TPEN Exerts Antitumor Efficacy in Murine Mammary Adenocarcinoma Through an H2O2 Signaling Mechanism Dependent on Caspase-3

Anti-Cancer Agents in Medicinal Chemistry - Tập 18 Số 11 - Trang 1617-1628 - 2019
Viviana Soto‐Mercado1,2, Miguel Mendivil‐Perez1,2, Claudia Urueña-Pinzon3, Susana Fiorentino3, Carlos Vélez-Pardo1,2, Marlene Jiménez-Del-Río1,2
1Neuroscience Research Group, Medical Research Institute, Faculty of Medicine, University of Antioquia (UdeA), Calle 70 No. 52-21, and Calle 62 # 52-59,Building 1, Room 412, SIU Medellin, Colombia
2SIU Medellin,Colombia
3Grupo de Inmunobiologia y Biologia Celular, Facultad de Ciencias, Departamento de Microbiología, Pontificia Universidad Javeriana, Carrera 7 No. 40 - 62, Bogota, Colombia

Tóm tắt

Background: Breast cancer is the second most common cancer worldwide. N, N, N’, N’-Tetrakis (2-pyridylmethyl)-ethylenediamine (TPEN) is a lipid-soluble zinc metal chelator that induces apoptosis in cancer cells through oxidative stress (OS). However, the effectiveness and the mechanisms involved in TPENinduced cell death in mammary adenocarcinoma cells in vitro and in vivo are still unclear. Objective: This study aimed to evaluate the cytotoxic effect of TPEN in mouse embryonic fibroblasts (MEFs, as normal control cells) and mammary adenocarcinoma cancer cells (TS/A cells) in vitro and in a mammary tumor model in vivo. Methods: Cells were treated with TPEN (0-3 µM), and changes in nuclear chromatin and DNA, mitochondrial membrane potential (ΔΨm), and intracellular reactive oxygen species (ROS) levels were determined by both fluorescence microscopy and flow cytometry. Cell proliferation and the cell cycle were also analyzed. Cellular markers of apoptosis were evaluated by Western blot. Finally, the effect of TPEN in a mammary adenocarcinoma tumor model in vivo was determined by immunohistological analyses. Results: TPEN induced apoptosis in TS/A cells in a dose-dependent manner, increasing nuclear chromatin condensation, DNA fragmentation, cell cycle arrest and ΔΨm loss. Additionally, TPEN increased dichlorofluorescein fluorescence (DCF+) intensity, indicative of ROS production; increased DJ-1-Cys106-sulfonate expression, a marker of intracellular H2O2 stress; induced p53 and PUMA upregulation; and activated caspase-3. Moreover, TPEN induced mammary cancer cell elimination and tumor size reduction in vivo 48 h after treatment through an OS-induced apoptotic mechanism. Conclusion: TPEN selectively induces apoptosis in TS/A cells through an H2O2-mediated signaling pathway. Our findings support the use of TPEN as a potential treatment for breast cancer.

Từ khóa


Tài liệu tham khảo

Weinberg RA. A perspective on cancer cell metastasis.

Kumar A, Pal JK, Sharma NK. Breast cancer stem cells as last soldiers eluding therapeutic burn: A hard nut to crack.

Li ZL, He ZX, Qiu JX, Zhou SF. Molecular mechanisms for tumour resistance to chemotherapy.

Weinberg RA. Hallmarks of cancer: the next generation.

Bravo-San Pedro JM, Vitale I, Aaronson SA, Abrams JM, Adam D, Alnemri ES, Altucci L, Andrews D, Annicchiarico-Petruzzelli M, Baehrecke EH, Bazan NG, Bertrand MJ, Bianchi K, Blagosklonny MV, Blomgren K, Borner C, Bredesen DE, Brenner C, Campanella M, Candi E, Cecconi F, Chan FK, Chandel NS, Cheng EH, Chipuk JE, Cidlowski JA, Ciechanover A, Dawson TM, Dawson VL, De Laurenzi V, De Maria R, Debatin KM, Di Daniele N, Dixit VM, Dynlacht BD, El-Deiry WS, Fimia GM, Flavell RA, Fulda S, Garrido C, Gougeon ML, Green DR, Gronemeyer H, Hajnoczky G, Hardwick JM, Hengartner MO, Ichijo H, Joseph B, Jost PJ, Kaufmann T, Kepp O, Klionsky DJ, Knight RA, Kumar S, Lemasters JJ, Levine B, Linkermann A, Lipton SA, Lockshin RA, Lopez-Otin C, Lugli E, Madeo F, Malorni W, Marine JC, Martin SJ, Martinou JC, Medema JP, Meier P, Melino S, Mizushima N, Moll U, Munoz-Pinedo C, Nunez G, Oberst A, Panaretakis T, Penninger JM, Peter ME, Piacentini M, Pinton P, Prehn JH, Puthalakath H, Rabinovich GA, Ravichandran KS, Rizzuto R, Rodrigues CM, Rubinsztein DC, Rudel T, Shi Y, Simon HU, Stockwell BR, Szabadkai G, Tait SW, Tang HL, Tavernarakis N, Tsujimoto Y, Vanden Berghe T, Vandenabeele P, Villunger A, Wagner EF, Walczak H, White E, Wood WG, Yuan J, Zakeri Z, Zhivotovsky B, Melino G, Kroemer G. Essential versus accessory aspects of cell death: recommendations of the NCCD 2015.

Merhi RA, Rahal O, Stoyanovsky DA, Zaki A, Haidar H, Kagan VE, Gali-Muhtasib H, Machaca K. Copper chelation selectively kills colon cancer cells through redox cycling and generation of reactive oxygen species.

Fatfat M, Hankache C, Osman B, Khalife H, Machaca K, Muhtasib HG. Chk1 and DNA-PK mediate TPEN-induced DNA damage in a ROS dependent manner in human colon cancer cells.

Singh R, Smith TL, D’Agostino R, Caudell D, Balaji KC, Gmeiner WH. Prostate-specific membrane antigen-targeted liposomes specifically deliver the Zn(2+) chelator TPEN inducing oxidative stress in prostate cancer cells.

Velez-Pardo C, Jimenez-Del-Rio M. TPEN induces apoptosis independently of zinc chelator activity in a model of acute lymphoblastic leukemia and acute leukemia cells through oxidative stress and mitochondria caspase-3- and AIF-dependent pathways.

Velez-Pardo C, Jimenez-Del-Rio M. Metal chelator TPEN selectively induces apoptosis in K562 cells through reactive oxygen species signaling mechanism: implications for chronic myeloid leukemia.

Wang J, Zhou F, Liu Y, Lai Y, Chen X, Chen D, Luo L, Hua ZC. Zinc Depletion by TPEN Induces Apoptosis in Human Acute Promyelocytic NB4 Cells.

Ghavami S, Eshraghi M, Booy EP, Los M. Cytotoxic effects of intra and extracellular zinc chelation on human breast cancer cells.

de Giovanni C, Lollini PL, Nicoletti G, Prodi G. TS/A: a new metastasizing cell line from a BALB/c spontaneous mammary adenocarcinoma.

Drews K, Adjaye J. Preparation of mouse embryonic fibroblast cells suitable for culturing human embryonic and induced pluripotent stem cells.

Rodermond HM, Stap J, Haveman J, van-Bree C. Clonogenic assay of cells

Uruena CP, Llano M, Gomez-Cadena A, Hernandez JF, Sequeda LG, Loaiza AE, Barreto A, Li S, Fiorentino S. Standardized Extract from Caesalpinia spinosa is Cytotoxic Over Cancer Stem Cells and Enhance Anticancer Activity of Doxorubicin.

Hwang JJ, Han SH, Chung SJ, Koh JY, Lee JY. Endogenous zinc mediates apoptotic programmed cell death in the developing brain.

Matsunaga Y, Zhanghui W, Noda N, Asai Y, Moriwaki A, Matsumoto T, Nakano T, Matsumoto K, Nakanishi Y, Inoue H. A zinc chelator TPEN attenuates airway hyperresponsiveness and airway inflammation in mice

. Oxidized DJ-1 as a possible biomarker of Parkinson’s disease.

Rahn J, Lichtenfels R, Wessjohann LA, Seliger B. Hydrogen peroxide - production, fate and role in redox signaling of tumor cells.

Kimata J, Taira T, Ariga H, Niki E. Cysteine-106 of DJ-1 is the most sensitive cysteine residue to hydrogen peroxide-mediated oxidation in human umbilical vein endothelial cells.

Nishitoh H, Fujii M, Takeda K, Tobiume K, Sawada Y, Kawabata M, Miyazono K, Ichijo H. Mammalian thioredoxin is a direct inhibitor of apoptosis signal-regulating kinase (ASK) 1.

Peters RT, Dang LC, Maniatis T. MEKK1 activates both IkappaB kinase alpha and IkappaB kinase beta.

Hou G, Zhang Y, Dai Y, Zhao H. c-Jun transactivates Puma gene expression to promote osteoarthritis.

Schmidt C, Sclabas GM, Li Z, Pelicano H, Peng B, Yao A, Niu J, Zhang W, Evans DB, Abbruzzese JL, Huang P, Chiao PJ. Stabilization of p53 is a novel mechanism for proapoptotic function of NF-kappaB.

Attardi LD. p53 at a glance.

Zhang L, Hwang PM, Kinzler KW, Vogelstein B. PUMA induces the rapid apoptosis of colorectal cancer cells.

Kilianska ZM. PUMA, a critical mediator of cell death--one decade on from its discovery.

Chipuk JE. The deadly landscape of pro-apoptotic BCL-2 proteins in the outer mitochondrial membrane.

Sprengart ML, Wati MR, Porter AG. Caspase-3 is required for DNA fragmentation and morphological changes associated with apoptosis.

. MCF-7 breast carcinoma cells do not express caspase-3.

Jimenez-Del-Rio M, Sierra-Garcia L, Lopez-Osorio B, Velez-Pardo C. Vitamin E synthetic derivate-TPGS-selectively induces apoptosis in jurkat t cells oxidative stress signaling pathways: implications for acute lymphoblastic leukemia.