1
|
Shang Q, Jiang Y, Wan Z, Peng J, Xu Z, Li W, Yang D, Zhao H, Xu X, Zhou Y, Zeng X, Chen Q, Xu H. The clinical implication and translational research of OSCC differentiation. Br J Cancer 2024; 130:660-670. [PMID: 38177661 PMCID: PMC10876927 DOI: 10.1038/s41416-023-02566-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 12/13/2023] [Accepted: 12/19/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND The clinical value and molecular characteristics of tumor differentiation in oral squamous cell carcinoma (OSCC) remain unclear. There is a lack of a related molecular classification prediction system based on pathological images for precision medicine. METHODS Integration of epidemiology, genomics, experiments, and deep learning to clarify the clinical value and molecular characteristics, and develop a novel OSCC molecular classification prediction system. RESULTS Large-scale epidemiology data (n = 118,817) demonstrated OSCC differentiation was a significant prognosis indicator (p < 0.001), and well-differentiated OSCC was more chemo-resistant than poorly differentiated OSCC. These results were confirmed in the TCGA database and in vitro. Furthermore, we found chemo-resistant related pathways and cell cycle-related pathways were up-regulated in well- and poorly differentiated OSCC, respectively. Based on the characteristics of OSCC differentiation, a molecular grade of OSCC was obtained and combined with pathological images to establish a novel prediction system through deep learning, named ShuffleNetV2-based Molecular Grade of OSCC (SMGO). Importantly, our independent multi-center cohort of OSCC (n = 340) confirmed the high accuracy of SMGO. CONCLUSIONS OSCC differentiation was a significant indicator of prognosis and chemotherapy selection. Importantly, SMGO could be an indispensable reference for OSCC differentiation and assist the decision-making of chemotherapy.
Collapse
Affiliation(s)
- Qianhui Shang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, PR China
| | - Yuchen Jiang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, PR China
| | - Zixin Wan
- Department of Pathology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, PR China
| | - Jiakuan Peng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, PR China
| | - Ziang Xu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, PR China
| | - Weiqi Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, PR China
| | - Dan Yang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, PR China
| | - Hang Zhao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, PR China
| | - Xiaoping Xu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, PR China
| | - Yu Zhou
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, PR China
| | - Xin Zeng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, PR China
| | - Qianming Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, PR China.
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, Affiliated Stomatology Hospital, Zhejiang University School of Stomatology, Hangzhou, Zhejiang, 310006, PR China.
| | - Hao Xu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, PR China.
| |
Collapse
|
2
|
Wahdan-Alaswad RS, Edgerton SM, Kim HM, Tan AC, Haugen BR, Liu B, Thor AD. Thyroid hormone enhances estrogen-mediated proliferation and cell cycle regulatory pathways in steroid receptor-positive breast Cancer. Cell Cycle 2023:1-20. [PMID: 37723865 DOI: 10.1080/15384101.2023.2249702] [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: 01/24/2023] [Accepted: 08/15/2023] [Indexed: 09/20/2023] Open
Abstract
Estrogen receptor (ER) α expression and associated signaling is a major driver of over two-thirds of all breast cancers (BC). ER targeting strategies are typically used as a first-line therapy in patients with steroid receptor positive (SR+) disease. Secondary resistance to anti-estrogenic agents may occur with clonal expansion and disease progression. Mechanisms underlying hormone resistance are an expanding field of significant translational importance. Cross-talk with other nuclear hormones, receptors, and signaling pathways, including thyroid hormones (TH) and their receptors (THRs), have been shown to promote endocrine therapy resistance in some studies. We have shown that TH replacement therapy (THRT) was independently and significantly associated with higher rates of relapse and mortality in SR positive (+), node-negative (LN-) BC patients, whereas it showed no association with outcomes in SR negative (-) patients. LN-, SR+ patients receiving THRT and tamoxifen had the worst outcomes, suggesting a pro-carcinogenic interaction that significantly and independently shortened survival and increased mortality. Using in vivo and in vitro models, we previously showed hormonal cross-talk, altered gene signaling, target gene activation, and resistance to tamoxifen in the presence of TH. In this report, we show TH ± E2 ± tamoxifen inhibits cell cycle control signaling, reduces apoptosis, and enhances cell proliferation, tumor growth, tamoxifen resistance, and clonal expansion. Mechanistically these changes involve numerous genes and pathways, including critical cell cycle regulatory proteins and genes identified using various molecular methods. These studies facilitate a greater mechanistic understanding of the biological and molecular impact of TH on SR+ BC.
Collapse
Affiliation(s)
- Reema S Wahdan-Alaswad
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- University of Colorado Cancer Center, Aurora, CO, USA
| | - Susan M Edgerton
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- University of Colorado Cancer Center, Aurora, CO, USA
| | - Hyun Min Kim
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Aik Choon Tan
- Department of Oncological Sciences and Biomedical Informatics, Huntsman Cancer Institute, Salt Lake City, Utah, USA
| | - Bryan R Haugen
- University of Colorado Cancer Center, Aurora, CO, USA
- Division of Endocrinology, Metabolism, & Diabetes, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, USA
| | - Bolin Liu
- Department of Genetics, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University (LSU) Health Sciences Center, New Orleans, LA, USA
| | - Ann D Thor
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- University of Colorado Cancer Center, Aurora, CO, USA
| |
Collapse
|
3
|
Olson AT, Kang Y, Ladha AM, Zhu S, Lim CB, Nabet B, Lagunoff M, Gujral TS, Geballe AP. Polypharmacology-based kinome screen identifies new regulators of KSHV reactivation. PLoS Pathog 2023; 19:e1011169. [PMID: 37669313 PMCID: PMC10503724 DOI: 10.1371/journal.ppat.1011169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 09/15/2023] [Accepted: 08/16/2023] [Indexed: 09/07/2023] Open
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) causes several human diseases including Kaposi's sarcoma (KS), a leading cause of cancer in Africa and in patients with AIDS. KS tumor cells harbor KSHV predominantly in a latent form, while typically <5% contain lytic replicating virus. Because both latent and lytic stages likely contribute to cancer initiation and progression, continued dissection of host regulators of this biological switch will provide insights into fundamental pathways controlling the KSHV life cycle and related disease pathogenesis. Several cellular protein kinases have been reported to promote or restrict KSHV reactivation, but our knowledge of these signaling mediators and pathways is incomplete. We employed a polypharmacology-based kinome screen to identify specific kinases that regulate KSHV reactivation. Those identified by the screen and validated by knockdown experiments included several kinases that enhance lytic reactivation: ERBB2 (HER2 or neu), ERBB3 (HER3), ERBB4 (HER4), MKNK2 (MNK2), ITK, TEC, and DSTYK (RIPK5). Conversely, ERBB1 (EGFR1 or HER1), MKNK1 (MNK1) and FRK (PTK5) were found to promote the maintenance of latency. Mechanistic characterization of ERBB2 pro-lytic functions revealed a signaling connection between ERBB2 and the activation of CREB1, a transcription factor that drives KSHV lytic gene expression. These studies provided a proof-of-principle application of a polypharmacology-based kinome screen for the study of KSHV reactivation and enabled the discovery of both kinase inhibitors and specific kinases that regulate the KSHV latent-to-lytic replication switch.
Collapse
Affiliation(s)
- Annabel T. Olson
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Yuqi Kang
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Anushka M. Ladha
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
| | - Songli Zhu
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Chuan Bian Lim
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Behnam Nabet
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Michael Lagunoff
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
| | - Taranjit S. Gujral
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
- Department of Pharmacology, University of Washington, Seattle, Washington, United States of America
| | - Adam P. Geballe
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
- Department of Microbiology, University of Washington, Seattle, Washington, United States of America
- Division of Clinical Research, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
4
|
Hamm C, Fifield BA, Kay A, Kulkarni S, Gupta R, Mathews J, Ferraiuolo RM, Al-Wahsh H, Mailloux E, Hussein A, Porter LA. A prospective phase II clinical trial identifying the optimal regimen for carboplatin plus standard backbone of anthracycline and taxane-based chemotherapy in triple negative breast cancer. Med Oncol 2022; 39:49. [PMID: 35103812 DOI: 10.1007/s12032-021-01637-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 12/23/2021] [Indexed: 12/31/2022]
Abstract
Addition of platinums to combination chemotherapy for triple negative breast cancer (TNBC) has shown efficacy and is increasingly accepted in the clinic, yet optimal delivery is unknown. A prospective clinical trial with TNBC patients was conducted to determine the optimal chemotherapy regimen to deliver carboplatin with standard dose dense ACT. Tissue microarray was conducted to isolate markers indicative of response to treatment. 90 TNBC patients were enrolled onto our trial. The most successful version placed the carboplatin on the second and final paclitaxel treatment with liberal hematological parameters. Our final regimen had the lowest grade 3 or 4 toxicities, no delays, no dose reductions of carboplatin, and 32% reduction in paclitaxel doses. Stage I (AJCC7) patients did well with carboplatin-based chemotherapy with zero relapse rate. Reduction in protein levels of androgen receptor and PD-L1 were found to be potential indicators of patient relapse. We have optimized a protocol for the addition of carboplatin to standard of care chemotherapy in TNBC patients. Early data indicates reduced protein levels of androgen receptor and PD-L1 as indicators of response to treatment.Trial registration This trial was registered at Canadian Cancer Trials. http://www.canadiancancertrials.ca/.
Collapse
Affiliation(s)
- Caroline Hamm
- University of Windsor, Windsor, ON, N9B 3P4, Canada.
- Western University, Windsor, ON, N9B 3P4, Canada.
- Windsor Cancer Research Group, Windsor, ON, N9B 3P4, Canada.
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, 48201, USA.
- WE-SPARK Health Institute, Windsor, ON, N9B 3P4, Canada.
| | - Bre-Anne Fifield
- University of Windsor, Windsor, ON, N9B 3P4, Canada
- Windsor Cancer Research Group, Windsor, ON, N9B 3P4, Canada
| | - Amin Kay
- University of Windsor, Windsor, ON, N9B 3P4, Canada
- Western University, Windsor, ON, N9B 3P4, Canada
| | - Swati Kulkarni
- University of Windsor, Windsor, ON, N9B 3P4, Canada
- Western University, Windsor, ON, N9B 3P4, Canada
- Windsor Cancer Research Group, Windsor, ON, N9B 3P4, Canada
- WE-SPARK Health Institute, Windsor, ON, N9B 3P4, Canada
| | - Rasna Gupta
- University of Windsor, Windsor, ON, N9B 3P4, Canada
- Western University, Windsor, ON, N9B 3P4, Canada
- Windsor Cancer Research Group, Windsor, ON, N9B 3P4, Canada
| | - John Mathews
- University of Windsor, Windsor, ON, N9B 3P4, Canada
- Western University, Windsor, ON, N9B 3P4, Canada
| | - Rosa-Maria Ferraiuolo
- Windsor Cancer Research Group, Windsor, ON, N9B 3P4, Canada
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | | | - Emily Mailloux
- University of Windsor, Windsor, ON, N9B 3P4, Canada
- Windsor Cancer Research Group, Windsor, ON, N9B 3P4, Canada
| | | | - Lisa A Porter
- University of Windsor, Windsor, ON, N9B 3P4, Canada.
- Windsor Cancer Research Group, Windsor, ON, N9B 3P4, Canada.
- WE-SPARK Health Institute, Windsor, ON, N9B 3P4, Canada.
| |
Collapse
|
5
|
Tanriover G, Dilmac S, Aytac G, Farooqi AA, Sindel M. Effects of melatonin and doxorubicin on primary tumor and metastasis in breast cancer model. Anticancer Agents Med Chem 2021; 22:1970-1983. [PMID: 34961467 DOI: 10.2174/1871520621666211213094258] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 08/11/2021] [Accepted: 11/01/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Melatonin exerts oncostatic effects on breast cancer via immunomodulation and anti-oxidation. Doxorubicin is an effective chemotherapeutic agent, but parallel studies also provide ample evidence of an off-target effect of Doxorubicin in breast cancer patients. OBJECTIVE Combinatorial use of doxorubicin and melatonin has not been comprehensively analyzed in breast cancer models. We hypothesized that the anti-oxidative, anti-proliferative and anti-inflammatory effects of melatonin could ameliorate the off-target effects of doxorubicin in breast cancer patients and enhance the anti-tumoral effects of doxorubicin. The goal of the study is to test this hypothesis in cancer cell lines and xenografted mice. METHODS The effects of Melatonin and doxorubicin on the cell viability were evaluated in 4T1-Brain Metastatic Tumor (4TBM). Furthermore, the effects of melatonin and doxorubicin on the primary tumors and systemic metastasis were evaluated in the xenografted mice. Lung and liver tissues were removed and metastasis analyses were performed. The levels of p65, phospho-STAT3, CD11b+, GR1+, Ki67, and cleaved caspase-3 proteins were determined with immunohistochemistry and western blot analysis. We examined the effects of melatonin and Melatonin+Doxorubicin combination therapy on 4TBM cells. RESULTS Our results showed that doxorubicin inhibited the proliferation of metastatic breast cancer cells while melatonin did not affect cells. Tumor growth and metastasis were markedly suppressed in melatonin alone and combination with doxorubicin. The expression of CD11b+ and GR1+ proteins which are indicators of myeloid-derived suppressor cells (MDSCs) were noted to be reduced in both primary tumor and metastatic tissues in melatonin and doxorubicin groups. CONCLUSION The combination of melatonin with doxorubicin reduced primary tumor growth and distant metastasis. Based on these results, melatonin is a promising candidate for combinatory use with conventional chemotherapeutics for breast cancer treatment.
Collapse
Affiliation(s)
- Gamze Tanriover
- Akdeniz University, Faculty of Medicine Department of Histology and Embryology, Antalya. Turkey
| | - Sayra Dilmac
- Akdeniz University, Faculty of Medicine Department of Histology and Embryology, Antalya. Turkey
| | - Gunes Aytac
- TOBB University of Economics & Technology, Faculty of Medicine, Department of Anatomy, Ankara. Turkey
| | | | - Muzaffer Sindel
- Akdeniz University, Faculty of Medicine Department of Anatomy, Antalya. Turkey
| |
Collapse
|
6
|
Wang X, Kokabee L, Kokabee M, Conklin DS. Bruton's Tyrosine Kinase and Its Isoforms in Cancer. Front Cell Dev Biol 2021; 9:668996. [PMID: 34307353 PMCID: PMC8297165 DOI: 10.3389/fcell.2021.668996] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 05/25/2021] [Indexed: 01/04/2023] Open
Abstract
Bruton’s tyrosine kinase (BTK) is a soluble tyrosine kinase with central roles in the development, maturation, and signaling of B cells. BTK has been found to regulate cell proliferation, survival, and migration in various B-cell malignancies. Targeting BTK with recently developed BTK inhibitors has been approved by the Food and Drug Administration (FDA) for the treatment of several hematological malignancies and has transformed the treatment of several B-cell malignancies. The roles that BTK plays in B cells have been appreciated for some time. Recent studies have established that BTK is expressed and plays pro-tumorigenic roles in several epithelial cancers. In this review, we focus on novel isoforms of the BTK protein expressed in epithelial cancers. We review recent work on the expression, function, and signaling of these isoforms and their value as potential therapeutic targets in epithelial tumors.
Collapse
Affiliation(s)
- Xianhui Wang
- Department of Biomedical Sciences, Cancer Research Center, State University of New York, Rensselaer, NY, United States
| | - Leila Kokabee
- Department of Biomedical Sciences, Cancer Research Center, State University of New York, Rensselaer, NY, United States
| | - Mostafa Kokabee
- Department of Biomedical Sciences, Cancer Research Center, State University of New York, Rensselaer, NY, United States
| | - Douglas S Conklin
- Department of Biomedical Sciences, Cancer Research Center, State University of New York, Rensselaer, NY, United States
| |
Collapse
|
7
|
Korobeinikova E, Ugenskiene R, Insodaite R, Rudzianskas V, Gudaitiene J, Juozaityte E. The role of functional polymorphisms in oxidative stress-related genes on early-stage breast cancer survival. Int J Biol Markers 2021; 36:14-21. [PMID: 33885357 DOI: 10.1177/17246008211011177] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Genetic variations in oxidative stress-related genes may alter the coded protein level and impact the pathogenesis of breast cancer. METHODS The current study investigated the associations of functional single nucleotide polymorphisms in the NFE2L2, HMOX1, P21, TXNRD2, and ATF3 genes with the early-stage breast cancer clinicopathological characteristics and disease-free survival, metastasis-free survival, and overall survival. A total of 202 Eastern European (Lithuanian) women with primary I-II stage breast cancer were involved. Genotyping of the single nucleotide polymorphisms was performed using TaqMan single nucleotide polymorphisms genotyping assays. RESULTS The CA+AA genotypes of P21 rs1801270 were significantly less frequent in patients with lymph node metastasis and larger tumor size (P=0.041 and P=0.022, respectively). The TT genotype in ATF3 rs3125289 had significantly lower risk of estrogen receptor (ER), progesterone receptor (PR) negative, and human epidermal growth factor receptor 2 (HER2) positive status (P=0.023, P=0.046, and P=0.040, respectively). In both, univariate and multivariate Cox analysis, TXNRD2 rs1139793 GG genotype vs. GA+AA was a negative prognostic factor for disease-free survival (multivariate hazard ratio (HR) 2.248; P=0.025) and overall survival (multivariate HR 2.248; P=0.029). The ATF3 rs11119982 CC genotype in the genotype model was a negative prognostic factor for disease-free survival (multivariate HR 5.878; P=0.006), metastasis-free survival (multivariate HR 4.759; P=0.018), and overall survival (multivariate HR 3.280; P=0.048). CONCLUSION Our findings suggest that P21 rs1801270 is associated with lymph node metastasis and larger tumor size, and ATF3 rs3125289 is associated with ER, PR, and HER2 status. Two potential, novel, early-stage breast cancer survival biomarkers, TXNRD2 rs1139793 and ATF3 rs11119982, were detected. Further investigations are needed to confirm the results of the current study.
Collapse
Affiliation(s)
| | | | - Ruta Insodaite
- Lithuanian University of Health Sciences, Kaunas, Lithuania
| | | | | | | |
Collapse
|
8
|
Lai TC, Fang CY, Jan YH, Hsieh HL, Yang YF, Liu CY, Chang PMH, Hsiao M. Kinase shRNA screening reveals that TAOK3 enhances microtubule-targeted drug resistance of breast cancer cells via the NF-κB signaling pathway. Cell Commun Signal 2020; 18:164. [PMID: 33087151 PMCID: PMC7579951 DOI: 10.1186/s12964-020-00600-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 05/25/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Chemotherapy is currently one of the most effective treatments for advanced breast cancer. Anti-microtubule agents, including taxanes, eribulin and vinca-alkaloids are one of the primary major anti-breast cancer chemotherapies; however, chemoresistance remains a problem that is difficult to solve. We aimed to discover novel candidate protein targets to combat chemoresistance in breast cancer. METHODS A lentiviral shRNA-based high-throughput screening platform was designed and developed to screen the global kinome to find new therapeutic targets in paclitaxel-resistant breast cancer cells. The phenotypes were confirmed with alternative expression in vitro and in vivo. Molecular mechanisms were investigated using global phosphoprotein arrays and expression microarrays. Global microarray analysis was performed to determine TAOK3 and genes that induced paclitaxel resistance. RESULTS A serine/threonine kinase gene, TAOK3, was identified from 724 screened kinase genes. TAOK3 shRNA exhibited the most significant reduction in IC50 values in response to paclitaxel treatment. Ectopic downregulation of TAOK3 resulted in paclitaxel-resistant breast cancer cells sensitize to paclitaxel treatment in vitro and in vivo. The expression of TAOK3 also was correlated to sensitivity to two other anti-microtubule drugs, eribulin and vinorelbine. Our TAOK3-modulated microarray analysis indicated that NF-κB signaling played a major upstream regulation role. TAOK3 inhibitor, CP43, and shRNA of NF-κB both reduced the paclitaxel resistance in TAOK3 overexpressed cells. In clinical microarray databases, high TAOK3 expressed breast cancer patients had poorer prognoses after adjuvant chemotherapy. CONCLUSIONS Here we identified TAOK3 overexpression increased anti-microtubule drug resistance through upregulation of NF-κB signaling, which reduced cell death in breast cancer. Therefore, inhibition of the interaction between TAOK3 and NF-κB signaling may have therapeutic implications for breast cancer patients treated with anti-microtubule drugs. Video abstract.
Collapse
Affiliation(s)
- Tsung-Ching Lai
- Division of Pulmonary Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, 116, Taiwan
- Pulmonary Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, 116, Taiwan
- Genomics Research Center, Academia Sinica, Taipei, 115, Taiwan
| | - Chih-Yeu Fang
- Genomics Research Center, Academia Sinica, Taipei, 115, Taiwan
| | - Yi-Hua Jan
- Genomics Research Center, Academia Sinica, Taipei, 115, Taiwan
| | | | - Yi-Fang Yang
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, 81362, Taiwan
| | - Chun-Yu Liu
- Department of Oncology, Taipei Veterans General Hospital, Taipei, 11217, Taiwan
- Division of Transfusion Medicine, Department of Medicine, Taipei Veterans General Hospital, Taipei, 11217, Taiwan
- Comprehensive Breast Health Center, Taipei Veterans General Hospital, Taipei, 11217, Taiwan
| | - Peter Mu-Hsin Chang
- Department of Oncology, Taipei Veterans General Hospital, Taipei, 11217, Taiwan.
- Faculty of Medicine, National Yang Ming University, Taipei, 112, Taiwan.
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei, 115, Taiwan.
- Faculty of Medicine, National Yang Ming University, Taipei, 112, Taiwan.
- Department of Biochemistry, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
- The Ph.D.Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, 11031, Taiwan.
| |
Collapse
|
9
|
Jiang L, Ren L, Chen H, Pan J, Zhang Z, Kuang X, Chen X, Bao W, Lin C, Zhou Z, Huang D, Yang J, Huang H, Wang L, Hou N, Song L. NCAPG confers trastuzumab resistance via activating SRC/STAT3 signaling pathway in HER2-positive breast cancer. Cell Death Dis 2020; 11:547. [PMID: 32683421 PMCID: PMC7368860 DOI: 10.1038/s41419-020-02753-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 06/30/2020] [Accepted: 07/03/2020] [Indexed: 12/24/2022]
Abstract
HER2+ breast cancer (BC) is characterized by rapid growth, early recurrence, early metastasis, and chemoresistance. Trastuzumab is the most effective treatment for HER2+ BC and effectively reduces the risk of recurrence and death of patients. Resistance to trastuzumab results in cancer recurrence and metastasis, leading to poor prognosis of HER2+ BC. In the present study, we found that non-structural maintenance of chromosome condensin 1 complex subunit G (NCAPG) expression was highly upregulated in trastuzumab-resistant HER2+ BC. Ectopic NCAPG was positively correlated with tumor relapse and shorter survival in HER2+ BC patients. Moreover, overexpression of NCAPG promoted, while silencing of NCAPG reduced, the proliferative and anti-apoptotic capacity of HER2+ BC cells both in vitro and in vivo, indicating NCAPG reduces the sensitivity of HER2+ BC cells to trastuzumab and may confer trastuzumab resistance. Furthermore, our results suggest that NCAPG triggers a series of biological cascades by phosphorylating SRC and enhancing nuclear localization and activation of STAT3. To summarize, our study explores a crucial role for NCAPG in trastuzumab resistance and its underlying mechanisms in HER2+ BC, and suggests that NCAPG could be both a potential prognostic marker as well as a therapeutic target to effectively overcome trastuzumab resistance.
Collapse
Affiliation(s)
- Lili Jiang
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, 510095, Guangzhou, China. .,Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Science, Guangzhou Medical University, 511436, Guangzhou, China.
| | - Liangliang Ren
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, 510080, Guangzhou, China
| | - Han Chen
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, 510095, Guangzhou, China.,Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Science, Guangzhou Medical University, 511436, Guangzhou, China
| | - Jinyuan Pan
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, 510095, Guangzhou, China.,Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Science, Guangzhou Medical University, 511436, Guangzhou, China
| | - Zhuojun Zhang
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, 510095, Guangzhou, China.,Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Science, Guangzhou Medical University, 511436, Guangzhou, China
| | - Xiangqin Kuang
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, 510095, Guangzhou, China.,Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Science, Guangzhou Medical University, 511436, Guangzhou, China
| | - Xuhong Chen
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, 510095, Guangzhou, China.,Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Science, Guangzhou Medical University, 511436, Guangzhou, China
| | - Wenhao Bao
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, 510095, Guangzhou, China.,Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Science, Guangzhou Medical University, 511436, Guangzhou, China
| | - Chun Lin
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, 510095, Guangzhou, China.,Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Science, Guangzhou Medical University, 511436, Guangzhou, China
| | - Zhongqiu Zhou
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, 510095, Guangzhou, China.,Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Science, Guangzhou Medical University, 511436, Guangzhou, China
| | - Danping Huang
- Department of Ultrasonography, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 510623, Guangzhou, China
| | - Jianan Yang
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, 510095, Guangzhou, China.,Department of Urologic Oncosurgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, 510095, Guangzhou, China
| | - Hongbiao Huang
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, 510095, Guangzhou, China.,Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Science, Guangzhou Medical University, 511436, Guangzhou, China
| | - Lan Wang
- Department of Pathogen Biology and Immunology, School of Basic Courses, Guangdong Pharmaceutical University, 510006, Guangzhou, China
| | - Ning Hou
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, 511436, Guangzhou, China
| | - Libing Song
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 510060, Guangzhou, China.
| |
Collapse
|
10
|
Hwang S, Seong H, Ryu J, Jeong JY, Kang TS, Nam KY, Seo SW, Kim SJ, Kang SS, Han YS. Phosphorylation of STAT3 and ERBB2 mediates hypoxia‑induced VEGF release in ARPE‑19 cells. Mol Med Rep 2020; 22:2733-2740. [PMID: 32945388 PMCID: PMC7453508 DOI: 10.3892/mmr.2020.11344] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 06/15/2020] [Indexed: 12/14/2022] Open
Abstract
Neovascularization in the retina can cause loss of vision. Vascular endothelial growth factor (VEGF) serves an important role in the pathogenesis of retinal vascular diseases. Hypoxia is a notable cause of VEGF release and both STAT3 and ERBB2 are known to be associated with VEGF. In addition, STAT3 and ERBB2 interact with each other. In the present study, it was hypothesized that signal transducer and activator of transcription 3 (STAT3) and erbB-2 receptor tyrosine kinase 2 (ERBB2) may be involved in the regulation of hypoxia-induced VEGF in the retina. Cells of the retinal pigment epithelium (RPE) are an important source of VEGF. Therefore, the RPE-derived human cell line ARPE-19 was exposed to hypoxia. Hypoxia-induced phosphorylation of STAT3 and ERBB2 in ARPE-19 cells was decreased by AG490, an inhibitor of Janus kinase 2, as were hypoxia-induced VEGF release and tube formation in human umbilical vein endothelial cells. Thus, phosphorylation of ERBB2 and STAT3 regulates hypoxia-induced VEGF release in ARPE-19 cells. The results of the present study suggested that inhibition of ERBB2 and STAT3-mediated pathways under hypoxia may represent a new strategy for treating retinal vascular disease.
Collapse
Affiliation(s)
- Soohyun Hwang
- Department of Anatomy and Convergence Medical Science, College of Medicine, Gyeongsang National University, Jinju, South Gyeongsang 52727, Republic of Korea
| | - Hyemin Seong
- Department of Anatomy and Convergence Medical Science, College of Medicine, Gyeongsang National University, Jinju, South Gyeongsang 52727, Republic of Korea
| | - Jinhyun Ryu
- Department of Anatomy and Convergence Medical Science, College of Medicine, Gyeongsang National University, Jinju, South Gyeongsang 52727, Republic of Korea
| | - Joo Yeon Jeong
- Department of Anatomy and Convergence Medical Science, College of Medicine, Gyeongsang National University, Jinju, South Gyeongsang 52727, Republic of Korea
| | - Tae Seen Kang
- Department of Ophthalmology, Gyeongsang National University Changwon Hospital, Changwon, Gyeongsangnam‑do 51472, Republic of Korea
| | - Ki Yup Nam
- Department of Ophthalmology, Gyeongsang National University Changwon Hospital, Changwon, Gyeongsangnam‑do 51472, Republic of Korea
| | - Seong Wook Seo
- Department of Ophthalmology, College of Medicine, Gyeongsang National University, Jinju, South Gyeongsang 52727, Republic of Korea
| | - Seong Jae Kim
- Department of Ophthalmology, College of Medicine, Gyeongsang National University, Jinju, South Gyeongsang 52727, Republic of Korea
| | - Sang Soo Kang
- Department of Anatomy and Convergence Medical Science, College of Medicine, Gyeongsang National University, Jinju, South Gyeongsang 52727, Republic of Korea
| | - Yong Seop Han
- Department of Ophthalmology, Gyeongsang National University Changwon Hospital, Changwon, Gyeongsangnam‑do 51472, Republic of Korea
| |
Collapse
|
11
|
Hermawan A, Putri H, Utomo RY. Comprehensive bioinformatics study reveals targets and molecular mechanism of hesperetin in overcoming breast cancer chemoresistance. Mol Divers 2019; 24:933-947. [PMID: 31659695 DOI: 10.1007/s11030-019-10003-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 10/03/2019] [Indexed: 02/06/2023]
Abstract
The effectiveness of chemotherapy in breast cancer treatment can be increased using a combinatorial agent. Hesperetin has been reported to increase the sensitivity of doxorubicin in breast cancer cells; however, the underlying molecular mechanism remains unclear. This present study was conducted to identify the potential target and molecular mechanism of hesperetin in circumventing breast cancer chemoresistance using a bioinformatics approach. Microarray data obtained after hesperetin treatment in the NCI-60 cell line panel collection were retrieved from the COMPARE public library. These data were then compared with the list of the regulatory genes of breast cancer resistance obtained from PubMed and further analyzed for gene ontology and KEGG pathway enrichment, as well as protein-protein interaction network. A Venn diagram of COMPARE microarray data and the gene list from PubMed generated 56 genes (potential therapeutic target genes/PTTGs). These PTTGs participate in the biological process of the JAK-STAT cascade and are located in the nucleus, exert a molecular function in protein serine/threonine kinase activity, and regulate the erbB signaling pathway. Drug association analysis demonstrated that both hesperetin and the erbB receptor inhibitors, i.e., monoclonal antibody and tyrosine kinase inhibitor, target the same mRNA expression. Furthermore, results of the molecular docking study revealed that hesperetin is a promising inhibitor that targets ABL1, DNMT3B, and MLH1 due to the similarity of binding properties with its native ligand. In conclusion, the possible pathways and the regulatory genes identified in this study may offer new insights into the mechanism by which hesperetin overcomes breast cancer chemoresistance. A combinatorial therapy with hesperetin targeting ABL1, DNMT3B, and MLH1 may be effective in circumventing chemoresistance in breast cancer.
Collapse
Affiliation(s)
- Adam Hermawan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Universitas Gadjah Mada Sekip Utara II, Yogyakarta, 55281, Indonesia.
| | - Herwandhani Putri
- Cancer Chemoprevention Research Center, Faculty of Pharmacy, Universitas Gadjah Mada Sekip Utara II, Yogyakarta, 55281, Indonesia
| | - Rohmad Yudi Utomo
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Universitas Gadjah Mada Sekip Utara II, Yogyakarta, 55281, Indonesia
| |
Collapse
|
12
|
Xiang S, Dauchy RT, Hoffman AE, Pointer D, Frasch T, Blask DE, Hill SM. Epigenetic inhibition of the tumor suppressor ARHI by light at night-induced circadian melatonin disruption mediates STAT3-driven paclitaxel resistance in breast cancer. J Pineal Res 2019; 67:e12586. [PMID: 31077613 PMCID: PMC6750268 DOI: 10.1111/jpi.12586] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 01/23/2019] [Accepted: 01/25/2019] [Indexed: 12/20/2022]
Abstract
Disruption of circadian time structure and suppression of circadian nocturnal melatonin (MLT) production by exposure to dim light at night (dLAN), as occurs with night shift work and/or disturbed sleep-wake cycles, is associated with a significantly increased risk of breast cancer and resistance to tamoxifen and doxorubicin. Melatonin inhibition of human breast cancer chemoresistance involves mechanisms including suppression of tumor metabolism and inhibition of kinases and transcription factors which are often activated in drug-resistant breast cancer. Signal transducer and activator of transcription 3 (STAT3), frequently overexpressed and activated in paclitaxel (PTX)-resistant breast cancer, promotes the expression of DNA methyltransferase one (DNMT1) to epigenetically suppress the transcription of tumor suppressor Aplasia Ras homolog one (ARHI) which can sequester STAT3 in the cytoplasm to block PTX resistance. We demonstrate that breast tumor xenografts in rats exposed to dLAN and circadian MLT disrupted express elevated levels of phosphorylated and acetylated STAT3, increased DNMT1, but reduced sirtuin 1 (SIRT1) and ARHI. Furthermore, MLT and/or SIRT1 administration blocked/reversed interleukin 6 (IL-6)-induced acetylation of STAT3 and its methylation of ARH1 to increase ARH1 mRNA expression in MCF-7 breast cancer cells. Finally, analyses of the I-SPY 1 trial demonstrate that elevated MT1 receptor expression is significantly correlated with pathologic complete response following neo-adjuvant therapy in breast cancer patients. This is the first study to demonstrate circadian disruption of MLT by dLAN driving intrinsic resistance to PTX via epigenetic mechanisms increasing STAT3 expression and that MLT administration can reestablish sensitivity of breast tumors to PTX and drive tumor regression.
Collapse
Affiliation(s)
- Shulin Xiang
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana
- Tulane Center for Circadian Biology, Tulane University School of Medicine, New Orleans, Louisiana
- Tulane Cancer Center and Louisiana Cancer Research Consortium, New Orleans, Louisiana
- Tulane Circadian Cancer Biology Group, New Orleans, Louisiana
| | - Robert T Dauchy
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana
- Tulane Center for Circadian Biology, Tulane University School of Medicine, New Orleans, Louisiana
- Tulane Cancer Center and Louisiana Cancer Research Consortium, New Orleans, Louisiana
- Tulane Circadian Cancer Biology Group, New Orleans, Louisiana
| | - Aaron E Hoffman
- Tulane Center for Circadian Biology, Tulane University School of Medicine, New Orleans, Louisiana
- Tulane Cancer Center and Louisiana Cancer Research Consortium, New Orleans, Louisiana
- Tulane Circadian Cancer Biology Group, New Orleans, Louisiana
- Department of Epidemiology, Tulane School of Public Health, New Orleans, Louisiana
| | - David Pointer
- Department of Surgery, Tulane University School of Medicine, New Orleans, Louisiana
| | - Tripp Frasch
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana
- Tulane Center for Circadian Biology, Tulane University School of Medicine, New Orleans, Louisiana
| | - David E Blask
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana
- Tulane Center for Circadian Biology, Tulane University School of Medicine, New Orleans, Louisiana
- Tulane Cancer Center and Louisiana Cancer Research Consortium, New Orleans, Louisiana
- Tulane Circadian Cancer Biology Group, New Orleans, Louisiana
| | - Steven M Hill
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana
- Tulane Center for Circadian Biology, Tulane University School of Medicine, New Orleans, Louisiana
- Tulane Cancer Center and Louisiana Cancer Research Consortium, New Orleans, Louisiana
- Tulane Circadian Cancer Biology Group, New Orleans, Louisiana
| |
Collapse
|
13
|
Manu KA, Cao PHA, Chai TF, Casey PJ, Wang M. p21cip1/waf1 Coordinate Autophagy, Proliferation and Apoptosis in Response to Metabolic Stress. Cancers (Basel) 2019; 11:cancers11081112. [PMID: 31382612 PMCID: PMC6721591 DOI: 10.3390/cancers11081112] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 07/25/2019] [Accepted: 07/30/2019] [Indexed: 12/12/2022] Open
Abstract
Cancer cells possess metabolic properties that are different from benign cells. These unique characteristics have become attractive targets that are being actively investigated for cancer therapy. p21cip1/waf1, also known as Cyclin-Dependent Kinase inhibitor 1A, is encoded by the CDKN1A gene. It is a major p53 target gene involved in cell cycle progression that has been extensively evaluated. To date, p21 has been reported to regulate various cell functions, both dependent and independent of p53. Besides regulating the cell cycle, p21 also modulates apoptosis, induces senescence, and maintains cellular quiescence in response to various stimuli. p21 transcription is induced in response to stresses, including those from oxidative and chemotherapeutic treatment. A recent study has shown that in response to metabolic stresses such as nutrient and energy depletion, p21 expression is induced to regulate various cell functions. Despite the biological significance, the mechanism of p21 regulation in cancer adaptation to metabolic stress is underexplored and thus represents an exciting field. This review focuses on the recent development of p21 regulation in response to metabolic stress and its impact in inducing cell cycle arrest and death in cancer cells.
Collapse
Affiliation(s)
- Kanjoormana Aryan Manu
- Program in Cancer and Stem Cell Biology, Duke-NUS Graduate Medical School, Singapore 169857, Singapore
| | - Pham Hong Anh Cao
- Program in Cancer and Stem Cell Biology, Duke-NUS Graduate Medical School, Singapore 169857, Singapore
| | - Tin Fan Chai
- Program in Cancer and Stem Cell Biology, Duke-NUS Graduate Medical School, Singapore 169857, Singapore
| | - Patrick J Casey
- Program in Cancer and Stem Cell Biology, Duke-NUS Graduate Medical School, Singapore 169857, Singapore
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Mei Wang
- Program in Cancer and Stem Cell Biology, Duke-NUS Graduate Medical School, Singapore 169857, Singapore.
- Department of Biochemistry, National University of Singapore, Singapore 117596, Singapore.
| |
Collapse
|
14
|
Wang P, Zhang Z, Ma Y, Lu J, Zhao H, Wang S, Tan J, Li B. Prognostic values of GMPS, PR, CD40, and p21 in ovarian cancer. PeerJ 2019; 7:e6301. [PMID: 30701134 PMCID: PMC6348951 DOI: 10.7717/peerj.6301] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 12/14/2018] [Indexed: 12/11/2022] Open
Abstract
Early detection and prediction of prognosis and treatment responses are all the keys in improving survival of ovarian cancer patients. This study profiled an ovarian cancer progression model to identify prognostic biomarkers for ovarian cancer patients. Mouse ovarian surface epithelial cells (MOSECs) can undergo spontaneous malignant transformation in vitro cell culture. These were used as a model of ovarian cancer progression for alterations in gene expression and signaling detected using the Illumina HiSeq2000 Next-Generation Sequencing platform and bioinformatical analyses. The differential expression of four selected genes was identified using the gene expression profiling interaction analysis (http://gepia.cancer-pku.cn/) and then associated with survival in ovarian cancer patients using the Cancer Genome Atlas dataset and the online Kaplan–Meier Plotter (http://www.kmplot.com) data. The data showed 263 aberrantly expressed genes, including 182 up-regulated and 81 down-regulated genes between the early and late stages of tumor progression in MOSECs. The bioinformatic data revealed four genes (i.e., guanosine 5′-monophosphate synthase (GMPS), progesterone receptor (PR), CD40, and p21 (cyclin-dependent kinase inhibitor 1A)) to play an important role in ovarian cancer progression. Furthermore, the Cancer Genome Atlas dataset validated the differential expression of these four genes, which were associated with prognosis in ovarian cancer patients. In conclusion, this study profiled differentially expressed genes using the ovarian cancer progression model and identified four (i.e., GMPS, PR, CD40, and p21) as prognostic markers for ovarian cancer patients. Future studies of prospective patients could further verify the clinical usefulness of this four-gene signature.
Collapse
Affiliation(s)
- Ping Wang
- Fujian Key Laboratory of Transplant Biology, Fuzhou General Hospital, Fuzhou, Fujian, China
| | - Zengli Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, Suzhou, Jiangsu, China
| | - Yujie Ma
- Fujian Key Laboratory of Transplant Biology, Fuzhou General Hospital, Fuzhou, Fujian, China
| | - Jun Lu
- Fujian Key Laboratory of Transplant Biology, Fuzhou General Hospital, Fuzhou, Fujian, China
| | - Hu Zhao
- Fujian Key Laboratory of Transplant Biology, Fuzhou General Hospital, Fuzhou, Fujian, China
| | - Shuiliang Wang
- Fujian Key Laboratory of Transplant Biology, Fuzhou General Hospital, Fuzhou, Fujian, China
| | - Jianming Tan
- Fujian Key Laboratory of Transplant Biology, Fuzhou General Hospital, Fuzhou, Fujian, China.,Fujian Hongyi Health Institute, Fuzhou, Fujian, China
| | - Bingyan Li
- Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
15
|
Wang X, Goldstein D, Crowe PJ, Yang JL. Antitumour effects and mechanisms of action of the panHER inhibitor, dacomitinib, alone and in combination with the STAT3 inhibitor, S3I-201, in human sarcoma cell lines. Int J Oncol 2018; 52:2143-2154. [PMID: 29620166 DOI: 10.3892/ijo.2018.4337] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 03/12/2018] [Indexed: 11/06/2022] Open
Abstract
The 5-year survival rate for metastatic sarcoma is 16%. Although the phosphorylated human epidermal growth factor receptor (pEGFR/HER1) has been shown to be an independent predictor of overall survival in patients with sarcoma, we have previously demonstrated that sarcoma cell lines exhibit resistance, despite gefitinib blocking p-EGFR and signal transducers in EGFR downstream pathways. Gefitinib failed to decrease the ratio of phosphorylated (p-)signal transducer and activator of transcription (STAT3)/p-STAT1, suggesting that relative STAT3 abundance and activation may be involved in drug resistance. In this study, we used the panHER inhibitor, dacomitinib, to further block HER2-dependent activation, applying multiple methods, such as proliferation assay, clonogenic survival assay, anti-anoikis assay and western blot analysis. Although dacomitinib inhibited EGFR, HER2, AKT and Erk activation more effectively than gefitinib, it still only exerted minimal anti-proliferative effects on sarcoma cell lines due to the STAT3 escape pathway. However, the addition of the STAT3 inhibitor, S3I-201, to dacomitinib achieved a significant enhancement in growth inhibition, by perturbing p-STAT3/p-STAT1. Using a panel of sarcoma cell lines with different histological types, we identified that the addition of the STAT3 inhibitor enhanced the growth inhibitory effects of the panHER inhibitor, dacomitinib, on sarcoma cells. Our findings may have clinical implications on overcoming the resistance caused by the STAT3 escape pathway and optimising EGFR/panHER-targeted therapy in sarcoma.
Collapse
Affiliation(s)
- Xiaochun Wang
- Sarcoma and Nano-Oncology Group, Adult Cancer Program, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW 2052, Australia
| | - David Goldstein
- Department of Medical Oncology, Prince of Wales Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia
| | - Philip J Crowe
- Sarcoma and Nano-Oncology Group, Adult Cancer Program, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW 2052, Australia
| | - Jia-Lin Yang
- Sarcoma and Nano-Oncology Group, Adult Cancer Program, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW 2052, Australia
| |
Collapse
|
16
|
Bontempo A, Ugalde-Villanueva B, Delgado-González E, Rodríguez ÁL, Aceves C. Molecular iodine impairs chemoresistance mechanisms, enhances doxorubicin retention and induces downregulation of the CD44+/CD24+ and E-cadherin+/vimentin+ subpopulations in MCF-7 cells resistant to low doses of doxorubicin. Oncol Rep 2017; 38:2867-2876. [PMID: 28901484 DOI: 10.3892/or.2017.5934] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 07/10/2017] [Indexed: 11/05/2022] Open
Abstract
One of the most dreaded clinical events for an oncology patient is resistance to treatment. Chemoresistance is a complex phenomenon based on alterations in apoptosis, the cell cycle and drug metabolism, and it correlates with the cancer stem cell phenotype and/or epithelial-mesenchymal transition. Molecular iodine (I2) exerts an antitumor effect on different types of iodine-capturing neoplasms by its oxidant/antioxidant properties and formation of iodolipids. In the present study, wild-type breast carcinoma cells (MCF-7/W) were treated chronically with 10 nM doxorubicin (DOX) to establish a low-dose DOX-resistant mammary cancer model (MCF-7/D). MCF-7/D cells were established after 30 days of treatment when the culture showed a proliferation rate similar to that of MCF-7/W. These DOX-resistant cells also showed increases in p21, Bcl-2 and MDR-1 expression. Supplementation with 200 µM I2 exerted similar effects in both cell lines: it decreased the proliferation rate by ~40%, and I2 co-administration with DOX significantly increased the inhibitory effect (to ~60%) and also increased apoptosis (BAX/Bcl-2 index), principally by inhibiting Bcl-2 expression. The inhibition by I2 + DOX was also accompanied by impaired MDR-1 induction as well as by a significant increase in PPARγ expression. All of these changes could be attributed to enhanced DOX retention and differential down-selection of CD44+/CD24+ and E-cadherin+/vimentin+ subpopulations. I2 + DOX-selected cells showed a weak induction of xenografts in Foxn1nu/nu mice, indicating that the iodine supplements reversed the tumorogenic capacity of the MCF-7/D cells. In conclusion, I2 is able to reduce the drug resistance and invasive capacity of mammary cancer cells exposed to DOX and represents an anti-chemoresistance agent with clinical potential.
Collapse
Affiliation(s)
- Alexander Bontempo
- Instituto de Neurobiologa, Universidad Nacional Autónoma de México (UNAM), Campus Juriquilla, Juriquilla, Santiago de Querétaro 76230, Mexico
| | - Brenda Ugalde-Villanueva
- Instituto de Neurobiologa, Universidad Nacional Autónoma de México (UNAM), Campus Juriquilla, Juriquilla, Santiago de Querétaro 76230, Mexico
| | - Evangelina Delgado-González
- Instituto de Neurobiologa, Universidad Nacional Autónoma de México (UNAM), Campus Juriquilla, Juriquilla, Santiago de Querétaro 76230, Mexico
| | - Ángel Luis Rodríguez
- Centro de Física Aplicada y Tecnologa Avanzada, Universidad Nacional Autónoma de México (UNAM), Campus Juriquilla, Juriquilla, Santiago de Querétaro 76230, Mexico
| | - Carmen Aceves
- Instituto de Neurobiologa, Universidad Nacional Autónoma de México (UNAM), Campus Juriquilla, Juriquilla, Santiago de Querétaro 76230, Mexico
| |
Collapse
|
17
|
Liao XH, Xiang Y, Yu CX, Li JP, Li H, Nie Q, Hu P, Zhou J, Zhang TC. STAT3 is required for MiR-17-5p-mediated sensitization to chemotherapy-induced apoptosis in breast cancer cells. Oncotarget 2017; 8:15763-15774. [PMID: 28178652 PMCID: PMC5362521 DOI: 10.18632/oncotarget.15000] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Accepted: 01/05/2017] [Indexed: 01/07/2023] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) controls cell survival, growth, migration, and invasion. Here, we observed that STAT3 exerted anti-apoptotic effects in breast cancer cells. On the other hand, miR-17-5p induced apoptosis in breast cancer cells, and overexpression of miR-17-5p sensitized MCF-7 cells to paclitaxel-induced apoptosis via STAT3. Overexpression of STAT3 in MCF-7 cells decreased paclitaxel-induced apoptosis, but STAT3 knockout abolished the miR-17-5p-induced increases in apoptosis. Finally, miR-17-5p promoted apoptosis by increasing p53 expression, which was inhibited by STAT3. These results demonstrate a novel pathway via which miR-17-5p inhibits STAT3 and increases p53 expression to promote apoptosis in breast cancer cells.
Collapse
Affiliation(s)
- Xing-Hua Liao
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Hubei, 430081, P.R. China
| | - Yuan Xiang
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Hubei, 430081, P.R. China
| | - Cheng-Xi Yu
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Hubei, 430081, P.R. China
| | - Jia-Peng Li
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Hubei, 430081, P.R. China
| | - Hui Li
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Hubei, 430081, P.R. China
| | - Qi Nie
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Hubei, 430081, P.R. China.,Wuhan Medical Treatment Center, Hubei, 430023, P.R. China
| | - Peng Hu
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Hubei, 430081, P.R. China
| | - Jun Zhou
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Hubei, 430081, P.R. China.,School of Medicine, Wuhan University of Science and Technology, Wuhan, 430065, P.R. China
| | - Tong-Cun Zhang
- Institute of Biology and Medicine, Wuhan University of Science and Technology, Hubei, 430081, P.R. China.,Key Laboratory of Industrial Fermentation Microbiology, Ministry of Education and Tianjin, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, 300457, P.R. China
| |
Collapse
|
18
|
HER2 in Breast Cancer Stemness: A Negative Feedback Loop towards Trastuzumab Resistance. Cancers (Basel) 2017; 9:cancers9050040. [PMID: 28445439 PMCID: PMC5447950 DOI: 10.3390/cancers9050040] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 04/10/2017] [Accepted: 04/21/2017] [Indexed: 12/19/2022] Open
Abstract
HER2 receptor tyrosine kinase that is overexpressed in approximately 20% of all breast cancers (BCs) is a poor prognosis factor and a precious target for BC therapy. Trastuzumab is approved by FDA to specifically target HER2 for treating HER2+ BC. However, about 60% of patients with HER2+ breast tumor develop de novo resistance to trastuzumab, partially due to the loss of expression of HER2 extracellular domain on their tumor cells. This is due to shedding/cleavage of HER2 by metalloproteinases (ADAMs and MMPs). HER2 shedding results in the accumulation of intracellular carboxyl-terminal HER2 (p95HER2), which is a common phenomenon in trastuzumab-resistant tumors and is suggested as a predictive marker for trastuzumab resistance. Up-regulation of the metalloproteinases is a poor prognosis factor and is commonly seen in mesenchymal-like cancer stem cells that are risen during epithelial to mesenchymal transition (EMT) of tumor cells. HER2 cleavage during EMT can explain why secondary metastatic tumors with high percentage of mesenchymal-like cancer stem cells are mostly resistant to trastuzumab but still sensitive to lapatinib. Importantly, many studies report HER2 interaction with oncogenic/stemness signaling pathways including TGF-β/Smad, Wnt/β-catenin, Notch, JAK/STAT and Hedgehog. HER2 overexpression promotes EMT and the emergence of cancer stem cell properties in BC. Increased expression and activation of metalloproteinases during EMT leads to proteolytic cleavage and shedding of HER2 receptor, which downregulates HER2 extracellular domain and eventually increases trastuzumab resistance. Here, we review the hypothesis that a negative feedback loop between HER2 and stemness signaling drives resistance of BC to trastuzumab.
Collapse
|
19
|
Mendoza-Rodríguez M, Arévalo Romero H, Fuentes-Pananá EM, Ayala-Sumuano JT, Meza I. IL-1β induces up-regulation of BIRC3, a gene involved in chemoresistance to doxorubicin in breast cancer cells. Cancer Lett 2017; 390:39-44. [DOI: 10.1016/j.canlet.2017.01.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 12/14/2016] [Accepted: 01/06/2017] [Indexed: 02/01/2023]
|
20
|
Reyes-Zárate E, Sánchez-Pérez Y, Gutiérrez-Ruiz MC, Chirino YI, Osornio-Vargas ÁR, Morales-Bárcenas R, Souza-Arroyo V, García-Cuellar CM. Atmospheric particulate matter (PM10) exposure-induced cell cycle arrest and apoptosis evasion through STAT3 activation via PKCζ and Src kinases in lung cells. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2016; 214:646-656. [PMID: 27131825 DOI: 10.1016/j.envpol.2016.04.072] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 04/18/2016] [Accepted: 04/19/2016] [Indexed: 06/05/2023]
Abstract
Atmospheric particulate matter with aerodynamic diameter ≤10 μm (PM10) is a risk factor for the development of lung cancer, but cellular pathways are not completely understood. STAT3 is a p21(Waf1/Cip1) transcription factor and is associated with proliferation and cell survival and is upregulated in lung cancer. PM10 exposure induces p21(Waf1/Cip1) expression, which could be related to STAT3 activation. The aims of this work were to investigate whether STAT3 was activated on lung epithelial cells after PM10 exposure and to determine whether or not STAT3 could have an impact on cell cycle distribution and cell survival. Our results showed that PM10 induced STAT3 activation through Src and PKCζ kinases, and it is partially responsible for the p21(Waf1/Cip1) induction that was also observed. Moreover, PM10 induced G1-G0 cell cycle arrest. The inhibition of STAT3 phosphorylation prevented cell cycle arrest and triggered apoptosis. These results suggest that PM10 exposure might activate a survival pathway related to STAT3 activation, similar to what has been described as part of the immune system and apoptosis evasion during tumor promotion and development.
Collapse
Affiliation(s)
- Elizabeth Reyes-Zárate
- Posgrado en Biología Experimental, Universidad Autónoma Metropolitana-Iztapalapa, Mexico; Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, 09340, México DF, Mexico
| | - Yesennia Sánchez-Pérez
- Instituto Nacional de Cancerología, Subdirección de Investigación Básica, San Fernando No. 22, Tlalpan, 14080, México DF, Mexico
| | - María Concepción Gutiérrez-Ruiz
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, 09340, México DF, Mexico
| | - Yolanda I Chirino
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, UNAM, Tlalnepantla, 54059, Estado de México, Mexico
| | | | - Rocío Morales-Bárcenas
- Instituto Nacional de Cancerología, Subdirección de Investigación Básica, San Fernando No. 22, Tlalpan, 14080, México DF, Mexico
| | - Verónica Souza-Arroyo
- Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, 09340, México DF, Mexico.
| | - Claudia María García-Cuellar
- Instituto Nacional de Cancerología, Subdirección de Investigación Básica, San Fernando No. 22, Tlalpan, 14080, México DF, Mexico.
| |
Collapse
|
21
|
Banerjee K, Resat H. Constitutive activation of STAT3 in breast cancer cells: A review. Int J Cancer 2015; 138:2570-8. [PMID: 26559373 DOI: 10.1002/ijc.29923] [Citation(s) in RCA: 450] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 10/29/2015] [Indexed: 12/19/2022]
Abstract
Signal transducer and activator of transcription 3 (STAT3) is constitutively activated in numerous cancer types, including more than 40% of breast cancers. In contrast to tight regulation of STAT3 as a latent transcription factor in normal cells, its signaling in breast cancer oncogenesis is multifaceted. Signaling through the IL-6/JAK/STAT3 pathway initiated by the binding of IL-6 family of cytokines (i.e., IL-6 and IL-11) to their receptors have been implicated in breast cancer development. Receptors with intrinsic kinase activity such as EGFR and VEGFR directly or indirectly induce STAT3 activation in various breast cancer types. Aberrant STAT3 signaling promotes breast tumor progression through deregulation of the expression of downstream target genes which control proliferation (Bcl-2, Bcl-xL, Survivin, Cyclin D1, c-Myc and Mcl-1), angiogenesis (Hif1α and VEGF) and epithelial-mesenchymal transition (Vimentin, TWIST, MMP-9 and MMP-7). These multiple modes of STAT3 regulation therefore make it a central linking point for a multitude of signaling processes. Extensive efforts to target STAT3 activation in breast cancer had no remarkable success in the past because the highly interconnected nature of STAT3 signaling introduces lack of selectivity in pathway identification for STAT3 targeted molecular therapies or because its role in tumorigenesis may not be as critical as it was thought. This review provides a full spectrum of STAT3's involvement in breast cancer by consolidating the knowledge about its role in breast cancer development at multiple levels: its differential regulation by different receptor signaling pathways, its downstream target genes, and modification of its transcriptional activity by its coregulatory transcription factors.
Collapse
Affiliation(s)
- Kasturi Banerjee
- The Gene and Linda Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, WA
| | - Haluk Resat
- The Gene and Linda Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, WA
| |
Collapse
|
22
|
Wei CY, Tan QX, Zhu X, Qin QH, Zhu FB, Mo QG, Yang WP. Expression of CDKN1A/p21 and TGFBR2 in breast cancer and their prognostic significance. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:14619-14629. [PMID: 26823785 PMCID: PMC4713571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 10/28/2015] [Indexed: 06/05/2023]
Abstract
BACKGROUND A new diagnostic and prognostic biomarker may be of value in cancer diseases. Our study aimed to evaluate the CDKN1A/p21 and TGFBR2 level measurable in a cohort of patients with breast cancer after mastectomy, and to confirm their suitability to serve as prognostic biomarkers of the cancer. METHODS The expression levels of CDKN1A/p21 and TGFBR2 were detected by reverse transcription-PCR (RT-PCR), western blot assay and immunohistochemical staining for 65 primary tumor samples and paired adjacent noncancerous breast tissues. Their relations to clinicopathologic parameters and to the prognosis of patients with breast cancer were analyzed. RESULTS We found the mRNA and protein expression levels of CDKN1A/p21 were significantly upregulated in breast cancer tissues compared with adjacent nontumorous breast tissues. Increased CDKN1A/p21 expression showed a significant correlation with larger tumor size (P=0.014), higher tumor dedifferentiation grade (P=0.021), lymph node metastasis (P=0.019) and a shorter disease-free survival (P=0.044). Contrarily, the expression levels of TGFBR2 mRNA and protein were significantly decreased in breast cancer tissues compared with adjacent nontumorous breast tissues. Underexpression of TGFBR2 in breast cancer was correlated with larger tumor size (P=0.034), lymph node metastasis (P=0.039) and a shorter disease-free survival (P=0.035). Statistical analysis suggested that there was no significant association between CDKN1A/p21 and TGFBR2 expression. CONCLUSIONS in summary, our results suggested that high CDKN1A/p21 and low TGFBR2 expression was closely correlated with adverse pathological parameters and poor prognosis in breast cancer. Both CDKN1A/p21 and TGFBR2 are presented as possible candidates for breast cancer biomarkers.
Collapse
Affiliation(s)
- Chang-Yuan Wei
- Department of Breast Surgery, The Affiliated Tumor Hospital of Guangxi Medical UniversityNanning 530021, China
| | - Qi-Xing Tan
- Department of Breast Surgery, The Affiliated Tumor Hospital of Guangxi Medical UniversityNanning 530021, China
| | - Xiao Zhu
- Department of Breast Surgery, The Affiliated Tumor Hospital of Guangxi Medical UniversityNanning 530021, China
| | - Qing-Hong Qin
- Department of Breast Surgery, The Affiliated Tumor Hospital of Guangxi Medical UniversityNanning 530021, China
| | - Fei-Bai Zhu
- Department of Obstetrics and Gynecology, Wenzhou Central HospitalWenzhou 325000, China
| | - Qin-Guo Mo
- Department of Breast Surgery, The Affiliated Tumor Hospital of Guangxi Medical UniversityNanning 530021, China
| | - Wei-Ping Yang
- Department of Ultrasound Diagnosis, The Affiliated Tumor Hospital of Guangxi Medical UniversityNanning 530021, China
| |
Collapse
|
23
|
Proietti CJ, Izzo F, Díaz Flaqué MC, Cordo Russo R, Venturutti L, Mercogliano MF, De Martino M, Pineda V, Muñoz S, Guzmán P, Roa JC, Schillaci R, Elizalde PV. Heregulin Co-opts PR Transcriptional Action Via Stat3 Role As a Coregulator to Drive Cancer Growth. Mol Endocrinol 2015; 29:1468-85. [PMID: 26340407 DOI: 10.1210/me.2015-1170] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Accumulated findings have demonstrated the presence of bidirectional interactions between progesterone receptor (PR) and the ErbB family of receptor tyrosine kinases signaling pathways in breast cancer. We previously revealed signal transducer and activator of transcription 3 (Stat3) as a nodal convergence point between said signaling pathways proving that Stat3 is activated by one of the ErbBs' ligands, heregulin (HRG)β1 via ErbB2 and through the co-option of PR as a signaling molecule. Here, we found that HRGβ1 induced Stat3 recruitment to the promoters of the progestin-regulated cell cycle modulators Bcl-XL and p21(CIP1) and also stimulated Stat3 binding to the mouse mammary tumor virus promoter, which carries consensus progesterone response elements. Interestingly, HRGβ1-activated Stat3 displayed differential functions on PR activity depending on the promoter bound. Indeed, Stat3 was required for PR binding in bcl-X, p21(CIP1), and c-myc promoters while exerting a PR coactivator function on the mouse mammary tumor virus promoter. Stat3 also proved to be necessary for HRGβ1-induced in vivo tumor growth. Our results endow Stat3 a novel function as a coregulator of HRGβ1-activated PR to promote breast cancer growth. These findings underscore the importance of understanding the complex interactions between PR and other regulatory factors, such as Stat3, that contribute to determine the context-dependent transcriptional actions of PR.
Collapse
Affiliation(s)
- Cecilia J Proietti
- Instituto de Biología y Medicina Experimental (C.J.P., F.I., M.C.D.F., R.C.R., L.V., M.F.M., M.D.M., R.S., P.V.E.), National Council of Scientific Research, Buenos Aires, 1428 ADN Argentina; Departamento de Anatomía Patológica (Scientific and Technological Bioresource Nucleus) (V.P., S.M., P.G., J.C.R.), Universidad de La Frontera, Temuco, 8330024 Chile; Departamento de Anatomía Patológica (J.C.R.), Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile; and Advanced Center for Chronic Diseases (J.C.R.), Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile
| | - Franco Izzo
- Instituto de Biología y Medicina Experimental (C.J.P., F.I., M.C.D.F., R.C.R., L.V., M.F.M., M.D.M., R.S., P.V.E.), National Council of Scientific Research, Buenos Aires, 1428 ADN Argentina; Departamento de Anatomía Patológica (Scientific and Technological Bioresource Nucleus) (V.P., S.M., P.G., J.C.R.), Universidad de La Frontera, Temuco, 8330024 Chile; Departamento de Anatomía Patológica (J.C.R.), Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile; and Advanced Center for Chronic Diseases (J.C.R.), Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile
| | - María Celeste Díaz Flaqué
- Instituto de Biología y Medicina Experimental (C.J.P., F.I., M.C.D.F., R.C.R., L.V., M.F.M., M.D.M., R.S., P.V.E.), National Council of Scientific Research, Buenos Aires, 1428 ADN Argentina; Departamento de Anatomía Patológica (Scientific and Technological Bioresource Nucleus) (V.P., S.M., P.G., J.C.R.), Universidad de La Frontera, Temuco, 8330024 Chile; Departamento de Anatomía Patológica (J.C.R.), Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile; and Advanced Center for Chronic Diseases (J.C.R.), Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile
| | - Rosalía Cordo Russo
- Instituto de Biología y Medicina Experimental (C.J.P., F.I., M.C.D.F., R.C.R., L.V., M.F.M., M.D.M., R.S., P.V.E.), National Council of Scientific Research, Buenos Aires, 1428 ADN Argentina; Departamento de Anatomía Patológica (Scientific and Technological Bioresource Nucleus) (V.P., S.M., P.G., J.C.R.), Universidad de La Frontera, Temuco, 8330024 Chile; Departamento de Anatomía Patológica (J.C.R.), Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile; and Advanced Center for Chronic Diseases (J.C.R.), Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile
| | - Leandro Venturutti
- Instituto de Biología y Medicina Experimental (C.J.P., F.I., M.C.D.F., R.C.R., L.V., M.F.M., M.D.M., R.S., P.V.E.), National Council of Scientific Research, Buenos Aires, 1428 ADN Argentina; Departamento de Anatomía Patológica (Scientific and Technological Bioresource Nucleus) (V.P., S.M., P.G., J.C.R.), Universidad de La Frontera, Temuco, 8330024 Chile; Departamento de Anatomía Patológica (J.C.R.), Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile; and Advanced Center for Chronic Diseases (J.C.R.), Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile
| | - María Florencia Mercogliano
- Instituto de Biología y Medicina Experimental (C.J.P., F.I., M.C.D.F., R.C.R., L.V., M.F.M., M.D.M., R.S., P.V.E.), National Council of Scientific Research, Buenos Aires, 1428 ADN Argentina; Departamento de Anatomía Patológica (Scientific and Technological Bioresource Nucleus) (V.P., S.M., P.G., J.C.R.), Universidad de La Frontera, Temuco, 8330024 Chile; Departamento de Anatomía Patológica (J.C.R.), Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile; and Advanced Center for Chronic Diseases (J.C.R.), Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile
| | - Mara De Martino
- Instituto de Biología y Medicina Experimental (C.J.P., F.I., M.C.D.F., R.C.R., L.V., M.F.M., M.D.M., R.S., P.V.E.), National Council of Scientific Research, Buenos Aires, 1428 ADN Argentina; Departamento de Anatomía Patológica (Scientific and Technological Bioresource Nucleus) (V.P., S.M., P.G., J.C.R.), Universidad de La Frontera, Temuco, 8330024 Chile; Departamento de Anatomía Patológica (J.C.R.), Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile; and Advanced Center for Chronic Diseases (J.C.R.), Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile
| | - Viviana Pineda
- Instituto de Biología y Medicina Experimental (C.J.P., F.I., M.C.D.F., R.C.R., L.V., M.F.M., M.D.M., R.S., P.V.E.), National Council of Scientific Research, Buenos Aires, 1428 ADN Argentina; Departamento de Anatomía Patológica (Scientific and Technological Bioresource Nucleus) (V.P., S.M., P.G., J.C.R.), Universidad de La Frontera, Temuco, 8330024 Chile; Departamento de Anatomía Patológica (J.C.R.), Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile; and Advanced Center for Chronic Diseases (J.C.R.), Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile
| | - Sergio Muñoz
- Instituto de Biología y Medicina Experimental (C.J.P., F.I., M.C.D.F., R.C.R., L.V., M.F.M., M.D.M., R.S., P.V.E.), National Council of Scientific Research, Buenos Aires, 1428 ADN Argentina; Departamento de Anatomía Patológica (Scientific and Technological Bioresource Nucleus) (V.P., S.M., P.G., J.C.R.), Universidad de La Frontera, Temuco, 8330024 Chile; Departamento de Anatomía Patológica (J.C.R.), Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile; and Advanced Center for Chronic Diseases (J.C.R.), Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile
| | - Pablo Guzmán
- Instituto de Biología y Medicina Experimental (C.J.P., F.I., M.C.D.F., R.C.R., L.V., M.F.M., M.D.M., R.S., P.V.E.), National Council of Scientific Research, Buenos Aires, 1428 ADN Argentina; Departamento de Anatomía Patológica (Scientific and Technological Bioresource Nucleus) (V.P., S.M., P.G., J.C.R.), Universidad de La Frontera, Temuco, 8330024 Chile; Departamento de Anatomía Patológica (J.C.R.), Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile; and Advanced Center for Chronic Diseases (J.C.R.), Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile
| | - Juan C Roa
- Instituto de Biología y Medicina Experimental (C.J.P., F.I., M.C.D.F., R.C.R., L.V., M.F.M., M.D.M., R.S., P.V.E.), National Council of Scientific Research, Buenos Aires, 1428 ADN Argentina; Departamento de Anatomía Patológica (Scientific and Technological Bioresource Nucleus) (V.P., S.M., P.G., J.C.R.), Universidad de La Frontera, Temuco, 8330024 Chile; Departamento de Anatomía Patológica (J.C.R.), Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile; and Advanced Center for Chronic Diseases (J.C.R.), Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile
| | - Roxana Schillaci
- Instituto de Biología y Medicina Experimental (C.J.P., F.I., M.C.D.F., R.C.R., L.V., M.F.M., M.D.M., R.S., P.V.E.), National Council of Scientific Research, Buenos Aires, 1428 ADN Argentina; Departamento de Anatomía Patológica (Scientific and Technological Bioresource Nucleus) (V.P., S.M., P.G., J.C.R.), Universidad de La Frontera, Temuco, 8330024 Chile; Departamento de Anatomía Patológica (J.C.R.), Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile; and Advanced Center for Chronic Diseases (J.C.R.), Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile
| | - Patricia V Elizalde
- Instituto de Biología y Medicina Experimental (C.J.P., F.I., M.C.D.F., R.C.R., L.V., M.F.M., M.D.M., R.S., P.V.E.), National Council of Scientific Research, Buenos Aires, 1428 ADN Argentina; Departamento de Anatomía Patológica (Scientific and Technological Bioresource Nucleus) (V.P., S.M., P.G., J.C.R.), Universidad de La Frontera, Temuco, 8330024 Chile; Departamento de Anatomía Patológica (J.C.R.), Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile; and Advanced Center for Chronic Diseases (J.C.R.), Pontificia Universidad Católica de Chile, Santiago de Chile, 8330024 Chile
| |
Collapse
|
24
|
Wu X, Zahari MS, Renuse S, Nirujogi RS, Kim MS, Manda SS, Stearns V, Gabrielson E, Sukumar S, Pandey A. Phosphoproteomic Analysis Identifies Focal Adhesion Kinase 2 (FAK2) as a Potential Therapeutic Target for Tamoxifen Resistance in Breast Cancer. Mol Cell Proteomics 2015; 14:2887-900. [PMID: 26330541 DOI: 10.1074/mcp.m115.050484] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Indexed: 01/13/2023] Open
Abstract
Tamoxifen, an estrogen receptor-α (ER) antagonist, is an important agent for the treatment of breast cancer. However, this therapy is complicated by the fact that a substantial number of patients exhibit either de novo or acquired resistance. To characterize the signaling mechanisms underlying this resistance, we treated the MCF7 breast cancer cell line with tamoxifen for over six months and showed that this cell line acquired resistance to tamoxifen in vitro and in vivo. We performed SILAC-based quantitative phosphoproteomic profiling on the tamoxifen resistant and vehicle-treated sensitive cell lines to quantify the phosphorylation alterations associated with tamoxifen resistance. From >5600 unique phosphopeptides identified, 1529 peptides exhibited hyperphosphorylation and 409 peptides showed hypophosphorylation in the tamoxifen resistant cells. Gene set enrichment analysis revealed that focal adhesion pathway was one of the most enriched signaling pathways activated in tamoxifen resistant cells. Significantly, we showed that the focal adhesion kinase FAK2 was not only hyperphosphorylated but also transcriptionally up-regulated in tamoxifen resistant cells. FAK2 suppression by specific siRNA knockdown or a small molecule inhibitor repressed cellular proliferation in vitro and tumor formation in vivo. More importantly, our survival analysis revealed that high expression of FAK2 is significantly associated with shorter metastasis-free survival in estrogen receptor-positive breast cancer patients treated with tamoxifen. Our studies suggest that FAK2 is a potential therapeutic target for the management of hormone-refractory breast cancers.
Collapse
Affiliation(s)
- Xinyan Wu
- From the ‡McKusick-Nathans Institute of Genetic Medicine and Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Muhammad Saddiq Zahari
- From the ‡McKusick-Nathans Institute of Genetic Medicine and Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Santosh Renuse
- §Institute of Bioinformatics, International Technology Park, Bangalore, 560066 India
| | - Raja Sekhar Nirujogi
- §Institute of Bioinformatics, International Technology Park, Bangalore, 560066 India
| | - Min-Sik Kim
- From the ‡McKusick-Nathans Institute of Genetic Medicine and Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Srikanth S Manda
- §Institute of Bioinformatics, International Technology Park, Bangalore, 560066 India
| | | | - Edward Gabrielson
- ‖Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231
| | | | - Akhilesh Pandey
- From the ‡McKusick-Nathans Institute of Genetic Medicine and Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205; ¶Department of Oncology; ‖Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231
| |
Collapse
|
25
|
Yaglom JA, McFarland C, Mirny L, Sherman MY. Oncogene-triggered suppression of DNA repair leads to DNA instability in cancer. Oncotarget 2015; 5:8367-78. [PMID: 25252808 PMCID: PMC4226689 DOI: 10.18632/oncotarget.2259] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
DNA instability is an important contributor to cancer development. Previously, defects in the chromosome segregation and excessive DNA double strand breaks due to the replication or oxidative stresses were implicated in DNA instability in cancer. Here, we demonstrate that DNA instability can directly result from the oncogene-induced senescence signaling. Expression of the activated form of Her2 oncogene, NeuT, in immortalized breast epithelial cells led to downregulation of the major DNA repair factor histone H2AX and a number of other components of the HR and NHEJ double strand DNA breaks repair pathways. H2AX expression was regulated at the transcriptional level via a senescence pathway involving p21-mediated regulation of CDK and Rb1. The p21-dependent downregulation of H2AX was seen both in cell culture and the MMTV-neu mouse model of Her2-positive breast cancer. Importantly, downregulation of H2AX upon Her2/NeuT expression impaired repair of double strand DNA breaks. This impairment resulted in both increased DNA instability in the form of somatic copy number alterations, and in increased sensitivity to the chemotherapeutic drug doxorubicin. Overall, these findings indicate that the Her2/NeuT oncogene signaling directly potentiates DNA instability and increases sensitivity to DNA damaging treatments.
Collapse
Affiliation(s)
- Julia A Yaglom
- Department Biochemistry, Boston University School of Medicine, Boston, MA
| | | | - Leonid Mirny
- Institute for Medical Engineering and Sciences, Massachusetts Institute of Technology, Cambridge, MA
| | - Michael Y Sherman
- Department Biochemistry, Boston University School of Medicine, Boston, MA
| |
Collapse
|
26
|
Chouchana L, Fernández-Ramos AA, Dumont F, Marchetti C, Ceballos-Picot I, Beaune P, Gurwitz D, Loriot MA. Molecular insight into thiopurine resistance: transcriptomic signature in lymphoblastoid cell lines. Genome Med 2015; 7:37. [PMID: 26015807 PMCID: PMC4443628 DOI: 10.1186/s13073-015-0150-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 03/03/2015] [Indexed: 12/15/2022] Open
Abstract
Background There has been considerable progress in the management of acute lymphoblastic leukemia (ALL) but further improvement is needed to increase long-term survival. The thiopurine agent 6-mercaptopurine (6-MP) used for ALL maintenance therapy has a key influence on clinical outcomes and relapse prevention. Genetic inheritance in thiopurine metabolism plays a major role in interindividual clinical response variability to thiopurines; however, most cases of thiopurine resistance remain unexplained. Methods We used lymphoblastoid cell lines (LCLs) from healthy donors, selected for their extreme thiopurine susceptibility. Thiopurine metabolism was characterized by the determination of TPMT and HPRT activity. We performed genome-wide expression profiling in resistant and sensitive cell lines with the goal of elucidating the mechanisms of thiopurine resistance. Results We determined a higher TPMT activity (+44%; P = 0.024) in resistant compared to sensitive cell lines, although there was no difference in HPRT activity. We identified a 32-gene transcriptomic signature that predicts thiopurine resistance. This signature includes the GTPBP4 gene coding for a GTP-binding protein that interacts with p53. A comprehensive pathway analysis of the genes differentially expressed between resistant and sensitive cell lines indicated a role for cell cycle and DNA mismatch repair system in thiopurine resistance. It also revealed overexpression of the ATM/p53/p21 pathway, which is activated in response to DNA damage and induces cell cycle arrest in thiopurine resistant LCLs. Furthermore, overexpression of the p53 target gene TNFRSF10D or the negative cell cycle regulator CCNG2 induces cell cycle arrest and may also contribute to thiopurine resistance. ARHGDIA under-expression in resistant cell lines may constitute a novel molecular mechanism contributing to thiopurine resistance based on Rac1 inhibition induced apoptosis and in relation with thiopurine pharmacodynamics. Conclusion Our study provides new insights into the molecular mechanisms underlying thiopurine resistance and suggests a potential research focus for developing tailored medicine. Electronic supplementary material The online version of this article (doi:10.1186/s13073-015-0150-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Laurent Chouchana
- INSERM UMR-S 1147, 45 rue des Saints-Pères, Paris, 75006 France ; Université Paris Descartes, Sorbonne Paris Cité, 45 rue des Saints-Pères, Paris, 75006 France
| | - Ana Aurora Fernández-Ramos
- INSERM UMR-S 1147, 45 rue des Saints-Pères, Paris, 75006 France ; Université Paris Descartes, Sorbonne Paris Cité, 45 rue des Saints-Pères, Paris, 75006 France
| | - Florent Dumont
- Université Paris Descartes, Sorbonne Paris Cité, 45 rue des Saints-Pères, Paris, 75006 France ; INSERM U1016, Institut Cochin, 22 Rue Mechain, Paris, 75014 France
| | - Catherine Marchetti
- INSERM UMR-S 1147, 45 rue des Saints-Pères, Paris, 75006 France ; Université Paris Descartes, Sorbonne Paris Cité, 45 rue des Saints-Pères, Paris, 75006 France
| | - Irène Ceballos-Picot
- Université Paris Descartes, Sorbonne Paris Cité, 45 rue des Saints-Pères, Paris, 75006 France ; Assistance Publique-Hôpitaux de Paris, Hôpital Necker-Enfants Malades, Biochimie Métabolique, 149 Rue de Sèvres, Paris, 75015 France
| | - Philippe Beaune
- INSERM UMR-S 1147, 45 rue des Saints-Pères, Paris, 75006 France ; Université Paris Descartes, Sorbonne Paris Cité, 45 rue des Saints-Pères, Paris, 75006 France ; Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Biochimie Pharmacogénétique et Oncologie Moléculaire, 20 rue Leblanc, Paris, 75015 France
| | - David Gurwitz
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Marie-Anne Loriot
- INSERM UMR-S 1147, 45 rue des Saints-Pères, Paris, 75006 France ; Université Paris Descartes, Sorbonne Paris Cité, 45 rue des Saints-Pères, Paris, 75006 France ; Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Biochimie Pharmacogénétique et Oncologie Moléculaire, 20 rue Leblanc, Paris, 75015 France
| |
Collapse
|
27
|
Zhao Y, Yang X. The Hippo pathway in chemotherapeutic drug resistance. Int J Cancer 2014; 137:2767-73. [DOI: 10.1002/ijc.29293] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 10/23/2014] [Indexed: 12/11/2022]
Affiliation(s)
- Yulei Zhao
- Department of Pathology and Molecular Medicine; Queen's University; Kingston Ontario Canada
| | - Xiaolong Yang
- Department of Pathology and Molecular Medicine; Queen's University; Kingston Ontario Canada
| |
Collapse
|
28
|
Sigurðsson HH, Olesen CW, Dybboe R, Lauritzen G, Pedersen SF. Constitutively active ErbB2 regulates cisplatin-induced cell death in breast cancer cells via pro- and antiapoptotic mechanisms. Mol Cancer Res 2014; 13:63-77. [PMID: 25143433 DOI: 10.1158/1541-7786.mcr-14-0011] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
UNLABELLED Despite the frequent expression of N-terminally truncated ErbB2 (ΔNErbB2/p95HER2) in breast cancer and its association with Herceptin resistance and poor prognosis, it remains poorly understood how ΔNErbB2 affects chemotherapy-induced cell death. Previously it was shown that ΔNErbB2 upregulates acid extrusion from MCF-7 breast cancer cells and that inhibition of the Na(+)/H(+) exchanger (SLC9A1/NHE1) strongly sensitizes ΔNErbB2-expressing MCF-7 cells to cisplatin chemotherapy. The aim of this study was to identify the mechanism through which ΔNErbB2 regulates cisplatin-induced breast cancer cell death, and determine how NHE1 regulates this process. Cisplatin treatment elicited apoptosis, ATM phosphorylation, upregulation of p53, Noxa (PMAIP1), and PUMA (BBC3), and cleavage of caspase-9, -7, fodrin, and PARP-1 in MCF-7 cells. Inducible ΔNErbB2 expression strongly reduced cisplatin-induced ATM- and p53-phosphorylation, augmented Noxa upregulation and caspase-9 and -7 cleavage, doubled p21(WAF1/Cip1) (CDKN1A) expression, and nearly abolished Bcl-2 expression. LC3-GFP analysis demonstrated that autophagic flux was reduced by cisplatin in a manner augmented by ΔNErbB2, yet did not contribute to cisplatin-induced death. Using knockdown approaches, it was shown that cisplatin-induced caspase-7 cleavage in ΔNErbB2-MCF-7 cells was Noxa- and caspase-9 dependent. This pathway was augmented by NHE1 inhibition, while the Na(+)/HCO3 (-) cotransporter (SLC4A7/NBCn1) was internalized following cisplatin exposure. IMPLICATIONS This work reveals that ΔNErbB2 strongly affects several major pro- and antiapoptotic pathways and provides mechanistic insight into the role of NHE1 in chemotherapy resistance. These findings have relevance for defining therapy regimens in breast cancers with ΔNErbB2 and/or NHE1 overexpression.
Collapse
Affiliation(s)
- Haraldur H Sigurðsson
- Department of Biology, Section for Cell and Developmental Biology, University of Copenhagen, Copenhagen, Denmark
| | - Christina W Olesen
- Department of Biology, Section for Cell and Developmental Biology, University of Copenhagen, Copenhagen, Denmark
| | - Rie Dybboe
- Department of Biology, Section for Cell and Developmental Biology, University of Copenhagen, Copenhagen, Denmark
| | - Gitte Lauritzen
- Department of Biology, Section for Cell and Developmental Biology, University of Copenhagen, Copenhagen, Denmark
| | - Stine F Pedersen
- Department of Biology, Section for Cell and Developmental Biology, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
29
|
Withacnistin inhibits recruitment of STAT3 and STAT5 to growth factor and cytokine receptors and induces regression of breast tumours. Br J Cancer 2014; 111:894-902. [PMID: 24983364 PMCID: PMC4150266 DOI: 10.1038/bjc.2014.349] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 04/14/2014] [Accepted: 05/14/2014] [Indexed: 01/05/2023] Open
Abstract
Background: The binding of STAT3 and STAT5 to growth factor and cytokine receptors such as EGFR and IL-6 receptor gp130 is critical to their activation and ability to contribute to malignant transformation. Therefore, interfering with these biochemical processes could lead to the discovery of novel anticancer agents. Methods: Co-immunoprecipitation, western blotting, microscopy, DNA binding, invasion, and soft agar assays as well as a mouse model were used to investigate the mechanism by which the natural product Withacnistin (Wit) inhibits STAT 3/5 tyrosine phosphoryaltion and activation. Results: Wit blocks EGF- and IL-6-stimulated binding of STAT3 and STAT5 to EGFR and gp130. Wit inhibits EGF-, PDGF-, IL-6-, IFNβ-, and GM-CSF-stimulation of tyrosine phosphorylation of STAT3 and STAT5 but not of EGFR or PDGFR. The inhibition of P-STAT3 and P-STAT5 occurred rapidly, within minutes of Wit treatment and growth factor stimulation. Wit also inhibits STAT3 nuclear translocation, DNA binding, promoter transcriptional activation, and it suppresses the expression levels of STAT3 target genes such as Bcl-xL and Mcl-1. Finally, Wit induces apoptosis, inhibits anchorage-dependent and -independent growth and invasion, and causes breast tumour regression in an ErbB2-driven transgenic mouse model. Conclusions: These data warrant further development of Wit as a novel anticancer drug for targeting tumours that harbour hyperactivated STAT3 and STAT5.
Collapse
|
30
|
Malik S, Saha R, Seth P. Involvement of extracellular signal-regulated kinase (ERK1/2)-p53-p21 axis in mediating neural stem/progenitor cell cycle arrest in co-morbid HIV-drug abuse exposure. J Neuroimmune Pharmacol 2014; 9:340-53. [PMID: 24469921 DOI: 10.1007/s11481-014-9523-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Accepted: 01/14/2014] [Indexed: 10/25/2022]
Abstract
Neurological complications in opioid abusing Human Immunodeficiency Virus-1 (HIV-1) patients suggest enhanced neurodegeneration as compared to non-drug abusing HIV-1 infected population. Neural precursor cells (NPCs), the multipotent cells of the mammalian brain, are susceptible to HIV-1 infection and as opiates also perturb their growth kinetics, detailed mechanistic studies for their co-morbid exposure are highly warranted. Using a well characterized in vitro model of human fetal brain-derived neural precursor cells, we investigated alterations in NPC properties at both acute and chronic durations. Chronic morphine and Tat treatment attenuated proliferation in NPCs, with cells stalled at G1-phase of the cell cycle. Furthermore HIV-Tat and morphine exposure increased activation of extracellular signal-regulated kinase-1/2 (ERK1/2), enhanced levels of p53 and p21, and decreased cyclin D1 and Akt levels in NPCs. Regulated by ERK1/2 and p53, p21 was found to be indispensible for Tat and morphine mediated cell cycle arrest. Our study elaborates on the cellular and molecular machinery in NPCs and provides significant mechanistic details into HIV-drug abuse co-morbidity that may have far reaching clinical consequences both in pediatric as well as adult neuroAIDS.
Collapse
Affiliation(s)
- Shaily Malik
- Cellular and Molecular Neuroscience, National Brain Research Centre (NBRC), NH-8, Nainwal Road, Manesar, Gurgaon, Haryana, 122051, India
| | | | | |
Collapse
|
31
|
Gorbatenko A, Olesen CW, Mørup N, Thiel G, Kallunki T, Valen E, Pedersen SF. ErbB2 upregulates the Na+,HCO3(-)-cotransporter NBCn1/SLC4A7 in human breast cancer cells via Akt, ERK, Src, and Kruppel-like factor 4. FASEB J 2013; 28:350-63. [PMID: 24088818 DOI: 10.1096/fj.13-233288] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Misregulation of acid-base transport plays central roles in cancer development. We previously demonstrated the strong up-regulation of the Na(+),HCO3(-) cotransporter NBCn1 (SLC4A7) in MCF-7 breast cancer cells by a truncated, constitutively active ErbB2 (HER2) receptor, ΔNErbB2, and showed that NBCn1 expression and activity are increased in breast cancer tissue from patients. Here, we present the first in-depth characterization of an SLC4A7 promoter and identify its minimal ΔNErbB2-sensitive region. Inhibition or siRNA-mediated knockdown of PI3K, Akt1, ERK1/2, or Src decreased the NBCn1 protein level in ΔNErbB2-expressing MCF-7 cells by ~50, 60, 30 and 35%, respectively. Further, knockdown of the transcription factor Krüppel-like factor 4 (KLF4) reduced NBCn1 protein expression by ~40%, and KLF4 overexpression increased NBCn1 expression by 50-80%. In contrast, knockdown of the closely related transcription factor specificity protein 1 (Sp1) or transfection with dominant-negative Sp1 increased NBCn1 expression by ~35 and ~50%, respectively. NBCn1 expression was also increased by stimulation of full-length ErbB1, -2, and -3 receptors in SKBr3 cells (1.5- and 2-fold by NRG1 or EGF, respectively) or after their exogenous expression in MCF-7 cells. Finally, stimulation with NRG1 or EGF more than doubled acid extrusion capacity in SKBr3 cells. In conclusion, NBCn1 is strongly upregulated by ErbB receptor signaling in a manner involving opposite effects of KLF4 and Sp1, transcription factors with central roles in cancer development. ErbB-induced up-regulation of NBCn1-mediated acid extrusion may play important physiological and pathophysiological roles in the breast epithelium and other tissues with high ErbB receptor levels.
Collapse
Affiliation(s)
- Andrej Gorbatenko
- 1Department of Biology, University of Copenhagen, 13, Universitetsparken, DK-2100 Copenhagen, Denmark.
| | | | | | | | | | | | | |
Collapse
|
32
|
Zhang S, Huang WC, Zhang L, Zhang C, Lowery FJ, Ding Z, Guo H, Wang H, Huang S, Sahin AA, Aldape KD, Steeg PS, Yu D. SRC family kinases as novel therapeutic targets to treat breast cancer brain metastases. Cancer Res 2013; 73:5764-74. [PMID: 23913825 DOI: 10.1158/0008-5472.can-12-1803] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Despite better control of early-stage disease and improved overall survival of patients with breast cancer, the incidence of life-threatening brain metastases continues to increase in some of these patients. Unfortunately, other than palliative treatments there is no effective therapy for this condition. In this study, we reveal a critical role for Src activation in promoting brain metastasis in a preclinical model of breast cancer and we show how Src-targeting combinatorial regimens can treat HER2(+) brain metastases in this model. We found that Src was hyperactivated in brain-seeking breast cancer cells derived from human cell lines or from patients' brain metastases. Mechanistically, Src activation promoted tumor cell extravasation into the brain parenchyma via permeabilization of the blood-brain barrier. When combined with the EGFR/HER2 dual-targeting drug lapatinib, an Src-targeting combinatorial regimen prevented outgrowth of disseminated breast cancer cells through the induction of cell-cycle arrest. More importantly, this combinatorial regimen inhibited the outgrowth of established experimental brain metastases, prolonging the survival of metastases-bearing mice. Our results provide a rationale for clinical evaluation of Src-targeting regimens to treat patients with breast cancer suffering from brain metastasis.
Collapse
Affiliation(s)
- Siyuan Zhang
- Authors' Affiliations: Departments of Molecular and Cellular Oncology, Neurosurgery, and Pathology, The University of Texas MD Anderson Cancer Center; Cancer Biology Program, Graduate School of Biomedical Sciences-Houston, Houston, Texas; Women's Cancers Section, Laboratory of Molecular Pharmacology, National Cancer Institute, Bethesda, Maryland; and Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Diaz Flaqué MC, Vicario R, Proietti CJ, Izzo F, Schillaci R, Elizalde PV. Progestin drives breast cancer growth by inducing p21(CIP1) expression through the assembly of a transcriptional complex among Stat3, progesterone receptor and ErbB-2. Steroids 2013. [PMID: 23178160 DOI: 10.1016/j.steroids.2012.11.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Cell cycle regulator p21(CIP1) has controversial biological effects in breast cancer since in spite of its role as cell cycle inhibitor and promoter of cellular senescence, it also induces cell proliferation and chemoteraphy resistance. We here explored the molecular mechanisms involved in progestin regulation of p21(CIP1) expression. We also investigated the biological effects of p21(CIP1) in breast cancer cells. We found that the synthetic progestin medroxyprogesterone acetate (MPA) upregulates p21(CIP1) protein expression via c-Src, signal transducer and activator of transcription 3 (Stat3) and ErbB-2 phosphorylation. Notably, we also found that ErbB-2 nuclear function plays a key role in MPA-induction of p21(CIP1) expression. Interestingly, we determined that progestin drives p21(CIP1) transcriptional activation via a novel nonclassical transcriptional mechanism in which progesterone receptor is recruited along with Stat3 and ErbB-2 to a Stat3 binding site at p21(CIP1) promoter. Our findings revealed that ErbB-2 functions as a coactivator of Stat3 in progestin induction of p21(CIP1) transcriptional activation. Furthermore, we demonstrated that blockage of p21(CIP1) expression strongly inhibited in vitro and in vivo progestin-induced breast cancer cell proliferation. These results further support the hypothesis that according to cell context and type of stimulus, p21(CIP1) is capable of inducing cell cycle progression. Moreover, we provided evidence that Stat3 and nuclear ErbB-2 are key players in progestin-induced p21(CIP1) regulation.
Collapse
Affiliation(s)
- María C Diaz Flaqué
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Vuelta de Obligado 2490, Buenos Aires C1428ADN, Argentina
| | | | | | | | | | | |
Collapse
|
34
|
Wei J, Yuan Y, Jin C, Chen H, Leng L, He F, Wang J. The ubiquitin ligase TRAF6 negatively regulates the JAK-STAT signaling pathway by binding to STAT3 and mediating its ubiquitination. PLoS One 2012. [PMID: 23185365 PMCID: PMC3501508 DOI: 10.1371/journal.pone.0049567] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
STAT3 is a key transcription factor that mediates various cellular and organismal processes, such as cell growth, apoptosis, immune response and cancer. However, the molecular mechanisms of STAT3 regulation remain poorly understood. Here, we identified TRAF6 as a new STAT3 interactor. TRAF6 augmented the ubiquitination of STAT3 and deactivated its transcriptional activity induced by IFNα stimulation or overexpressed with JAK2. Both the RING domain and the TRAF-type zinc finger domain of TRAF6 were indispensable for STAT3 deactivation. Accordingly, TRAF6 also down-regulated the expression of two known STAT3 target genes, CRP and ACT. Therefore, we showed that TRAF6 is a new regulator of JAK/STAT signaling and provide a new mechanistic explanation for the crosstalk between the NF-κB and the JAK-STAT pathways.
Collapse
Affiliation(s)
- Juncheng Wei
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
- Institute of Basic Medical Science, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yanzhi Yuan
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Chaozhi Jin
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Hui Chen
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Ling Leng
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
| | - Fuchu He
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
- Institute of Basic Medical Science, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- * E-mail: (J. Wang); (FH)
| | - Jian Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, China
- * E-mail: (J. Wang); (FH)
| |
Collapse
|
35
|
Nam HJ, Im SA, Oh DY, Elvin P, Kim HP, Yoon YK, Min A, Song SH, Han SW, Kim TY, Bang YJ. Antitumor activity of saracatinib (AZD0530), a c-Src/Abl kinase inhibitor, alone or in combination with chemotherapeutic agents in gastric cancer. Mol Cancer Ther 2012; 12:16-26. [PMID: 23144237 DOI: 10.1158/1535-7163.mct-12-0109] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Src is a nonreceptor tyrosine kinase involved in the cross-talk and mediation of many signaling pathways that promote cell proliferation, adhesion, invasion, migration, and tumorigenesis. Increased Src activity has been reported in many types of human cancer, including gastric cancer. Therefore, this factor has been identified as a promising therapeutic target for cancer treatments, and targeting Src in gastric cancer is predicted to have potent effects. We evaluated the antitumor effect of a c-Src/Abl kinase inhibitor, saracatinib (AZD0530), alone or combined with chemotherapeutic agents in gastric cancer cell lines and a NCI-N87 xenograft model. Among 10 gastric cancer cell lines, saracatinib specifically inhibited the growth and migration/invasion of SNU216 and NCI-N87 cells. Saracatinib blocked the Src/FAK, HER family, and oncogenic signaling pathways, and it induced G(1) arrest and apoptosis in SNU216 and NCI-N87 cells. Apoptosis required induction of the proapoptotic BCL2 family member Bim. Knockdown of Bim using siRNA decreased apoptosis induced by treatment with saracatinib, suggesting that Bim has an important role in saracatinib-induced apoptosis. Saracatinib enhanced the effects of lapatinib, an EGFR/HER2 dual inhibitor, in SNU216 and NCI-N87 cells. Furthermore, combined treatment with saracatinib and 5-fluorouracil (5-FU) or cisplatin exerted synergistic effects in both saracatinib-sensitive and saracatinib-resistant cells. Consistent with our in vitro findings, cotreatment with saracatinib and 5-FU resulted in enhanced antitumor activity in the NCI-N87 xenografts. These data indicate that the inhibition of Src kinase activity by saracatinib alone or in combination with other agents can be a strategy to target gastric cancer.
Collapse
Affiliation(s)
- Hyun-Jin Nam
- Cancer Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
Since cancer is one of the leading causes of death worldwide, there is an urgent need to find better treatments. Currently, the use of chemotherapeutics remains the predominant option for cancer therapy. However, one of the major obstacles for successful cancer therapy using these chemotherapeutics is that patients often do not respond or eventually develop resistance after initial treatment. Therefore identification of genes involved in chemotherapeutic response is critical for predicting tumour response and treating drug-resistant cancer patients. A group of genes commonly lost or inactivated are tumour suppressor genes, which can promote the initiation and progression of cancer through regulation of various biological processes such as cell proliferation, cell death and cell migration/invasion. Recently, mounting evidence suggests that these tumour suppressor genes also play a very important role in the response of cancers to a variety of chemotherapeutic drugs. In the present review, we will provide a comprehensive overview on how major tumour suppressor genes [Rb (retinoblastoma), p53 family, cyclin-dependent kinase inhibitors, BRCA1 (breast-cancer susceptibility gene 1), PTEN (phosphatase and tensin homologue deleted on chromosome 10), Hippo pathway, etc.] are involved in chemotherapeutic drug response and discuss their applications in predicting the clinical outcome of chemotherapy for cancer patients. We also propose that tumour suppressor genes are critical chemotherapeutic targets for the successful treatment of drug-resistant cancer patients in future applications.
Collapse
|
37
|
Du L, Subauste MC, DeSevo C, Zhao Z, Baker M, Borkowski R, Schageman JJ, Greer R, Yang CR, Suraokar M, Wistuba II, Gazdar AF, Minna JD, Pertsemlidis A. miR-337-3p and its targets STAT3 and RAP1A modulate taxane sensitivity in non-small cell lung cancers. PLoS One 2012; 7:e39167. [PMID: 22723956 PMCID: PMC3377607 DOI: 10.1371/journal.pone.0039167] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Accepted: 05/17/2012] [Indexed: 12/14/2022] Open
Abstract
NSCLC (non-small cell lung cancer) often exhibits resistance to paclitaxel treatment. Identifying the elements regulating paclitaxel response will advance efforts to overcome such resistance in NSCLC therapy. Using in vitro approaches, we demonstrated that over-expression of the microRNA miR-337-3p sensitizes NCI-H1155 cells to paclitaxel, and that miR-337-3p mimic has a general effect on paclitaxel response in NSCLC cell lines, which may provide a novel adjuvant strategy to paclitaxel in the treatment of lung cancer. By combining in vitro and in silico approaches, we identified STAT3 and RAP1A as direct targets that mediate the effect of miR-337-3p on paclitaxel sensitivity. Further investigation showed that miR-337-3p mimic also sensitizes cells to docetaxel, another member of the taxane family, and that STAT3 levels are significantly correlated with taxane resistance in lung cancer cell lines, suggesting that endogenous STAT3 expression is a determinant of intrinsic taxane resistance in lung cancer. The identification of a miR-337-3p as a modulator of cellular response to taxanes, and STAT3 and RAP1A as regulatory targets which mediate that response, defines a novel regulatory pathway modulating paclitaxel sensitivity in lung cancer cells, which may provide novel adjuvant strategies along with paclitaxel in the treatment of lung cancer and may also provide biomarkers for predicting paclitaxel response in NSCLC.
Collapse
Affiliation(s)
- Liqin Du
- Greehey Children's Cancer Research Institute, UT Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Department of Cellular and Structural Biology, UT Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Maria C. Subauste
- McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, Texas, United States of America
| | - Christopher DeSevo
- Division of Basic Sciences, Southwestern Graduate School of Biomedical Sciences, UT Southwestern Medical Center, Dallas, Texas, United States of America
| | - Zhenze Zhao
- Greehey Children's Cancer Research Institute, UT Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Michael Baker
- Division of Basic Sciences, Southwestern Graduate School of Biomedical Sciences, UT Southwestern Medical Center, Dallas, Texas, United States of America
| | - Robert Borkowski
- Division of Basic Sciences, Southwestern Graduate School of Biomedical Sciences, UT Southwestern Medical Center, Dallas, Texas, United States of America
| | - Jeoffrey J. Schageman
- McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, Texas, United States of America
| | - Rachel Greer
- Division of Basic Sciences, Southwestern Graduate School of Biomedical Sciences, UT Southwestern Medical Center, Dallas, Texas, United States of America
| | - Chin-Rang Yang
- Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, Texas, United States of America
| | - Milind Suraokar
- Department of Pathology, UT MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Ignacio I. Wistuba
- Department of Pathology, UT MD Anderson Cancer Center, Houston, Texas, United States of America
- Department of Thoracic/Head and Neck Medical Oncology, UT MD Anderson Cancer Center, Houston, Texas, United States of America
| | - Adi F. Gazdar
- Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, Texas, United States of America
- Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Dallas, Texas, United States of America
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas, United States of America
| | - John D. Minna
- Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, Texas, United States of America
- Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Dallas, Texas, United States of America
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, Texas, United States of America
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, United States of America
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas, United States of America
| | - Alexander Pertsemlidis
- Greehey Children's Cancer Research Institute, UT Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Department of Cellular and Structural Biology, UT Health Science Center at San Antonio, San Antonio, Texas, United States of America
- Department of Pediatrics, UT Health Science Center at San Antonio, San Antonio, Texas, United States of America
- * E-mail:
| |
Collapse
|
38
|
Rivas MA, Venturutti L, Huang YW, Schillaci R, Huang THM, Elizalde PV. Downregulation of the tumor-suppressor miR-16 via progestin-mediated oncogenic signaling contributes to breast cancer development. Breast Cancer Res 2012; 14:R77. [PMID: 22583478 PMCID: PMC3446340 DOI: 10.1186/bcr3187] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2011] [Revised: 04/29/2012] [Accepted: 05/14/2012] [Indexed: 12/19/2022] Open
Abstract
Introduction Experimental and clinical evidence points to a critical role of progesterone and the nuclear progesterone receptor (PR) in controlling mammary gland tumorigenesis. However, the molecular mechanisms of progesterone action in breast cancer still remain elusive. On the other hand, micro RNAs (miRNAs) are short ribonucleic acids which have also been found to play a pivotal role in cancer pathogenesis. The role of miRNA in progestin-induced breast cancer is poorly explored. In this study we explored progestin modulation of miRNA expression in mammary tumorigenesis. Methods We performed a genome-wide study to explore progestin-mediated regulation of miRNA expression in breast cancer. miR-16 expression was studied by RT-qPCR in cancer cell lines with silenced PR, signal transducer and activator of transcription 3 (Stat3) or c-Myc, treated or not with progestins. Breast cancer cells were transfected with the precursor of miR-16 and proliferation assays, Western blots or in vivo experiments were performed. Target genes of miR-16 were searched through a bioinformatical approach, and the study was focused on cyclin E. Reporter gene assays were performed to confirm that cyclin E 3'UTR is a direct target of miR-16. Results We found that nine miRNAs were upregulated and seven were downregulated by progestin in mammary tumor cells. miR-16, whose function as a tumor suppressor in leukemia has already been shown, was identified as one of the downregulated miRNAs in murine and human breast cancer cells. Progestin induced a decrease in miR-16 levels via the classical PR and through a hierarchical interplay between Stat3 and the oncogenic transcription factor c-Myc. A search for miR-16 targets showed that the CCNE1 gene, encoding the cell cycle regulator cyclin E, contains conserved putative miR-16 target sites in its mRNA 3' UTR region. We found that, similar to the molecular mechanism underlying progestin-modulated miR-16 expression, Stat3 and c-Myc participated in the induction of cyclin E expression by progestin. Moreover, overexpression of miR-16 abrogated the ability of progestin to induce cyclin E upregulation, revealing that cyclin E is a novel target of miR-16 in breast cancer. Overexpression of miR-16 also inhibited progestin-induced breast tumor growth in vitro and in vivo, demonstrating for the first time, a role for miR-16 as a tumor suppressor in mammary tumorigenesis. We also found that the ErbB ligand heregulin (HRG) downregulated the expression of miR-16, which then participates in the proliferative activity of HRG in breast tumor cells. Conclusions In this study, we reveal the first progestin-regulated miRNA expression profile and identify a novel role for miR-16 as a tumor suppressor in progestin- and growth factor-induced growth in breast cancer.
Collapse
Affiliation(s)
- Martin A Rivas
- Laboratory of Molecular Mechanisms of Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Vuelta de Obligado 2490, C1428ADN Buenos Aires, Argentina
| | | | | | | | | | | |
Collapse
|
39
|
Kaye S, Aamdal S, Jones R, Freyer G, Pujade-Lauraine E, de Vries EGE, Barriuso J, Sandhu S, Tan DSW, Hartog V, Kuenen B, Ruijter R, Kristensen GB, Nyakas M, Barrett S, Burke W, Pietersma D, Stuart M, Emeribe U, Boven E. Phase I study of saracatinib (AZD0530) in combination with paclitaxel and/or carboplatin in patients with solid tumours. Br J Cancer 2012; 106:1728-34. [PMID: 22531637 PMCID: PMC3364128 DOI: 10.1038/bjc.2012.158] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Background: As a prelude to combination studies aimed at resistance reversal, this dose-escalation/dose-expansion study investigated the selective Src kinase inhibitor saracatinib (AZD0530) in combination with carboplatin and/or paclitaxel. Methods: Patients with advanced solid tumours received saracatinib once-daily oral tablets in combination with either carboplatin AUC 5 every 3 weeks (q3w), paclitaxel 175 mg m−2 q3w, paclitaxel 80 mg m−2 every 1 week (q1w), or carboplatin AUC 5 plus paclitaxel 175 mg m−2 q3w. The primary endpoint was safety/tolerability. Results: A total of 116 patients received saracatinib 125 (N=20), 175 (N=44), 225 (N=40), 250 (N=9), or 300 mg (N=3). There were no clear dose-related trends within each chemotherapy regimen group in number or severity of adverse events (AEs). However, combining all groups, the occurrence of grade ⩾3 asthenic AEs (all causality) was dose-related (125 mg, 10% 175 mg, 20% ⩾225 mg, 33%), and grade ⩾3 neutropenia occurred more commonly at doses ⩾225 mg. There was no evidence that saracatinib affected exposure to carboplatin or paclitaxel, or vice versa. Objective responses were seen in 5 out of 44 patients (11%) receiving carboplatin plus paclitaxel q3w, and 5 out of 24 (21%) receiving paclitaxel q1w. Conclusion: Saracatinib doses up to 175 mg with paclitaxel with/without carboplatin showed acceptable toxicity in most patients, and are suitable for further trials.
Collapse
Affiliation(s)
- S Kaye
- Drug Development Unit, Royal Marsden Hospital, Downs Road, Sutton, Surrey SM2 5PT, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
de Hoon JPJ, Veeck J, Vriens BEPJ, Calon TGA, van Engeland M, Tjan-Heijnen VCG. Taxane resistance in breast cancer: a closed HER2 circuit? Biochim Biophys Acta Rev Cancer 2012; 1825:197-206. [PMID: 22280939 DOI: 10.1016/j.bbcan.2012.01.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2011] [Revised: 01/09/2012] [Accepted: 01/10/2012] [Indexed: 12/11/2022]
Abstract
Microtubule inhibitors, such as the taxanes docetaxel and paclitaxel, are commonly used drugs for the treatment of breast cancer. Although highly active in a large fraction of individuals a considerable number of patients show poor response due to either intrinsic or acquired drug resistance. Extensive research in the past identified several taxane resistance-related mechanisms being activated by pathologically altered single gene function. To date, however, a clinically relevant predictive biomarker for taxanes has not been derived yet from this knowledge, most likely due to the manifold of resistance mechanisms that may combine in one tumor, thereby fostering escape from taxane cytotoxicity. Here, we aimed to comprehensively review the current literature on taxane resistance mechanisms in breast cancer. Interestingly, besides altered microtubule physiology we identified the HER2 signaling cascade as a major dominator influencing several routes of cytotoxicity escape, such as cell survival, apoptosis, drug efflux, and drug metabolism. Furthermore, the transcription factor YBX-1, activated by HER2, facilitates a sustaining HER2 signaling feedback loop contributing to the establishment of cellular survival detours. In conclusion, taxane resistance in breast cancer follows a multiplex establishment of drug cytotoxicity escape routes, which may be most efficiently therapeutically targeted by interference with their mutually governing signaling nodes.
Collapse
Affiliation(s)
- Joep P J de Hoon
- Division of Medical Oncology, Department of Internal Medicine, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | | | | | | | | | | |
Collapse
|
41
|
Proietti CJ, Béguelin W, Flaqué MCD, Cayrol F, Rivas MA, Tkach M, Charreau EH, Schillaci R, Elizalde PV. Novel role of signal transducer and activator of transcription 3 as a progesterone receptor coactivator in breast cancer. Steroids 2011; 76:381-92. [PMID: 21184768 DOI: 10.1016/j.steroids.2010.12.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2010] [Revised: 12/13/2010] [Accepted: 12/14/2010] [Indexed: 12/19/2022]
Abstract
Interactions between progesterone receptor (PR) and signal transducer and activator of transcription 3 (Stat3)-mediated signaling pathways have already been described. In the present study, we explored the capacity of Stat3 to functionally interact with progesterone receptor (PR) and modulate PR transcriptional activation in breast cancer cells. We found that the synthetic progestin medroxyprogesterone acetate (MPA) induced the association of a PR/Stat3 complex in which Stat3 acts as a coactivator of PR. We demonstrated that Stat3 activation is required for MPA modulation of the endogenous genes bcl-X and p21(CIP1) which are involved in MPA-induced cell cycle regulation. Stat3 activity as a coactivator of PR was observed in both the classical and nonclassical ligand activated-PR transcriptional mechanisms, since the effects described were identified in the bcl-X promoter which contains a progesterone responsive element and in the p21(CIP1) promoter which carries Sp1 binding sites where PR is recruited via the transcription factor Sp1. The data herein presented identifies a potential therapeutic intervention for PR-positive breast tumors consisting of targeting Stat3 function or PR/Stat3 interaction which will result in the inhibition of PR function.
Collapse
Affiliation(s)
- Cecilia J Proietti
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Vuelta de Obligado 2490, Buenos Aires C1428ADN, Argentina.
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Chandra-Kuntal K, Singh SV. Diallyl trisulfide inhibits activation of signal transducer and activator of transcription 3 in prostate cancer cells in culture and in vivo. Cancer Prev Res (Phila) 2010; 3:1473-83. [PMID: 20959517 DOI: 10.1158/1940-6207.capr-10-0123] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Signal transducer and activator of transcription 3 (STAT3) is an oncogenic transcription factor implicated in prostate carcinogenesis. The present study shows that diallyl trisulfide (DATS), a promising cancer-chemopreventive constituent of processed garlic, inhibits phosphorylation of STAT3 in prostate cancer cells in culture and in vivo. Exposure of DU145 and LNCaP human prostate cancer cells to growth-suppressive and pharmacologically relevant concentrations of DATS (20 and 40 μmol/L) resulted in suppression of constitutive (DU145) as well as interleukin-6 (IL-6)-induced (LNCaP) phosphorylation of STAT3 (Tyr(705)), which correlated with inhibition of Janus-activated kinase 2 phosphorylation. Constitutive and/or IL-6-induced nuclear translocation of pSTAT3 and STAT3 dimerization was also markedly inhibited on treatment with DATS in both cell lines. Inhibition of prostate cancer development in transgenic adenocarcinoma of mouse prostate mice by gavage of DATS correlated with a visible decrease in the levels of pSTAT3. Interestingly, the IL-6-mediated activation of STAT3 largely failed to confer protection against proapoptotic response to DATS in both cells. Likewise, DATS-mediated inhibition of cell migration was either not affected or minimally reversed by IL-6 treatment or ectopic expression of constitutively active STAT3. In conclusion, the present study indicates that DATS treatment suppresses STAT3 phosphorylation in prostate cancer cells in culture and in vivo, but activation of this oncogenic transcription factor is largely dispensable for cellular responses to DATS. Ability of DATS to overcome STAT3 activation is a therapeutic advantage for this chemopreventive agent.
Collapse
Affiliation(s)
- Kumar Chandra-Kuntal
- Department of Pharmacology and Chemical Biology, and University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, USA
| | | |
Collapse
|
43
|
Progesterone receptor induces ErbB-2 nuclear translocation to promote breast cancer growth via a novel transcriptional effect: ErbB-2 function as a coactivator of Stat3. Mol Cell Biol 2010; 30:5456-72. [PMID: 20876300 DOI: 10.1128/mcb.00012-10] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Progesterone receptor (PR) and ErbB-2 bidirectional cross talk participates in breast cancer development. Here, we identified a new mechanism of the PR and ErbB-2 interaction involving the PR induction of ErbB-2 nuclear translocation and the assembly of a transcriptional complex in which ErbB-2 acts as a coactivator of Stat3. We also highlighted that the function of ErbB-2 as a Stat3 coactivator drives progestin-induced cyclin D1 promoter activation. Notably, PR is also recruited together with Stat3 and ErbB-2 to the cyclin D1 promoter, unraveling a new and unexpected nonclassical PR genomic mechanism. The assembly of the nuclear Stat3/ErbB-2 transcriptional complex plays a key role in the proliferation of breast tumors with functional PR and ErbB-2. Our findings reveal a novel therapeutic intervention for PR- and ErbB-2-positive breast tumors via the specific blockage of ErbB-2 nuclear translocation.
Collapse
|
44
|
Lee J, Hahm ER, Singh SV. Withaferin A inhibits activation of signal transducer and activator of transcription 3 in human breast cancer cells. Carcinogenesis 2010; 31:1991-8. [PMID: 20724373 DOI: 10.1093/carcin/bgq175] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
We have shown previously that withaferin A (WA), a promising anticancer constituent of Ayurvedic medicine plant Withania somnifera, inhibits growth of human breast cancer cells in culture and in vivo in association with apoptosis induction. The present study builds on these observations and demonstrates that WA inhibits constitutive as well as interleukin-6 (IL-6)-inducible activation of signal transducer and activator of transcription 3 (STAT3), which is an oncogenic transcription factor activated in many human malignancies including breast cancer. The WA treatment (2 and 4 μM) decreased constitutive (MDA-MB-231) and/or IL-6-inducible (MDA-MB-231 and MCF-7) phosphorylation of STAT3 (Tyr(705)) and its upstream regulator Janus-activated kinase 2 (JAK2; Tyr(1007/1008)) in MDA-MB-231, which was accompanied by suppression of their protein levels especially at the higher concentration. Exposure of MDA-MB-231 or MCF-7 cells to WA also resulted in suppression of (i) transcriptional activity of STAT3 with or without IL-6 stimulation in both cells; (ii) dimerization of STAT3 (MDA-MB-231) and (iii) nuclear translocation of Tyr(705)-phosphorylated STAT3 in both cells. To our surprise, the IL-6-stimulation, either before or after WA treatment, did not have an appreciable effect on WA-mediated apoptosis in MDA-MB-231 or MCF-7 cell line. The IL-6-stimulated activation of STAT3 conferred a modest protection against WA-mediated suppression of MDA-MB-231 cell invasion. General implication of these findings is that WA can trigger apoptosis and largely inhibit cell migration/invasion of breast cancer cells even after IL-6-induced activation of STAT3, which should be viewed as a therapeutic advantage for this agent.
Collapse
Affiliation(s)
- Joomin Lee
- Department of Pharmacology & Chemical Biology, and University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, 5117 Centre Avenue, Pittsburgh, PA 15213, USA
| | | | | |
Collapse
|
45
|
Elevated expression of erbB3 confers paclitaxel resistance in erbB2-overexpressing breast cancer cells via upregulation of Survivin. Oncogene 2010; 29:4225-36. [DOI: 10.1038/onc.2010.180] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
46
|
Identification of an ovarian clear cell carcinoma gene signature that reflects inherent disease biology and the carcinogenic processes. Oncogene 2010; 29:1741-52. [PMID: 20062075 DOI: 10.1038/onc.2009.470] [Citation(s) in RCA: 138] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Ovarian clear cell carcinoma (OCCC) shows unique clinical features including an association with endometriosis and poor prognosis. We previously reported that the contents of endometriotic cysts, especially high concentrations of free iron, are a possible cause of OCCC carcinogenesis through iron-induced persistent oxidative stress. In this study, we conducted gene expression microarray analysis using 38 ovarian cancer cell lines and identified genes commonly expressed in both OCCC cell lines and clinical samples, which comprise an OCCC gene signature. The OCCC signature reproducibly predicts OCCC specimens in other microarray data sets, suggesting that this gene profile reflects the inherent biological characteristics of OCCC. The OCCC signature contains known markers of OCCC, such as hepatocyte nuclear factor-1beta (HNF-1beta) and versican (VCAN), and other genes that reflect oxidative stress. Expression of OCCC signature genes was induced by treatment of immortalized ovarian surface epithelial cells with the contents of endometriotic cysts, indicating that the OCCC signature is largely dependent on the tumor microenvironment. Induction of OCCC signature genes is at least in part epigenetically regulated, as we found hypomethylation of HNF-1beta and VCAN in OCCC cell lines. This genome-wide study indicates that the tumor microenvironment induces specific gene expression profiles that contribute to the development of distinct cancer subtypes.
Collapse
|
47
|
Li SH, Hawthorne VS, Neal CL, Sanghera S, Xu J, Yang J, Guo H, Steeg PS, Yu D. Upregulation of neutrophil gelatinase-associated lipocalin by ErbB2 through nuclear factor-kappaB activation. Cancer Res 2010; 69:9163-8. [PMID: 19951994 DOI: 10.1158/0008-5472.can-09-2483] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
ErbB2 (HER2, neu) is a receptor tyrosine kinase overexpressed in about 25% of invasive breast carcinomas. Neutrophil gelatinase-associated lipocalin (NGAL) is a secreted glycoprotein expressed in a variety of cancers, including breast carcinomas. NGAL can inhibit erythroid cell production, leading to anemia. Anemia usually occurs in cancer patients and negatively affects quality of life. However, current treatment for cancer-related anemia has potential complications. ErbB2, NGAL, and anemia have all been associated with increased metastasis and poor prognosis in breast cancer patients, although the relationship between ErbB2 and NGAL expression is not clear. Here, using breast cancer cell lines in vitro and transgenic mice carrying the activated c-neu oncogene driven by a mouse mammary tumor virus (MMTV-neu) in vivo, we show that ErbB2 overexpression leads to NGAL upregulation, which is dependent on nuclear factor-kappaB (NF-kappaB) activity. MMTV-neu transgenic mice developed anemia after tumor onset, and anemia progression could be partially arrested by a NF-kappaB inhibitor and ErbB2-targeted therapy. Taken together, upregulation of NGAL by ErbB2 through NF-kappaB activation is involved in cancer-related anemia, and the ErbB2, NF-kappaB, and NGAL pathways may serve as potential therapeutic targets for cancer-related anemia.
Collapse
Affiliation(s)
- Shau-Hsuan Li
- Department of Molecular and Cellular Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Bonner JA, Trummell HQ, Willey CD, Plants BA, Raisch KP. Inhibition of STAT-3 results in radiosensitization of human squamous cell carcinoma. Radiother Oncol 2009; 92:339-44. [PMID: 19616333 DOI: 10.1016/j.radonc.2009.06.022] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2009] [Revised: 06/15/2009] [Accepted: 06/24/2009] [Indexed: 11/17/2022]
Abstract
BACKGROUND Signal transducer and activator of transcription-3 (STAT-3) is a downstream component of the Epidermal Growth Factor Receptor (EGFr) signaling process that may facilitate the resistance of tumor cells to conventional cancer treatments. Studies were performed to determine if inhibition of this downstream protein produces radiosensitization. METHODS/RESULTS A431 cells (human squamous cell carcinoma cells with EGFr overexpression) were found to be sensitized to radiation after treatment with STAT-3 small interfering RNA (siRNA). Therefore, a short hairpin RNA (shRNA) against STAT-3 was designed and cloned into a pBABE vector system modified for shRNA expression. Following transfection, clone 2.1 was selected for further study as it showed a dramatic reduction of STAT-3 protein (and mRNA) when compared to A431 parental cells or a negative control shRNA cell line (transfected with STAT-3 shRNA with 2 base pairs mutated). A431 2.1 showed doubling times of 25-31h as compared to 18-24h for the parental cell line. The A431 shRNA knockdown STAT-3 cells A431 were more sensitive to radiation than A431 parental or negative STAT-3 control cells. CONCLUSION A431 cells stably transfected with shRNA against STAT-3 resulted in enhanced radiosensitivity. Further work will be necessary to determine whether the inhibition of STAT-3 phosphorylation is a necessary step for the radiosensitization that is induced by the inhibition of EGFr.
Collapse
Affiliation(s)
- James A Bonner
- Department of Radiation Oncology, The University of Alabama at Birmingham, AL 35233, USA.
| | | | | | | | | |
Collapse
|