• Title/Summary/Keyword: lung epithelial cells

Search Result 243, Processing Time 0.027 seconds

Fine Structure of Book Lung in the Wolf Spider, Pardosa astrigera (별늑대거미 (Pardosa astrigera L. Koch) 서폐의 미세구조)

  • Lim, Hyoung-Soo;Moon, Myung-Jin
    • Applied Microscopy
    • /
    • v.24 no.2
    • /
    • pp.1-11
    • /
    • 1994
  • The book lung in the wolf spider, Pardosa astrigera was consisted of a series of flattened triangular-shaped air sacs, stacked with about 70 sheets, and was located in the ventrolateral region of opisthosoma. Each hemolymph spaces (average $8{\mu}m$ in thickness) surrounded by the air sacs (average $6{\mu}m$ in thickness). The air sacs was supported by cylindrical cuticular spikes of microfibril bundles. Epithelial cell processes surrounded the hemolymph spaces. The nuclei of the epithelial cells were concentrated near the atrium. In the middle portion of air sac, the epithelial cells formed pillars across the hemolymph spaces and spot desmosome and zonula adherens were seen between the plasma membranes. In the hemolymph space of this spider, granular hemocytes (average diameter $8{\mu}m$) were the most dominant type of hemocytes. In the medial sinus, the hemolymph flow between the air sacs of a paired book lungs and then flow out of the lung vein. The air comes in the atrium through the ventral lung slit and makes a tidal wave in and out of the air sacs.

  • PDF

Transforming Growth Factor-β-Induced RBFOX3 Inhibition Promotes Epithelial-Mesenchymal Transition of Lung Cancer Cells

  • Kim, Yong-Eun;Kim, Jong Ok;Park, Ki-Sun;Won, Minho;Kim, Kyoon Eon;Kim, Kee K.
    • Molecules and Cells
    • /
    • v.39 no.8
    • /
    • pp.625-630
    • /
    • 2016
  • The RNA-binding protein Rbfox3 is a well-known splicing regulator that is used as a marker for post-mitotic neurons in various vertebrate species. Although recent studies indicate a variable expression of Rbfox3 in non-neuronal tissues, including lung tissue, its cellular function in lung cancer remains largely unknown. Here, we report that the number of RBFOX3-positive cells in tumorous lung tissue is lower than that in normal lung tissue. As the transforming growth factor-${\beta}$ (TGF-${\beta}$) signaling pathway is important in cancer progression, we investigated its role in RBFOX3 expression in A549 lung adenocarcinoma cells. TGF-${\beta}1$ treatment inhibited RBFOX3 expression at the transcriptional level. Further, RBFOX3 depletion led to a change in the expression levels of a subset of proteins related to epithelial-mesenchymal transition (EMT), such as E-cadherin and Claudin-1, during TGF-${\beta}1$-induced EMT. In immunofluorescence microscopic analysis, mesenchymal morphology was more prominent in RBFOX3-depleted cells than in control cells. These findings show that TGF-${\beta}$-induced RBFOX3 inhibition plays an important role in EMT and propose a novel role for RBFOX3 in cancer progression.

Effects of Particulate Matters on A549 and RAW 264.7 Cells (대도시의 입자상 물질이 A549와 RAW 264.7 세포에 미치는 영향)

  • Baak, Young-Mann;Kim, Ji-Hong;Kim, Kyoung-Ah;Ro, Chul-Un;Kim, Hyung-Jung;Lim, Young
    • Journal of Preventive Medicine and Public Health
    • /
    • v.34 no.1
    • /
    • pp.41-46
    • /
    • 2001
  • Objectives : To investigate the effects of particulate matter (PM), a marker of environmental pollution derived from combustion sources, on lung epithelial cells (A549) and macrophage (RAW 264.7). Methods : The production of reactive radicals from lung cells, the lipid peroxidation of cell membrane, and the cytotoxicity of PM were measured using an in vitro model. The results were compared with a control group. Results : The presence of PM significantly increased the production of reactive oxygen species and reactive nitrogen species with time and in a dose dependent pattern and also increased the malondialdehyde concentration in lung epithelial cells. The cytotoxicity of PM was increased with increasing concentration of PM. Conclusions : It has been suggested that urban particulate matter causes an inflammatory reaction in lung tissue through the production of hydroxyl radicals, nitric oxides and numerous cytokines. The causal chemical determinant responsible for these biologic effects are not well understood, but the bioavailable metal in PM seems to determine the tonicity of inhaled PM.

  • PDF

Effects of 3'-isovaleryl-4'-senecioylkhellactone from Peucedanum japonicum Thunberg on PMA-Stimulated Inflammatory Response in A549 Human Lung Epithelial Cells

  • Hwang, Daseul;Ryu, Hyung Won;Park, Ji-Won;Kim, Jung-Hee;Kim, Doo-Young;Oh, Jae-Hoon;Kwon, Ok-Kyoung;Han, Sang-Bae;Ahn, Kyung-Seop
    • Journal of Microbiology and Biotechnology
    • /
    • v.32 no.1
    • /
    • pp.81-90
    • /
    • 2022
  • Peucedanum japonicum Thunberg (PJT) has been used in traditional medicine to treat colds, coughs, fevers, and other inflammatory diseases. The goal of this study was to investigate whether 3'-isovaleryl-4'-senecioylkhellactone (IVSK) from PJT has anti-inflammatory effects on lung epithelial cells. The anti-inflammatory effects of IVSK were evaluated using phorbol 12-myristate 13-acetate (PMA)-stimulated A549 cells and regular human lung epithelial cells as a reference. IVSK reduced the secretion of the inflammatory mediators interleukin (IL)-8 and monocyte chemoattractant protein-1 (MCP-1), and the mRNA expression of IL-6, IL-8, MCP-1, and IL-1β. Additionally, it inhibited the phosphorylation of IκB kinase (IKK), p65, Iκ-Bα, and mitogen-activated protein kinases (MAPKs) p38, JNK, and ERK in A549 cells stimulated with PMA. Moreover, the binding affinity of activator protein-1 (AP-1) and nuclear factor-κB (NF-κB) was significantly reduced in the luciferase assay, while nuclear translocation was markedly inhibited by IVSK in the immunocytochemistry. These findings indicate that IVSK can protect against inflammation through the AP-1 and NF-κB pathway and could possibly be used as a lead compound for the treatment of inflammatory lung diseases.

Metastatic Lung Carcinoma Involving the Periodontium : Report of a case (폐암이 치주 조직에 전이된 증례보고)

  • Shin, Ji-Yearn;Han, Soo-Boo;Hwang, Kwang-Se;Kye, Seung-Beom
    • Journal of Periodontal and Implant Science
    • /
    • v.27 no.1
    • /
    • pp.111-116
    • /
    • 1997
  • The oral cavity is easily accessible for direct exposure of a malignant disease. 1 percent of the oral malignant tumors are of metastatic origin and approximately 10 percent to 25 percent of the 1 percent fraction originate from the lungs. A case of metastatic lung carcinoma to the gingiva in a 88-year-old male is reported. He complained of pain and swelling between right maxillary 1st premolar and 2nd molar. Although surgical excision of the lesion has been done, the gingival lesion developed as a quickly growing mass and recurred 2 weeks after surgical excision. The gingival mass was histopathologically diagnosed as an undifferentiated carcinoma. Epithelial layer was continuous without ulceration and it seems that the cancer cells are originated from primary tumor. Infiltrated cancer cells were pleomorphic and dyskeratotic. The cells had 2 or more nuclei, not showing squamous or glandular differentiation. Immunohistochemical study revealed the cells originated from the epithelial cells. The prognosis is poor, because prognosis depends on surgical elimination of the primary tumor.

  • PDF

MUC1-C influences cell survival in lung adenocarcinoma Calu-3 cells after SARS-CoV-2 infection

  • Kim, Dongbum;Maharjan, Sony;Kim, Jinsoo;Park, Sangkyu;Park, Jeong-A;Park, Byoung Kwon;Lee, Younghee;Kwon, Hyung-Joo
    • BMB Reports
    • /
    • v.54 no.8
    • /
    • pp.425-430
    • /
    • 2021
  • Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) induces coronavirus disease 2019 (COVID-19) and may increase the risk of adverse outcomes in lung cancer patients. In this study, we investigated the expression and function of mucin 1 (MUC1) after SARS-CoV-2 infection in the lung epithelial cancer cell line Calu-3. MUC1 is a major constituent of the mucus layer in the respiratory tract and contributes to pathogen defense. SARS-CoV-2 infection induced MUC1 C-terminal subunit (MUC1-C) expression in a STAT3 activation-dependent manner. Inhibition of MUC1-C signaling increased apoptosis-related protein levels and reduced proliferation-related protein levels; however, SARS-CoV-2 replication was not affected. Together, these results suggest that increased MUC1-C expression in response to SARS-CoV-2 infection may trigger the growth of lung cancer cells, and COVID-19 may be a risk factor for lung cancer patients.

Expression of Tiam1 in Lung Cancer and its Clinical Significance

  • Wang, Hong-Ming;Wang, Jing
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.13 no.2
    • /
    • pp.613-615
    • /
    • 2012
  • The aim of this study was to ana1yze T-cell lymphoma invasion and metastasis-inducing factor 1 (Tiam1) expression in 1ung cancer patients. A total of 204 patients with lung cancer tissue lesions were enrolled in the present study, along with 40 cases of normal lung tissue and 40 of normal fetal lung tissue. Tiam1 protein expression level was determined using intensity quantitative analysis, for comparison in lung cancer, metastatic, normal lung, and fetal lung tissue. The positive unit (PU) of Tiam1 was $13.5{\pm}5.42$ in lung cancer,$5.67{\pm}1.56$ in norma1 epithelial cells, and $5.89{\pm}1.45$ in fetal lung epithelial cells. The value in the lung cancer tissue was significantly higher than that in the normal lung tissue and the fetal lung tissue (P<0.01). The Tiam1 PU values with lymph node metastasis and without 1ymph node metastasis were $15.2{\pm}4.34$ and $12.5{\pm}4.23$, respectively, and the difference was statistically significant (P<0.05). The Tiam1 PU values in different tumor, nodes, metastasis (TNM) stages, III-IV period, and I-II phase were $14.7{\pm}4.14$ and $11.0{\pm}5.34$ (P<0.05). A correlation was found between Tiam1 expression and the age of patient, tumor size, tumor type, and tumor differentiation. Tiam1 protein expression in the lung tumor tissue is significantly higher than that in the normal lung tissue and fetal lung tissue. Tiam1 expression may be closely related to lung cancer development and metastasis.

Promising Therapeutic Effects of Embryonic Stem Cells-Origin Mesenchymal Stem Cells in Experimental Pulmonary Fibrosis Models: Immunomodulatory and Anti-Apoptotic Mechanisms

  • Hanna Lee;Ok-Yi Jeong;Hee Jin Park;Sung-Lim Lee;Eun-yeong Bok;Mingyo Kim;Young Sun Suh;Yun-Hong Cheon;Hyun-Ok Kim;Suhee Kim;Sung Hak Chun;Jung Min Park;Young Jin Lee;Sang-Il Lee
    • IMMUNE NETWORK
    • /
    • v.23 no.6
    • /
    • pp.45.1-45.22
    • /
    • 2023
  • Interstitial lung disease (ILD) involves persistent inflammation and fibrosis, leading to respiratory failure and even death. Adult tissue-derived mesenchymal stem cells (MSCs) show potential in ILD therapeutics but obtaining an adequate quantity of cells for drug application is difficult. Daewoong Pharmaceutical's MSCs (DW-MSCs) derived from embryonic stem cells sustain a high proliferative capacity following long-term culture and expansion. The aim of this study was to investigate the therapeutic potential of DW-MSCs in experimental mouse models of ILD. DW-MSCs were expanded up to 12 passages for in vivo application in bleomycin-induced pulmonary fibrosis and collagen-induced connective tissue disease-ILD mouse models. We assessed lung inflammation and fibrosis, lung tissue immune cells, fibrosis-related gene/protein expression, apoptosis and mitochondrial function of alveolar epithelial cells, and mitochondrial transfer ability. Intravenous administration of DWMSCs consistently improved lung fibrosis and reduced inflammatory and fibrotic markers expression in both models across various disease stages. The therapeutic effect of DW-MSCs was comparable to that following daily oral administration of nintedanib or pirfenidone. Mechanistically, DW-MSCs exhibited immunomodulatory effects by reducing the number of B cells during the early phase and increasing the ratio of Tregs to Th17 cells during the late phase of bleomycin-induced pulmonary fibrosis. Furthermore, DW-MSCs exhibited anti-apoptotic effects, increased cell viability, and improved mitochondrial respiration in alveolar epithelial cells by transferring their mitochondria to alveolar epithelial cells. Our findings indicate the strong potential of DW-MSCs in the treatment of ILD owing to their high efficacy and immunomodulatory and anti-apoptotic effects.

FBW7 Upregulation Enhances Cisplatin Cytotoxicity in Non-small Cell Lung Cancer Cells

  • Yu, Hao-Gang;Wei, Wei;Xia, Li-Hong;Han, Wei-Li;Zhao, Peng;Wu, Sheng-Jun;Li, Wei-Dong;Chen, Wei
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.14 no.11
    • /
    • pp.6321-6326
    • /
    • 2013
  • Introduction: Lung cancer is extremely harmful to human health and has one of the highest worldwide incidences of all malignant tumors. Approximately 80% of lung cancers are classified as non-small cell lung cancers (NSCLCs). Cisplatin-based multidrug chemotherapy regimen is standard for such lesions, but drug resistance is an increasing problem. F-box/WD repeat-containing protein 7 (FBW7) is a member of the F-box protein family that regulates cell cycle progression, and cell growth and differentiation. FBW7 also functions as a tumor suppressor. Methods: We used cell viability assays, Western blotting, and immunofluorescence combined with siRNA interference or plasmid transfection to investigate the underlying mechanism of cisplatin resistance in NSCLC cells. Results: We found that FBW7 upregulation significantly increased cisplatin chemosensitivity and that cells expressing low levels of FBW7, such as NCI-H1299 cells, have a mesenchymal phenotype. Furthermore, siRNA-mediated silencing or plasmid-mediated upregulation of FBW7 resulted in altered epithelial-mesenchymal transition (EMT) patterns in NSCLC cells. These data support a role for FBW7 in regulating the EMT in NSCLC cells. Conclusion: FBW7 is a potential drug target for combating drug resistance and regulating the EMT in NSCLC cells.

Ginsenosides Rk1 and Rg5 inhibit transforming growth factor-β1-induced epithelial-mesenchymal transition and suppress migration, invasion, anoikis resistance, and development of stem-like features in lung cancer

  • Kim, Hyunhee;Choi, Pilju;Kim, Taejung;Kim, Youngseok;Song, Bong Geun;Park, Young-Tae;Choi, Seon-Jun;Yoon, Cheol Hee;Lim, Won-Chul;Ko, Hyeonseok;Ham, Jungyeob
    • Journal of Ginseng Research
    • /
    • v.45 no.1
    • /
    • pp.134-148
    • /
    • 2021
  • Background: Lung cancer has a high incidence worldwide, and most lung cancer-associated deaths are attributable to cancer metastasis. Although several medicinal properties of Panax ginseng Meyer have been reported, the effect of ginsenosides Rk1 and Rg5 on epithelial-mesenchymal transition (EMT) stimulated by transforming growth factor beta 1 (TGF-β1) and self-renewal in A549 cells is relatively unknown. Methods: We treated TGF-β1 or alternatively Rk1 and Rg5 in A549 cells. We used western blot analysis, real-time polymerase chain reaction (qPCR), wound healing assay, Matrigel invasion assay, and anoikis assays to determine the effect of Rk1 and Rg5 on TGF-mediated EMT in lung cancer cell. In addition, we performed tumorsphere formation assays and real-time PCR to evaluate the stem-like properties. Results: EMT is induced by TGF-β1 in A549 cells causing the development of cancer stem-like features. Expression of E-cadherin, an epithelial marker, decreased and an increase in vimentin expression was noted. Cell mobility, invasiveness, and anoikis resistance were enhanced with TGF-β1 treatment. In addition, the expression of stem cell markers, CD44, and CD133, was also increased. Treatment with Rk1 and Rg5 suppressed EMT by TGF-β1 and the development of stemness in a dose-dependent manner. Additionally, Rk1 and Rg5 markedly suppressed TGF-β1-induced metalloproteinase-2/9 (MMP2/9) activity, and activation of Smad2/3 and nuclear factor kappa B/extra-cellular signal regulated kinases (NF-kB/ERK) pathways in lung cancer cells. Conclusions: Rk1 and Rg5 regulate the EMT inducing TGF-β1 by suppressing the Smad and NF-κB/ERK pathways (non-Smad pathway).