27
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Pectolinarigenin Induced Cell Cycle Arrest, Autophagy, and Apoptosis in Gastric Cancer Cell via PI3K/AKT/mTOR Signaling Pathway

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Pectolinarigenin (PEC), a natural flavonoid present in Cirsium chanroenicum and in some species of Citrus fruits, has various pharmacological benefits such as anti-inflammatory and anti-cancer activities. In the present study, we investigated the anti-cancer mechanism of PEC induced cell death caused by autophagy and apoptosis in AGS and MKN28 human gastric cancer cells. The PEC treatment significantly inhibited the AGS and MKN28 cell growth in a dose-dependent manner. Further, PEC significantly elevated sub-G1 phase in AGS cells and G2/M phase cell cycle arrest in both AGS and MKN28 cells. Apoptosis was confirmed by Annexin V and Hoechst 33342 fluorescent staining. Moreover, Immunoblotting results revealed that PEC treatment down-regulated the inhibitor of apoptosis protein (IAP) family protein XIAP that leads to the activation of caspase-3 thereby cleavage of PARP (poly-ADP-ribose polymerase) in both AGS and MKN28 cells in a dose-dependent manner. The autophagy-inducing effect was indicated by the increased formation of acidic vesicular organelles (AVOs) and increased protein levels of LC3-II conversion in both AGS and MKN28 cells. PEC shows the down regulation of PI3K/AKT/mTOR pathway which is a major regulator of autophagic and apoptotic cell death in cancer cells that leads to the down-regulation of p-4EBP1, p-p70S6K, and p-eIF4E in PEC treated cells when compared with the untreated cells. In conclusion, PEC treatment might have anti-cancer effect by down-regulation of PI3K/AKT/mTOR pathway leading to G2/M phase cell cycle arrest, autophagic and apoptotic cell death in human gastric cancer cells. Further studies of PEC treatment can support to develop as a potential alternative therapeutic agent for human gastric carcinoma.

          Related collections

          Most cited references25

          • Record: found
          • Abstract: not found
          • Article: not found

          Epidemiology of gastric cancer

            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Targeting Apoptosis Pathways in Cancer Therapy

            Apoptosis, or programmed cell death, is a mechanism by which cells undergo death to control cell proliferation or in response to DNA damage. The understanding of apoptosis has provided the basis for novel targeted therapies that can induce death in cancer cells or sensitize them to established cytotoxic agents and radiation therapy. These novel agents include those targeting the extrinsic pathway such as tumor necrosis factor-related apoptosis-inducing ligand receptor 1, and those targeting the intrinsic Bcl-2 family pathway such as antisense bcl-2 oligonucleotides. Many pathways and proteins control the apoptosis machinery. Examples include p53, the nuclear factor kappa B, the phosphatidylinositol 3 kinase pathway, and the ubiquitin/proteosome pathway. These can be targeted by specific modulators such as bortezomib, and mammalian target of rapamycin inhibitors such as CCI-779 and RAD 001. Because these pathways may be preferentially altered in tumor cells, there is potential for a selective effect in tumors sparing normal tissue. This article reviews the current understanding of the apoptotic pathways, including the extrinsic (cytoplasmic) and intrinsic (mitochondrial) pathways, and the agents being developed to target these pathways.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Targeting the PI3K-AKT-mTOR pathway: progress, pitfalls, and promises.

              The strategy of 'drugging the cancer kinome' has led to the successful development and regulatory approval of several novel molecular targeted agents. The spotlight is now shifting to the phosphatidylinositide 3-kinase (PI3K)-AKT-mammalian target of rapamycin (mTOR) pathway as a key potential target. This review details the role of the pathway in oncogenesis and the rationale for inhibiting its vital components. The focus will be on the progress made in the development of novel therapies for cancer treatment, with emphasis placed on agents that have entered clinical development. Strategies involving horizontal and vertical blockade of the pathway, as well as the use of biomarkers to select appropriate patients and to provide proof of target modulation will also be highlighted. Finally, we discuss the issues and limitations involved with targeting the PI3K-AKT-mTOR pathway, and predict what the future may hold for these novel anticancer therapeutics.
                Bookmark

                Author and article information

                Journal
                Nutrients
                Nutrients
                nutrients
                Nutrients
                MDPI
                2072-6643
                08 August 2018
                August 2018
                : 10
                : 8
                : 1043
                Affiliations
                [1 ]Research Institute of Life science and College of Veterinary Medicine, Gyeongsang National University, 501 Jinju-daero, Jinju 52828, Korea; lhj1990y@ 123456gmail.com (H.J.L.); gowdavenu27@ 123456gmail.com (V.V.G.S.); ksm4234@ 123456naver.com (S.M.K.); sangdis2@ 123456naver.com (S.E.H.)
                [2 ]Gyeongnam Department of Environment Toxicology and Chemistry, Biological Resources Research Group, Korea Institute of Toxicology, 17 Jegok-gil, Jinju 52834, Korea; sjlee@ 123456kitox.re.kr (S.J.L.); jdher@ 123456kitox.re.kr (J.D.H.)
                [3 ]Department of Internal Medicine, Gyeongsang National University Cancer Center, School of Medicine, Gyeongsang National University, 15 Jinju-daero, Jinju 52727, Korea; suchibiodash@ 123456gmail.com (S.R.); lwshmo@ 123456gnu.ac.kr (W.S.L.)
                [4 ]Department of Nursing Science, International University of Korea, 965 Dongbu-ro, Jinju 52833, Korea; iuknurse@ 123456nate.com
                Author notes
                [* ]Correspondence: gonskim@ 123456gnu.ac.kr ; Tel.: +82-55-772-2346
                Author information
                https://orcid.org/0000-0003-3359-7504
                https://orcid.org/0000-0002-0844-1009
                Article
                nutrients-10-01043
                10.3390/nu10081043
                6115855
                30096805
                25d134d3-d2b1-45ad-a879-87d692401894
                © 2018 by the authors.

                Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license ( http://creativecommons.org/licenses/by/4.0/).

                History
                : 29 June 2018
                : 06 August 2018
                Categories
                Article

                Nutrition & Dietetics
                pectolinarigenin,gastric cancer,apoptosis,autophagy,pi3k/akt/mtor
                Nutrition & Dietetics
                pectolinarigenin, gastric cancer, apoptosis, autophagy, pi3k/akt/mtor

                Comments

                Comment on this article