1
|
Liu J, Gao L, Ji B, Geng R, Chen J, Tao X, Cai Q, Chen Z. BCL7A as a novel prognostic biomarker for glioma patients. J Transl Med 2021; 19:335. [PMID: 34362400 PMCID: PMC8348860 DOI: 10.1186/s12967-021-03003-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 07/23/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Glioma is the most common primary brain tumor and represents one of the most aggressive and lethal types of human cancer. BCL7 family has been found in several cancer types and could be involved in tumor progression. While the role of BCL7 family in human glioma has remained to be elucidated. METHODS Paraffin-embedded tumor samples were obtained to detect BCL7 expression by performing in glioma. Data (including normalized gene expression and corresponding clinical data) were obtained from Gliovis, CGGA, GEO, cBioportal and Oncomine and were used to investigate BCL7 genes expression in glioma. Survival analyses were calculated by Kaplan-Meier methods and Cox regression analysis in TCGA and CGGA. Gene Set Enrichment Analyses (GSEA) and gene ontology (GO) analysis was employed to perform the biological processes enrichment. RESULTS BCL7A expression in glioma tissues was lower compared to non-tumor brain tissues (NBT), and exhibited a negative correlation with glioma grades. Results from immunohistochemical (IHC) staining and public dataset validation demonstrated that BCL7B and BCL7C were highly expressed in glioma tissues compared to NBT. Cox regression analysis identified BCL7A as the only gene in the BCL7 family that was independently associated with the prognosis of lower-grade glioma (LGG) and glioblastoma (GBM). GO and GSEA analyses revealed the potential contribution of BCL7A in adaptive immune response and neutrophil activation in the tumor microenvironment. Moreover, we found that BCL7A had no prognostic effect on the overall survival of GBM patients who received IR only; however, patients who received chemotherapy (TMZ) combined with IR in the high BCL7A group survived longer than patients in the low BCL7A group (HR = 0.346, p < 0.05). CONCLUSION BCL7A is a new tumor suppressor gene and can be adopted as a biomarker for independent prognosis in glioma and to evaluate response to TMZ.
Collapse
Affiliation(s)
- Junhui Liu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, No.238, Jiefang Road, Wuchang District, Wuhan, 430060, Hubei, China.,Central Laboratory, Renmin Hospital of Wuhan University, No.238, Jiefang Road, Wuchang District, Wuhan, 430060, Hubei, China
| | - Lun Gao
- Department of Neurosurgery, Renmin Hospital of Wuhan University, No.238, Jiefang Road, Wuchang District, Wuhan, 430060, Hubei, China.,Central Laboratory, Renmin Hospital of Wuhan University, No.238, Jiefang Road, Wuchang District, Wuhan, 430060, Hubei, China
| | - Baowei Ji
- Department of Neurosurgery, Renmin Hospital of Wuhan University, No.238, Jiefang Road, Wuchang District, Wuhan, 430060, Hubei, China
| | - Rongxin Geng
- Department of Neurosurgery, Renmin Hospital of Wuhan University, No.238, Jiefang Road, Wuchang District, Wuhan, 430060, Hubei, China
| | - Jing Chen
- Department of Neurosurgery, Renmin Hospital of Wuhan University, No.238, Jiefang Road, Wuchang District, Wuhan, 430060, Hubei, China
| | - Xiang Tao
- Department of Neurosurgery, Renmin Hospital of Wuhan University, No.238, Jiefang Road, Wuchang District, Wuhan, 430060, Hubei, China
| | - Qiang Cai
- Department of Neurosurgery, Renmin Hospital of Wuhan University, No.238, Jiefang Road, Wuchang District, Wuhan, 430060, Hubei, China.
| | - Zhibiao Chen
- Department of Neurosurgery, Renmin Hospital of Wuhan University, No.238, Jiefang Road, Wuchang District, Wuhan, 430060, Hubei, China.
| |
Collapse
|
2
|
Molavi O, Narimani F, Asiaee F, Sharifi S, Tarhriz V, Shayanfar A, Hejazi M, Lai R. Silibinin sensitizes chemo-resistant breast cancer cells to chemotherapy. PHARMACEUTICAL BIOLOGY 2017; 55:729-739. [PMID: 28027688 PMCID: PMC6130726 DOI: 10.1080/13880209.2016.1270972] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 10/30/2016] [Accepted: 12/03/2016] [Indexed: 05/22/2023]
Abstract
CONTEXT Multiple drug resistance is the major obstacle to conventional chemotherapy. Silibinin, a nontoxic naturally occurring compound, has anticancer activity and can increase the cytotoxic effects of chemotherapy in various cancer models. OBJECTIVE To evaluate the effects of silibinin on enhancing the sensitivity of chemo-resistant human breast cell lines to doxorubicin (DOX) and paclitaxel (PAC). MATERIALS AND METHODS The cells were treated with silibinin (at 50 to 600 μM concentrations) and/or chemo drugs for 24 and 48 h, then cell viability and changes in oncogenic proteins were determined by MTT assay and Western blotting/RT-PCR, respectively. Flow cytometry was used to study apoptosis in the cells receiving different treatments. The antitumorigenic effects of silibinin (at 200 to 400 μM concentration) were evaluated by mammosphere assay. RESULTS Silibinin exerted significant growth inhibitory effects with IC50 ranging from 200 to 570 μM in different cell lines. Treatment of DOX-resistant MDA-MB-435 cells with silibinin at 200 μM reduced DOX IC50 from 71 to 10 μg/mL and significantly suppressed the key oncogenic pathways including STAT3, AKT, and ERK in these cells. Interestingly treatment of DOX-resistant MDA-MB-435 cells with silibinin at 400 μM concentration for 48 h induced a 50% decrease in the numbers of colonies as compared with DMSO-treated cells. Treatment of PAC-resistant MCF-7 cells with silibinin at 400 μM concentration generated synergistic effects when it was used in combination with PAC at 250 nM concentration (CI = 0.81). CONCLUSION Silibinin sensitizes chemo-resistant cells to chemotherapeutic agents and can be useful in treating breast cancers.
Collapse
Affiliation(s)
- Ommoleila Molavi
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Laboratory Medicine and Pathology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
- CONTACT Ommoleila MolaviDepartment of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, 111-Faculty of Pharmacy, Tabriz University, Tabriz, Iran
| | - Farzaneh Narimani
- Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farshid Asiaee
- Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Simin Sharifi
- Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahideh Tarhriz
- Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Shayanfar
- Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Raymond Lai
- Department of Laboratory Medicine and Pathology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
3
|
Zhao H, Zhang Y, Sun J, Zhan C, Zhao L. Raltitrexed Inhibits HepG2 Cell Proliferation via G0/G1 Cell Cycle Arrest. Oncol Res 2017; 23:237-48. [PMID: 27098147 PMCID: PMC7838684 DOI: 10.3727/096504016x14562725373671] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Raltitrexed (RTX) is an antimetabolite drug used as a chemotherapeutic agent for treating colorectal cancer, malignant mesothelioma, and gastric cancer. The antitumor capacity of RTX is attributed to its inhibitory activity on thymidylate synthase (TS), a key enzyme in the synthesis of DNA precursors. The current study is aimed at investigating the potential antitumor effects of RTX in liver cancer. Using the HepG2 cell line as an in vitro model of liver cancer, we evaluated the effects of RTX on cell proliferation employing both a WST-8 assay and a clone formation efficiency assay. In addition, we monitored the ultrastructure changes of HepG2 cells in response to RTX with transmission electric microscopy. To investigate the mechanism underlying the regulation of cell proliferation by RTX, we analyzed cell cycle using cell flow cytometry. Moreover, real-time PCR and Western blot analyses were conducted to examine expression levels of cell cycle regulatory proteins cyclin A and cyclin-dependent kinase 2 (CDK2), as well as their mediators tumor suppressor genes p53 and p16. Our results demonstrate that RTX inhibits HepG2 proliferation by arresting the cell cycle at G0/G1. This cell cycle arrest function was mediated via downregulation of cyclin A and CDK2. The observed elevated expression of p53 and p16 by RTX may contribute to the reduction of cyclin A/CDK2. Our study indicates that RTX could serve as a potential chemotherapeutic agent in the treatment of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Hongwei Zhao
- Department of Hepatopancreatobiliary Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | | | | | | | | |
Collapse
|
4
|
Yokoo M, Kubota Y, Motoyama K, Higashi T, Taniyoshi M, Tokumaru H, Nishiyama R, Tabe Y, Mochinaga S, Sato A, Sueoka-Aragane N, Sueoka E, Arima H, Irie T, Kimura S. 2-Hydroxypropyl-β-Cyclodextrin Acts as a Novel Anticancer Agent. PLoS One 2015; 10:e0141946. [PMID: 26535909 PMCID: PMC4633159 DOI: 10.1371/journal.pone.0141946] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 10/15/2015] [Indexed: 12/11/2022] Open
Abstract
2-Hydroxypropyl-β-cyclodextrin (HP-β-CyD) is a cyclic oligosaccharide that is widely used as an enabling excipient in pharmaceutical formulations, but also as a cholesterol modifier. HP-β-CyD has recently been approved for the treatment of Niemann-Pick Type C disease, a lysosomal lipid storage disorder, and is used in clinical practice. Since cholesterol accumulation and/or dysregulated cholesterol metabolism has been described in various malignancies, including leukemia, we hypothesized that HP-β-CyD itself might have anticancer effects. This study provides evidence that HP-β-CyD inhibits leukemic cell proliferation at physiologically available doses. First, we identified the potency of HP-β-CyD in vitro against various leukemic cell lines derived from acute myeloid leukemia (AML), acute lymphoblastic leukemia and chronic myeloid leukemia (CML). HP-β-CyD treatment reduced intracellular cholesterol resulting in significant leukemic cell growth inhibition through G2/M cell-cycle arrest and apoptosis. Intraperitoneal injection of HP-β-CyD significantly improved survival in leukemia mouse models. Importantly, HP-β-CyD also showed anticancer effects against CML cells expressing a T315I BCR-ABL mutation (that confers resistance to most ABL tyrosine kinase inhibitors), and hypoxia-adapted CML cells that have characteristics of leukemic stem cells. In addition, colony forming ability of human primary AML and CML cells was inhibited by HP-β-CyD. Systemic administration of HP-β-CyD to mice had no significant adverse effects. These data suggest that HP-β-CyD is a promising anticancer agent regardless of disease or cellular characteristics.
Collapse
MESH Headings
- 2-Hydroxypropyl-beta-cyclodextrin
- Animals
- Antineoplastic Agents/therapeutic use
- Antineoplastic Agents/toxicity
- Apoptosis/drug effects
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Cholesterol/analysis
- Cholesterol/metabolism
- Colorimetry
- Drug Resistance, Neoplasm/drug effects
- Fusion Proteins, bcr-abl/genetics
- Fusion Proteins, bcr-abl/metabolism
- G2 Phase Cell Cycle Checkpoints/drug effects
- Humans
- K562 Cells
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myeloid, Acute/drug therapy
- Lung/pathology
- M Phase Cell Cycle Checkpoints/drug effects
- Mice
- Mice, Inbred BALB C
- Mice, Inbred NOD
- Mice, Nude
- Mice, SCID
- Signal Transduction/drug effects
- Transplantation, Heterologous
- beta-Cyclodextrins/therapeutic use
- beta-Cyclodextrins/toxicity
Collapse
Affiliation(s)
- Masako Yokoo
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Yasushi Kubota
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
- Department of Transfusion Medicine, Saga University Hospital, Saga, Japan
- * E-mail:
| | - Keiichi Motoyama
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Taishi Higashi
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Masatoshi Taniyoshi
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hiroko Tokumaru
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Rena Nishiyama
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yoko Tabe
- Department of Clinical Laboratory Medicine, Juntendo University School of Medicine, Tokyo, Japan
| | | | - Akemi Sato
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Naoko Sueoka-Aragane
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Eisaburo Sueoka
- Department of Transfusion Medicine, Saga University Hospital, Saga, Japan
- Department of Clinical Laboratory Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Hidetoshi Arima
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
- Program for Leading Graduate Schools “HIGO (Health life science: Interdisciplinary and Global Oriented) Program”, Kumamoto University, Kumamoto, Japan
| | - Tetsumi Irie
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
- Program for Leading Graduate Schools “HIGO (Health life science: Interdisciplinary and Global Oriented) Program”, Kumamoto University, Kumamoto, Japan
| | - Shinya Kimura
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| |
Collapse
|
5
|
Licciardi PV, Kwa FAA, Ververis K, Di Costanzo N, Balcerczyk A, Tang ML, El-Osta A, Karagiannis TC. Influence of natural and synthetic histone deacetylase inhibitors on chromatin. Antioxid Redox Signal 2012; 17:340-54. [PMID: 22229817 DOI: 10.1089/ars.2011.4480] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
SIGNIFICANCE Histone deacetylase inhibitors (HDACIs) have emerged as a new class of anticancer therapeutics. The hydroxamic acid, suberoylanilide hydroxamic acid (Vorinostat, Zolinza™), and the cyclic peptide, depsipeptide (Romidepsin, Istodax™), were approved by the U.S. Food and Drug Administration (FDA) for the treatment of cutaneous T-cell lymphoma in 2006 and 2009, respectively. At least 15 HDACIs are currently undergoing clinical trials either alone or in combination with other therapeutic modalities for the treatment of numerous hematological and solid malignancies. RECENT ADVANCES The potential utility of HDACIs has been extended to nononcologic applications, including autoimmune disorders, inflammation, diseases of the central nervous system, and malaria. CRITICAL ISSUES Given the promise of HDACIs, there is growing interest in the potential of dietary compounds that possess HDAC inhibition activity. This review is focused on the identification of and recent findings with HDACIs from dietary, medicinal plant, and microbial sources. We discuss the mechanisms of action and clinical potential of natural HDACIs. FUTURE DIRECTIONS Apart from identification of further HDACI compounds from dietary sources, further research will be aimed at understanding the effects on gene regulation on lifetime exposure to these compounds. Another important issue that requires clarification.
Collapse
Affiliation(s)
- Paul V Licciardi
- Allergy and Immune Disorders, Murdoch Childrens Research Institute, Royal Children's Hospital, Parkville, Victoria, Australia
| | | | | | | | | | | | | | | |
Collapse
|
6
|
Li LS, Morales JC, Veigl M, Sedwick D, Greer S, Meyers M, Wagner M, Fishel R, Boothman DA. DNA mismatch repair (MMR)-dependent 5-fluorouracil cytotoxicity and the potential for new therapeutic targets. Br J Pharmacol 2009; 158:679-92. [PMID: 19775280 DOI: 10.1111/j.1476-5381.2009.00423.x] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The metabolism and efficacy of 5-fluorouracil (FUra) and other fluorinated pyrimidine (FP) derivatives have been intensively investigated for over fifty years. FUra and its antimetabolites can be incorporated at RNA- and DNA-levels, with RNA level incorporation provoking toxic responses in human normal tissue, and DNA-level antimetabolite formation and incorporation believed primarily responsible for tumour-selective responses. Attempts to direct FUra into DNA-level antimetabolites, based on mechanism-of-action studies, have led to gradual improvements in tumour therapy. These include the use of leukovorin to stabilize the inhibitory thymidylate synthase-5-fluoro-2'-deoxyuridine 5' monophoshate (FdUMP)-5,10-methylene tetrahydrofolate (5,10-CH(2)FH(4)) trimeric complex. FUra incorporated into DNA also contributes to antitumour activity in preclinical and clinical studies. This review examines our current state of knowledge regarding the mechanistic aspects of FUra:Gua lesion detection by DNA mismatch repair (MMR) machinery that ultimately results in lethality. MMR-dependent direct cell death signalling or futile cycle responses will be discussed. As 10-30% of sporadic colon and endometrial tumours display MMR defects as a result of human MutL homologue-1 (hMLH1) promoter hypermethylation, we discuss the use and manipulation of the hypomethylating agent, 5-fluorodeoxycytidine (FdCyd), and our ability to manipulate its metabolism using the cytidine or deoxycytidylate (dCMP) deaminase inhibitors, tetrahydrouridine or deoxytetrahydrouridine, respectively, as a method for re-expression of hMLH1 and re-sensitization of tumours to FP therapy.
Collapse
Affiliation(s)
- Long Shan Li
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Sarkar S, Mandal M. Growth factor receptors and apoptosis regulators: signaling pathways, prognosis, chemosensitivity and treatment outcomes of breast cancer. BREAST CANCER-BASIC AND CLINICAL RESEARCH 2009; 3:47-60. [PMID: 21556249 PMCID: PMC3086304 DOI: 10.4137/bcbcr.s2492] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Biomarkers of breast cancer are necessary for prognosis and prediction to chemotherapy. Prognostic biomarkers provide information regarding outcome irrespective of therapy, while predictive biomarkers provide information regarding response to therapy. Candidate prognostic biomarkers for breast cancers are growth factor receptors, steroid receptors, Ki-67, cyclins, urokinase plasminogen activator, p53, p21, pro- and anti-apoptotic factors, BRCA1 and BRCA2. But currently, the predictive markers are Estrogen and Progesterone receptors responding to endocrine therapy, and HER-2 responding to herceptin. But there are numerous breast cancer cases, where tamoxifen is ineffective even after estrogen receptor positivity. This lead to search of new prognostic and predictive markers and the number of potential markers is constantly increasing due to proteomics and genomics studies. However, most biomarkers individually have poor sensitivity or specificity, or other clinical value. It can be resolved by studying various biomarkers simultaneously, which will help in better prognosis and increasing sensitivity for chemotherapeutic agents. This review is focusing on growth factor receptors, apoptosis markers, signaling cascades, and their correlation with other associated biomarkers in breast cancers. As our knowledge regarding molecular biomarkers for breast cancer increases, prognostic indices will be developed that combine the predictive power of individual molecular biomarkers with specific clinical and pathologic factors. Rigorous comparison of these existing as well as emerging markers with current treatment selection is likely to see an escalation in an era of personalized medicines to ensure the breast cancer patients receive optimal treatment. This will also solve the treatment modalities and complications related to chemotherapeutic regimens.
Collapse
Affiliation(s)
- Siddik Sarkar
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur-721302, India
| | | |
Collapse
|
8
|
Meyers M, Wagner MW, Mazurek A, Schmutte C, Fishel R, Boothman DA. DNA mismatch repair-dependent response to fluoropyrimidine-generated damage. J Biol Chem 2004; 280:5516-26. [PMID: 15611052 DOI: 10.1074/jbc.m412105200] [Citation(s) in RCA: 98] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Previous studies from our laboratory indicated that expression of the MLH1 DNA mismatch repair (MMR) gene was necessary to restore cytotoxicity and an efficient G(2) arrest in HCT116 human colon cancer cells, as well as Mlh1(-/-) murine embryonic fibroblasts, after treatment with 5-fluoro-2'-deoxyuridine (FdUrd). Here, we show that an identical phenomenon occurred when expression of MSH2, the other major MMR gene, was restored in HEC59 human endometrial carcinoma cells or was present in adenovirus E1A-immortalized Msh2(+/+) (compared with isogenic Msh2(-/-)) murine embryonic stem cells. Because MMR status had little effect on cellular responses (i.e. G(2) arrest and lethality) to the thymidylate synthase inhibitor, Tomudex, and a greater level of [(3)H]FdUrd incorporation into DNA was found in MMR-deficient cells, we concluded that the differential FdUrd cytotoxicity between MMR-competent and MMR-deficient cells was mediated at the level of DNA incorporation. Analyses of ATPase activation suggested that the hMSH2-hMSH6 heterodimer only recognized FdUrd moieties (as the base 5-fluorouracil (FU) in DNA) when mispaired with guanine, but not paired with adenine. Furthermore, analyses of incorporated FdUrd using methyl-CpG-binding domain 4 glycosylase indicated that there was more misincorporated FU:Gua in the DNA of MMR-deficient HCT116 cells. Our data provide the first demonstration that MMR specifically detects FU:Gua (in the first round of DNA replication), signaling a sustained G(2) arrest and lethality.
Collapse
Affiliation(s)
- Mark Meyers
- Department of Radiation Oncology and Case Comprehensive Cancer Center, Laboratory of Molecular Stress Responses, Case Western Reserve University, Cleveland, OH 44106, USA
| | | | | | | | | | | |
Collapse
|
9
|
Geller JI, Szekely-Szucs K, Petak I, Doyle B, Houghton JA. P21Cip1 is a critical mediator of the cytotoxic action of thymidylate synthase inhibitors in colorectal carcinoma cells. Cancer Res 2004; 64:6296-303. [PMID: 15342418 DOI: 10.1158/0008-5472.can-04-0863] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We have demonstrated previously that interferon (IFN)-gamma sensitizes human colon carcinoma cell lines to the cytotoxic effects of 5-fluorouracil combined with leucovorin and to the thymidylate synthase inhibitor, ZD9331, dependent on thymineless stress-induced DNA damage, independent of p53. Here we demonstrate that the cyclin-dependent kinase (CDK) inhibitor p21(Cip1) regulates thymineless stress-induced cytotoxicity in these cells. HCT116 wild-type (wt) and p53-/- cells underwent apoptosis and loss in clonogenic survival when exposed to ZD9331, whereas p21Cip1-/- cells were resistant. In contrast, IFN-gamma induced marked cytotoxicity in p21Cip1-/- cells only. ZD9331 induced p21Cip1 up-regulation in all of the cell lines examined, as did thymidine deprivation in thymidylate synthase-deficient (thymidylate synthase-) cells. Furthermore, selective induction of p21Cip1 in RKO was sufficient to induce apoptosis. P21Cip1, cdk1, cdk2, and cyclin E mRNA expression increased coincident with S-phase accumulation in HT29 cells treated with ZD9331 or 5fluorouracil/leucovorin, as demonstrated by cDNA microarray analyses. Cell cycle analyses revealed that HCT116 wt and p21Cip1 -/- cells accumulated in S phase within 24 h of ZD9331 exposure; however, wt cells exited S-phase more rapidly, where apoptosis occurred before mitosis, either in late S or G2. Finally, the CDK inhibitor roscovitine potentiated the cytotoxic activity of ZD9331 in both wt and p21Cip1-/- cells, strongly suggesting a role for p21Cip1-dependent CDK inhibition in cytotoxicity induced by thymidylate synthase inhibition. In summary, p21Cip1 positively regulates the cytotoxic action of thymidylate synthase inhibitors, negatively regulates the cytotoxic action of IFN-gamma, and enhances S-phase exit after thymineless stress, possibly via interaction with CDK-cyclin complexes.
Collapse
Affiliation(s)
- James I Geller
- Division of Molecular Therapeutics, Department of Hematology-Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | | | | | | | | |
Collapse
|
10
|
Liu S, Bishop WR, Liu M. Differential effects of cell cycle regulatory protein p21(WAF1/Cip1) on apoptosis and sensitivity to cancer chemotherapy. Drug Resist Updat 2003; 6:183-95. [PMID: 12962684 DOI: 10.1016/s1368-7646(03)00044-x] [Citation(s) in RCA: 126] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
p21(WAF1/Cip1) was initially identified as a cell cycle regulatory protein that can cause cell cycle arrest. It is induced by both p53-dependent and p53-independent mechanisms. This mini-review briefly discusses its currently known functions in apoptosis and drug sensitivity. As an inhibitor of cell proliferation, p21(WAF1/Cip1) plays an important role in drug-induced tumor suppression. Nevertheless, a number of recent studies have shown that p21(WAF1/Cip1) can assume both pro- or anti-apoptotic functions in response to anti-tumor agents depending on cell type and cellular context. This dual role of p21(WAF1/Cip1) in cancer cells complicates using p21(WAF1/Cip1) status to predict response to anti-tumor agents. However, it is possible to develop p21(WAF1/Cip1)-targeted reagents or p21(WAF1/Cip1) gene transfer techniques to have a beneficial effect within a well-defined therapeutic context. Better understanding of the roles of p21(WAF1/Cip1) in tumors should enable a more rational approach to anti-tumor drug design and therapy.
Collapse
Affiliation(s)
- Suxing Liu
- Biological Research-Oncology, Schering-Plough Research Institute, 2015 Galloping Hill Road, Kenilworth, NJ 07033, USA.
| | | | | |
Collapse
|
11
|
Liby K, Neltner B, Mohamet L, Menchen L, Ben-Jonathan N. Prolactin overexpression by MDA-MB-435 human breast cancer cells accelerates tumor growth. Breast Cancer Res Treat 2003; 79:241-52. [PMID: 12825859 DOI: 10.1023/a:1023956223037] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Prolactin (PRL) is an important hormone in mammary tumorigenesis in rodents but its involvement in human breast cancer has been controversial. A role for locally produced PRL in breast carcinogenesis is suggested by its mitogenic action on breast cancer cells and the expression of both PRL and its receptor (PRL-R) in breast carcinomas. Our objective was to examine whether PRL, overexpressed by breast cancer cells, forms an autocrine/paracrine loop that confers a growth advantage for tumors. MDA-MB-435 breast cancer cells overexpressing 23K human PRL were generated, and PRL production and secretion by the clones were confirmed by RT-PCR, western blotting, and the Nb2 bioassay; control clones contain vector only. In vitro the 23K PRL clones proliferated faster and expressed higher levels of the PRL-R protein than controls only when incubated in charcoal-stripped serum (CSS) devoid of lactogenic hormones. When injected into the mammary fatpad of female nude mice or subcutaneously into males, the PRL-overexpressing clones formed tumors that grew 2-4-fold faster than tumors derived from control clones or wild type MDA-MB-435 cells. Western analysis demonstrated significantly higher PRL, PRL-R, and bcl-2 levels in the tumors overexpressing PRL compared to control tumors. These data support a role for breast PRL as a growth/anti-apoptotic factor and suggest that it may serve as a novel therapeutic target for the treatment of breast cancer.
Collapse
Affiliation(s)
- Karen Liby
- Department of Cell Biology, University of Cincinnati Medical School, Cincinnati, OH 45267-0521, USA
| | | | | | | | | |
Collapse
|
12
|
Kim R, Osaki A, Hirai T, Toge T. Utility of technetium-99m methoxyisobutyl isonitrile uptake analysis for prediction of the response to chemotherapy in advanced and relapsed breast cancer. Breast Cancer 2003; 9:240-7. [PMID: 12185336 DOI: 10.1007/bf02967596] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
BACKGROUND Technetium-99m methoxyisobutyl isonitrile (Tc-SESTAMIBI) is a substrate of P-glycoprotein and multidrug-resistance associated protein in drug-resistant cells. To assess the clinical effectiveness of Tc-SESTAMIBI for predicting the chemotherapy response to treatment with anthracyclines and vinca alkaloids, we retrospectively evaluated the relationship between the accumulation of Tc-SESTAMIBI and the tumor response. METHODS Thirteen patients, including 12 advanced cases and 1 relapsed case, were investigated, all of whom had been treated with anthracyclines or a vinca alkaloid regimen. The accumulation of Tc-SESTAMIBI was compared at 10 min and 2 h after Tc-SESTAMIBI administration. The relationship between the accumulation of Tc-SESTAMIBI and the tumor response following treatment with anthracyclines and vinca alkaloids was assessed. RESULTS Eight of 13 patients responded to treatment with anthracyclines and vinca alkaloids, whereas 5 patients did not respond to treatment. At 10 min, 6 (75.0% ) of the 8 responding patients had a high accumulation of Tc-SESTAMIBI, whereas 4 (80.0% ) of the 5 non-responding patients had a low accumulation of Tc-SESTAMIBI. The overall predictive value was 76.9%. The relationship was not statistically significant (Fisher's test). The difference in the decrease of accumulation of Tc-SESTAMIBI between 10 min and 2 h was not associated with tumor response to treatment in 6 of the responding patients with high accumulation. Two false negative cases and one false positive case were observed, suggesting the presence of another factor contributing to drug sensitivity in tumor response, such as apoptosis-related genes. CONCLUSIONS Assessment of the initial accumulation of Tc-SESTAMIBI can be a predictive marker of tumor response to treatment with anthracyclines and vinca alkaloids in patients with advanced and relapsed breast cancer. Further studies are required to explore other factors involved in the tumor response to treatment with anthracyclines and vinca alkaloids.
Collapse
Affiliation(s)
- Ryungsa Kim
- Department of Surgical Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan.
| | | | | | | |
Collapse
|
13
|
Orlandi L, Binda M, Folini M, Bearzatto A, Villa R, Daidone MG, Zaffaroni N. Ribozyme-mediated inhibition of PKCalpha sensitizes androgen-independent human prostate cancer cells to cisplatin-induced apoptosis. Prostate 2003; 54:133-43. [PMID: 12497586 DOI: 10.1002/pros.10181] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Therapeutic strategies to target the molecular basis of hormone and drug resistance of prostate cancer cells are needed. Since protein kinase Calpha (PKCalpha) is thought to have a role in the development of the androgen-independent phenotype of prostate cancer cells and in apoptosis suppression, the objective of the present study was to test whether specific inhibition of PKCalpha by a hammerhead ribozyme was able to sensitize androgen-independent prostate cancer cells the effects of apoptosis-inducing anticancer drugs. METHODS An active ribozyme (PKCalphaRZ) targeting codon 4 in human PKCalpha mRNA was synthesized by in vitro transcription. A mutant ribozyme (PKCalphamutRZ) was also made by deleting G(12) from the catalytic core of the active ribozyme and used as a control throughout the study. The double-stranded, ribozyme-encoding sequences were then inserted into an expression vector under the control of the cytomegalovirus promoter and delivered to growing prostate cancer cells (DU145 and PC-3) by a DOTAP-mediated transfer. A neomycin resistance gene on the vector was used to select ribozyme-expressing clones. The clones were analyzed for PKCalpha expression, sensitivity to anticancer drugs and ability to undergo drug-induced apoptosis. RESULTS Two DU145-derived cell clones expressing the active ribozyme (DURZ 2 and DURZ 12) and one clone expressing the catalytically inactive ribozyme (DUmutRZ) were selected for the study. DURZ 2 and DURZ 12 were characterized by a markedly (about 40-50%) lower PKCalpha protein level than parental DU145 cells, whereas no reduction in PKCalpha expression was observed in DUmutRZ cells. Results of cytotoxicity experiments indicated that DURZ 2 and DURZ 12 but not DUmutRZ cells were significantly more sensitive than parental DU145 cells to a 1 hr exposure to the mononuclear platinum compounds (cisplatin and oxaliplatin) and showed an increased susceptibility to undergo cisplatin-induced apoptosis. A significantly enhanced apoptotic response to cisplatin was also observed in a PC-3-derived polyclonal cell population endogenously expressing the active ribozyme. CONCLUSIONS Results of the study highlight the importance of PKCalpha in the response of prostate cancer cells to mononuclear platinum compounds and indicate specific inhibition of the enzyme as a potential therapeutic strategy to sensitize androgen-independent prostate cancer cells to these drugs.
Collapse
Affiliation(s)
- Linda Orlandi
- Dipartimento di Oncologia Sperimentale, Unita' Operativa 10, Istituto Nazionale per lo Studio e la Cura dei Tumori, 20133 Milano, Italy
| | | | | | | | | | | | | |
Collapse
|
14
|
Orlandi L, Bertoli G, Abolafio G, Daidone MG, Zaffaroni N. Effects of liposome-entrapped annamycin in human breast cancer cells: interference with cell cycle progression and induction of apoptosis. J Cell Biochem 2001; 81:9-22. [PMID: 11180394 DOI: 10.1002/1097-4644(20010401)81:1<9::aid-jcb1020>3.0.co;2-c] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The effects of liposome-encapsulated annamycin (L-Ann) were investigated in two human breast cancer cell lines, MCF7 and MDA-MB-435. For comparative purposes, doxorubicin (Dx) was used throughout the study. A 4-hour treatment with L-Ann was significantly more active in MDA-MB-435 than in MCF7 cells (IC(50) values of 0.03 and 0.08 microg/ml, respectively), whereas Dx was equally active in the two cell lines (IC(50) 0.12 microg/ml). L-Ann induced an accumulation of cells in G2M phases which was dose-dependent in MDA-MB-435 but not in MCF7 cells. Dx also caused a dose-dependent increase of G2M cell fraction in MDA-MB-435 cells, whereas a G2M cell accumulation was observed only after treatment with the highest Dx concentration in MCF7 cells. G2M phase cell accumulations induced in MCF7 cells by L-Ann or Dx were accompanied by a decrease in cdc2 kinase activity and in cyclin B1 and cdc2 expression. Conversely, in MDA-MB-435 cells exposed to L-Ann or Dx, cdc2 kinase activity, cyclin B1 and cdc2 expression increased in parallel to the increase in the number of cells accumulated in the G2M phase. L-Ann and Dx induced apoptosis in MDA-MB-435 but not in MCF7 cells. In MDA-MB-435 cells exposed to L-Ann or Dx, no change was observed in the expression of bax, but there was a p53-independent increase in p21(waf1) expression. In MCF7 cells, treatment with L-Ann or Dx induced an increase in p53 expression with a consequent transactivation of p21(waf1) and bax. Our results indicate that L-Ann is more cytotoxic than Dx in breast cancer cells and is able to induce apoptosis through p53-independent mechanisms.
Collapse
Affiliation(s)
- L Orlandi
- Dipartimento di Oncologia Sperimentale, Unita' Operativa # 10, Istituto Nazionale per lo Studio e la Cura dei Tumori, 20133 Milano, Italy
| | | | | | | | | |
Collapse
|
15
|
Orlandi L, Colella G, Bearzatto A, Abolafio G, Manzotti C, Daidone MG, Zaffaroni N. Effects of a novel trinuclear platinum complex in cisplatin-sensitive and cisplatin-resistant human ovarian cancer cell lines: interference with cell cycle progression and induction of apoptosis. Eur J Cancer 2001; 37:649-59. [PMID: 11290441 DOI: 10.1016/s0959-8049(00)00445-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
We evaluated the effects of the trinuclear platinum complex, BBR 3464, in a human ovarian carcinoma cell line (OAW42) and in its cisplatin (CDDP)-resistant counterpart (OAW42MER). A 14-fold increased sensitivity to a 1-h BBR 3464 exposure was found in OAW42MER cells compared with their parental cell line. Flow cytometric experiments showed that BBR 3464 was able to induce a persistent block of OAW42 and OAW42MER cells in the G2M phase, whereas CDDP caused an initial accumulation of cells in the S phase followed by an increase in the G2M cell fraction in both cell lines. Exposure to equitoxic (IC(50)) drug concentrations induced programmed cell death in both cell lines. However, the percentage of cells with an apoptotic nuclear morphology was slightly higher after CDDP than BBR 3464 treatment in OAW42 cells, whereas the opposite pattern was observed in OAW42MER cells. Degradation of the nuclear lamin B was detected in OAW42 cells after exposure to each drug. Conversely, in OAW42MER cells lamin B cleavage was only appreciable after BBR 3464 exposure. In OAW42 cells, CDDP and BBR 3464 did not appreciably affect the mitochondrial membrane potential (Deltapsi(mt)), whereas in the OAW42MER cell line a marked Deltapsi(mt) reduction was observed after exposure to BBR 3464, but not to CDDP. The results of the study would suggest that the sensitivity to BBR 3464 observed in the CDDP-resistant OAW42MER cell line might be attributable to the ability of the trinuclear platinum complex to modify DNA in a way which is different from that of CDDP and, as a consequence, to induce different cellular responses to DNA damage such as the triggering of specific apoptotic pathways.
Collapse
Affiliation(s)
- L Orlandi
- Dipartimento di Oncologia Sperimentale, Unita' Operativa # 10, Istituto Nazionale per lo Studio e la Cura dei Tumori, 20133, Milan, Italy
| | | | | | | | | | | | | |
Collapse
|
16
|
Paradiso A, Simone G, Lena MD, Leone B, Vallejo C, Lacava J, Dellapasqua S, Daidone MG, Costa A. Expression of apoptosis-related markers and clinical outcome in patients with advanced colorectal cancer. Br J Cancer 2001; 84:651-8. [PMID: 11237386 PMCID: PMC2363782 DOI: 10.1054/bjoc.2000.1658] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The clinical relevance of bax and bcl-2 protein expression has been investigated in 84 patients with recurrent or metastatic colorectal cancer submitted to a chemotherapy regimen including methotrexate and fluorouracil/leucovorin. Cytoplasmic immunostaining of bax and bcl-2 was present in 65.5% and 38%, respectively, of the tumours. No association was found between bax and bcl-2 or between p53 and bax or bcl-2 protein expression. Moreover, the biomarkers were unrelated to patient and tumour characteristics known to affect the clinical outcome of colorectal cancer patients. In general, the apoptosis-related markers did not appear indicative of short- and long-term clinical response nor of prognosis. Bcl-2-negative lesions were more frequent among patients who reached an objective clinical response, which is in agreement with previously reported data regarding other tumour types. When the interrelationship between p53 and bax expression was examined, a better response rate (40%) was found for patients whose tumours did not express p53 and bax, and a better prognosis (2-year probability of overall survival 75%) for patients with p53-positive and bax-negative tumours. In the present series of patients with advanced colorectal cancer submitted to systemic chemotherapy we did not find a clear association between expression of apoptosis-related markers and clinical outcome, even in the subset of patients in which the apoptotic index as determined by the TUNEL approach was investigated.
Collapse
Affiliation(s)
- A Paradiso
- Clinical Experimental Oncology Laboratory, Oncology Institute, Bari, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|