• 제목/요약/키워드: Cancer therapeutics

검색결과 561건 처리시간 0.022초

생식생물학에세 프로테오믹스의 응용 (Potential Importance of Proteomics in Research of Reproductive Biology)

  • 김호승;윤용달
    • 한국발생생물학회지:발생과생식
    • /
    • 제8권1호
    • /
    • pp.1-9
    • /
    • 2004
  • 프로테오믹스(proteomics, 단백질체학이라고도 함)의 잠재적 중요성은 간질환, 심장질환, 몇몇 종류의 암 등의 의학, 생식 독성, 발생 독성, 생체 독성 연구 분야에서도 명백하게 제시되었다. 그러나 단백질을 대상으로 연구하여 유전자 기능을 연구하는 프로테오믹스 연구를 각각의 분야에 접목시키려는 노력은 아직까지 빈약하다. 프로테오믹스는 기능을 갖는 단백질들의 발현을 종합적이고 정량적으로 측정하는 가장 직접적인 수단이고, 질병, 약물투여, 쇼크, 내분비계 장애물질 등 생물학적인 동요(perturbation)에 의하여 변하는 단백질들의 발현 양상 변화를 정확하게 관찰할 수 있게 한다. 그리고 생체내 유전자 발현의 궁극적인 양상을 규명할 수 있으며, 또한 유전자, 단백질 및 질병간의 연결고리를 제공한다. 기존의 biomarker는 다른 질병 표지자와 연관성이 높아 직접적인 biomaker와 정확한 연관을 판정하기 어렵다. 따라서 대량 발굴 탐색(high-throughput screening)이 가능한 2차원 전기 영동 분석과 MALDI-TOF또는 protein chip array와 SELDI-TOF에 의한 단백질 분자 구조 분석 기술 및 이들을 지원하는 생물정보학(bioinformatics)의 발전을 이용하여, 생식학 연구에 이용할 수 있는 표적 단백질 발굴 및 정성 정량적 연구에 적절한 이용이 가능할 것이다. 이러한 연구는 생식과정 중 배아 발생 및 조직 기관 발생 중 유전자 발현의 변화, 내분비계 장애물질 등 호르몬 및 독성 물질의 작용 기전, ecotoxicogenomics지표 marker의 변동 분석, 중간대사물질체학(metabolomics)에의 이용 등등의 연구에 필수적인 방법으로 발전할 것이다.

  • PDF

Selective Estrogen Receptor Modulation by Larrea nitida on MCF-7 Cell Proliferation and Immature Rat Uterus

  • Ahn, Hye-Na;Jeong, Si-Yeon;Bae, Gyu-Un;Chang, Minsun;Zhang, Dongwei;Liu, Xiyuan;Pei, Yihua;Chin, Young-Won;Lee, Joongku;Oh, Sei-Ryang;Song, Yun Seon
    • Biomolecules & Therapeutics
    • /
    • 제22권4호
    • /
    • pp.347-354
    • /
    • 2014
  • Larrea nitida is a plant that belongs to the Zygophyllaceae family and is widely used in South America to treat inflammatory diseases, tumors and menstrual pain. However, its pharmacological activity remains unclear. In this study we evaluated the property of selective estrogen receptor modulator (SERM) of Larrea nitida extracts (LNE) as a phytoestrogen that can mimic, modulate or disrupt the actions of endogenous estrogens, depending on the tissue and relative amount of other SERMs. To investigate the property of SERM of LNE, we performed MCF-7 cell proliferation assays, estrogen response element (ERE)-luciferase reporter gene assay, human estrogen receptor (hER) binding assays and in vivo uterotrophic assay. To gain insight into the active principles, we performed a bioassay-guided analysis of LNE employing solvents of various polarities and using classical column chromatography, which yielded 16 fractions (LNs). LNE showed high binding affinities for $hER{\alpha}$ and $hER{\beta}$ with $IC_{50}$ values of $1.20{\times}10^{-7}$ g/ml and $1.00{\times}10^{-7}$ g/ml, respectively. LNE induced $17{\beta}$-estradiol (E2)-induced MCF-7 cell proliferation, however, it reduced the proliferation in the presence of E2. Furthermore, LNE had an atrophic effect in the uterus of immature rats through reducing the expression level of progesterone receptor (PR) proteins. LN08 and LN10 had more potent affinities for binding on $hER{\alpha}$ and ${\beta}$ than other fractions. Our results indicate that LNE had higher binding affinities for $hER{\beta}$ than $hER{\alpha}$, and showed SERM properties in MCF-7 breast cancer cells and the rat uterus. LNE may be useful for the treatment of estrogen-related conditions, such as female cancers and menopause.

대장암과 선종 병변에서 mTOR 신호 단백질의 면역조직화학 염색성 평가 (Evaluation of the Immunohistochemical Staining Pattern of the mTOR Signaling Proteins in Colorectal Cancers and Adenoma Lesions)

  • 김진목;이현욱
    • 대한임상검사과학회지
    • /
    • 제49권4호
    • /
    • pp.470-476
    • /
    • 2017
  • mTOR 신호전달 단백질의 변화는 다양한 종류의 암에서 관찰 되었다. 따라서 이들 단백질은 최근에 암 치료제에 대한 새롭고 흥미로운 표적이 되고 있다. 우리는 대장암과 선종 환자의 mTOR 세포신호의 활성도를 조사하였다. 면역조직화학적 방법으로 대장암과 선종의 세포신호 단백질 성분인 mTOR, p70-S6K, S6, 4EBP1 발현을 분석하였다. 이번 연구는 모두 100개의 예를 악성(Colorectal Adenocarcinoma, CRAC) 40건, 고등급 선종(Adenoma with High grade intraepithelial neoplasms, HIN) 30건, 저등급 선종(Adenoma with Low-grade intraepithelial neoplasms, LIN) 30건으로 분류하여 진행하였다. p-mTOR의 발현률은 LIN 7%, HIN 30%, CRAC 75%였고 p-S6의 발현률 또한 LIN 10%, HIN 27%, CRAC 55%였다. p-mTOR, p-S6의 발현과 선종-선암 연속성은 중요한 상관관계 있다는 것이 발견되었다. 그리고 흥미롭게도 p-S6 발현률은 진행암보다 초기암에서 더 높았다.

β-lapachone-Induced Apoptosis of Human Gastric Carcinoma AGS Cells Is Caspase-Dependent and Regulated by the PI3K/Akt Pathway

  • Yu, Hai Yang;Kim, Sung Ok;Jin, Cheng-Yun;Kim, Gi-Young;Kim, Wun-Jae;Yoo, Young Hyun;Choi, Yung Hyun
    • Biomolecules & Therapeutics
    • /
    • 제22권3호
    • /
    • pp.184-192
    • /
    • 2014
  • ${\beta}$-lapachone is a naturally occurring quinone that selectively induces apoptotic cell death in a variety of human cancer cells in vitro and in vivo; however, its mechanism of action needs to be further elaborated. In this study, we investigated the effects of ${\beta}$-lapachone on the induction of apoptosis in human gastric carcinoma AGS cells. ${\beta}$-lapachone significantly inhibited cellular proliferation, and some typical apoptotic characteristics such as chromatin condensation and an increase in the population of sub-G1 hypodiploid cells were observed in ${\beta}$-lapachone-treated AGS cells. Treatment with ${\beta}$-lapachone caused mitochondrial transmembrane potential dissipation, stimulated the mitochondria-mediated intrinsic apoptotic pathway, as indicated by caspase-9 activation, cytochrome c release, Bcl-2 downregulation and Bax upregulation, as well as death receptor-mediated extrinsic apoptotic pathway, as indicated by activation of caspase-8 and truncation of Bid. This process was accompanied by activation of caspase-3 and concomitant with cleavage of poly(ADP-ribose) polymerase. The general caspase inhibitor, z-VAD-fmk, significantly abolished ${\beta}$-lapachone-induced cell death and inhibited growth. Further analysis demonstrated that the induction of apoptosis by ${\beta}$-lapachone was accompanied by inactivation of the phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathway. The PI3K inhibitor LY29004 significantly increased ${\beta}$-lapachone-induced apoptosis and growth inhibition. Taken together, these findings indicate that the apoptotic activity of ${\beta}$-lapachone is probably regulated by a caspase-dependent cascade through activation of both intrinsic and extrinsic signaling pathways, and that inhibition of the PI3K/Akt signaling may contribute to ${\beta}$-lapachone-mediated AGS cell growth inhibition and apoptosis induction.

Baicalein Inhibits the Migration and Invasion of B16F10 Mouse Melanoma Cells through Inactivation of the PI3K/Akt Signaling Pathway

  • Choi, Eun-Ok;Cho, Eun-Ju;Jeong, Jin-Woo;Park, Cheol;Hong, Su-Hyun;Hwang, Hye-Jin;Moon, Sung-Kwon;Son, Chang Gue;Kim, Wun-Jae;Choi, Yung Hyun
    • Biomolecules & Therapeutics
    • /
    • 제25권2호
    • /
    • pp.213-221
    • /
    • 2017
  • Baicalein, a natural flavonoid obtained from the rhizome of Scutellaria baicalensis Georgi, has been reported to have anticancer activities in several human cancer cell lines. However, its antimetastatic effects and associated mechanisms in melanoma cells have not been extensively studied. The current study examined the effects of baicalein on cell motility and anti-invasive activity using mouse melanoma B16F10 cells. Within the noncytotoxic concentration range, baicalein significantly inhibited the cell motility and invasiveness of B16F10 cells in a concentration-dependent manner. Baicalein also reduced the activity and expression of matrix metalloproteinase (MMP)-2 and -9; however, the levels of tissue inhibitor of metalloproteinase-1 and -2 were concomitantly increased. The inhibitory effects of baicalein on cell motility and invasiveness were found to be associated with its tightening of tight junction (TJ), which was demonstrated by an increase in transepithelial electrical resistance and downregulation of the claudin family of proteins. Additionally, treatment with baicalein markedly reduced the expression levels of lipopolysaccharide-induced phosphorylated Akt and the invasive activity in B16F10 cells. Taken together, these results suggest that baicalein inhibits B16F10 melanoma cell migration and invasion by reducing the expression of MMPs and tightening TJ through the suppression of claudin expression, possibly in association with a suppression of the phosphoinositide 3-kinase/Akt signaling pathway.

위축성 위염과 장상피화생의 호전에 영향을 미치는 인자에 대한 전향적 연구 (Predictive Factors for Improvement of Atrophic Gastritis and Intestinal Metaplasia: A Long-term Prospective Clinical Study)

  • 황영재;김나영;윤창용;권민구;백성민;권영재;이혜승;이제봉;최윤진;윤혁;신철민;박영수;이동호
    • 대한상부위장관⦁헬리코박터학회지
    • /
    • 제18권3호
    • /
    • pp.186-197
    • /
    • 2018
  • Background/Aims: To investigate the predictive factors for improvement of atrophic gastritis (AG) and intestinal metaplasia (IM). Materials and Methods: A total of 778 subjects were prospectively enrolled and followed up for 10 years. Histological analysis of AG and IM was performed by using the updated Sydney system. To find the predictive factors for reversibility of AG and IM, 24 factors including genetic polymorphisms and bacterial and environmental factors were analyzed. Results: In all subjects, the predictive factor by multivariate analysis for improvement of both antral and corpus AG was successful eradication. The predictive factors for improvement of antral IM were age and successful eradication. The predictive factor for improvement of corpus IM was successful eradication. In patients with Helicobacter pylori infection, age and cagA were predictive factors for improvement of AG and IM. In patients with H. pylori eradication, monthly income and cagA were predictive factors for improvement of AG and IM. Conclusions: H. pylori eradication is an important predictive factor of regression of AG and IM and would be beneficial for the prevention of intestinal-type gastric cancer. Young age, high income, and cagA are additional predictive factors for improving AG and IM status. Thus, various factors affect the improvement of AG and IM.

A Novel Anti-PD-L1 Antibody Exhibits Antitumor Effects on Multiple Myeloma in Murine Models via Antibody-Dependent Cellular Cytotoxicity

  • Ahn, Jae-Hee;Lee, Byung-Hyun;Kim, Seong-Eun;Kwon, Bo-Eun;Jeong, Hyunjin;Choi, Jong Rip;Kim, Min Jung;Park, Yong;Kim, Byung Soo;Kim, Dae Hee;Ko, Hyun-Jeong
    • Biomolecules & Therapeutics
    • /
    • 제29권2호
    • /
    • pp.166-174
    • /
    • 2021
  • Multiple myeloma is a malignant cancer of plasma cells. Despite recent progress with immunomodulatory drugs and proteasome inhibitors, it remains an incurable disease that requires other strategies to overcome its recurrence and non-response. Based on the high expression levels of programmed death-ligand 1 (PD-L1) in human multiple myeloma isolated from bone marrow and the murine myeloma cell lines, NS-1 and MOPC-315, we propose PD-L1 molecule as a target of anti-multiple myeloma therapy. We developed a novel anti-PD-L1 antibody containing a murine immunoglobulin G subclass 2a (IgG2a) fragment crystallizable (Fc) domain that can induce antibody-dependent cellular cytotoxicity. The newly developed anti-PD-L1 antibody showed significant antitumor effects against multiple myeloma in mice subcutaneously, intraperitoneally, or intravenously inoculated with NS-1 and MOPC-315 cells. The anti-PD-L1 effects on multiple myeloma may be related to a decrease in the immunosuppressive myeloid-derived suppressor cells (MDSCs), but there were no changes in the splenic MDSCs after combined treatment with lenalidomide and the anti-PD-L1 antibody. Interestingly, the newly developed anti-PD-L1 antibody can induce antibody-dependent cellular cytotoxicity in the myeloma cells, which differs from the existing anti-PD-L1 antibodies. Collectively, we have developed a new anti-PD-L1 antibody that binds to mouse and human PD-L1 and demonstrated the antitumor effects of the antibody in several syngeneic murine myeloma models. Thus, PD-L1 is a promising target to treat multiple myeloma, and the novel anti-PD-L1 antibody may be an effective anti-myeloma drug via antibody-dependent cellular cytotoxicity effects.

Bioactive Lipids and Their Derivatives in Biomedical Applications

  • Park, Jinwon;Choi, Jaehyun;Kim, Dae-Duk;Lee, Seunghee;Lee, Bongjin;Lee, Yunhee;Kim, Sanghee;Kwon, Sungwon;Noh, Minsoo;Lee, Mi-Ock;Le, Quoc-Viet;Oh, Yu-Kyoung
    • Biomolecules & Therapeutics
    • /
    • 제29권5호
    • /
    • pp.465-482
    • /
    • 2021
  • Lipids, which along with carbohydrates and proteins are among the most important nutrients for the living organism, have a variety of biological functions that can be applied widely in biomedicine. A fatty acid, the most fundamental biological lipid, may be classified by length of its aliphatic chain, and the short-, medium-, and long-chain fatty acids and each have distinct biological activities with therapeutic relevance. For example, short-chain fatty acids have immune regulatory activities and could be useful against autoimmune disease; medium-chain fatty acids generate ketogenic metabolites and may be used to control seizure; and some metabolites oxidized from long-chain fatty acids could be used to treat metabolic disorders. Glycerolipids play important roles in pathological environments, such as those of cancers or metabolic disorders, and thus are regarded as a potential therapeutic target. Phospholipids represent the main building unit of the plasma membrane of cells, and play key roles in cellular signaling. Due to their physical properties, glycerophospholipids are frequently used as pharmaceutical ingredients, in addition to being potential novel drug targets for treating disease. Sphingolipids, which comprise another component of the plasma membrane, have their own distinct biological functions and have been investigated in nanotechnological applications such as drug delivery systems. Saccharolipids, which are derived from bacteria, have endotoxin effects that stimulate the immune system. Chemically modified saccharolipids might be useful for cancer immunotherapy or as vaccine adjuvants. This review will address the important biological function of several key lipids and offer critical insights into their potential therapeutic applications.

Analysis of SARS-CoV-2 Mutations after Nirmatrelvir Treatment in a Lung Cancer Xenograft Mouse Model

  • Bo Min Kang;Dongbum Kim;Jinsoo Kim;Kyeongbin Baek;Sangkyu Park;Ha-Eun Shin;Myeong-Heon Lee;Minyoung Kim;Suyeon Kim;Younghee Lee;Hyung-Joo Kwon
    • Biomolecules & Therapeutics
    • /
    • 제32권4호
    • /
    • pp.481-491
    • /
    • 2024
  • Paxlovid is the first approved oral treatment for coronavirus disease 2019 and includes nirmatrelvir, a protease inhibitor targeting the main protease (Mpro) of SARS-CoV-2, as one of the key components. While some specific mutations emerged in Mpro were revealed to significantly reduce viral susceptibility to nirmatrelvir in vitro, there is no report regarding resistance to nirmatrelvir in patients and animal models for SARS-CoV-2 infection yet. We recently developed xenograft tumors derived from Calu-3 cells in immunodeficient mice and demonstrated extended replication of SARS-CoV-2 in the tumors. In this study, we investigated the effect of nirmatrelvir administration on SARS-CoV-2 replication. Treatment with nirmatrelvir after virus infection significantly reduced the replication of the parental SARS-CoV-2 and SARS-CoV-2 Omicron at 5 days post-infection (dpi). However, the virus titers were completely recovered at the time points of 15 and 30 dpi. The virus genomes in the tumors at 30 dpi were analyzed to investigate whether nirmatrelvir-resistant mutant viruses had emerged during the extended replication of SARS-CoV-2. Various mutations in several genes including ORF1ab, ORF3a, ORF7a, ORF7b, ORF8, and N occurred in the SARS-CoV-2 genome; however, no mutations were induced in the Mpro sequence by a single round of nirmatrelvir treatment, and none were observed even after two rounds of treatment. The parental SARS-CoV-2 and its sublineage isolates showed similar IC50 values of nirmatrelvir in Vero E6 cells. Therefore, it is probable that inducing viral resistance to nirmatrelvir in vivo is challenging differently from in vitro passage.

The Combination of Gefitinib and Acetaminophen Exacerbates Hepatotoxicity via ROS-Mediated Apoptosis

  • Jiangxin Xu;Xiangliang Huang;Yourong Zhou;Zhifei Xu;Xinjun Cai;Bo Yang;Qiaojun He;Peihua Luo;Hao Yan;Jie Jin
    • Biomolecules & Therapeutics
    • /
    • 제32권5호
    • /
    • pp.647-657
    • /
    • 2024
  • Gefitinib is the well-tolerated first-line treatment of non-small cell lung cancer. As it needs analgesics during oncology treatment, particularly in the context of the coronavirus disease, where patients are more susceptible to contract high fever and sore throat. This has increased the likelihood of taking both gefitinib and antipyretic analgesic acetaminophen (APAP). Given that gefitinib and APAP overdose can predispose patients to liver injury or even acute liver failure, there is a risk of severe hepatotoxicity when these two drugs are used concomitantly. However, little is known regarding their safety at therapeutic doses. This study simulated the administration of gefitinib and APAP at clinically relevant doses in an animal model and confirmed that gefitinib in combination with APAP exhibited additional hepatotoxicity. We found that gefitinib plus APAP significantly exacerbated cell death, whereas each drug by itself had little or minor effect on hepatocyte survival. Mechanistically, combination of gefitinib and APAP induces hepatocyte death via the apoptotic pathway obviously. Reactive oxygen species (ROS) generation and DNA damage accumulation are involved in hepatocyte apoptosis. Gefitinib plus APAP also promotes the expression of Kelch-like ECH-associated protein 1 (Keap1) and downregulated the antioxidant factor, Nuclear factor erythroid 2-related factor 2 (Nrf2), by inhibiting p62 expression. Taken together, this study revealed the potential ROS-mediated apoptosis-dependent hepatotoxicity effect of the combination of gefitinib and APAP, in which the p62/Keap1/Nrf2 signaling pathway participates and plays an important regulatory role.