3
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      mTOR inhibitor Everolimus-induced apoptosis in melanoma cells

      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

          Melanoma is the most aggressive, therapy-resistant skin cancer. The mammalian target of rapamycin (mTOR), the serine/threonine kinase which integrates both intracellular and extracellular signals, plays a crucial role in coordinating the balance between the growth and death of cells. The object of this study is a comparison of the influence of mTOR inhibitor everolimus in the concentration range between 20 nM and 10 μM, used individually and in combination with selected downstream protein kinases inhibitors: LY294002 (PI3K), U0126 (ERK1/2), AS-703026 (MEK) and MK-2206 (AKT) on the expression of pro-survival proteins: p-Bcl-2 (S70), p-Bcl-2 (T56), Bcl-2, Bcl-xL, Mcl-1, activity of caspase-3, proliferation and induction of apoptosis in melanoma cells. Current results clearly show that the nanomolar concentration of the mTOR inhibitor everolimus in combination with the inhibitor of MAP kinase (AS-703026) or AKT kinase (MK-2206) is effective in inducing apoptosis and reducing proliferation of melanoma cells. The herein research results confirm the hypothesis on the important role of mTOR signaling in cancer progression, and gives hope that implementation of successful combination of its inhibitors will find recognition and application in cancer treatment in the near future.

          Related collections

          Most cited references29

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

          Mechanisms of apoptosis sensitivity and resistance to the BH3 mimetic ABT-737 in acute myeloid leukemia.

          BCL-2 proteins are critical for cell survival and are overexpressed in many tumors. ABT-737 is a small-molecule BH3 mimetic that exhibits single-agent activity against lymphoma and small-cell lung cancer in preclinical studies. We here report that ABT-737 effectively kills acute myeloid leukemia blast, progenitor, and stem cells without affecting normal hematopoietic cells. ABT-737 induced the disruption of the BCL-2/BAX complex and BAK-dependent but BIM-independent activation of the intrinsic apoptotic pathway. In cells with phosphorylated BCL-2 or increased MCL-1, ABT-737 was inactive. Inhibition of BCL-2 phosphorylation and reduction of MCL-1 expression restored sensitivity to ABT-737. These data suggest that ABT-737 could be a highly effective antileukemia agent when the mechanisms of resistance identified here are considered.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Anticancer therapy targeting the apoptotic pathway.

            Apoptosis, or programmed cell death, has an essential role in controlling cell number in many developmental and physiological settings and in chemotherapy-induced tumour-cell killing. It is a genetically regulated biological process, guided by the ratio of proapoptotic and antiapoptotic proteins. Recently, inducers of apoptosis have been used in cancer therapy. Several studies have attempted to induce apoptosis by triggering the tumour-necrosis-factor-related apoptosis-inducing ligand receptor and the BCL2 family of proteins, and others have targeted the caspases, and proteins that inhibit apoptosis. Most of these therapies are still in preclinical development because of their low efficacy and susceptibility to drug resistance, but some of them have shown promising results. In this article, we review the development and clinical efficacy of proapoptotic drugs that have shown promise.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Mechanisms and context underlying the role of autophagy in cancer metastasis

              Macroautophagy/autophagy is a fundamental cellular degradation mechanism that maintains cell homeostasis, regulates cell signaling, and promotes cell survival. Its role in promoting tumor cell survival in stress conditions is well characterized, and makes autophagy an attractive target for cancer therapy. Emerging research indicates that autophagy also influences cancer metastasis, which is the primary cause of cancer-associated mortality. However, data demonstrate that the regulatory role of autophagy in metastasis is multifaceted, and includes both metastasis-suppressing and -promoting functions. The metastasis-suppressing functions of autophagy, in particular, have important implications for autophagy-based treatments, as inhibition of autophagy may increase the risk of metastasis. In this review, we discuss the mechanisms and context underlying the role of autophagy in metastasis, which include autophagy-mediated regulation of focal adhesion dynamics, integrin signaling and trafficking, Rho GTPase-mediated cytoskeleton remodeling, anoikis resistance, extracellular matrix remodeling, epithelial-to-mesenchymal transition signaling, and tumor-stromal cell interactions. Through this, we aim to clarify the context-dependent nature of autophagy-mediated metastasis and provide direction for further research investigating the role of autophagy in cancer metastasis.
                Bookmark

                Author and article information

                Contributors
                +4812 4227400 , mbciolcz@cyf-kr.edu.pl
                Journal
                J Cell Commun Signal
                J Cell Commun Signal
                Journal of Cell Communication and Signaling
                Springer Netherlands (Dordrecht )
                1873-9601
                1873-961X
                8 March 2019
                8 March 2019
                September 2019
                : 13
                : 3
                : 357-368
                Affiliations
                ISNI 0000 0001 2162 9631, GRID grid.5522.0, Medical Biochemistry, , Jagiellonian University Medical College, ; ul. Kopernika 7, 31-034 Kraków, Poland
                Author information
                http://orcid.org/0000-0003-2631-7308
                Article
                510
                10.1007/s12079-019-00510-0
                6732148
                30848427
                3686f922-bca8-440a-92b4-54cd9b8a0493
                © The Author(s) 2019

                Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License ( http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

                History
                : 2 February 2019
                : 22 February 2019
                Funding
                Funded by: FundRef http://dx.doi.org/10.13039/100009045, Uniwersytet Jagielloński Collegium Medicum;
                Award ID: CCBY
                Award Recipient :
                Categories
                Article
                Custom metadata
                © The International CCN Society 2019

                Cell biology
                melanoma,apoptosis,caspase-3 activity,proliferation,protein kinase inhibitors,mtor
                Cell biology
                melanoma, apoptosis, caspase-3 activity, proliferation, protein kinase inhibitors, mtor

                Comments

                Comment on this article