• 제목/요약/키워드: EGFR- Erk1/2

검색결과 21건 처리시간 0.019초

CUG2 유전자에 의하여 감소된 FBXW7 E3 ligase 발현이 유사-종양줄기세포 표현형을 유도 (The Decreased Expression of Fbxw7 E3 Ligase Mediated by Cancer Upregulated Gene 2 Confers Cancer Stem Cell-like Phenotypes)

  • 야웃 낫파판;김남욱;붓루앙 파차라폰;조일래;카오윈 시리차트;고상석;강호영;정영화
    • 생명과학회지
    • /
    • 제32권4호
    • /
    • pp.271-278
    • /
    • 2022
  • 신규 종양 유전자 Cancer Upregulated Gene (CUG) 2가 어떻게 유사-종양줄기세포 표현형을 유도하는지 잘 알려져 있지 않다. Cyclin E, c-Myc, Notch, 그리고 Yap1와 같은 종양단백질를 분해하여 그 발현을 조절하는 FBXW7 E3 ligase의 발현이 대장암, 자궁경부암, 그리고 위암 등 여러 암조직에서 낮아져 있음이 보고되고 있다. 그래서 우리는 이 FBXW7 단백질이 CUG2에 의한 종양형성에 관여할 수 있다는 가설을 세웠다. 이 연구에서 우리는 각 대조구 세포주보다 CUG2가 과발현된 A549 폐암 세포주와 BEAS-2B 기관지 세포주에서 FBXW7 단백질 발현이 낮게 나왔다. 여기서 MG132를 처리하게 되면 감소된 FBXW7과 FBXW7 기질로 알려진 Yap1 단백질 발현이 증가되는 결과를 관찰하였다. 종양줄기세포 현상에서 FBXW7의 역할을 규명하기 위하여, FBXW7 siRNA를 처리하였다. 대조구 세포주에서 감소된 FBXW7의 조건은 세포 이동 침습, 그리고 구형 형성이 증가되는 종양줄기세포 현상이 촉진되는 것을 관찰하였고, CUG2가 과발현된 두 세포주에서 FBXW7 발현 벡타 도입으로 FBXW7 발현 증가는 종양줄기세포 현상이 억제됨을 알 수 있었다. 또한 FBXW7의 감소는 EGFR-Akt-ERK1/2와 β-catenin-Yap1-NEK2 신호 경로를 활성화시키고, 반대로 FBXW7 발현 증가는 이 두 경로의 활성이 억제됨을 알 수 있었다. 이들 결과를 종합해 보면, CUG2 과발현은 FBXW7의 발현 감소로 이어지고, 이는 EGFR-Akt-ERK1/2와 β-catenin-Yap1-NEK2 신호경로를 활성화시켜 유사-종양줄기세포 현상을 촉진하는 것으로 생각된다.

Apigenin and Wogonin Regulate Epidermal Growth Factor Receptor Signaling Pathway Involved in MUC5AC Mucin Gene Expression and Production from Cultured Airway Epithelial Cells

  • Sikder, Md. Asaduzzaman;Lee, Hyun Jae;Ryu, Jiho;Park, Su Hyun;Kim, Ju-Ock;Hong, Jang-Hee;Seok, Jeong Ho;Lee, Choong Jae
    • Tuberculosis and Respiratory Diseases
    • /
    • 제76권3호
    • /
    • pp.120-126
    • /
    • 2014
  • Background: We investigated whether wogonin and apigenin significantly affect the epidermal growth factor receptor (EGFR) signaling pathway involved in MUC5AC mucin gene expression, and production from cultured airway epithelial cells; this was based on our previous report that apigenin and wogonin suppressed MUC5AC mucin gene expression and production from human airway epithelial cells. Methods: Confluent NCI-H292 cells were pretreated with wogonin or apigenin for 15 minutes or 24 hours and then stimulated with epidermal growth factor (EGF) for 24 hours or the indicated periods. Results: We found that incubation of NCI-H292 cells with wogonin or apigenin inhibited the phosphorylation of EGFR. The downstream signals of EGFR such as phosphorylation of MEK1/2 and ERK1/2 were also inhibited by wogonin or apigenin. Conclusion: The results suggest that wogonin and apigenin inhibits EGFR signaling pathway, which may explain how they inhibit MUC5AC mucin gene expression and production induced by EGF.

Common Docking Domain Mutation E322K of the ERK2 Gene is Infrequent in Oral Squamous Cell Carcinomas

  • Valiathan, Gopalakrishnan Mohan;Thenumgal, Siji Jacob;Jayaraman, Bhaskar;Palaniyandi, Arunmozhi;Ramkumar, Hemalatha;Jayakumar, Keerthivasan;Bhaskaran, Sajeev;Ramanathan, Arvind
    • Asian Pacific Journal of Cancer Prevention
    • /
    • 제13권12호
    • /
    • pp.6155-6157
    • /
    • 2012
  • Background: Mutations in the MAPK (Mitogen Activated Protein Kinase) signaling pathway - EGFR/Ras/RAF/MEK have been associated with the development of several carcinomas. ERK2, a downstream target of the MAPK pathway and a founding member of the MAPK family is activated by cellular signals emanating at the cell membrane. Activated ERK2 translocates into the nucleus to transactivate genes that promote cell proliferation. MKP - a dual specific phosphatase - interacts with activated ERK2 via the common docking (CD) domain of the later to inactivate (dephosphorylate) and effectively terminate further cell proliferation. A constitutively active form of ERK2 carrying a single point mutation - E322K in its CD domain, was earlier reported by our laboratory. In the present study, we investigated the prevalence of this CD domain E322K mutation in 88 well differentiated OSCC tissue samples. Materials and Method: Genomic DNA specimens isolated from 88 oral squamous cell carcinoma tissue samples were amplified with primers flanking the CD domain of the ERK2 gene. Subsequently, PCR amplicons were gel purified and subjected to direct sequencing to screen for mutations. Results: Direct sequencing of eighty eight OSCC samples identified an E322K CD domain mutation in only one (1.1%) OSCC sample. Conclusions: Our result indicates that mutation in the CD domain of ERK2 is rare in OSCC patients, which suggests the role of genetic alterations in other mitogenic genes in the development of carcinoma in the rest of the patients. Nevertheless, the finding is clinically significant, as the relatively rare prevalence of the E322K mutation in OSCC suggests that ERK2, being a common end point signal in the multi-hierarchical mitogen activated signaling pathway may be explored as a viable drug target in the treatment of OSCC.

Epidermal Growth Factor Induces Vasoconstriction Through the Phosphatidylinositol 3-Kinase-Mediated Mitogen-Activated Protein Kinase Pathway in Hypertensive Rats

  • Kim, Jung-Hwan;Lee, Chang-Kwon;Park, Hyo-Jun;Kim, Hyo-Jin;So, Hyun-Ha;Lee, Keun-Sang;Lee, Hwan-Myung;Roh, Hui-Yul;Choi, Wahn-Soo;Park, Tae-Kyu;Kim, Bo-Kyung
    • 대한물리치료과학회지
    • /
    • 제13권2호
    • /
    • pp.137-145
    • /
    • 2006
  • We investigated whether increased contractile responsiveness to epidermal growth factor (EGF) is associated with altered activation of mitogen-activated protein kinase (MAPK) in the aortic smooth muscle of deoxycorticosterone acetate (DOCA)-salt hypertensive rats. EGF induced contraction and MAPK activity in aortic smooth muscle strips, which were significantly increased in tissues from the DOCA-salt hypertensive rats compared with those from sham-operated rats. AG1478, PD98059, and LY294002, inhibitors of EGF receptor (EGFR) tyrosine kinase, MAPK/extracellular signal-regulated kinase (ERK) kinase, and phosphatidylinositol 3-kinase (PI3K), respectively, inhibited the contraction and the activity of ERK1/2 that were elevated by EGF. Y27632 and GF109203X, inhibitors of Rho kinase and protein kinase C, respectively, attenuated EGF-induced contraction, with no diminution of ERK1/2 activity. Although EGF also elevated the activity of EGFR tyrosine kinase in both sham-operated and DOCA-salt hypertensive rats, the expression and the magnitude of activation did not differ between strips. These results strongly suggest that EGF induces contraction by the activation of ERK1/2, which is regulated by the PI3K pathway in the aortic smooth muscle of DOCA-salt hypertensive rats.

  • PDF

Clinical implications of the Hippo-YAP pathway in multiple cancer contexts

  • Kim, Han-Byul;Myung, Seung-Jae
    • BMB Reports
    • /
    • 제51권3호
    • /
    • pp.119-125
    • /
    • 2018
  • The Hippo pathway plays prominent and widespread roles in various forms of human carcinogenesis. Specifically, the Yes-associated protein (YAP), a downstream effector of the Hippo pathway, can lead to excessive cell proliferation and the inhibition of apoptosis, resulting in tumorigenesis. It was reported that the YAP is strongly elevated in multiple types of human malignancies such as breast, lung, small intestine, colon, and liver cancers. Recent work indicates that, surprisingly, Hippo signaling components' (SAV1, MST1/2, Lats1/2) mutations are virtually absent in human cancer, rendering this signaling an unlikely candidate to explain the vigorous activation of the YAP in most, if not all human tumors and an activated YAP promotes the resistance to RAF-, MAPK/ERK Kinase (MEK)-, and Epidermal growth factor receptor (EGFR)-targeted inhibitor therapy. The analysis of YAP expressions can facilitate the identification of patients who respond better to an anti-cancer drug treatment comprising RAF-, MEK-, and EGFR-targeted inhibitors. The prominence of YAP for those aspects of cancer biology denotes that these factors are ideal targets for the development of anti-cancer medications. Therefore, our report strongly indicates that the YAP is of potential prognostic utility and druggability in various human cancers.

3-Deoxysappanchalcone Inhibits Cell Growth of Gefitinib-Resistant Lung Cancer Cells by Simultaneous Targeting of EGFR and MET Kinases

  • Jin-Young Lee;Seung-On Lee;Ah-Won Kwak;Seon-Bin Chae;Seung-Sik Cho;Goo Yoon;Ki-Taek Kim;Yung Hyun Choi;Mee-Hyun Lee;Sang Hoon Joo;Jin Woo Park;Jung-Hyun Shim
    • Biomolecules & Therapeutics
    • /
    • 제31권4호
    • /
    • pp.446-455
    • /
    • 2023
  • The mechanistic functions of 3-deoxysappanchalcone (3-DSC), a chalcone compound known to have many pharmacological effects on lung cancer, have not yet been elucidated. In this study, we identified the comprehensive anti-cancer mechanism of 3-DSC, which targets EGFR and MET kinase in drug-resistant lung cancer cells. 3-DSC directly targets both EGFR and MET, thereby inhibiting the growth of drug-resistant lung cancer cells. Mechanistically, 3-DSC induced cell cycle arrest by modulating cell cycle regulatory proteins, including cyclin B1, cdc2, and p27. In addition, concomitant EGFR downstream signaling proteins such as MET, AKT, and ERK were affected by 3-DSC and contributed to the inhibition of cancer cell growth. Furthermore, our results show that 3-DSC increased redox homeostasis disruption, ER stress, mitochondrial depolarization, and caspase activation in gefitinib-resistant lung cancer cells, thereby abrogating cancer cell growth. 3-DSC induced apoptotic cell death which is regulated by Mcl-1, Bax, Apaf-1, and PARP in gefitinib-resistant lung cancer cells. 3-DSC also initiated the activation of caspases, and the pan-caspase inhibitor, Z-VAD-FMK, abrogated 3-DSC induced-apoptosis in lung cancer cells. These data imply that 3-DSC mainly increased mitochondria-associated intrinsic apoptosis in lung cancer cells to reduce lung cancer cell growth. Overall, 3-DSC inhibited the growth of drug-resistant lung cancer cells by simultaneously targeting EGFR and MET, which exerted anti-cancer effects through cell cycle arrest, mitochondrial homeostasis collapse, and increased ROS generation, eventually triggering anti-cancer mechanisms. 3-DSC could potentially be used as an effective anti-cancer strategy to overcome EGFR and MET target drug-resistant lung cancer.

담배 연기에 의한 Muc5ac 유전자 발현에 관여하는 세포 내 신호 전달 경로로서의 ERK1/2와 p38 MAPK (Muc5ac Gene Expression Induced by Cigarette Smoke is Mediated Via a Pathway Involving ERK1/2 and p38 MAPK)

  • 김용현;윤형규;김치홍;안중현;권순석;김영균;김관형;문화식;박성학;송정섭;조경숙
    • Tuberculosis and Respiratory Diseases
    • /
    • 제58권6호
    • /
    • pp.590-599
    • /
    • 2005
  • 연구배경 : 만성폐쇄성폐질환에서 나타나는 기도점액의 과다분비는 이 질환의 중요한 병리학적 소견이며 호흡곤란 등 환자의 증상을 악화시키는 요인 중의 하나이다. 기도 점액을 구성하는 여러 성분 중 Muc 유전자에 의해 만들어지는 당 단백이 흡연에 의해 생성이 증가하는데 이에 관여하는 세포 내 신호전달 과정에 대하여 확실히 밝혀진 바가 없다. 저자는 Muc 유전자 중 인체의 기도에 가장 많이 분비되는 Muc5ac 점액 생성을 담당하는 Muc5ac 유전자의 발현이 흡연에 의하여 증가하는데 관여하는 세포 내 신호전달 과정을 알아보고자 하였다. 재료 및 방법 : 사람 폐선암 세포주인 A549 세포를 배양하여 Muc5ac 유전자의 promotor를 luciferase reporter plasmid를 사용하여 세포 내에 transfection시키고 5% 담배연기 추출물로 자극하여 배양하였다. 또 세포 내 신호전달에 관여하는 표피성장인자 수용체 kinase의 억제제인 AG1478, mitogen-activated protein kinase kinase(MAPKK) 억제제인 PD98059, p38 mitogen-activated protein kinase 억제제인 SB203580으로 각각 전 처치 후 역시 5% 담배연기 추출물로 자극 배양하였다. 배양된 세포에서 단백질을 추출하여 luciferase 분석을 통하여 Muc5ac promoter 활성도를 측정하고 Western blot을 이용하여 표피성장인자 수용체와 mitogen-activated protein kinase (MAPK)인 extracellular signalrelated kinase (ERK)1/2, p38 MAPK, c-Jun N-terminal kinase (JNK)의 발현을 확인하였다. 또 세포에서 RNA를 추출한 후 Muc5ac primer를 이용하여 역전사효소 중합연쇄반응을 수행하여 Muc5ac mRNA 발현을 관찰하였다. 결 과 : 1. Muc5ac promoter를 삽입한 A549 세포를 5% 담배연기 추출물로 자극하였을 때 의의 있게 luciferase 활성도가 증가하였고(P<0.001) 자극하는 시간이 3시간이었을 때 luciferase 활성도가 최고치를 보였다(P<0.01). 또 담배연기 추출물 자극은 표피성장인자 수용체를 인산화시켰으며 인산화는 AG1478과 PD98059에 의하여 억제되었다. 2. AG1478 혹은 PD98059로 전 처치 후 5% 담배연기 추출물로 자극한 경우 5% 담배연기 추출물 단독으로 자극한 것에 비하여 유의하게 lucifearse 활성도가 억제되었고 (P<0.01) 세 가지 종류의 MAPK 중 ERK1/2와 p38 MAPK의 인산화는 관찰되었으나 JNK의 인산화는 관찰되지 않았다. 역전사효소 중합연쇄반응을 이용하여 관찰한 Muc5ac mRNA 발현은 담배연기 추출물에 의해 증가되었고 PD98059와 AG1478에 의하여 역시 억제되었다. 3. 담배연기 추출물에 의하여 인산화 된 ERK1/2는 PD98059에 의하여 인산화가 감소하였고 p38 MAPK의 인산화는 PD98059와 SB203580에 의하여 감소하였으며 이 두 가지 억제제는 모두 luciferase 활성도를 유의하게 억제시켰다(P<0.0001). 결 론 : 담배연기 추출물은 Muc5ac 유전자의 발현을 증가시켜 기도 내 점액 분비를 증가시키며 이는 표피성장인자 수용체를 매개로 ERK1/2와 p38 MAPK를 경유하여 세포 내 신호전달이 이루어진다고 생각된다. 따라서 점액 유전자 활성화를 매개하는 신호전달 과정을 차단하는 약제나 방법이 개발된다면 과도한 점액분비를 치료할 수 있을 것으로 생각한다.

PKHD1 Gene Silencing May Cause Cell Abnormal Proliferation through Modulation of Intracellular Calcium in Autosomal Recessive Polycystic Kidney Disease

  • Yang, Ji-Yun;Zhang, Sizhong;Zhou, Qin;Guo, Hong;Zhang, Ke;Zheng, Rong;Xiao, Cuiying
    • BMB Reports
    • /
    • 제40권4호
    • /
    • pp.467-474
    • /
    • 2007
  • Autosomal recessive polycystic kidney disease (ARPKD) is one of the important genetic disorders in pediatric practice. Mutation of the polycystic kidney and hepatic disease gene 1 (PKHD1) was identified as the cause of ARPKD. The gene encodes a 67-exon transcript for a large protein of 4074 amino acids termed fibrocystin, but its function remains unknown. The neoplastic-like in cystic epithelial proliferation and the epidermal growth factor/epidermal growth factor receptor (EGF/EGFR) axis overactivity are known as the most important characteristics of ARPKD. Since the misregulation of $Ca^{2+}$ signaling may lead to aberrant structure and function of the collecting ducts in kidney of rat with ARPKD, present study aimed to investigate the further mechanisms of abnormal proliferation of cystic cells by inhibition of PKHD1 expression. For this, a stable PKHD1-silenced HEK-293T cell line was established. Then cell proliferation rates, intracellular $Ca^{2+}$ concentration and extracellular signal-regulated kinase 1/2 (ERK1/2) activity were assessed after treatment with EGF, a calcium channel blocker and agonist, verapamil and Bay K8644. It was found that PKHD1-silenced HEK-293T cell lines were hyperproliferative to EGF stimulation. Also PKHD1-silencing lowered the intracellular $Ca^{2+}$ and caused EGF-induced ERK1/2 overactivation in the cells. An increase of intracellular $Ca^{2+}$ in PKHD1-silenced cells repressed the EGF-dependent ERK1/2 activation and the hyperproliferative response to EGF stimulation. Thus, inhibition of PKHD1 can cause EGF-induced excessive proliferation through decreasing intracellular $Ca^{2+}$ resulting in EGF-induced ERK1/2 activation. Our results suggest that the loss of fibrocystin may lead to abnormal proliferation in kidney epithelial cells and cyst formation in ARPKD by modulation of intracellular $Ca^{2+}$.

Kaempferol Regulates the Expression of Airway MUC5AC Mucin Gene via IκBα-NF-κB p65 and p38-p44/42-Sp1 Signaling Pathways

  • Li, Xin;Jin, Fengri;Lee, Hyun Jae;Lee, Choong Jae
    • Biomolecules & Therapeutics
    • /
    • 제29권3호
    • /
    • pp.303-310
    • /
    • 2021
  • In the present study, kaempferol, a flavonoidal natural compound found in Polygonati Rhizoma, was investigated for its potential effect on the gene expression and production of airway MUC5AC mucin. A human respiratory epithelial NCI-H292 cells was pretreated with kaempferol for 30 min and stimulated with epidermal growth factor (EGF) or phorbol 12-myristate 13-acetate (PMA), for the following 24 h. The effect on PMA-induced nuclear factor kappa B (NF-κB) signaling pathway or EGF-induced mitogen-activated protein kinase (MAPK) signaling pathway was investigated. Kaempferol suppressed the production and gene expression of MUC5AC mucins, induced by PMA through the inhibition of degradation of inhibitory kappa Bα (IκBα), and NF-κB p65 nuclear translocation. Also, kaempferol inhibited EGF-induced gene expression and production of MUC5AC mucin through regulating the phosphorylation of EGFR, phosphorylation of p38 MAPK and extracellular signal-regulated kinase (ERK) 1/2 (p44/42), and the nuclear expression of specificity protein-1 (Sp1). These results suggest kaempferol regulates the gene expression and production of mucin through regulation of NF-κB and MAPK signaling pathways, in human airway epithelial cells.

Translocalization of enhanced PKM2 protein into the nucleus induced by cancer upregulated gene 2 confers cancer stem cell-like phenotypes

  • Yawut, Natpaphan;Kaowinn, Sirichat;Cho, Il-Rae;Budluang, Phatcharaporn;Kim, Seonghye;Kim, Suhkmann;Youn, So Eun;Koh, Sang Seok;Chung, Young-Hwa
    • BMB Reports
    • /
    • 제55권2호
    • /
    • pp.98-103
    • /
    • 2022
  • Increased mRNA levels of cancer upregulated gene (CUG)2 have been detected in many different tumor tissues using Affymetrix microarray. Oncogenic capability of the CUG2 gene has been further reported. However, the mechanism by which CUG2 overexpression promotes cancer stem cell (CSC)-like phenotypes remains unknown. With recent studies showing that pyruvate kinase muscle 2 (PKM2) is overexpressed in clinical tissues from gastric, lung, and cervical cancer patients, we hypothesized that PKM2 might play an important role in CSC-like phenotypes caused by CUG2 overexpression. The present study revealed that PKM2 protein levels and translocation of PKM2 into the nucleus were enhanced in CUG2-overexpressing lung carcinoma A549 and immortalized bronchial BEAS-2B cells than in control cells. Expression levels of c-Myc, CyclinD1, and PKM2 were increased in CUG2-overexpressing cells than in control cells. Furthermore, EGFR and ERK inhibitors as well as suppression of Yap1 and NEK2 expression reduced PKM2 protein levels. Interestingly, knockdown of β-catenin expression failed to reduce PKM2 protein levels. Furthermore, reduction of PKM2 expression with its siRNA hindered CSC-like phenotypes such as faster wound healing, aggressive transwell migration, and increased size/number of sphere formation. The introduction of mutant S37A PKM2-green fluorescence protein (GFP) into cells without ability to move to the nucleus did not confer CSC-like phenotypes, whereas forced expression of wild-type PKM2 promoted such phenotypes. Overall, CUG2-induced increase in the expression of nuclear PKM2 contributes to CSC-like phenotypes by upregulating c-Myc and CyclinD1 as a co-activator.