide important molecular evidence that multipotency of human MSCs by hypoxic conditioning is determined by the mitotic cell cycle duration. Thus, the activation of mitotic cyclins could be a potential strategy to the application of stem cell therapy.

Hypoxia Inducible Factor-1α Directly Induces the Expression of Receptor Activator of Nuclear Factor-κB Ligand in Chondrocytes

  • Baek, Kyunghwa;Park, Hyun-Jung;Baek, Jeong-Hwa
    • International Journal of Oral Biology
    • /
    • 제41권1호
    • /
    • pp.9-15
    • /
    • 2016
  • Receptor activator of nuclear factor-${\kappa}B$ ligand (RANKL) is an osteoblast/stromal cell-derived essential factor for osteoclastogenesis. During endochondral bone formation, hypertrophic chondrocytes calcify cartilage matrix that is subsequently resorbed by osteoclasts in order to be replaced by new bone. Hypoxia-induced upregulation of RANKL expression has been previously demonstrated in an in vitro system using osteoblasts; however, the involved mechanism remains unclear in chondrocytes. In the present study, we investigated whether hypoxia regulates RANKL expression in ATDC5 cells, a murine chondrogenic cell line, and hypoxia-inducible factor-$1{\alpha}$ (HIF-$1{\alpha}$) mediates hypoxia-induced RANKL expression by transactivating the RANKL promoter. The expression levels of RANKL mRNA and protein, as well as HIF-$1{\alpha}$ protein, were significantly increased in ATDC5 cells under hypoxic condition. Constitutively active HIF-$1{\alpha}$ alone significantly increased the levels of RANKL expression under normoxic conditions, whereas dominant negative HIF-$1{\alpha}$ reduced hypoxia-induced RANKL expression. HIF-$1{\alpha}$ increased RANKL promoter reporter activity in a HIF-$1{\alpha}$ binding element-dependent manner in ATDC5 cells. Hypoxia-induced RANKL levels were much higher in differentiated ATDC5 cells, as compared to proliferating ATDC5 cells. These results suggested that under hypoxic conditions, HIF-$1{\alpha}$ mediates induction of RANKL expression in chondrocytes; in addition, hypoxia plays a role in osteoclastogenesis during endochondral bone formation, at least in part, through the induction of RANKL expression in hypertrophic chondrocytes.

Inhibition of hypoxia-induced cyclooxygenase-2 by Korean Red Ginseng is dependent on peroxisome proliferator-activated receptor gamma

  • Song, Heewon;Lee, Young Joo
    • Journal of Ginseng Research
    • /
    • 제41권3호
    • /
    • pp.240-246
    • /
    • 2017
  • Background: Korean Red Ginseng (KRG) is a traditional herbal medicine made by steaming and drying fresh ginseng. It strengthens the endocrine and immune systems to ameliorate various inflammatory responses. The cyclooxygenase-2 (COX-2)/prostaglandin E2 pathway has important implications for inflammation responses and tumorigenesis. Peroxisome proliferator-activated receptor gamma ($PPAR{\gamma}$) is a transcription factor that regulates not only adipogenesis and lipid homeostasis, but also angiogenesis and inflammatory responses. Methods: The effects of the KRG on inhibition of hypoxia-induced COX-2 via $PPAR{\gamma}$ in A549 cells were determined by luciferase assay, Western blot, and/or quantitative reverse transcription-polymerase chain reaction (qRT-PCR). The antimigration and invasive effects of KRG were evaluated on A549 cells using migration and matrigel invasion assays. Results and conclusion: We previously reported that hypoxia-induced COX-2 protein and mRNA levels were suppressed by KRG. This study examines the possibility of $PPAR{\gamma}$ as a cellular target of KRG for the suppression of hypoxia-induced COX-2. $PPAR{\gamma}$ protein levels and $PPAR{\gamma}$-responsive element (PPRE)-driven reporter activities were increased by KRG. Reduction of hypoxia-induced COX-2 by KRG was abolished by the $PPAR{\gamma}$ inhibitor GW9662. In addition, the inhibition of $PPAR{\gamma}$ abolished the effect of KRG on hypoxia-induced cell migration and invasion. Discussion: Our results show that KRG inhibition of hypoxia-induced COX-2 expression and cell invasion is dependent on $PPAR{\gamma}$ activation, supporting the therapeutic potential for suppression of inflammation under hypoxia. Further studies are required to demonstrate whether KRG activates directly $PPAR{\gamma}$ and to identify the constituents responsible for this activity.

Biodistribution and PET imaging of [18F]FMISO in mousecolon cancer xenografted mice

  • Seelam, Sudhakara Reddy;Lee, Ji Youn;Kim, Young Joo;Lee, Yun-Sang;Jeong, Jae Min
    • 대한방사성의약품학회지
    • /
    • 제1권2호
    • /
    • pp.137-144
    • /
    • 2015
  • Hypoxia is an important adverse prognostic factor for tumor progression and is a major cause of failure of radiation therapy. In case of short-term hypoxia, the metabolism can recover to normal, but if hypoxia persists, it causes irreversible cell damage and finally leads to death. So a hypoxia marker would be very useful in oncology. In particular, 2-nitroimidazole can be reduced to form a reactive chemical species, which can bind irreversibly to cell components in the absence of sufficient oxygen, thus, the development of radiolabeled nitroimidazole derivatives for the imaging of hypoxia remains an active field of research to improve cancer therapy result. 2-nitroimidazole based hypoxia marker, [$^{18}F$]FMISO holds promise for the evaluation of tumor hypoxia by Positron emission tomography (PET), at both global and local levels. In the present study, [$^{18}F$]FMISO was synthesized using an automatic synthesis module with high radiochemical purity (>99%) in 60 min. Immunohistochemical analysis using pimonidazole confirmed the presence of hypoxia in xenografted CT-26 tumor tissue. A biodistribution study in CT-26 xenografted mice showed that the increased tumor-to-muscle ratio and tumor-to-blood ratios from 10 to 120 min post-injection. In the PET study, [$^{18}F$]FMISO also showed increased tumor-to-muscle ratios from 10 to 120 min post-injection. In conclusion, this study demonstrates the feasibility and utility of [$^{18}F$]FMISO for imaging hypoxiain mouse colon cancer model using small animal PET.

Aryl Hydrocarbon Receptor Nuclear Translocator is Involved in ATP Homeostasis in Both Normoxic and Hypoxic Monolayer Mouse Hepatoma Cells

  • Choi, Su-Mi;Park, Hyun-Sung
    • Biomolecules & Therapeutics
    • /
    • 제14권3호
    • /
    • pp.132-136
    • /
    • 2006
  • Aryl hydrocarbon receptor nuclear translocator (Arnt) belongs to bHLH-PAS protein family. Here, we study the role of Arnt in both cell growth and glucose metabolism. Our results demonstrated that the absence of Arnt does affect ATP homeostasis but not cell growth in monolayer-cultured mouse hepatoma cells. ATP level of Arnt defective BpRc1 hepatoma cells is less than that of wild type hepatoma cells in both normoxia and hypoxia. BpRc1 cells also fail to increase the expression of glycolytic enzymes in response to hypoxia. Our results suggest that Arnt is essential for glucose metabolism and ATP production but not for cell growth.

Acute Hypoxia Activates an ENaC-like Channel in Rat Pheochromocytoma (PC12) Cells

  • Bae, Yeon Ju;Yoo, Jae-Cheal;Park, Nammi;Kang, Dawon;Han, Jaehee;Hwang, Eunmi;Park, Jae-Yong;Hong, Seong-Geun
    • The Korean Journal of Physiology and Pharmacology
    • /
    • 제17권1호
    • /
    • pp.57-64
    • /
    • 2013
  • Cells can resist and even recover from stress induced by acute hypoxia, whereas chronic hypoxia often leads to irreversible damage and eventually death. Although little is known about the response(s) to acute hypoxia in neuronal cells, alterations in ion channel activity could be preferential. This study aimed to elucidate which channel type is involved in the response to acute hypoxia in rat pheochromocytomal (PC12) cells as a neuronal cell model. Using perfusing solution saturated with 95% $N_2$ and 5% $CO_2$, induction of cell hypoxia was confirmed based on increased intracellular $Ca^{2+}$ with diminished oxygen content in the perfusate. During acute hypoxia, one channel type with a conductance of about 30 pS (2.5 pA at -80 mV) was activated within the first 2~3 min following onset of hypoxia and was long-lived for more than 300 ms with high open probability ($P_o$, up to 0.8). This channel was permeable to $Na^+$ ions, but not to $K^+$, $Ca^+$, and $Cl^-$ ions, and was sensitively blocked by amiloride (200 nM). These characteristics and behaviors were quite similar to those of epithelial sodium channel (ENaC). RT-PCR and Western blot analyses confirmed that ENaC channel was endogenously expressed in PC12 cells. Taken together, a 30-pS ENaC-like channel was activated in response to acute hypoxia in PC12 cells. This is the first evidence of an acute hypoxia-activated $Na^+$ channel that can contribute to depolarization of the cell.

인간 제대혈 유래 혈관내피세포의 혈관 튜브 형성능에 미치는 Sirtuin-2 (SIRT2)의 역활 (The Role of Sirtuin-2 in Tubular Forming Activity of Human Umbilical Vein Endothelial Cells)

  • 정석윤;김철민;김다연;이동형;이규섭;권상모
    • 생명과학회지
    • /
    • 제23권1호
    • /
    • pp.131-136
    • /
    • 2013
  • Sirtuin family 단백질은 암, 당뇨, 심혈관 질환, 뇌신경계 질환과 같은 노화와 연관된 질병들의 발병에 중요한 역할을 하는 것으로 알려져 있다. 우선적으로 혈관내피세포를 이용하여 Hypoxia에서의 sirtuin family의 발현을 확인한 결과, SIRT2 mRNA가 가장 강하게 발현되는 결과를 얻었다. 혈관신생과정에서의 SIRT2 단백질의 생리학적 역할을 규명하고자, 본 실험실에서는 저산소상태에서의 SIRT2의 생리학적인 의미에 주안점을 두었다. Normoxia에서 SIRT2는 세포질에 존재하고 있으나, hypoxia에 의해서 SIRT2 단백질이 핵 내로 이동이 일어남을 확인하였다. 또한 hypoxia에 의해서 SIRT2와 혈관신생인자인 VEGF의 발현이 증가하며, Normoxia와 hypoxia 두 환경에서 혈관내피세포의 혈관형성능력이 SIRT2 inhibitor인 AK-1에 의해서 억제된 것을 확인하였다. 본 연구결과를 통해서 SIRT2가 혈관신생과정을 조절하는 중요한 역할을 함을 시사하였다.

Overexpression of CD44 Standard Isoform Upregulates HIF-1α Signaling in Hypoxic Breast Cancer Cells

  • Ryu, Dayoung;Ryoo, In-geun;Kwak, Mi-Kyoung
    • Biomolecules & Therapeutics
    • /
    • 제26권5호
    • /
    • pp.487-493
    • /
    • 2018
  • Cluster of differentiation 44 (CD44), a cell surface receptor for hyaluronic acid (HA), is involved in aggressive cancer phenotypes. Herein, we investigated the role of the CD44 standard isoform (CD44s) in hypoxia-inducible $factor-1{\alpha}$ ($HIF-1{\alpha}$) regulation using MCF7 overexpressing CD44s (pCD44s-MCF7). When pCD44s-MCF7 was incubated under hypoxia, levels of $HIF-1{\alpha}$, vascular endothelial growth factor, and the $HIF-1{\alpha}$ response element-derived luciferase activity were significantly increased compared to those in the control MCF7. Incubation of pCD44s-MCF7 cells with HA further increased $HIF-1{\alpha}$ accumulation, and the silencing of CD44s attenuated $HIF-1{\alpha}$ elevation, which verifies the role of CD44s in $HIF-1{\alpha}$ regulation. In addition, the levels of phosphorylated extracellular signal-regulated kinase (ERK) was higher in hypoxic pCD44s-MCF7 cells, and $HIF-1{\alpha}$ accumulation was diminished by the pharmacological inhibitors of ERK. CD44s-mediated $HIF-1{\alpha}$ augmentation resulted in two functional outcomes. First, pCD44s-MCF7 cells showed facilitated cell motility under hypoxia via the upregulation of proteins associated with epithelial-mesenchymal transition, such as SNAIL1 and ZEB1. Second, pCD44s-MCF7 cells exhibited higher levels of glycolytic proteins, such as glucose transporter-1, and produced higher levels of lactate under hypoxa. As a consequence of the enhanced glycolytic adaptation to hypoxia, pCD44s-MCF7 cells exhibited a higher rate of cell survival under hypoxia than that of the control MCF7, and glucose deprivation abolished these differential responses of the two cell lines. Taken together, these results suggest that CD44s activates hypoxia-inducible $HIF-1{\alpha}$ signaling via ERK pathway, and the $CD44s-ERK-HIF-1{\alpha}$ pathway is involved in facilitated cancer cell viability and motility under hypoxic conditions.