Skip to main content
Top
Published in: Breast Cancer Research and Treatment 1/2009

01-09-2009 | Letter to the editor

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

Author: Reiner U. Jänicke

Published in: Breast Cancer Research and Treatment | Issue 1/2009

Login to get access

Excerpt

Apoptosis, a fundamental process essential for normal tissue homeostasis and development, is closely associated with the activation of a class of aspartate-specific cysteine-dependent proteases, called caspases, that lead to the demise of the cell via limited proteolysis of a multitude of cellular substrates [1, 2]. Caspases are expressed as inactive zymogens that become activated upon cleavage by other caspases in a so-called caspase activation cascade, or by mere oligomerization instigated by the formation of large multi-protein complexes such as the death-inducing signaling complex (DISC) or the apoptosome [3, 4]. Whereas DISC formation occurs via the so-called extrinsic death pathway that is instigated by activation of members of the death receptor family such as CD95, tumor necrosis factor receptor (TNF-R) or the receptors of the TNF-R-related apoptosis-inducing ligand (TRAIL), the apoptosome is formed following activation of mitochondria and is hence termed the intrinsic or mitochondrial death pathway. Based on their order in cell death pathways, caspases can be divided into initiator (caspase-2, -8, -9, and -10) and effector (caspase-3, -6, and -7) caspases. Among them, caspase-3, a member of the latter group, is absolutely crucial for apoptosis induction, as this enzyme is not only activated downstream of both the extrinsic and intrinsic death pathway, it is also responsible for the cleavage of the majority of substrates known so far [1, 5]. More importantly, with the proteolysis of discrete substrates, caspase-3 evokes some of the typical morphological and biochemical alterations associated with apoptosis. For instance, whereas the caspase-3-mediated cleavages of α-fodrin, gelsolin, rho-associated kinase-1 (ROCK-1) and p21-activated kinase 2 (PAK2) contribute to membrane blebbing, cleavage of the inhibitor of the caspase-activated DNase (ICAD) leads to the typical DNA fragmentation pattern observed in apoptosis [1]. Furthermore, with the cleavage-mediated activation of the calcium-independent phospholipase A2 and subsequent production of the chemotactic phospholipid lysophosphatidylcholine, caspase-3 appears to be also responsible for the generation of so-called “eat-me” signals that induce migration of phagocytes to the site of apoptotic cell death [6]. Thus, caspase-3 not only instigates and pursues the demise of a cell, but, in addition, makes sure that the corpse is properly disposed, a function crucial for avoiding inflammatory processes. …
Literature
6.
go back to reference Lauber K, Bohn E, Kröber SM, Xiao YJ, Blumenthal SG, Lindemann RK, Marini P, Wiedig C, Zobywalski A, Baksh S, Xu Y, Autenrieth IB, Schulze-Osthoff K, Belka C, Stuhler G, Wesselborg S (2003) Apoptotic cells induce migration of phagocytes via caspase-3-mediated release of a lipid attraction signal. Cell 113:717–730. doi:10.1016/S0092-8674(03)00422-7 PubMedCrossRef Lauber K, Bohn E, Kröber SM, Xiao YJ, Blumenthal SG, Lindemann RK, Marini P, Wiedig C, Zobywalski A, Baksh S, Xu Y, Autenrieth IB, Schulze-Osthoff K, Belka C, Stuhler G, Wesselborg S (2003) Apoptotic cells induce migration of phagocytes via caspase-3-mediated release of a lipid attraction signal. Cell 113:717–730. doi:10.​1016/​S0092-8674(03)00422-7 PubMedCrossRef
7.
go back to reference Wang R, Wang X, Li B, Lin F, Dong K, Gao P, Zhang H-Z (2008) Tumor-specific adenovirus-mediated Puma gene transfer using the survivin promoter enhances radiosensitivity of breast cancer cells in vitro and in vivo. Breast Cancer Res Treat [Epub ahead of print] PMID: 18791823 Wang R, Wang X, Li B, Lin F, Dong K, Gao P, Zhang H-Z (2008) Tumor-specific adenovirus-mediated Puma gene transfer using the survivin promoter enhances radiosensitivity of breast cancer cells in vitro and in vivo. Breast Cancer Res Treat [Epub ahead of print] PMID: 18791823
9.
go back to reference Nizamutdinova IT, Lee GW, Son KH, Jeon SJ, Kang SS, Kim YS, Lee JH, Seo HG, Chang KC, Kim HJ (2008) Tanshione I effectively induces apoptosis in estrogen receptor-positive (MCF-7) and estrogen receptor-negative (MDA-MB-231) breast cancer cells. Int J Oncol 33:485–491PubMed Nizamutdinova IT, Lee GW, Son KH, Jeon SJ, Kang SS, Kim YS, Lee JH, Seo HG, Chang KC, Kim HJ (2008) Tanshione I effectively induces apoptosis in estrogen receptor-positive (MCF-7) and estrogen receptor-negative (MDA-MB-231) breast cancer cells. Int J Oncol 33:485–491PubMed
10.
go back to reference Kumar A, D’Souza SS, Gaonkar SL, Rai KML, Salimath BP (2008) Growth inhibition and induction of apoptosis in MCF-7 breast cancer cells by a new series of substituted-1,3,4-oxadiazole derivatives. Invest New Drugs 26:425–435. doi:10.1007/s10637-008-9116-5 PubMedCrossRef Kumar A, D’Souza SS, Gaonkar SL, Rai KML, Salimath BP (2008) Growth inhibition and induction of apoptosis in MCF-7 breast cancer cells by a new series of substituted-1,3,4-oxadiazole derivatives. Invest New Drugs 26:425–435. doi:10.​1007/​s10637-008-9116-5 PubMedCrossRef
11.
go back to reference Cui Q, Yu J-h, Wu J-n, Tashiro S-i, Onodera S, Minami M, Ikejima T (2007) p53-mediated cell cycle arrest and apoptosis through a caspase-3-independent, but caspase-9-dependent pathway in oridonin-treated MCF-7 human breast cancer cells. Acta Pharmacol Sin 28:1057–1066. doi:10.1111/j.1745-7254.2007.00588.x PubMedCrossRef Cui Q, Yu J-h, Wu J-n, Tashiro S-i, Onodera S, Minami M, Ikejima T (2007) p53-mediated cell cycle arrest and apoptosis through a caspase-3-independent, but caspase-9-dependent pathway in oridonin-treated MCF-7 human breast cancer cells. Acta Pharmacol Sin 28:1057–1066. doi:10.​1111/​j.​1745-7254.​2007.​00588.​x PubMedCrossRef
13.
go back to reference Zhang GP, Lu YY, Lv JC, Ou HJ (2006) Effect of ursolic acid on caspase-3 and PARP expression of human MCF-7 cells. Zhongguo Zhong Yao Za Zhi 31:141–144PubMed Zhang GP, Lu YY, Lv JC, Ou HJ (2006) Effect of ursolic acid on caspase-3 and PARP expression of human MCF-7 cells. Zhongguo Zhong Yao Za Zhi 31:141–144PubMed
14.
15.
go back to reference Chen JS, Konopleva M, Andreeff M, Multani AS, Pathak S, Mehta K (2004) Drug-resistant breast carcinoma (MCF-7) cells are paradoxically sensitive to apoptosis. J Cell Physiol 200:223–234. doi:10.1002/jcp.20014 PubMedCrossRef Chen JS, Konopleva M, Andreeff M, Multani AS, Pathak S, Mehta K (2004) Drug-resistant breast carcinoma (MCF-7) cells are paradoxically sensitive to apoptosis. J Cell Physiol 200:223–234. doi:10.​1002/​jcp.​20014 PubMedCrossRef
16.
19.
go back to reference Jänicke RU, Engels IH, Dunkern T, Kaina B, Schulze-Osthoff K, Porter AG (2001) Ionizing radiation but not anticancer drugs causes cell cycle arrest and failure to activate the mitochondrial death pathway in MCF-7 breast carcinoma cells. Oncogene 20:5043–5053. doi:10.1038/sj.onc.1204659 PubMedCrossRef Jänicke RU, Engels IH, Dunkern T, Kaina B, Schulze-Osthoff K, Porter AG (2001) Ionizing radiation but not anticancer drugs causes cell cycle arrest and failure to activate the mitochondrial death pathway in MCF-7 breast carcinoma cells. Oncogene 20:5043–5053. doi:10.​1038/​sj.​onc.​1204659 PubMedCrossRef
20.
go back to reference Essmann F, Engels IH, Totzke G, Schulze-Osthoff K, Jänicke RU (2004) Apoptosis resistance of MCF-7 breast carcinoma cells to ionizing radiation is independent of p53 and cell cycle control but caused by the lack of caspase-3 and a caffeine-inhibitable event. Cancer Res 64:7065–7072. doi:10.1158/0008-5472.CAN-04-1082 PubMedCrossRef Essmann F, Engels IH, Totzke G, Schulze-Osthoff K, Jänicke RU (2004) Apoptosis resistance of MCF-7 breast carcinoma cells to ionizing radiation is independent of p53 and cell cycle control but caused by the lack of caspase-3 and a caffeine-inhibitable event. Cancer Res 64:7065–7072. doi:10.​1158/​0008-5472.​CAN-04-1082 PubMedCrossRef
22.
go back to reference Wolf BB, Schuler M, Echeverri F, Green DR (1999) Caspase-3 is the primary activator of apoptotic DNA fragmentation via DNA fragmentation factor-45/inhibitor of caspase-activated DNase inactivation. J Biol Chem 274:30651–30656. doi:10.1074/jbc.274.43.30651 PubMedCrossRef Wolf BB, Schuler M, Echeverri F, Green DR (1999) Caspase-3 is the primary activator of apoptotic DNA fragmentation via DNA fragmentation factor-45/inhibitor of caspase-activated DNase inactivation. J Biol Chem 274:30651–30656. doi:10.​1074/​jbc.​274.​43.​30651 PubMedCrossRef
Metadata
Title
MCF-7 breast carcinoma cells do not express caspase-3
Author
Reiner U. Jänicke
Publication date
01-09-2009
Publisher
Springer US
Published in
Breast Cancer Research and Treatment / Issue 1/2009
Print ISSN: 0167-6806
Electronic ISSN: 1573-7217
DOI
https://doi.org/10.1007/s10549-008-0217-9

Other articles of this Issue 1/2009

Breast Cancer Research and Treatment 1/2009 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine