1
|
Hou M, Yue M, Han X, Sun T, Zhu Y, Li Z, Han J, Zhao B, Tu M, An Y. Comparative analysis of BAG1 and BAG2: Insights into their structures, functions and implications in disease pathogenesis. Int Immunopharmacol 2024; 143:113369. [PMID: 39405938 DOI: 10.1016/j.intimp.2024.113369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/22/2024] [Accepted: 10/06/2024] [Indexed: 10/30/2024]
Abstract
As BAG family members, Bcl-2 associated athanogene family protein 1 (BAG1) and 2 (BAG2) are implicated in multiple cellular processes, including apoptosis, autophagy, protein folding and homeostasis. Although structurally similar, they considerably differ in many ways. Unlike BAG2, BAG1 has four isoforms (BAG1L, BAG1M, BAG1S and BAG1 p29) displaying different expression features and functional patterns. BAG1 and BAG2 play different cellular functions by interacting with different molecules to participate in the regulation of various diseases, including cancer/tumor and neurodegenerative diseases. Commonly, BAG1 acts as a protective factor to predict a good prognosis of patients with some types of cancer or a risk factor in some other cancers, while BAG2 is regarded as a risk factor to promote cancer/tumor progression. In neurodegenerative diseases, BAG2 commonly acts as a neuroprotective factor. In this review, we summarized the differences in molacular structure and biological function between BAG1 and BAG2, as well as the influences of them on pathogenesis of diseases, and explore the prospects for their clinical therapy application by specifying the activators and inhibitors of BAG1 and BAG2, which might provide a better understanding of the underlying pathogenesis and developing the targeted therapy strategies for diseases.
Collapse
Affiliation(s)
- Mengwen Hou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; School of Stomatology, Henan University, Kaifeng 475004, China
| | - Man Yue
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; School of Stomatology, Henan University, Kaifeng 475004, China
| | - Xu Han
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; School of Stomatology, Henan University, Kaifeng 475004, China
| | - Tiantian Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; School of Stomatology, Henan University, Kaifeng 475004, China
| | - Yonghao Zhu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; School of Stomatology, Henan University, Kaifeng 475004, China
| | - Zhihao Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng 475004, China
| | - Jiayang Han
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; School of Stomatology, Henan University, Kaifeng 475004, China
| | - Binbin Zhao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; School of Stomatology, Henan University, Kaifeng 475004, China
| | - Mengjie Tu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; School of Stomatology, Henan University, Kaifeng 475004, China
| | - Yang An
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng 475004, China.
| |
Collapse
|
2
|
Parsian M, Mutlu P, Taghavi Pourianazar N, Yalcin Azarkan S, Gunduz U. Investigation of the Therapeutic Effects of Palbociclib Conjugated Magnetic Nanoparticles on Different Types of Breast Cancer Cell Lines. Cell Mol Bioeng 2023; 16:143-157. [PMID: 37096074 PMCID: PMC10121994 DOI: 10.1007/s12195-022-00758-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 12/14/2022] [Indexed: 01/09/2023] Open
Abstract
Introduction Drug targeting and controlled drug release systems in cancer treatment have many advantages over conventional chemotherapy in terms of limiting systemic toxicity, side effects, and overcoming drug resistance. Methods and Results In this paper, fabricating nanoscale delivery system composed of magnetic nanoparticles (MNPs) covered with poly-amidoamine (PAMAM) dendrimers and using its advantages were fully used to help the chemotherapeutic drug, Palbociclib, effectively reach tumors, specifically and stay stable in the circulation longer. In order to determine whether conjugate selectivity can be increased for the specific drug type, we have reported different strategies for loading and conjugation of Palbociclib to different generations of magnetic PAMAM dendrimers. The best method leading to the highest amount of Palbociclib conjugation was chosen, and the characterization of the Palbociclib conjugated dendrimeric magnetic nanoparticles (PAL-DcMNPs) were performed. In vitro pharmacological activity of the conjugation was demonstrated by measuring the cell viability and lactate dehydrogenase (LHD) release. Obtained results indicated that PAL-DcMNPs treatment of the breast cancer cell lines, leads to an increase in cell toxicity compared to free Palbociclib. The observed effects were more evident for MCF-7 cells than for MDA-MB231 and SKBR3 cells, considering that viability decreased to 30% at 2.5 µM treatment of PAL-DcMNPs at MCF-7 cells. Finally, in Palbociclib and PAL-DcMNPs treated breast cancer cells, the expression levels of some pro-apoptotic and drug resistance related genes were performed by RT-PCR analysis. Conclusion Our knowledge indicates that the proposed approach is novel, and it can provide new insight into the development of Palbociclib targeting delivery system for cancer treatment.
Collapse
Affiliation(s)
- Maryam Parsian
- Department of Biotechnology, Middle East Technical University, Ankara, Turkey
| | - Pelin Mutlu
- Department of Biotechnology, Biotechnology Institute, Ankara University, Ankara, Turkey
| | | | - Serap Yalcin Azarkan
- Department of Molecular Biology and Genetics, Ahi Evran University, Kirsehir, Turkey
| | - Ufuk Gunduz
- Department of Biotechnology, Middle East Technical University, Ankara, Turkey
- Department of Biological Sciences, Middle East Technical University, Ankara, Turkey
| |
Collapse
|
3
|
Evidence for heterogeneity in response to treatment in mammary tumors of dogs as happens in humans. Vet Res Commun 2023; 47:111-120. [PMID: 35570257 DOI: 10.1007/s11259-022-09934-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 05/04/2022] [Indexed: 01/27/2023]
Abstract
Tumors are formed by various clones developed over a long time. This gives rise to a heterogeneous nature. This heterogeneity is the hardest challenge in the treatment of cancers because it is the main reason for drug resistance. This is a well-known fact in human cancer. Therefore, we have reasoned that if the tumor heterogeneity in canine mammary gland tumors (CMGTs) could be shown by an ex vivo assay, which will be used first time in veterinary oncology practice, this could be used further in clinics. To achieve this, twenty-six patients were included in the study. Tumor tissues were obtained from animals during routine surgery. Tumor cells were isolated and seeded ex vivo. The cells were exposed to anticancer drugs that are clinically used. Seven days after the treatment, chemosensitivity has luminometrically been assayed by ATP-tumor chemosensitivity assay (ATP-TCA). It has clearly been shown that all the tumor tissues have responded to treatment differently, implying that heterogeneity exists in mammary tumors. There has also been found that there was a weak to moderate statistically significant correlation between tumor size and drug index. However, there has been no correlation between drug index and metastasis to lymph nodes. Hyperplasic areas had relatively higher PCNA values. The results of our study demonstrate the heterogeneity in responses to in vitro drugs. Clinical trials based on test results and follow-up studies with large numbers of animals are needed to prove that such chemotherapeutic activity assessment tests can be clinically useful in predicting drug responses in CMGTs.
Collapse
|
4
|
Berger Fridman I, Kostas J, Gregus M, Ray S, Sullivan MR, Ivanov AR, Cohen S, Konry T. High-throughput microfluidic 3D biomimetic model enabling quantitative description of the human breast tumor microenvironment. Acta Biomater 2021; 132:473-488. [PMID: 34153511 PMCID: PMC8434998 DOI: 10.1016/j.actbio.2021.06.025] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/31/2021] [Accepted: 06/14/2021] [Indexed: 12/15/2022]
Abstract
Cancer is driven by both genetic aberrations in the tumor cells and fundamental changes in the tumor microenvironment (TME). These changes offer potential targets for novel therapeutics, yet lack of in vitro 3D models recapitulating this complex microenvironment impedes such progress. Here, we generated several tumor-stroma scaffolds reflecting the dynamic in vivo breast TME, using a high throughput microfluidic system. Alginate (Alg) or alginate-alginate sulfate (Alg/Alg-S) hydrogels were used as ECM-mimics, enabling the encapsulation and culture of tumor cells, fibroblasts and immune cells (macrophages and T cells, of the innate and adaptive immune systems, respectively). Specifically, Alg/Alg-S was shown capable of capturing and presenting growth factors and cytokines with binding affinity that is comparable to heparin. Viability and cytotoxicity were shown to strongly correlate with the dynamics of cellular milieu, as well as hydrogel type. Using on-chip immunofluorescence, production of reactive oxygen species and apoptosis were imaged and quantitatively analyzed. We then show how macrophages in our microfluidic system were shifted from a proinflammatory to an immunosuppressive phenotype when encapsulated in Alg/Alg-S, reflecting in vivo TME dynamics. LC-MS proteomic profiling of tumor cells sorted from the TME scaffolds revealed upregulation of proteins involved in cell-cell interactions and immunomodulation in Alg/Alg-S scaffolds, correlating with in vivo findings and demonstrating the appropriateness of Alg/Alg-S as an ECM biomimetic. Finally, we show the formation of large tumor-derived vesicles, formed exclusively in Alg/Alg-S scaffolds. Altogether, our system offers a robust platform for quantitative description of the breast TME that successfully recapitulates in vivo patterns. STATEMENT OF SIGNIFICANCE: Cancer progression is driven by profound changes in both tumor cells and surrounding stroma. Here, we present a high throughput microfluidic system for the generation and analysis of dynamic tumor-stroma scaffolds, that mimic the complex in vivo TME cell proportions and compositions, constructing robust in vitro models for the study of the TME. Utilizing Alg/Alg-S as a bioinspired ECM, mimicking heparin's in vivo capabilities of capturing and presenting signaling molecules, we show how Alg/Alg-S induces complex in vivo-like responses in our models. Alg/Alg-S is shown here to promote dynamic protein expression patterns, that can serve as potential therapeutic targets for breast cancer treatment. Formation of large tumor-derived vesicles, observed exclusively in the Alg/Alg-S scaffolds suggests a mechanism for tumor survival.
Collapse
Affiliation(s)
- Ilana Berger Fridman
- Department of Pharmaceutical Sciences, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA; Avram and Stella Goldstein-Goren Department of Biotechnology Engineering and Regenerative Medicine and Stem Cell Center, Ben-Gurion University of the Negev, POB 653, Beer-Sheva 84105, Israel.
| | - James Kostas
- Department of Chemistry and Chemical Biology, Barnett Institute of Chemical and Biological Analysis, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA.
| | - Michal Gregus
- Department of Chemistry and Chemical Biology, Barnett Institute of Chemical and Biological Analysis, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA.
| | - Somak Ray
- Department of Chemistry and Chemical Biology, Barnett Institute of Chemical and Biological Analysis, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA.
| | - Matthew R Sullivan
- Department of Pharmaceutical Sciences, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA.
| | - Alexander R Ivanov
- Department of Chemistry and Chemical Biology, Barnett Institute of Chemical and Biological Analysis, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA.
| | - Smadar Cohen
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering and Regenerative Medicine and Stem Cell Center, Ben-Gurion University of the Negev, POB 653, Beer-Sheva 84105, Israel.
| | - Tania Konry
- Department of Pharmaceutical Sciences, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA.
| |
Collapse
|
5
|
Petiti J, Lo Iacono M, Rosso V, Andreani G, Jovanovski A, Podestà M, Lame D, Gobbi MD, Fava C, Saglio G, Frassoni F, Cilloni D. Bcl-xL represents a therapeutic target in Philadelphia negative myeloproliferative neoplasms. J Cell Mol Med 2020; 24:10978-10986. [PMID: 32790151 PMCID: PMC7521327 DOI: 10.1111/jcmm.15730] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/14/2020] [Accepted: 07/16/2020] [Indexed: 12/19/2022] Open
Abstract
Myeloproliferative neoplasms are divided into essential thrombocythemia (ET), polycythemia vera (PV) and primary myelofibrosis (PMF). Although ruxolitinib was proven to be effective in reducing symptoms, patients rarely achieve complete molecular remission. Therefore, it is relevant to identify new therapeutic targets to improve the clinical outcome of patients. Bcl‐xL protein, the long isoform encoded by alternative splicing of the Bcl‐x gene, acts as an anti‐apoptotic regulator. Our study investigated the role of Bcl‐xL as a marker of severity of MPN and the possibility to target Bcl‐xL in patients. 129 MPN patients and 21 healthy patients were enrolled in the study. We analysed Bcl‐xL expression in leucocytes and in enriched CD34+ and CD235a+ cells. Furthermore, ABT‐737, a Bcl‐xL inhibitor, was tested in HEL cells and in leucocytes from MPN patients. Bcl‐xL was found progressively over‐expressed in cells from ET, PV and PMF patients, independently by JAK2 mutational status. Moreover, our data indicated that the combination of ABT‐737 and ruxolitinib resulted in a significantly higher apoptotic rate than the individual drug. Our study suggests that Bcl‐xL plays an important role in MPN independently from JAK2 V617F mutation. Furthermore, data demonstrate that targeting simultaneously JAK2 and Bcl‐xL might represent an interesting new approach.
Collapse
Affiliation(s)
- Jessica Petiti
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Marco Lo Iacono
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Valentina Rosso
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Giacomo Andreani
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | | | - Marina Podestà
- Department of Pediatric Hemato-Oncology and Stem Cell and Cellular Therapy Laboratory, Institute G. Gaslini, Genova, Italy
| | - Dorela Lame
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Marco De Gobbi
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Carmen Fava
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Giuseppe Saglio
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Francesco Frassoni
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Daniela Cilloni
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| |
Collapse
|
6
|
Aloe-Emodin Induces Breast Tumor Cell Apoptosis through Upregulation of miR-15a/miR-16-1 That Suppresses BCL2. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:5108298. [PMID: 32190086 PMCID: PMC7073502 DOI: 10.1155/2020/5108298] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 01/20/2020] [Accepted: 01/29/2020] [Indexed: 02/06/2023]
Abstract
PURPOSE Aloe-emodin (AE) is a natural compound derived from aloe vera and palmatum rhubarb and shows anticancer activities in various cancers. Bcl-2 family is the main regulator of cell death or cell survival. This study describes the effects of AE on proliferation of breast tumor (BT) cells. METHODS MCF-10A, MCF-10AT, MCF-7, and MDA-MB-231 cell lines were exposed to AE. Cell proliferation and apoptosis were assessed by CCK-8 and flow cytometry. Protein levels were measured by Western blotting. The levels of mRNA and miRNA were examined by RT-PCR. Bioinformatics was applied to screen miRNAs that bind to 3'-UTR of mRNA. RESULTS The results showed that AE selective activity inhibited the proliferation and induced apoptosis of MCF-10AT and MCF-7 cells but exhibited no significant inhibition in MCF10A and MDA-MB-231 cells. Mechanistically, AE dose-dependently decreased the protein expression of Bcl-2 and Bcl-xl, while it increased Bax protein expression in MCF-10AT and MCF-7 cells. The levels of Bcl-xl and Bax mRNA were altered by AE treatment, which was consistent with the protein expression results. However, Bcl-2 mRNA levels were not affected in either cell line, suggesting that AE may modulate the protein translation of Bcl-2 through miRNAs. In all candidate miRNAs that bind to 3'-UTR of Bcl-2, miR-15a and miR-16-1 were dose-dependently downregulated by AE. Moreover, inhibition of miR-15a/16-1 could eliminate the inhibition of MCF-10AT and MCF-7 cells growth by AE and could reverse the downregulation of AE-induced Bcl-2 protein level. CONCLUSION Our research provides an important basis that AE induces BT cell apoptosis through upregulation of miR-15a/miR-16-1 that suppresses BCL2.
Collapse
|
7
|
Boudreau MW, Peh J, Hergenrother PJ. Procaspase-3 Overexpression in Cancer: A Paradoxical Observation with Therapeutic Potential. ACS Chem Biol 2019; 14:2335-2348. [PMID: 31260254 PMCID: PMC6858495 DOI: 10.1021/acschembio.9b00338] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Many anticancer strategies rely on the promotion of apoptosis in cancer cells as a means to shrink tumors. Crucial for apoptotic function are executioner caspases, most notably caspase-3, that proteolyze a variety of proteins, inducing cell death. Paradoxically, overexpression of procaspase-3 (PC-3), the low-activity zymogen precursor to caspase-3, has been reported in a variety of cancer types. Until recently, this counterintuitive overexpression of a pro-apoptotic protein in cancer has been puzzling. Recent studies suggest subapoptotic caspase-3 activity may promote oncogenic transformation, a possible explanation for the enigmatic overexpression of PC-3. Herein, the overexpression of PC-3 in cancer and its mechanistic basis is reviewed; collectively, the data suggest the potential for exploitation of PC-3 overexpression with PC-3 activators as a targeted anticancer strategy.
Collapse
Affiliation(s)
- Matthew W. Boudreau
- Department of Chemistry and Institute for Genomic Biology, University of Illinois at Urbana–Champaign, Urbana, Illinois, United States
| | - Jessie Peh
- Department of Chemistry and Institute for Genomic Biology, University of Illinois at Urbana–Champaign, Urbana, Illinois, United States
| | - Paul J. Hergenrother
- Department of Chemistry and Institute for Genomic Biology, University of Illinois at Urbana–Champaign, Urbana, Illinois, United States
| |
Collapse
|
8
|
Bozdogan O, Atasoy P, Bozdogan N, Erekul S, Batislam E, Yilmaz E, Başar MM. Bag-1 Expression in Hyperplastic and Neoplastic Prostate Tissue: Is There Any Relationship with BCL-Related Proteins and Androgen Receptor Status? TUMORI JOURNAL 2019; 91:539-45. [PMID: 16457154 DOI: 10.1177/030089160509100615] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Aims and Background To evaluate the function and distribution of BAG-1 protein in hyperplastic and neoplastic prostate tissue and establish the relationship between this protein and BCL-related proteins (BCL-2 and BAX), androgen receptor (AR) expression and chromogranin A. Methods Twenty-eight prostatic adenocarcinomas and 16 prostate hyperplasias were included in this retrospective study. BAG-1, BCL-2, BAX, androgen receptor and chromogranin A immunostaining was performed by means of standard avidin-biotin peroxidase methods. The M30 antibody was used to identify preapoptotic and apoptotic cells. The immunohistochemical histological score (HSCORE) semi-quantative system was used to evaluate immunohistochemical staining. Results Statistical analysis showed a significant difference in HSCOREs of BAX, M30 and AR between the carcinoma and hyperplasia groups. Carcinomas expressed higher HSCOREs of these markers than hyperplasias. There were significant differences in nuclear and cytoplasmic BAG-1 positivity between high and low-grade carcinomas. BAG-1 expression was higher in low-grade carcinomas. In the carcinoma group there was a positive correlation (Pearson) between BCL-2 and cytoplasmic/nuclear BAG-1. In the hyperplasia group there was a negative correlation between BAX and BCL-2, and between AR and M30. We also detected a positive correlation between AR and nuclear/cytoplasmic BAG-1 and between nuclear and cytoplasmic BAG-1 in hyperplasias. BAG-1 showed the same specific basal cell localization as BCL-2 in hyperplastic and normal glands. Conclusions The BAG-1 protein showed a distinct distribution pattern in hyperplastic and neoplastic prostate. BAG-1 in association with BCL-2 inhibits apoptosis and may prolong the life of neoplastic cells and give them a chance to gain new oncogenic features in early carcinogenesis.
Collapse
Affiliation(s)
- Onder Bozdogan
- Department of Pathology, Kirikkale University Medical School, Turkey.
| | | | | | | | | | | | | |
Collapse
|
9
|
Than VT, Tran HTT, Ly DV, Dang HV, Nguyen MN, Truong AD. Bioinformatic identification and expression analysis of the chicken B cell lymphoma (BCL) gene. Genes Genomics 2019; 41:1195-1206. [PMID: 31313104 DOI: 10.1007/s13258-019-00849-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 07/03/2019] [Indexed: 01/02/2023]
Abstract
BACKGROUND B cell lymphoma (BCL) families play an important role in apoptosis as a growth factor, cell death programming, cytokine expression and immune-related genes expression. OBJECTIVES In this study, to investigate the roles of BCLs, we performed genome-wide identification, expression and functional analyses of the BCL family in chicken. METHODS Chicken BCLs genes were identified and analyzed by using bioinformatics approach. Expression profiles and Hierarchical cluster analysis of the BCLs genes in different chicken tissues were obtained from the genome-wide RNA-seq in the GEO, and Cluster and Java Treeview, respectively. RESULTS A total of 16 BCLs genes were identified from the chicken genome, which could be further classified into five distinct groups in the phylogenetic tree. On the other hand, the interaction among BCLs proteins and between BCLs proteins with NF-κB subunits are limited, indicating that the remaining the functions of BCLs protein could be investigated in chicken. Moreover, KEGG pathway analysis indicated that BCL gene family was involved in regulation of apoptotic and immune response. Finally, BCL gene family was differentially expressed in chicken tissues, pathogen infection and growth stages of early chicken early embryo. CONCLUSION This study provides significant insights into the potential functions of BCLs in chicken, including the regulation of apoptosis, cell death and expression of immune-related genes.
Collapse
Affiliation(s)
- Van Thai Than
- NTT Hi-Tech Institute, Nguyen Tat Thanh University, Ho Chi Minh City, 700000, Vietnam.
| | - Ha Thi Thanh Tran
- Department of Biochemistry and Immunology, National Institute of Veterinary Research, 86 Truong Chinh, Dong Da, Hanoi, 100000, Vietnam
| | - Duc Viet Ly
- Department of Biochemistry and Immunology, National Institute of Veterinary Research, 86 Truong Chinh, Dong Da, Hanoi, 100000, Vietnam
| | - Hoang Vu Dang
- Department of Biochemistry and Immunology, National Institute of Veterinary Research, 86 Truong Chinh, Dong Da, Hanoi, 100000, Vietnam
| | - Minh Nam Nguyen
- Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh City, Vietnam
| | - Anh Duc Truong
- Department of Biochemistry and Immunology, National Institute of Veterinary Research, 86 Truong Chinh, Dong Da, Hanoi, 100000, Vietnam.
| |
Collapse
|
10
|
Kilbas PO, Akcay IM, Doganay GD, Arisan ED. Bag-1 silencing enhanced chemotherapeutic drug-induced apoptosis in MCF-7 breast cancer cells affecting PI3K/Akt/mTOR and MAPK signaling pathways. Mol Biol Rep 2019; 46:847-860. [PMID: 30661182 DOI: 10.1007/s11033-018-4540-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 11/28/2018] [Indexed: 12/21/2022]
Abstract
The multifunctional anti-apoptotic Bag-1 protein has important roles in apoptosis, proteasome-mediated degradation, transcriptional regulation, and intracellular signaling. Bag-1 promotes cell survival and proliferation, and is overexpressed in breast cancer. Therefore, Bag-1-targeted therapy might be a promising strategy to treat breast cancer. However, the effects of Bag-1 silencing in combination with conventional chemotherapeutic drugs on cell viability and major signaling pathways have not yet been fully investigated in breast cancer cells. In this study, we investigated the cytotoxic effects of Bag-1 silencing, alone and in combination with cisplatin or paclitaxel treatment, in MCF-7 breast cancer cells. Bag-1 knockdown by shRNA or siRNA transfection sensitized MCF-7 cells to apoptosis induced by cisplatin or paclitaxel. Combination of Bag-1 silencing and drug treatment more potently downregulated the pro-survival PI3K/Akt/mTOR and p44/42 mitogen activated protein kinase (MAPK) pathways, and more potently upregulated the stress-activated p38 and SAPK/JNK MAPK pathways. Bag-1-silenced drug-treated cells had also highly reduced proliferative capacity, downregulated cyclin-cyclin dependent kinase complexes and upregulated tumor suppressors p21 and Rb. These results overall indicated that Bag-1 silencing enhanced cisplatin- or paclitaxel-induced cytotoxicity through multiple pathways. In conclusion, Bag-1 targeted therapy might enhance the therapeutic potential of conventional anti-cancer drugs in the treatment of breast cancer.
Collapse
Affiliation(s)
- Pelin Ozfiliz Kilbas
- Department of Molecular Biology-Genetics and Biotechnology, Istanbul Technical University, Istanbul, Turkey.,Department of Molecular Biology and Genetics, Istanbul Kultur University, Istanbul, Turkey
| | - Izzet Mehmet Akcay
- Department of Molecular Biology-Genetics and Biotechnology, Istanbul Technical University, Istanbul, Turkey
| | - Gizem Dinler Doganay
- Department of Molecular Biology-Genetics and Biotechnology, Istanbul Technical University, Istanbul, Turkey.
| | - Elif Damla Arisan
- Department of Molecular Biology and Genetics, Istanbul Kultur University, Istanbul, Turkey.
| |
Collapse
|
11
|
Wang W, Suga T, Hagimori M, Kuroda N, Fuchigami Y, Kawakami S. Investigation of Intracellular Delivery of NuBCP-9 by Conjugation with Oligoarginines Peptides in MDA-MB-231 Cells. Biol Pharm Bull 2018; 41:1448-1455. [PMID: 30175779 DOI: 10.1248/bpb.b18-00335] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Oligoarginines (Rn) are becoming promising tools for the intracellular delivery of biologically active molecules. NuBCP-9, a peptide that induces apoptosis in B-cell lymphoma 2 (Bcl-2)-expressing cancer cells, has been reported to promote the uptake and non-specific cytotoxicity of R8, also called octaarginine. However, it is unknown whether a similar synergistic effect can be seen with other Rn. In this study, we conjugated NuBCP-9 with various Rn (n=8, 10, 12, 14) to investigate and compare their cellular uptake characteristics. In addition, their non-specific cytotoxicity and apoptosis-inducing abilities were evaluated. We found that NuBCP-9 conjugated with Rn enhanced cellular uptake mainly through clathrin-mediated endocytosis and macropinocytosis, and that the uptake pathways were not different from those used by unconjugated Rn. However, the cytotoxicity study showed that NuBCP-9-R12 and NuBCP-9-R14 conjugates enhanced non-specific cytotoxicity. We found that NuBCP-9-R10 conjugate had the highest uptake efficiency and induced correspondingly high levels of apoptosis, while resulting in a tolerable degree of non-specific toxicity.
Collapse
Affiliation(s)
- Wei Wang
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University.,Department of Analytical Chemistry for Pharmaceutics, Graduate School of Biomedical Sciences, Nagasaki University
| | - Tadaharu Suga
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University.,Department of Analytical Chemistry for Pharmaceutics, Graduate School of Biomedical Sciences, Nagasaki University
| | - Masayori Hagimori
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University
| | - Naotaka Kuroda
- Department of Analytical Chemistry for Pharmaceutics, Graduate School of Biomedical Sciences, Nagasaki University
| | - Yuki Fuchigami
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University
| | - Shigeru Kawakami
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University
| |
Collapse
|
12
|
Aydogan A, Ozmen O, Haligur M, Sipahi C, Ileri D, Haligur A. Immunohistochemical evaluation of bcl-2, ER-alpha, caspase -3, -8, -9, PCNA and Ki-67 expressions in canine mammary carcinomas. Biotech Histochem 2018; 93:286-292. [PMID: 29565181 DOI: 10.1080/10520295.2018.1434680] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
Abstract
We investigated the expression of bcl-2, estrogen receptor alpha (ER-alpha), caspase-3, -8, -9, proliferating cell nuclear antigen (PCNA) and Ki-67 in canine mammary carcinomas. We used 65 paraffin embedded and re-diagnosed archival canine mammary tumor samples to which we applied the routine streptavidin-biotin-peroxidase technique. Seventeen cases were re-diagnosed as tubulopapillary carcinoma, 31 were re-diagnosed as complex carcinoma and 17 were re-diagnosed as carcinosarcoma. Differences of expression of bcl-2 and PCNA were statistically significant according to tumor type. Differences in expression of ER-alpha, caspase-3, -8, -9 and Ki-67 were not statistically significant. Differences of expression of bcl-2 and PCNA were statistically significant compared to ER-alpha, caspase-3, -8, -9 and Ki-67 in carcinosarcomas. We report the prognostic significance of bcl-2 and PCNA expression in canine mammary carcinosarcomas.
Collapse
Affiliation(s)
- A Aydogan
- a University of Cukurova , Faculty of Ceyhan Veterinary Medicine, Department of Pathology , Adana
| | - O Ozmen
- b University of Mehmet Akif Ersoy , Faculty of Veterinary Medicine, Department of Pathology , Burdur
| | - M Haligur
- a University of Cukurova , Faculty of Ceyhan Veterinary Medicine, Department of Pathology , Adana
| | - C Sipahi
- c University of Mehmet Akif Ersoy , Faculty of Veterinary Medicine, Department of Animal Health Economics and Management , Burdur
| | - D Ileri
- b University of Mehmet Akif Ersoy , Faculty of Veterinary Medicine, Department of Pathology , Burdur
| | - A Haligur
- d University of Cukurova , Faculty of Ceyhan Veterinary Medicine, Department of Anatomy , Adana , Turkey
| |
Collapse
|
13
|
Park SS, Lee DM, Lim JH, Lee D, Park SJ, Kim HM, Sohn S, Yoon G, Eom YW, Jeong SY, Choi EK, Choi KS. Pyrrolidine dithiocarbamate reverses Bcl-xL-mediated apoptotic resistance to doxorubicin by inducing paraptosis. Carcinogenesis 2018; 39:458-470. [DOI: 10.1093/carcin/bgy003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 01/08/2018] [Indexed: 02/07/2023] Open
Affiliation(s)
- Seok Soon Park
- Department of Biochemistry, Ajou University School of Medicine, Suwon, Korea
- Department of Biomedical Sciences, Ajou Graduate School, Suwon, Korea
- Asan Institute for Life Sciences, Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Dong Min Lee
- Department of Biochemistry, Ajou University School of Medicine, Suwon, Korea
- Department of Biomedical Sciences, Ajou Graduate School, Suwon, Korea
- Genomic Instability Center, Ajou University School of Medicine, Suwon, Korea
| | - Jun Hee Lim
- Genomic Instability Center, Ajou University School of Medicine, Suwon, Korea
| | - Dongjoo Lee
- Department of Pharmacy, Ajou University, Suwon, Korea
| | - Sang Jun Park
- Department of Energy Systems Research, Ajou University, Suwon, Korea
| | - Hwan Myung Kim
- Department of Energy Systems Research, Ajou University, Suwon, Korea
| | | | - Gyesoon Yoon
- Department of Biochemistry, Ajou University School of Medicine, Suwon, Korea
- Department of Biomedical Sciences, Ajou Graduate School, Suwon, Korea
| | - Young Woo Eom
- Cell therapy and Tissue Engineering Center, Wonju College of Medicine, Yonsei University, Wonju, Korea
| | - Seong-Yun Jeong
- Asan Institute for Life Sciences, Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Eun Kyung Choi
- Center for Advancing Cancer Therapeutics, Department of Radiation Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Kyeong Sook Choi
- Department of Biochemistry, Ajou University School of Medicine, Suwon, Korea
- Department of Biomedical Sciences, Ajou Graduate School, Suwon, Korea
- Genomic Instability Center, Ajou University School of Medicine, Suwon, Korea
| |
Collapse
|
14
|
Cato L, Neeb A, Sharp A, Buzón V, Ficarro SB, Yang L, Muhle-Goll C, Kuznik NC, Riisnaes R, Nava Rodrigues D, Armant O, Gourain V, Adelmant G, Ntim EA, Westerling T, Dolling D, Rescigno P, Figueiredo I, Fauser F, Wu J, Rottenberg JT, Shatkina L, Ester C, Luy B, Puchta H, Troppmair J, Jung N, Bräse S, Strähle U, Marto JA, Nienhaus GU, Al-Lazikani B, Salvatella X, de Bono JS, Cato ACB, Brown M. Development of Bag-1L as a therapeutic target in androgen receptor-dependent prostate cancer. eLife 2017; 6:e27159. [PMID: 28826504 PMCID: PMC5629025 DOI: 10.7554/elife.27159] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 08/07/2017] [Indexed: 12/12/2022] Open
Abstract
Targeting the activation function-1 (AF-1) domain located in the N-terminus of the androgen receptor (AR) is an attractive therapeutic alternative to the current approaches to inhibit AR action in prostate cancer (PCa). Here we show that the AR AF-1 is bound by the cochaperone Bag-1L. Mutations in the AR interaction domain or loss of Bag-1L abrogate AR signaling and reduce PCa growth. Clinically, Bag-1L protein levels increase with progression to castration-resistant PCa (CRPC) and high levels of Bag-1L in primary PCa associate with a reduced clinical benefit from abiraterone when these tumors progress. Intriguingly, residues in Bag-1L important for its interaction with the AR AF-1 are within a potentially druggable pocket, implicating Bag-1L as a potential therapeutic target in PCa.
Collapse
|
15
|
Silva MN, Leite JS, Mello MFV, Silva KVGC, Corgozinho KB, de Souza HJM, Cunha SCS, Ferreira AMR. Histologic evaluation of Ki-67 and cleaved caspase-3 expression in feline mammary carcinoma. J Feline Med Surg 2017; 19:440-445. [PMID: 26917536 PMCID: PMC11119657 DOI: 10.1177/1098612x16634150] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Objectives The purpose of this study was to evaluate histopathologic aspects of, and the expression of Ki-67 and cleaved caspase-3 in, feline mammary carcinoma (FMC). Methods Feline mammary tumors were surgically obtained by mastectomy from 30 female cats and were fixed with formalin and embedded in paraffin wax. Four-micron sections were stained with hematoxylin and eosin for histopathologic diagnosis. Ki-67 and cleaved caspase-3 were analyzed by immunohistochemistry. Results Samples were histologically confirmed as FMC. Positive immunostaining was observed in all cancer samples for both nuclear Ki-67 and cleaved caspase-3, with a mean positive staining percentage of 27.5% and 21.2%, respectively. No statistically significant correlations between Ki-67 and cleaved caspase-3 were observed within FMC. Conclusions and relevance A high proliferation index was found in feline mammary tumors. This is the first study evaluating cleaved caspase-3 expression in FMC.
Collapse
Affiliation(s)
- Millena N Silva
- Postgraduate Program in Pathology, Fluminense Federal University, Niterói, RJ, Brazil
| | - Juliana S Leite
- Department of Pathology and Veterinary Clinics, Faculty of Veterinary Medicine, Fluminense Federal University Niteroi, RJ, Brazil
| | - Marcela FV Mello
- Department of Pathology and Veterinary Clinics, Faculty of Veterinary Medicine, Fluminense Federal University Niteroi, RJ, Brazil
| | - Kassia VGC Silva
- Postgraduate Program in Clinical and Animal Reproduction, Faculty of Veterinary Medicine, Fluminense Federal University, Niteroi, RJ, Brazil
| | - Katia B Corgozinho
- Postgraduate Program in Clinical and Animal Reproduction, Faculty of Veterinary Medicine, Fluminense Federal University, Niteroi, RJ, Brazil
| | - Heloisa JM de Souza
- Department of Clinical and Surgery, Veterinary Institute, Federal Rural University of Rio de Janeiro, RJ, Brazil
| | - Simone CS Cunha
- Postgraduate Program in Clinical and Animal Reproduction, Faculty of Veterinary Medicine, Fluminense Federal University, Niteroi, RJ, Brazil
| | - Ana MR Ferreira
- Department of Pathology and Veterinary Clinics, Faculty of Veterinary Medicine, Fluminense Federal University Niteroi, RJ, Brazil
- Postgraduate Program in Clinical and Animal Reproduction, Faculty of Veterinary Medicine, Fluminense Federal University, Niteroi, RJ, Brazil
| |
Collapse
|
16
|
Taghavi Pourianazar N, Gunduz U. Changes in apoptosis-related gene expression and cytokine release in breast cancer cells treated with CpG-loaded magnetic PAMAM nanoparticles. Int J Pharm 2016; 515:11-19. [PMID: 27717915 DOI: 10.1016/j.ijpharm.2016.10.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 09/28/2016] [Accepted: 10/01/2016] [Indexed: 02/07/2023]
Abstract
CpG-oligodeoxynucleotide (CpG-ODN) can function as an immune adjuvant. Previously, we showed that stimulation of breast cancer cells with CpG-ODN conjugated with PAMAM dendrimer-coated magnetic nanoparticles (DcMNPs) has induced apoptosis. The aim of the current study was to evaluate the expression levels of some apoptosis-regulating genes in several human breast cancer cells treated with CpG/DcMNPs. Treated MDA-MB231 cells showed an increase in Noxa and Bax gene expression levels, whereas the expression level of Survivin decreased. Similarly, Noxa gene was overexpressed in treated MCF7 cells. In treated SKBR3 cells, a decline in the c-Flip mRNA level was determined. Furthermore, release of cytokines, IL-6, IL-10, and TNF-α, was determined in cell culture supernatants. CpG/DcMNP treatment leads to an increase in the release of IL-6 in MDA-MB231 and SKBR3 cells, whereas release of IL-10 and TNF-α did not change significantly. It is indicated that CpG-ODN may show its cytotoxic effect by regulating the expression of apoptosis-related genes and the release of cytokine in breast cancer cells.
Collapse
Affiliation(s)
| | - Ufuk Gunduz
- Middle East Technical University, Department of Biotechnology, 06800, Ankara, Turkey; Middle East Technical University, Department of Biological Sciences, 06800, Ankara, Turkey
| |
Collapse
|
17
|
Castro L, Gao X, Moore AB, Yu L, Di X, Kissling GE, Dixon D. A High Concentration of Genistein Induces Cell Death in Human Uterine Leiomyoma Cells by Autophagy. ACTA ACUST UNITED AC 2016; 5. [PMID: 27512718 DOI: 10.4172/2325-9655.s1-003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Genistein, an estrogenic, soy-derived isoflavone, may play a protective role against hormone-related cancers. We have reported that a high concentration of genistein inhibits cell proliferation and induces apoptosis in human uterine smooth muscle cells, but not in leiomyoma (fibroid) cells. To better understand the differential cell death responses of normal and tumor cells to a high concentration of genistein, we treated uterine smooth muscle cells and uterine leiomyoma cells with 50 μg/ml of genistein for 72 h and 168 h, and assessed for mediators of apoptosis, cytotoxicity and autophagy. We found that leiomyoma cells had increased protection from apoptosis by expressing an increased ratio of Bcl-2: bak at 72 h and 168 h; however, in smooth muscle cells, the Bcl-2: bak ratio was decreased at 72 h, but significantly rebounded by 168 h. The apoptosis extrinsic factors, Fas ligand and Fas receptor, were highly expressed in uterine smooth muscle cells following genistein treatment at both time points as evidenced by confocal microscopy. This was not seen in the uterine leiomyoma cells; however, cytotoxicity as indicated by elevated lactate dehydrogenase levels was significantly enhanced at 168 h. Increased immunoexpression of an autophagy/autophagosome marker was also observed in the leiomyoma cells, although minimally present in smooth muscle cells at 72 h. Ultrastructurally, there was evidence of autophagic vacuoles in the leiomyoma cells; whereas, the normal smooth muscle cells showed nuclear fragmentation indicative of apoptosis. In summary, our data show differential cell death pathways induced by genistein in tumor and normal uterine smooth muscle cells, and suggest novel cell death pathways that can be targeted for preventive and intervention strategies for inhibiting fibroid tumor cell growth in vivo.
Collapse
Affiliation(s)
- Lysandra Castro
- Molecular Pathogenesis Group, National Toxicology Program (NTP) Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Research Triangle Park, North Carolina 27709, USA
| | - Xioahua Gao
- Molecular Pathogenesis Group, National Toxicology Program (NTP) Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Research Triangle Park, North Carolina 27709, USA
| | - Alicia B Moore
- Molecular Pathogenesis Group, National Toxicology Program (NTP) Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Research Triangle Park, North Carolina 27709, USA
| | - Linda Yu
- Molecular Pathogenesis Group, National Toxicology Program (NTP) Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Research Triangle Park, North Carolina 27709, USA
| | - Xudong Di
- School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Grace E Kissling
- Biostatistics and Computational Biology Branch, Division of the Intramural Program (DIR) and NTP, National Institute of Environmental Health Sciences, National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Research Triangle Park, North Carolina 27709, USA
| | - Darlene Dixon
- Molecular Pathogenesis Group, National Toxicology Program (NTP) Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health (NIH), Department of Health and Human Services (DHHS), Research Triangle Park, North Carolina 27709, USA
| |
Collapse
|
18
|
Tiwari P, Khan MJ. Molecular and Computational Studies on Apoptotic Pathway Regulator, Bcl-2 Gene from Breast Cancer Cell Line MCF-7. Indian J Pharm Sci 2016; 78:87-93. [PMID: 27168686 PMCID: PMC4852581 DOI: 10.4103/0250-474x.180254] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cancer is a dreadful disease constituting abnormal growth and proliferation of malignant cells in the body. Next to lung cancer, breast cancer is the most common form of cancer affecting women. The apoptotic pathway regulators, B cell lymphoma family of protein, play a key role in various malignancies defining cancer and their constitutive expression plays an integral role in breast cancer chemotherapy. The research work discusses the identification and molecular cloning of a B cell lymphoma like gene from human breast cancer cell line. The open reading frame of the gene consisted of 965 nucleotides, encoding a protein of 380 amino acids with a predicted molecular weight of 42.5 kilodalton. The predicted physiochemical properties of the gene were as follows: Isoelectric point - 9.49, molecular formula - C1893H3004N534O548S16, total number of negatively charged residues, (Aspartate+Glutamate) - 26, total number of positively charged residues, (Arginine+Lysine)-39, instability index-42.08 (unstable protein) and grand average of hydropathicity is -0.202. Additionally, phobius prediction suggested non-cytoplasmic localization of the putative protein. The presence of secondary structure in the protein was determined by Memsat program. A 3 dimensional protein homology model was generated using threading based method of protein modeling for structural and functional annotation of the putative protein. Future prospects accounts for the biochemical characterization of the enzyme including in vitro assays on breast cancer cell line would establish the functional characteristics of the protein and its physiological mechanisms in breast cancer development and its therapeutic-target role in future.
Collapse
Affiliation(s)
- Pragya Tiwari
- Department of Metabolic and Structural Biology, Central Institute of Medicinal and Aromatic Plants, (CSIR-CIMAP), Lucknow-226 015, India
| | - M J Khan
- Department of Biochemistry, Aligarh Muslim University, Aligarh-202 002, India
| |
Collapse
|
19
|
Roth HS, Hergenrother PJ. Derivatives of Procaspase-Activating Compound 1 (PAC-1) and their Anticancer Activities. Curr Med Chem 2016; 23:201-41. [PMID: 26630918 PMCID: PMC4968085 DOI: 10.2174/0929867323666151127201829] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 11/04/2015] [Accepted: 11/27/2015] [Indexed: 01/26/2023]
Abstract
PAC-1 induces the activation of procaspase-3 in vitro and in cell culture by chelation of inhibitory labile zinc ions via its ortho-hydroxy-N-acylhydrazone moiety. First reported in 2006, PAC-1 has shown promise in cell culture and animal models of cancer, and a Phase I clinical trial in cancer patients began in March 2015 (NCT02355535). Because of the considerable interest in this compound and a well-defined structure-activity relationship, over 1000 PAC-1 derivatives have been synthesized in an effort to vary pharmacological properties such as potency and pharmacokinetics. This article provides a comprehensive examination of all PAC-1 derivatives reported to date. A survey of PAC-1 derivative libraries is provided, with an indepth discussion of four derivatives on which extensive studies have been performed.
Collapse
Affiliation(s)
| | - Paul J Hergenrother
- Department of Chemistry, University of Illinois, 261 Roger Adams Laboratory, Box 36-5, 600 S. Mathews Ave., Urbana, IL, 61801, USA.
| |
Collapse
|
20
|
Sharifi S, Barar J, Hejazi MS, Samadi N. Doxorubicin Changes Bax /Bcl-xL Ratio, Caspase-8 and 9 in Breast Cancer Cells. Adv Pharm Bull 2015; 5:351-9. [PMID: 26504757 DOI: 10.15171/apb.2015.049] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 11/20/2014] [Accepted: 03/01/2015] [Indexed: 12/16/2022] Open
Abstract
PURPOSE Doxorubicin is administrated as a single agent in first-line therapy of breast cancer to induce apoptosis in tumor cells. Bax, Bcl-xL, Caspase-8 and 9 proteins are involved in induction of apoptosis. The present study describes Bax, Bcl-xL gene expression and Caspase-8 and 9 protein levels in MCF-7 cells incubated with doxorubicin at different doses an incubation times. METHODS The cytotoxic effects of doxorubicin were studied using MTT assay. MCF-7 cells were treated with three concentrations of doxorubicin (0.1, 0.5, 1 μM) and incubated for 24, 48 and 72 hours then expression levels of Bax and Bcl-xL genes were elucidated by Real-time RT-PCR technique and protein levels of caspase-8 and caspase-9 proteins were measured using ELISA method. Morphological modifications of the cells were also monitored via light microscopic images. RESULTS Doxorubicin decreased the anti-apoptotic Bcl-xL and increased pro-apoptotic Bax mRNA levels. Doxorubicin induced a significant increase in Bax /Bcl-xL ratio in all doses and incubation times (p<0.05). Highest (more than 10 fold) increase in Bax /Bcl-xL ratio was revealed after 48 h incubation of the cells with in all doses of doxorubicin. Doxorubicin also increased caspase-9 level in a time and dose-dependent manner, while caspase-8 level didn't follow time and dose dependency pattern. CONCLUSION Our results confirm that doxorubicin induces mitochondrial-dependent apoptosis by down-regulation of Bcl-xL and up- regulation of Bax and caspase-9 expressions.
Collapse
Affiliation(s)
- Simin Sharifi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran. ; Students' Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jaleh Barar
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran. ; Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Saeid Hejazi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran. ; Faculty of Advanced Biomedical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nasser Samadi
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran. ; Faculty of Advanced Biomedical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran. ; Department of Biochemistry and Medical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
21
|
Roth HS, Botham RC, Schmid SC, Fan TM, Dirikolu L, Hergenrother PJ. Removal of Metabolic Liabilities Enables Development of Derivatives of Procaspase-Activating Compound 1 (PAC-1) with Improved Pharmacokinetics. J Med Chem 2015; 58:4046-65. [PMID: 25856364 DOI: 10.1021/acs.jmedchem.5b00413] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Procaspase-activating compound 1 (PAC-1) is an o-hydroxy-N-acylhydrazone that induces apoptosis in cancer cells by chelation of labile inhibitory zinc from procaspase-3. PAC-1 has been assessed in a wide variety of cell culture experiments and in vivo models of cancer, with promising results, and a phase 1 clinical trial in cancer patients has been initiated (NCT02355535). For certain applications, however, the in vivo half-life of PAC-1 could be limiting. Thus, with the goal of developing a compound with enhanced metabolic stability, a series of PAC-1 analogues were designed containing modifications that systematically block sites of metabolic vulnerability. Evaluation of the library of compounds identified four potentially superior candidates with comparable anticancer activity in cell culture, enhanced metabolic stability in liver microsomes, and improved tolerability in mice. In head-to-head experiments with PAC-1, pharmacokinetic evaluation in mice demonstrated extended elimination half-lives and greater area under the curve values for each of the four compounds, suggesting them as promising candidates for further development.
Collapse
Affiliation(s)
- Howard S Roth
- †Department of Chemistry, ‡Department of Veterinary Clinical Medicine, and §Department of Veterinary Biosciences, University of Illinois, Urbana, Illinois 61801, United States
| | - Rachel C Botham
- †Department of Chemistry, ‡Department of Veterinary Clinical Medicine, and §Department of Veterinary Biosciences, University of Illinois, Urbana, Illinois 61801, United States
| | - Steven C Schmid
- †Department of Chemistry, ‡Department of Veterinary Clinical Medicine, and §Department of Veterinary Biosciences, University of Illinois, Urbana, Illinois 61801, United States
| | - Timothy M Fan
- †Department of Chemistry, ‡Department of Veterinary Clinical Medicine, and §Department of Veterinary Biosciences, University of Illinois, Urbana, Illinois 61801, United States
| | - Levent Dirikolu
- †Department of Chemistry, ‡Department of Veterinary Clinical Medicine, and §Department of Veterinary Biosciences, University of Illinois, Urbana, Illinois 61801, United States
| | - Paul J Hergenrother
- †Department of Chemistry, ‡Department of Veterinary Clinical Medicine, and §Department of Veterinary Biosciences, University of Illinois, Urbana, Illinois 61801, United States
| |
Collapse
|
22
|
Duran GE, Wang YC, Francisco EB, Rose JC, Martinez FJ, Coller J, Brassard D, Vrignaud P, Sikic BI. Mechanisms of resistance to cabazitaxel. Mol Cancer Ther 2014; 14:193-201. [PMID: 25416788 DOI: 10.1158/1535-7163.mct-14-0155] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We studied mechanisms of resistance to the novel taxane cabazitaxel in established cellular models of taxane resistance. We also developed cabazitaxel-resistant variants from MCF-7 breast cancer cells by stepwise selection in drug alone (MCF-7/CTAX) or drug plus the transport inhibitor PSC-833 (MCF-7/CTAX-P). Among multidrug-resistant (MDR) variants, cabazitaxel was relatively less cross-resistant than paclitaxel and docetaxel (15- vs. 200-fold in MES-SA/Dx5 and 9- vs. 60-fold in MCF-7/TxT50, respectively). MCF-7/TxTP50 cells that were negative for MDR but had 9-fold resistance to paclitaxel were also 9-fold resistant to cabazitaxel. Selection with cabazitaxel alone (MCF-7/CTAX) yielded 33-fold resistance to cabazitaxel, 52-fold resistance to paclitaxel, activation of ABCB1, and 3-fold residual resistance to cabazitaxel with MDR inhibition. The MCF-7/CTAX-P variant did not express ABCB1, nor did it efflux rhodamine-123, BODIPY-labeled paclitaxel, and [(3)H]-docetaxel. These cells are hypersensitive to depolymerizing agents (vinca alkaloids and colchicine), have reduced baseline levels of stabilized microtubules, and impaired tubulin polymerization in response to taxanes (cabazitaxel or docetaxel) relative to MCF-7 parental cells. Class III β-tubulin (TUBB3) RNA and protein were elevated in both MCF-7/CTAX and MCF-7/CTAX-P. Decreased BRCA1 and altered epithelial-mesenchymal transition (EMT) markers are also associated with cabazitaxel resistance in these MCF-7 variants, and may serve as predictive biomarkers for its activity in the clinical setting. In summary, cabazitaxel resistance mechanisms include MDR (although at a lower level than paclitaxel and docetaxel), and alterations in microtubule dynamicity, as manifested by higher expression of TUBB3, decreased BRCA1, and by the induction of EMT.
Collapse
Affiliation(s)
- George E Duran
- Oncology Division, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Yan C Wang
- Oncology Division, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - E Brian Francisco
- Oncology Division, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - John C Rose
- Oncology Division, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Francisco J Martinez
- Oncology Division, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - John Coller
- Stanford Functional Genomics Facility, Stanford University School of Medicine, Stanford, California
| | | | | | - Branimir I Sikic
- Oncology Division, Department of Medicine, Stanford University School of Medicine, Stanford, California.
| |
Collapse
|
23
|
Botham RC, Fan TM, Im I, Borst LB, Dirikolu L, Hergenrother PJ. Dual small-molecule targeting of procaspase-3 dramatically enhances zymogen activation and anticancer activity. J Am Chem Soc 2014; 136:1312-9. [PMID: 24383395 PMCID: PMC3954530 DOI: 10.1021/ja4124303] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Combination anticancer therapy typically consists of drugs that target different biochemical pathways or those that act on different targets in the same pathway. Here we demonstrate a new concept in combination therapy, that of enzyme activation with two compounds that hit the same biological target, but through different mechanisms. Combinations of procaspase-3 activators PAC-1 and 1541B show considerable synergy in activating procaspase-3 in vitro, stimulate rapid and dramatic maturation of procaspase-3 in multiple cancer cell lines, and powerfully induce caspase-dependent apoptotic death to a degree well exceeding the additive effect. In addition, the combination of PAC-1 and 1541B effectively reduces tumor burden in a murine lymphoma model at dosages for which the compounds alone have minimal or no effect. These data suggest the potential of PAC-1/1541B combinations for the treatment of cancer and, more broadly, demonstrate that differentially acting enzyme activators can potently synergize to give a significantly heightened biological effect.
Collapse
Affiliation(s)
- Rachel C Botham
- Department of Chemistry, University of Illinois at Urbana-Champaign , Urbana, Illinois 61801, United States
| | | | | | | | | | | |
Collapse
|
24
|
Ni W, Chen B, Zhou G, Lu C, Xiao M, Guan C, Zhang Y, He S, Shen A, Ni R. Overexpressed nuclear BAG-1 in human hepatocellular carcinoma is associated with poor prognosis and resistance to doxorubicin. J Cell Biochem 2013; 114:2120-30. [PMID: 23553841 DOI: 10.1002/jcb.24560] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2013] [Accepted: 03/20/2013] [Indexed: 12/17/2022]
Abstract
Bcl-2-associated athanogene-1 (BAG-1) is a multifunctional anti-apoptotic protein which regulates an array of cellular processes, including apoptosis, signaling, proliferation, transcription, and cell motility and has been reported to be over-expressed in a number of human malignancies. To investigate the possible involvement of BAG-1 in tumorigenesis of hepatocellular carcinoma (HCC), we performed Western blot analysis in eight paired samples of HCC and adjacent peritumoral tissues and immunohistochemistry in 65 paraffin sections of HCC, which both showed an enhanced expression of nuclear BAG-1 isoform in HCC tissues. Statistical analysis confirmed that overexpression of nuclear BAG-1 in HCC tissues was significantly associated with histological grading (P < 0.001), poor prognosis (P = 0.004), and was found to be an independent prognostic indicator for HCC (P = 0.023). We also noted that BAG-1 was overexpressed in four HCC cell lines compared with a normal hepatocyte cell line, and BAG-1 overexpression increased resistance of HCC cells to doxorubicin, a common chemotherapeutic agent for HCC. Furthermore, we observed that knock down of BAG-1 with siRNA in HepG2 cells increased the chemosensitivity of cells, a process mediated through inhibition of doxorubicin-triggered NF-κB activation; and knock down of BAG-1 suppressed proliferation and cell cycle transition of HepG2 cells. In consequence, our results for the first time indicated that BAG-1 was dysregulated in HCC and suppression of BAG-1 expression which resulted in inhibiting of NF-κB signaling might be developed into a new strategy in HCC therapy.
Collapse
Affiliation(s)
- Wenkai Ni
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, 226001, P.R., China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Srivastava M, Nambiar M, Sharma S, Karki SS, Goldsmith G, Hegde M, Kumar S, Pandey M, Singh RK, Ray P, Natarajan R, Kelkar M, De A, Choudhary B, Raghavan SC. An inhibitor of nonhomologous end-joining abrogates double-strand break repair and impedes cancer progression. Cell 2013; 151:1474-87. [PMID: 23260137 DOI: 10.1016/j.cell.2012.11.054] [Citation(s) in RCA: 280] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Revised: 03/07/2012] [Accepted: 11/30/2012] [Indexed: 01/03/2023]
Abstract
DNA Ligase IV is responsible for sealing of double-strand breaks (DSBs) during nonhomologous end-joining (NHEJ). Inhibiting Ligase IV could result in amassing of DSBs, thereby serving as a strategy toward treatment of cancer. Here, we identify a molecule, SCR7 that inhibits joining of DSBs in cell-free repair system. SCR7 blocks Ligase IV-mediated joining by interfering with its DNA binding but not that of T4 DNA Ligase or Ligase I. SCR7 inhibits NHEJ in a Ligase IV-dependent manner within cells, and activates the intrinsic apoptotic pathway. More importantly, SCR7 impedes tumor progression in mouse models and when coadministered with DSB-inducing therapeutic modalities enhances their sensitivity significantly. This inhibitor to target NHEJ offers a strategy toward the treatment of cancer and improvement of existing regimens.
Collapse
Affiliation(s)
- Mrinal Srivastava
- Department of Biochemistry, Indian Institute of Science, Bangalore 560 012, India
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Delort L, Perrier S, Dubois V, Billard H, Mracek T, Bing C, Vasson MP, Caldefie-Chézet F. Zinc-α2-glycoprotein: a proliferative factor for breast cancer? In vitro study and molecular mechanisms. Oncol Rep 2013; 29:2025-9. [PMID: 23446778 DOI: 10.3892/or.2013.2311] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Accepted: 01/04/2013] [Indexed: 11/06/2022] Open
Abstract
Zinc-α2-glycoprotein (ZAG) is a new adipokine whose gene expression is downregulated in obese patients. We recently reported ZAG expression in breast tumor or healthy breast tissue and detected this expression at high levels in ductal carcinoma and in normal epithelial adjacent tissue but not in normal tissue of healthy women. In the present study, we used two human breast tumor cell lines (MCF-7 and MDA-MB‑231) and one fibrocystic breast cell line (MCF‑10a) to examine whether recombinant ZAG has an effect on proliferative/apoptotic response in breast cancer cell lines. ZAG seemed to exert a proliferative effect on breast cancer cell proliferation [+11 to 27% in MCF-7 with (ZAG) = 5-20 µg/ml; +13% in MDA-MB-231 with (ZAG) = 5 µg/ml] and, on the contrary, an anti-proliferative effect in the fibrocystic breast cell line [-5 to -8% in MCF-10a with (ZAG) = 5-10 µg/ml]. ZAG was able to modulate gene and protein expression involved in the apoptotic response. However, further studies are required to fully elucidate the effects of ZAG on the proliferation of mammary cells.
Collapse
Affiliation(s)
- Laetitia Delort
- Clermont University, University of Auvergne, UFR Pharmacy, Laboratory SVFp, F-63000 Clermont-Ferrand, France.
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Hsu DC, Roth HS, West DC, Botham RC, Novotny CJ, Schmid SC, Hergenrother PJ. Parallel synthesis and biological evaluation of 837 analogues of procaspase-activating compound 1 (PAC-1). ACS COMBINATORIAL SCIENCE 2012; 14:44-50. [PMID: 22007686 DOI: 10.1021/co2001372] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Procaspase-Activating Compound 1 (PAC-1) is an ortho-hydroxy N-acyl hydrazone that enhances the enzymatic activity of procaspase-3 in vitro and induces apoptosis in cancer cells. An analogue of PAC-1, called S-PAC-1, was evaluated in a veterinary clinical trial in pet dogs with lymphoma and found to have considerable potential as an anticancer agent. With the goal of identifying more potent compounds in this promising class of experimental therapeutics, a combinatorial library based on PAC-1 was created, and the compounds were evaluated for their ability to induce death of cancer cells in culture. For library construction, 31 hydrazides were condensed in parallel with 27 aldehydes to create 837 PAC-1 analogues, with an average purity of 91%. The compounds were evaluated for their ability to induce apoptosis in cancer cells, and through this work, six compounds were discovered to be substantially more potent than PAC-1 and S-PAC-1. These six hits were further evaluated for their ability to relieve zinc-mediated inhibition of procaspase-3 in vitro. In general, the newly identified hit compounds are two- to four-fold more potent than PAC-1 and S-PAC-1 in cell culture, and thus have promise as experimental therapeutics for treatment of the many cancers that have elevated expression levels of procaspase-3.
Collapse
Affiliation(s)
- Danny C. Hsu
- Department
of Chemistry and ‡Department of Biochemistry, Roger Adams Laboratory, University of Illinois, Urbana, Illinois 61801, United
States
| | - Howard S. Roth
- Department
of Chemistry and ‡Department of Biochemistry, Roger Adams Laboratory, University of Illinois, Urbana, Illinois 61801, United
States
| | - Diana C. West
- Department
of Chemistry and ‡Department of Biochemistry, Roger Adams Laboratory, University of Illinois, Urbana, Illinois 61801, United
States
| | - Rachel C. Botham
- Department
of Chemistry and ‡Department of Biochemistry, Roger Adams Laboratory, University of Illinois, Urbana, Illinois 61801, United
States
| | - Chris J. Novotny
- Department
of Chemistry and ‡Department of Biochemistry, Roger Adams Laboratory, University of Illinois, Urbana, Illinois 61801, United
States
| | - Steven C. Schmid
- Department
of Chemistry and ‡Department of Biochemistry, Roger Adams Laboratory, University of Illinois, Urbana, Illinois 61801, United
States
| | - Paul J. Hergenrother
- Department
of Chemistry and ‡Department of Biochemistry, Roger Adams Laboratory, University of Illinois, Urbana, Illinois 61801, United
States
| |
Collapse
|
28
|
Straten PT, Andersen MH. The anti-apoptotic members of the Bcl-2 family are attractive tumor-associated antigens. Oncotarget 2011; 1:239-45. [PMID: 21304176 DOI: 10.18632/oncotarget.100804] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Anti-apoptotic members of the Bcl-2 family (Bcl-2, Bcl-X(L) and Mcl-2) are pivotal regulators of apoptotic cell death. They are all highly overexpressed in cancers of different origin in which they enhance the survival of the cancer cells. Consequently, they represent prime candidates for anti-cancer therapy and specific antisense oligonucleotides or small molecule inhibitors have shown broad anti-cancer activities in pre-clinical models and are currently tested in clinical trials. In addition, immune-mediated tumor destruction is emerging as an interesting modality to treat cancer patients. Notably, spontaneous cellular immune responses against the Bcl-2 family proteins have been identified as frequent features in cancer patients underscoring that these proteins are natural targets for the immune system. Thus, Bcl-2 family may serve as an important and widely applicable target for anti-cancer immunotherapeutic strategies, alone or in the combination with conventional therapy. Here, we summarize the current knowledge of Bcl-2 family proteins as T-cell antigens, which has set the stage for the first explorative trial using these antigens in therapeutic vaccinations against cancer, and discuss future opportunities.
Collapse
Affiliation(s)
- Per thor Straten
- Center for Cancer Immune Therapy, Department of Hematology, Herlev University Hospital, Denmark
| | | |
Collapse
|
29
|
Jardé T, Perrier S, Vasson MP, Caldefie-Chézet F. Molecular mechanisms of leptin and adiponectin in breast cancer. Eur J Cancer 2011; 47:33-43. [PMID: 20889333 DOI: 10.1016/j.ejca.2010.09.005] [Citation(s) in RCA: 177] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2010] [Revised: 08/24/2010] [Accepted: 09/02/2010] [Indexed: 12/29/2022]
Abstract
Obesity is associated with an increased risk of breast cancer in postmenopausal women. Accumulating evidence suggests that adipose tissue, which is an endocrine organ producing a large range of factors, may interfere with breast cancer development. Leptin and adiponectin are two major adipocyte-secreted hormones. The pro-carcinogenic effect of leptin and conversely, the anti-carcinogenic effect of adiponectin result from two main mechanisms: a modulation in the signalling pathways involved in proliferation process and a subtle regulation of the apoptotic response. This review provides insight into recent findings on the molecular mechanisms of leptin and adiponectin in mammary tumours, and discusses the potential interplay between these two adipokines in breast cancer.
Collapse
Affiliation(s)
- Thierry Jardé
- Cardiff School of Biosciences, Cardiff University, S. Wales, Cardiff CF10 3US, United Kingdom.
| | | | | | | |
Collapse
|
30
|
Tiwari P, Kumar A, Balakrishnan S, Kushwaha HS, Mishra KP. Silibinin-induced apoptosis in MCF7 and T47D human breast carcinoma cells involves caspase-8 activation and mitochondrial pathway. Cancer Invest 2011; 29:12-20. [PMID: 21166494 DOI: 10.3109/07357907.2010.535053] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Silibinin, a natural flavonoid, under phase I/II clinical trial in prostate cancer patients was aimed to evaluate its chemotherapeutic potential in human breast cancer cell MCF7 and T47D. Results showed that T47D cells were found to be more sensitive to silibinin than MCF7 as observed by proliferation, clonogenic, and apoptotic assays, which was abrogated by pan-caspase inhibitor but remained unaffected by p53 inhibitor. Apoptotic events in both cell types differ temporally and also by magnitude that involved mitochondrial and caspase-8 activation pathway. These results have relevance in understanding silibinin treatment to breast tumor.
Collapse
Affiliation(s)
- Prabha Tiwari
- Radiological Physics and Advisory Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400 085, India
| | | | | | | | | |
Collapse
|
31
|
Yildiz Y, Yaylim-Eraltan I, Arikan S, Ergen HA, Küçücük S, Isbir T. Is there any correlation between TNF-related apoptosis-inducing ligand (TRAIL) genetic variants and breast cancer? Arch Med Sci 2010; 6:932-6. [PMID: 22427769 PMCID: PMC3302707 DOI: 10.5114/aoms.2010.19304] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2010] [Revised: 07/28/2010] [Accepted: 08/25/2010] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION TNF-related apoptosis-inducing ligand (TRAIL) is a death ligand and also a member of the TNF superfamily. We aimed to investigate the possible relationship between TRAIL and breast cancer. Here, we report the results of the first association study on genetic variation in the TRAIL gene and its effect on breast cancer susceptibility and prognosis. MATERIAL AND METHODS A C/T polymorphism at 1595 position in exon 5 of the TRAIL gene was genotyped in a Turkish breast cancer case-control population including 53 cases (mean age: 55.09 ±11.63 years) and 57 controls (mean age: 57.17 ±17.48 years) using polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) analysis. RESULTS There were no differences in the distribution of TRAIL genotypes and frequencies of the alleles in the breast cancer patients and controls. A heterozygous TRAIL CT polymorphism in exon 5 was present in 8.3% of tumour stage III-IV and 48.8% of stage I-II patients, and in 42.1% of controls. The reduced frequency of this genotype in patients who had advanced tumour stage was statistically significant (p = 0.017). CONCLUSIONS Our findings indicate that genetic variants of TRAIL at position 1595 in exon 5 might be associated with progression of breast cancer.
Collapse
Affiliation(s)
- Yemliha Yildiz
- Department of Molecular Medicine, Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | | | | | | | | | | |
Collapse
|
32
|
Thor Straten P, Andersen MH. The anti-apoptotic members of the Bcl-2 family are attractive tumor-associated antigens. Oncotarget 2010; 1:239-245. [PMID: 21304176 PMCID: PMC3248102 DOI: 10.18632/oncotarget.134] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2010] [Accepted: 08/09/2010] [Indexed: 11/25/2022] Open
Abstract
Anti-apoptotic members of the Bcl-2 family (Bcl-2, Bcl-X(L) and Mcl-2) are pivotal regulators of apoptotic cell death. They are all highly overexpressed in cancers of different origin in which they enhance the survival of the cancer cells. Consequently, they represent prime candidates for anti-cancer therapy and specific antisense oligonucleotides or small molecule inhibitors have shown broad anti-cancer activities in pre-clinical models and are currently tested in clinical trials. In addition, immune-mediated tumor destruction is emerging as an interesting modality to treat cancer patients. Notably, spontaneous cellular immune responses against the Bcl-2 family proteins have been identified as frequent features in cancer patients underscoring that these proteins are natural targets for the immune system. Thus, Bcl-2 family may serve as an important and widely applicable target for anti-cancer immunotherapeutic strategies, alone or in the combination with conventional therapy. Here, we summarize the current knowledge of Bcl-2 family proteins as T-cell antigens, which has set the stage for the first explorative trial using these antigens in therapeutic vaccinations against cancer, and discuss future opportunities.
Collapse
|
33
|
Placzek WJ, Wei J, Kitada S, Zhai D, Reed JC, Pellecchia M. A survey of the anti-apoptotic Bcl-2 subfamily expression in cancer types provides a platform to predict the efficacy of Bcl-2 antagonists in cancer therapy. Cell Death Dis 2010; 1:e40. [PMID: 21364647 PMCID: PMC3032312 DOI: 10.1038/cddis.2010.18] [Citation(s) in RCA: 229] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
We investigated the mRNA expression levels of all six antiapoptotic Bcl-2 subfamily members in 68 human cancer cell lines using qPCR techniques and measured the ability of known Bcl-2 inhibitors to induce cell death in 36 of the studied tumor cell lines. Our study reveals that Mcl-1 represents the anti-apoptotic Bcl-2 subfamily member with the highest mRNA levels in the lung, prostate, breast, ovarian, renal, and glioma cancer cell lines. In leukemia/lymphoma and melanoma cancer cell lines, Bcl-2 and Bfl-1 had the highest levels of mRNA, respectively. The observed correlation between the cell killing properties of known Bcl-2 inhibitors and the relative mRNA expression levels of anti-apoptotic Bcl-2 proteins provide critical insights into apoptosis-based anticancer strategies that target Bcl-2 proteins. Our data may explain current challenges of selective Bcl-2 inhibitors in the clinic, given that severe expression of Bcl-2 seems to be limited to leukemia cell lines. Furthermore, our data suggest that in most cancer types a strategy targeted to Mcl-1 inhibition, or combination of Bfl-1 and Mcl-1 inhibition for melanoma, may prove to be more successful than therapies targeting only Bcl-2.
Collapse
Affiliation(s)
- W J Placzek
- Sanford/Burnham Medical Research Institute, La Jolla, CA 92037, USA
| | | | | | | | | | | |
Collapse
|
34
|
Kim DJ, Kataoka K, Sano S, Connolly K, Kiguchi K, DiGiovanni J. Targeted disruption of Bcl-xL in mouse keratinocytes inhibits both UVB- and chemically induced skin carcinogenesis. Mol Carcinog 2009; 48:873-85. [PMID: 19309000 DOI: 10.1002/mc.20527] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Bcl-x(L) is one of several antiapoptotic proteins regulated by signal transducer and activator of transcription 3 (Stat3). We have recently shown that Stat3 is required for chemically induced and ultraviolet B (UVB)-induced skin carcinogenesis. In this study, the functional role of Bcl-x(L) in skin carcinogenesis was investigated using skin-specific Bcl-x(L)-deficient mice. In this model, Bcl-x(L) expression is disrupted in the basal compartment of mouse epidermis using the bovine keratin 5 (K5) promoter to drive expression of Cre recombinase (K5.Cre x Bcl-x(L) (fl/fl) mice). A significant increase in apoptosis induced by either UVB irradiation or 7,12-dimethylbenz[a]anthracene (DMBA) treatment was observed in the epidermis of Bcl-x(L)-deficient mice. Furthermore, an increase in apoptotic cells was noted in hair follicle keratinocytes, including those located in the bulge region. Cell proliferation was not affected by Bcl-x(L) deficiency following exposure to either UVB or 12-O-tetradecanoylphorbol-13-acetate (TPA). Bcl-x(L)-deficient mice were more resistant than wild-type controls to skin tumor development with delayed onset and reduced number of tumors using either UVB or the DMBA/TPA two-stage regimen. Moreover, Bcl-2, Mcl-1, and survivin protein levels were increased in the epidermis of Bcl-x(L)-deficient mice in the absence of stimuli. Furthermore, levels of these antiapoptotic proteins were also high in skin tumors from Bcl-x(L)-deficient mice that developed in response to either UVB or two-stage carcinogenesis protocols. Collectively, these studies demonstrate that Bcl-x(L) plays a role early in skin carcinogenesis through its anti-apoptotic functions to enhance survival of keratinocytes, including bulge region keratinocyte stem cells, following DNA damage.
Collapse
Affiliation(s)
- Dae Joon Kim
- Department of Carcinogenesis, The University of Texas M.D. Anderson Cancer Center, Science Park-Research Division, Smithville, Texas, USA
| | | | | | | | | | | |
Collapse
|
35
|
Cecka F, Hornychová H, Melichar B, Ryska A, Jandík P, Mergancová J, Klozová-Urminská H. Expression of bcl-2 in breast cancer: correlation with clinicopathological characteristics and survival. ACTA MEDICA (HRADEC KRÁLOVÉ) 2008; 51:107-12. [PMID: 18998362 DOI: 10.14712/18059694.2017.11] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Breast cancer is the most common malignancy in women. It is an immensely heterogeneous disease, characterised by a broad variety of clinical development. The research in recent years has focused on finding new markers of prognosis. This study investigates the role of expression of the bcl-2 protein in breast cancer. We analysed bcl-2 expression in 57 women with primary breast carcinoma who were treated with neoadjuvant (primary) chemotherapy, followed by a surgical procedure. The bcl-2 expression was correlated with other clinicopathological characteristics of the tumour- histological grade, stage, expression of hormonal receptors, proliferation rate, and with the survival of the patients. No significant association of bcl-2 expression with either overall survival or disease free survival was found.
Collapse
Affiliation(s)
- Filip Cecka
- Charles University in Prague, Faculty of Medicine and University Hospital Hradec Králové, Department of Surgery, Czech Republic.
| | | | | | | | | | | | | |
Collapse
|
36
|
Nadler Y, Camp RL, Giltnane JM, Moeder C, Rimm DL, Kluger HM, Kluger Y. Expression patterns and prognostic value of Bag-1 and Bcl-2 in breast cancer. Breast Cancer Res 2008; 10:R35. [PMID: 18430249 PMCID: PMC2397537 DOI: 10.1186/bcr1998] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2007] [Revised: 04/14/2008] [Accepted: 04/23/2008] [Indexed: 12/05/2022] Open
Abstract
Introduction Bcl-2 antanogene-1 (Bag-1) binds the anti-apoptotic mediator Bcl-2, and enhances its activity. Bcl-2 and Bag-1 are associated with chemotherapy resistance in cancer cells. Drugs that target Bcl-2 are currently in clinical development. The purpose of the present study was to examine expression patterns of Bag-1 in a large cohort of breast tumors and to assess the association with Bcl-2, estrogen receptor, progesterone receptor and Her2/neu, and other clinical/pathological variables. Methods Tissue microarrays containing primary specimens from 638 patients with 10-year follow-up were employed, and the expression of Bag-1, Bcl-2, estrogen receptor, progesterone receptor and Her2/neu was assessed using our automated quantitative analysis method. We used cytokeratin to define pixels as breast cancer (tumor mask) within the array spot, and we measured biomarker expression within the mask using Cy5 conjugated antibodies. Results High Bcl-2 expression was associated with improved survival in the entire cohort and in the node-positive subset (P = 0.008 and P = 0.002, respectively). High Bag-1 expression was associated with improved survival in the node-positive subset (P = 0.006). On multivariable analysis, neither Bcl-2 nor Bag-1 retained their independence as prognostic markers. Strong associations were found between Bag-1, Bcl-2, estrogen receptor and progesterone receptor. Conclusion Bag-1 and Bcl-2 expression in breast tumors is associated with improved outcome and steroid receptor positivity. Evaluation of Bcl-2 and Bag-1 expression in breast cancer may identify a subset of patients with a favorable prognosis, who might not benefit from chemotherapy or who might benefit from Bcl-2 targeting agents in addition to antihormonal therapy.
Collapse
Affiliation(s)
- Yasmine Nadler
- Department of Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Kallio A, Zheng A, Dahllund J, Heiskanen KM, Härkönen P. Role of mitochondria in tamoxifen-induced rapid death of MCF-7 breast cancer cells. Apoptosis 2008; 10:1395-410. [PMID: 16215679 DOI: 10.1007/s10495-005-2137-z] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Tamoxifen (Tam) is widely used in chemotherapy of estrogen receptor-positive breast cancer. It inhibits proliferation and induces apoptosis of breast cancer cells by estrogen receptor-dependent modulation of gene expression, but recent reports have shown that Tam (especially at pharmacological concentrations) has also rapid nongenomic effects. Here we studied the mechanisms by which Tam exerts rapid effects on breast cancer cell viability. In serum-free medium 5-7 microM Tam induced death of MCF-7 and MDA-MB-231 cells in a time-dependent manner in less than 60 min. This was associated with release of mitochondrial cytochrome c, a decrease of mitochondrial membrane potential and an increase in production of reactive oxygen species (ROS). This suggests that disruption of mitochondrial function has a primary role in the acute death response of the cells. Accordingly, bongkrekic acid, an inhibitor of mitochondrial permeability transition, was able to protect MCF-7 cells against Tam. Rapid cell death induction by Tam was not associated with immediate activation of caspase-9 or cleavage of poly (ADP-ribose) polymerase. It was not blocked by the caspase inhibitor z-Val-Ala-Asp-fluoromethylketone either. Diphenylene ionodium (DPI), an inhibitor of NADPH oxidase, was able to prevent Tam-induced cell death but not cytochrome c release, which suggests that ROS act distal to cytochrome c. The pure antiestrogen ICI 182780 (1 microM) could partly oppose the effect of Tam in estrogen receptor positive MCF-7 cells, but not in estrogen receptor negative MDA-MB-231 cells. Pre-culturing MCF-7 cells in the absence of 17beta-estradiol (E(2)) or in the presence of a low Tam concentration (1 microM) made the cells even more susceptible to rapid death induction by 5 or 7 microM Tam. This effect was associated with decreased levels of the anti-apoptotic proteins Bcl-X(L) and Bcl-2. In conclusion, our results demonstrate induction of a rapid mitochondrial cell death program in breast cancer cells at pharmacological concentrations of Tam, which are achievable in tumor tissue of Tam-treated breast cancer patients. These mechanisms may contribute to the ability of Tam therapy to induce death of breast cancer cells.
Collapse
Affiliation(s)
- A Kallio
- Department of Anatomy and Medicity Research Laboratory, Institute of Biomedicine, University of Turku, Tykistökatu 6A, 20520 Turku, Finland.
| | | | | | | | | |
Collapse
|
38
|
Clemo NK, Collard TJ, Southern SL, Edwards KD, Moorghen M, Packham G, Hague A, Paraskeva C, Williams AC. BAG-1 is up-regulated in colorectal tumour progression and promotes colorectal tumour cell survival through increased NF-κB activity. Carcinogenesis 2008; 29:849-57. [DOI: 10.1093/carcin/bgn004] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
39
|
Rabi T, Wang L, Banerjee S. Novel triterpenoid 25-hydroxy-3-oxoolean-12-en-28-oic acid induces growth arrest and apoptosis in breast cancer cells. Breast Cancer Res Treat 2007; 101:27-36. [PMID: 17028990 DOI: 10.1007/s10549-006-9275-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
25-Hydroxy-3-oxoolean-12-en-28-oic acid (Amooranin-AMR) is a triterpene acid isolated from the stem bark of a tropical tree (Amoora rohituka) grown wild in India. A herbal preparation used for the treatment of cancer by the Ayurvedic system of medicine contains the stem bark of Amoora rohituka as one of the ingredients. In this paper, we show that AMR displays a strong inhibitory effect on survival of human breast carcinoma MDA-468, breast adenocarcinoma MCF-7 cells compared to breast epithelial MCF-10A control cells. A 50% decrease in cells (IC50) ranged from 1.8 to 14.6 microM and cell growth was suppressed by arresting cell cycle at G2 + M phase. AMR effectively induces apoptosis and triggered a series of effects associated with apoptosis including cleavage of caspase-8, -9, -3, Bid and ER stress in MDA-468 cells and caspase- 8, -9, -6 and Bid in MCF-7 cells, release of cytochrome c from the mitochondria, cleavage of poly (ADP-ribose) polymerase (PARP) and DNA fragmentation with a concomitant upregulation of p53, Bax and down-regulation of Bcl-2 in MDA-468 cells, but Bax unchanged in MCF-7 cells. The use of caspase blocking peptides and acridine orange staining confirmed the involvement of primarily caspase-9 and -3 in MDA-468 cells with mutated p53 and primarily caspase-8, -9 and -6 in MCF-7 cells expressing wt p53. We also observed in MCF-7/p53siRNA cells AMR treatment caused reduced expression of Bcl-2 without affecting levels of Bax similar to MCF-7 cells treated with AMR and proteolytic activation of Bax in MDA-468 cells. These results suggest that AMR induces apoptosis in human breast carcinoma cells via caspase activation pathway and likely it is a p53-independent apoptosis.
Collapse
Affiliation(s)
- Thangaiyan Rabi
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA.
| | | | | |
Collapse
|
40
|
Rau KM, Day CP, Hung MC. Breast Cancer Gene Therapy. BREAST CANCER AND MOLECULAR MEDICINE 2007:705-740. [DOI: 10.1007/978-3-540-28266-2_34] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
41
|
Kobayashi S, Lee SH, Meng XW, Mott JL, Bronk SF, Werneburg NW, Craig RW, Kaufmann SH, Gores GJ. Serine 64 phosphorylation enhances the antiapoptotic function of Mcl-1. J Biol Chem 2007; 282:18407-18417. [PMID: 17463001 DOI: 10.1074/jbc.m610010200] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Mcl-1 is an antiapoptotic Bcl-2 family member that is highly regulated and when dysregulated contributes to cancer. The Mcl-1 protein is phosphorylated at multiple sites in response to different signaling events. Phosphorylations at Thr163 (by ERK) and Ser159 (by glycogen-synthase kinase 3beta) have recently been shown to slow and enhance, respectively, Mcl-1 protein turnover. Phosphorylation is also known to be stimulated at other, as-yet uncharacterized sites in the G2/M phase of the cell cycle. Using an S peptide-tagged Mcl-1 T163A mutant, Ser64 was identified as a novel Mcl-1 phosphorylation site by mass spectrometry. Immunoblotting demonstrated that phosphorylation at this site was maximal in cells in G2/M phase, was enhanced by tumor necrosis factor-alpha-related apoptosis-inducing ligand (TRAIL) treatment, was blocked by inhibitors of CDK (but not ERK or glycogen-synthase kinase 3beta), and was stimulated in vitro by CDK 1, CDK2, and JNK1. The half-life of a nonphosphorylatable S64A Mcl-1 mutant was indistinguishable from that of the wild type polypeptide. In contrast, this mutant failed to protect cells from TRAIL-mediated apoptosis, whereas reconstitution with the phosphomimetic S64E Mcl-1 mutant rendered cells TRAIL-resistant. This anti-apoptotic phenotype of the S64E Mcl-1 mutant was also associated with enhanced binding to the proapoptotic proteins Bim, Noxa, and Bak. A pharmacological CDK inhibitor that reduced Ser64 phosphorylation also sensitized cells to TRAIL cytotoxicity. Collectively, these observations not only identify G2/M-associated phosphorylation at Ser64 as a critical determinant of the antiapoptotic activity of Mcl-1 but also elucidate a novel mechanism by which CDK1/2 inhibitors can enhance the effectiveness of the cytotoxic cytokine TRAIL.
Collapse
Affiliation(s)
- Shogo Kobayashi
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, Minnesota 55905
| | - Sun-Hee Lee
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, Minnesota 55905
| | - Xue W Meng
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, Minnesota 55905; Division of Oncology Research, Mayo Clinic College of Medicine, Rochester, Minnesota 55905
| | - Justin L Mott
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, Minnesota 55905
| | - Steven F Bronk
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, Minnesota 55905
| | - Nathan W Werneburg
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, Minnesota 55905
| | - Ruth W Craig
- Department of Pharmacology and Toxicology, Dartmouth Medical School, Hanover, New Hampshire 03755
| | - Scott H Kaufmann
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, Minnesota 55905; Division of Oncology Research, Mayo Clinic College of Medicine, Rochester, Minnesota 55905
| | - Gregory J Gores
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, Minnesota 55905.
| |
Collapse
|
42
|
Lue H, Thiele M, Franz J, Dahl E, Speckgens S, Leng L, Fingerle-Rowson G, Bucala R, Lüscher B, Bernhagen J. Macrophage migration inhibitory factor (MIF) promotes cell survival by activation of the Akt pathway and role for CSN5/JAB1 in the control of autocrine MIF activity. Oncogene 2007; 26:5046-59. [PMID: 17310986 DOI: 10.1038/sj.onc.1210318] [Citation(s) in RCA: 287] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The phosphoinositide-3-kinase (PI3K)/Akt signaling pathway plays an important role in cell survival and the development of cancer. Macrophage migration inhibitory factor (MIF) is a critical inflammatory cytokine that was recently associated with tumorigenesis and that potently inhibits apoptosis. This may involve inhibition of p53-dependent genes, but the initiating molecular mechanism of how MIF controls survival/apoptosis is unknown. Here, we show that MIF prevents apoptosis and promotes tumor cell survival by directly activating the Akt pathway. MIF enhanced Akt activity in primary and immortalized fibroblasts (MEF and NIH/3T3), HeLa cervix carcinoma cells and various breast cancer cell lines. Activation was abolished by kinase inhibitors Ly294002 and PP2 and in Src/Yes/Fyn(SYF)(-/-) and CD74(-/-)(MEFs), while being enhanced in CD74-overexpressing MEFs, demonstrating that the MIF-induced Akt pathway encompasses signaling through the MIF receptor CD74 and the upstream kinases Src and PI3K. Akt was activated by exogenous rMIF and autocrine MIF action, as revealed by experiments in MIF(-/-)MEFs and antibody blockade. siRNA knockdown of CSN5/JAB1, a tumor marker and MIF-binding protein, showed that JAB1 controls autocrine MIF-mediated Akt signaling by inhibition of MIF secretion. Akt activation by MIF led to phosphorylation of the proapoptotic proteins BAD and Foxo3a. Apoptosis inhibition by MIF was functionally associated with Akt activation as it was abolished by overexpression of the Akt pathway inhibitor PTEN and occurred independently of p53. This was shown by studying DNA damage-induced apoptosis in fibroblasts, the Fas death pathway in HeLa cells that do not express functional p53, and etoposide-induced apoptosis in breast carcinoma cells expressing mutant p53. Importantly, dependence of breast cancer cell survival on MIF correlated with Akt activation and the PTEN status of these cells. Thus, MIF can directly promote cell survival through activation of the PI3K/Akt pathway and this effect is critical for tumor cell survival.
Collapse
Affiliation(s)
- H Lue
- Department of Biochemistry and Molecular Cell Biology, Institute of Biochemistry, University Hospital RWTH Aachen, Aachen, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Albert JM, Kim KW, Cao C, Lu B. Targeting the Akt/mammalian target of rapamycin pathway for radiosensitization of breast cancer. Mol Cancer Ther 2006; 5:1183-9. [PMID: 16731750 DOI: 10.1158/1535-7163.mct-05-0400] [Citation(s) in RCA: 141] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The phosphatidylinositol 3-kinase (PI3K)/Akt pathway is known to be activated by radiation. The mammalian target of rapamycin (mTOR) is downstream of Akt, and we investigated the effects of radiation on Akt/mTOR signaling in breast cancer cell models. RAD001 (everolimus), a potent derivative of the mTOR inhibitor rapamycin, was used to study the effects of mTOR inhibition, as the role of mTOR inhibition in enhancing radiation remains unexplored. RAD001 decreased clonogenic cell survival in both breast cancer cell lines MDA-MB-231 and MCF-7, although the effect is greater in MDA-MB-231 cells. Irradiation induced Akt and mTOR signaling, and this signaling is attenuated by RAD001. The radiation-induced signaling activation is mediated by PI3K because inhibition of PI3K with LY294002 inhibited the increase in downstream mTOR signaling. Additionally, caspase-dependent apoptosis is an important mechanism of cell death when RAD001 is combined with 3 Gy radiation, as shown by induction of caspase-3 cleavage. An increase in G(2)-M cell cycle arrest was seen in the combination treatment group when compared with controls, suggesting that cell cycle arrest may have been a contributing factor in the increased radiosensitization seen in this study. We conclude that RAD001 attenuates radiation-induced prosurvival Akt/mTOR signaling and enhances the cytotoxic effects of radiation in breast cancer cell models, showing promise as a method of radiosensitization of breast cancer.
Collapse
Affiliation(s)
- Jeffrey M Albert
- Department of Radiation Oncology, Vanderbilt University, B-902 The Vanderbilt Clinic, 1301 22nd Avenue South, Nashville, TN 37232-5671, USA
| | | | | | | |
Collapse
|
44
|
Sørensen RB, Nielsen OJ, Thor Straten P, Andersen MH. Functional capacity of Mcl-1-specific cytotoxic T-cells. Leukemia 2006; 20:1457-8. [PMID: 16728985 DOI: 10.1038/sj.leu.2404261] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
45
|
Roth W, Grimmel C, Rieger L, Strik H, Takayama S, Krajewski S, Meyermann R, Dichgans J, Reed JC, Weller M. Bag-1 and Bcl-2 gene transfer in malignant glioma: modulation of cell cycle regulation and apoptosis. Brain Pathol 2006; 10:223-34. [PMID: 10764042 PMCID: PMC8098428 DOI: 10.1111/j.1750-3639.2000.tb00256.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Bag-1 is a heat shock 70 kDa (Hsp70)-binding protein that can collaborate with Bcl-2 in suppressing apoptosis under some conditions. Here, we report that 11 of 12 human glioma cell lines express Bag-1 protein in vitro. Moreover, 15 of 19 human glioblastomas expressed Bag-1 as assessed by immunohistochemistry in primary tumor specimens. To examine the biological effects of Bag-1 in glioma cells, we expressed Bag-1 or Bcl-2 transgenes in 2 human malignant glioma cell lines, LN-18 and LN-229. Bag-1 significantly slowed glioma cell growth and reduced clonogenicity of both cell lines in vitro. Coexpressed Bcl-2 abrogated these effects of Bag-1. Intracranial LN-229 glioma xenografts implanted into nude mice revealed a substantial growth advantage afforded by Bcl-2. Bag-1 had no such effect, either in the absence or presence of Bcl-2. Upon serum starvation in vitro, Bcl-2 prevented cell death whereas Bag-1 did not. Both Bcl-2 and Bag-1 slowed proliferation of serum-starved cells when expressed alone. Importantly, coexpression of Bcl-2 and Bag-1 provided a distinct growth advantage under conditions of serum starvation that is probably the result of (i) the death-preventing activity of Bcl-2 and (ii) the property of Bag-1 to overcome a Bcl-2-mediated enhancement of exit from the cell cycle. In contrast to these Bcl-2/Bag-1 interactions observed under serum starvation conditions, Bag-1 did not further enhance the strong protection from staurosporine-, CD95 (Fas/Apo1) ligand-, Apo2 ligand (TRAIL)- or chemotherapeutic drug-induced apoptosis afforded by Bcl-2. Taken together, these results indicate a role for Bag-1/Bcl-2 interactions in providing a survival advantage to cancer cells in a deprived microenvironment that may be characteristic of ischemic/hypoxic tumors such as human glioblastoma multiforme, and suggest that Bcl-2/Bag-1 interactions also modulate cell proliferation.
Collapse
Affiliation(s)
- W Roth
- Department of Neurology, University of Tübingen, School of Medicine, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Giebel J, Woenckhaus C, Fabian M, Tost F. Age-related differential expression of apoptosis-related genes in conjunctival epithelial cells. ACTA ACUST UNITED AC 2005; 83:471-6. [PMID: 16029273 DOI: 10.1111/j.1600-0420.2005.00472.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
PURPOSE To investigate whether the expression of apoptosis-related genes in normal conjunctival epithelial cells is age-related (as a prerequisite to assessing whether dysregulation of apoptosis may be involved during degenerative diseases). METHODS Differential expression of apoptosis-related genes (e.g. apoptosis protease-activating factor 1 [Apaf-1]; caspases [casp] 3, 5, 8 and 9; Bad, Bax, Bcl-2, Bim, c-myc, Bag-1, as well as p53) was assessed by reverse transcription-polymerase chain reaction (RT-PCR). Samples were obtained from impression cytology (IC) specimens taken from 50 healthy subjects. Group A comprised 27 subjects aged 19-32 years and group B included 23 subjects aged 53-84 years. RESULTS Reverse transcription-PCR revealed the detection of apoptosis-related m-RNAs as follows (group A compared to group B): Apaf-1 0%/0%; Bcl-2 0%/35%; Bim 0%/9%; Bag-1 0%/9%; p53 0%/4%; casp-3 11%/52%; casp-5 59%/48%; casp-8 44%/22%; casp-9 4%/9%; Bax 81%/52%; Bad 96%/56%, and c-myc 89%/96%. CONCLUSION The data show an age-related expression of apoptosis-related genes such as casp-3, Bad, Bax and Bcl-2 in normal conjunctival cells. These results provide basic information which will help us understand the expression pattern of apoptotic genes during physiological ageing of the conjunctiva and the possible dysregulation of apoptotic genes during acute and chronic diseases such as dry eye disease, allergic conjunctivitis or cicatrizing conjunctivitis.
Collapse
Affiliation(s)
- Jürgen Giebel
- Institute of Anatomy, Ernst Moritz Arndt University, Greifswald, Germany
| | | | | | | |
Collapse
|
47
|
Andersen MH, Becker JC, Straten PT. Regulators of apoptosis: suitable targets for immune therapy of cancer. Nat Rev Drug Discov 2005; 4:399-409. [PMID: 15864269 DOI: 10.1038/nrd1717] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Harnessing the immune system in the battle against cancer has been the focus of tremendous research efforts during the past two decades. Several means for achieving this goal, including adoptive transfer of tumour-reactive T cells, systemic or localized administration of immune modulating cytokines and the use of 'therapeutic' vaccines, have been explored. Anti-apoptotic molecules that enhance the survival of cancer cells and facilitate their escape from cytotoxic therapies represent prime candidates as vaccination antigens. Notably, spontaneous cellular immune responses against these proteins have frequently been identified in cancer patients. Here, we summarize current knowledge of IAP and BCL2 family proteins as T-cell antigens, report the results of the first explorative trial using these antigens in therapeutic vaccinations against cancer and discuss future opportunities.
Collapse
Affiliation(s)
- Mads Hald Andersen
- Tumor Immunology Group, Institute of Cancer Biology, Danish Cancer Society, Strandboulevarden 49, Dk-2100 Copenhagen, Denmark.
| | | | | |
Collapse
|
48
|
Andersen MH, Becker JC, Thor Straten P. The antiapoptotic member of the Bcl-2 family Mcl-1 is a CTL target in cancer patients. Leukemia 2004; 19:484-5. [PMID: 15618955 DOI: 10.1038/sj.leu.2403621] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
49
|
Pusztai L, Krishnamurti S, Perez Cardona J, Sneige N, Esteva FJ, Volchenok M, Breitenfelder P, Kau SW, Takayama S, Krajewski S, Reed JC, Bast RC, Hortobagyi GN. Expression of BAG-1 and BcL-2 proteins before and after neoadjuvant chemotherapy of locally advanced breast cancer. Cancer Invest 2004; 22:248-56. [PMID: 15199607 DOI: 10.1081/cnv-120030213] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
It has been suggested that expression of anti-apoptotic proteins such as Bcl-2 or BAG-1 may confer cellular resistance to chemotherapy. A corollary of this hypothesis is that expression of these proteins may predict clinical response to treatment and that Bcl-2- or BAG-1-positive cells may selectively be enriched in postchemotherapy tissue specimens. The goal of this exploratory pilot study was to assess these two predictions by using immunohistochemistry in 29 paired pre- and postchemotherapy breast tissue specimens obtained from patients who underwent preoperative doxorubicin-based chemotherapy. All breast cancers expressed BAG-1 protein, and, in individual tumors, 40-100% of neoplastic cells stained positive for this protein. Homogenous cytoplasmic staining was typically observed, though neoplastic cells also showed nuclear staining in many specimens. We found no correlation between prechemotherapy expression of BAG-1 and subsequent pathological response to cytotoxic therapy. Paired pre- and posttreatment specimens showed similar levels of BAG-1 expression when residual tumor could be assessed. Bcl-2 was expressed in 55% of cancers and was localized to the cytoplasm. Absence of Bcl-2 expression in prechemotherapy specimens was associated with more frequent complete pathological response (58% vs. 20%; p = 0.04). However, similar to BAG-1, no difference between pre- and posttherapy expression of Bcl-2 was observed in neoplastic cells in paired tissue specimens. These observations suggest that BAG-1 contributes an important cellular function to breast epithelial cells, which is reflected by its ubiquitous expression in these tissues. However, it does not appear to determine response to doxorubicin-based chemotherapy. In contrast, lack of Bcl-2 expression was associated with a higher probability of complete pathological response to doxorubicin-based chemotherapy.
Collapse
Affiliation(s)
- Lajos Pusztai
- Department of Breast Medical Oncology, University of Texas M.D. Anderson Cancer Center, Box 424, 1515 Holcombe Blvd., Houston, TX 77030-4009, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Li D, Das S, Yamada T, Samuels HH. The NRIF3 family of transcriptional coregulators induces rapid and profound apoptosis in breast cancer cells. Mol Cell Biol 2004; 24:3838-48. [PMID: 15082778 PMCID: PMC387764 DOI: 10.1128/mcb.24.9.3838-3848.2004] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2003] [Revised: 12/16/2003] [Accepted: 02/04/2004] [Indexed: 11/20/2022] Open
Abstract
Many anticancer drugs kill cancer cells by inducing apoptosis. Despite the progress in understanding apoptosis, how to harness the cellular death machinery to selectively deliver tumor-specific cytotoxicity (while minimizing damage to other cells) remains an important challenge. We report here that expression of the NRIF3 family of transcriptional coregulators in a variety of breast cancer cell lines induces rapid and profound apoptosis (nearly 100% cell death within 24 h). A novel death domain (DD1) was mapped to a short 30-amino-acid region common to all members of the NRIF3 family. Mechanistic studies showed that DD1-induced apoptosis occurs through a novel caspase 2-mediated pathway that involves mitochondrial membrane permeabilization but does not require other caspases. Interestingly, the cytotoxicity of NRIF3 and DD1 appears to be cell type specific, as they selectively kill breast cancer or related cells but not other examined cells of different origins. Our study demonstrates the feasibility of selectively inducing cytotoxicity in a specific cancer and suggests that breast cancer cells contain a novel "death switch" that can be specifically triggered by NRIF3 or DD1. Strategies utilizing NRIF3 and/or DD1 and/or targeting this death switch may lead to the development of novel and more selective therapeutics against breast cancer.
Collapse
Affiliation(s)
- Dangsheng Li
- Department of Pharmacology, New York University School of Medicine, New York, New York 10016, USA.
| | | | | | | |
Collapse
|