1
|
Mohamed Allam D, Kasem H, Hegazy A, Mahmoud SF. Role of CTLA4 and pSTAT3 Immunostaining in Prognosis and Treatment of the Colorectal Carcinoma. IRANIAN JOURNAL OF PATHOLOGY 2024; 19:89-102. [PMID: 38864078 PMCID: PMC11164302 DOI: 10.30699/ijp.2024.2009619.3158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 12/07/2023] [Indexed: 06/13/2024]
Abstract
Background & Objective Colorectal carcinoma (CRC) is the third leading cause of cancer-caused death worldwide and constitutes about 6.48% of all malignancies in Egypt. Studying the molecular profile of CRC is essential for developing targeted therapies. STAT3 and CTLA4 expression are considered as molecular abnormalities involved in the CRC progression and chemo-resistance. Therefore, they could be used as potential therapeutic targets. This study aimed to evaluate pSTAT3 and CTLA4 expression levels and their possible roles as prognostic and predictive biomarkers in CRC using immunohistochemistry (IHC). Methods This retrospective study included 113 CRC patients. Tissue microarrays were constructed, followed by pSTAT3 and CTLA4 antibodies immunostaining. Their expression was assessed and compared with the clinicopathological parameters and survival data. Results Both pSTAT3 and CTLA4 overexpression were significantly associated with poor prognostic parameters, such as the presence of distant metastasis (P=0.02 & 0.03), high grade (P<0.001 & 0.03), high mitotic count (P<0.001 & 0.03), high tumor budding group (P=0.008 & 0.04), infiltrating tumor border (P<0.001 & 0.007) respectively, and advanced pathological stage with pSTAT3 (P=0.02). A significant association was found between overexpression of both markers and short overall survival. Correlations between the H-score of pSTAT3 and CTLA4 in CRC showed a significant positive correlation (P<0.001). Conclusion STAT3 and CTLA4 positivity may be linked to the development and progression of the CRC, and they may provide potential prognostic indicators and therapeutic targets for CRC patients.
Collapse
Affiliation(s)
- Dina Mohamed Allam
- Department of Pathology, Faculty of Medicine, Menoufia University, Shebin El Kom, Egypt
| | - Hend Kasem
- Department of Pathology, Faculty of Medicine, Menoufia University, Shebin El Kom, Egypt
| | - Amira Hegazy
- Department of Clinical Oncology and Nuclear Medicine, Faculty of Medicine, Menoufia University, Egypt
| | - Shereen F Mahmoud
- Department of Pathology, Faculty of Medicine, Menoufia University, Shebin El Kom, Egypt
| |
Collapse
|
2
|
Morato A, Accornero P, Hovey RC. ERBB Receptors and Their Ligands in the Developing Mammary Glands of Different Species: Fifteen Characters in Search of an Author. J Mammary Gland Biol Neoplasia 2023; 28:10. [PMID: 37219601 DOI: 10.1007/s10911-023-09538-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 04/26/2023] [Indexed: 05/24/2023] Open
Abstract
The ERBB tyrosine kinase receptors and their ligands belong to a complex family that has diverse biological effects and expression profiles in the developing mammary glands, where its members play an essential role in translating hormone signals into local effects. While our understanding of these processes stems mostly from mouse models, there is the potential for differences in how this family functions in the mammary glands of other species, particularly in light of their unique histomorphological features. Herein we review the postnatal distribution and function of ERBB receptors and their ligands in the mammary glands of rodents and humans, as well as for livestock and companion animals. Our analysis highlights the diverse biology for this family and its members across species, the regulation of their expression, and how their roles and functions might be modulated by varying stromal composition and hormone interactions. Given that ERBB receptors and their ligands have the potential to influence processes ranging from normal mammary development to diseased states such as cancer and/or mastitis, both in human and veterinary medicine, a more complete understanding of their biological functions should help to direct future research and the identification of new therapeutic targets.
Collapse
Affiliation(s)
- Alessia Morato
- Department of Animal Science, University of California, Davis, One Shields Avenue, Davis, CA, 95616, USA.
| | - Paolo Accornero
- Department of Veterinary Science, University of Turin, Largo Paolo Braccini 2, Grugliasco, TO, 10095, Italy
| | - Russell C Hovey
- Department of Animal Science, University of California, Davis, One Shields Avenue, Davis, CA, 95616, USA
| |
Collapse
|
3
|
Wright RHG, Vastolo V, Oliete JQ, Carbonell-Caballero J, Beato M. Global signalling network analysis of luminal T47D breast cancer cells in response to progesterone. Front Endocrinol (Lausanne) 2022; 13:888802. [PMID: 36034422 PMCID: PMC9403329 DOI: 10.3389/fendo.2022.888802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Breast cancer cells enter into the cell cycle following progestin exposure by the activation of signalling cascades involving a plethora of enzymes, transcription factors and co-factors that transmit the external signal from the cell membrane to chromatin, ultimately leading to a change of the gene expression program. Although many of the events within the signalling network have been described in isolation, how they globally team up to generate the final cell response is unclear. METHODS In this study we used antibody microarrays and phosphoproteomics to reveal a dynamic global signalling map that reveals new key regulated proteins and phosphor-sites and links between previously known and novel pathways. T47D breast cancer cells were used, and phospho-sites and pathways highlighted were validated using specific antibodies and phenotypic assays. Bioinformatic analysis revealed an enrichment in novel signalling pathways, a coordinated response between cellular compartments and protein complexes. RESULTS Detailed analysis of the data revealed intriguing changes in protein complexes involved in nuclear structure, epithelial to mesenchyme transition (EMT), cell adhesion, as well as transcription factors previously not associated with breast cancer cell proliferation. Pathway analysis confirmed the key role of the MAPK signalling cascade following progesterone and additional hormone regulated phospho-sites were identified. Full network analysis shows the activation of new signalling pathways previously not associated with progesterone signalling in T47D breast cancer cells such as ERBB and TRK. As different post-translational modifications can mediate complex crosstalk mechanisms and massive PARylation is also rapidly induced by progestins, we provide details of important chromatin regulatory complexes containing both phosphorylated and PARylated proteins. CONCLUSIONS This study contributes an important resource for the scientific community, as it identifies novel players and connections meaningful for breast cancer cell biology and potentially relevant for cancer management.
Collapse
Affiliation(s)
- Roni H. G. Wright
- Center for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Basic Sciences Department, Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, Barcelona, Spain
- *Correspondence: Roni H. G. Wright, ; Miguel Beato,
| | - Viviana Vastolo
- Center for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Javier Quilez Oliete
- Center for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - José Carbonell-Caballero
- Center for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Miguel Beato
- Center for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- *Correspondence: Roni H. G. Wright, ; Miguel Beato,
| |
Collapse
|
4
|
Mazumder A, Shiao S, Haricharan S. HER2 Activation and Endocrine Treatment Resistance in HER2-negative Breast Cancer. Endocrinology 2021; 162:6329618. [PMID: 34320193 PMCID: PMC8379900 DOI: 10.1210/endocr/bqab153] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Indexed: 11/19/2022]
Abstract
The lethality of estrogen receptor alpha positive (ER+) breast cancer, which is often considered to have better prognosis than other subtypes, is defined by resistance to the standard of care endocrine treatment. Relapse and metastasis are inevitable in almost every patient whose cancer is resistant to endocrine treatment. Therefore, understanding the underlying causes of treatment resistance remains an important biological and clinical focus of research in this area. Growth factor receptor pathway activation, specifically HER2 activation, has been identified as 1 mechanism of endocrine treatment resistance across a range of experimental model systems. However, clinical trials conducted to test whether targeting HER2 benefits patients with endocrine treatment-resistant ER+ breast cancer have consistently and disappointingly shown mixed results. One reason for the failure of these clinical trials could be the complexity of crosstalk between ER, HER2, and other growth factor receptors and the fluidity of HER2 activation in these cells, which makes it challenging to identify stratifiers for this targeted intervention. In the absence of stratifiers that can be assayed at diagnosis to allow prospective tailoring of HER2 inhibition to the right patients, clinical trials will continue to disappoint. To understand stratifiers, it is important that the field invests in key understudied areas of research including characterization of the tumor secretome and receptor activation in response to endocrine treatment, and mapping the ER-HER2 growth factor network in the normal and developing mammary gland. Understanding these mechanisms further is critical to improving outcomes for the hard-to-treat endocrine treatment-resistant ER+ breast cancer cohort.
Collapse
Affiliation(s)
- Aloran Mazumder
- Aging and Cancer Immuno-oncology, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Stephen Shiao
- Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Svasti Haricharan
- Aging and Cancer Immuno-oncology, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
- Correspondence: Svasti Haricharan, PhD, Sanford Burnham Prebys, 10901 N Torrey Pines Rd, La Jolla, CA, USA.
| |
Collapse
|
5
|
Abstract
Neuregulins, members of the largest subclass of growth factors of the epidermal growth factor family, mediate a myriad of cellular functions including survival, proliferation, and differentiation in normal tissues through binding to receptor tyrosine kinases of the ErbB family. However, aberrant neuregulin signaling in the tumor microenvironment is increasingly recognized as a key player in initiation and malignant progression of human cancers. In this chapter, we focus on the role of neuregulin signaling in the hallmarks of cancer, including cancer initiation and development, metastasis, as well as therapeutic resistance. Moreover, role of neuregulin signaling in the regulation of tumor microenvironment and targeting of neuregulin signaling in cancer from the therapeutic perspective are also briefly discussed.
Collapse
|
6
|
Hua G, Bergon A, Cauchy P, Kahn-Perlès B, Bertucci F, Birnbaum D, Benkirane-Jessel N, Imbert J. ERBB2b mRNA isoform encodes a nuclear variant of the ERBB2 oncogene in breast cancer. J Cell Biochem 2020; 121:4870-4886. [PMID: 32628295 DOI: 10.1002/jcb.29762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 05/05/2020] [Indexed: 11/10/2022]
Abstract
The presence of nuclear ERBB2 receptor-type tyrosine kinase is one of the causes of the resistance to membrane ERBB2-targeted therapy in breast cancers. It has been previously reported that this nuclear location arises through at least two different mechanisms: proteolytic shedding of the extracellular domain of the full-length receptor and translation of the messenger RNA (mRNA)-encoding ERBB2 from internal initiation codons. Here, we report a new mechanism and function where a significant portion of nuclear ERBB2 results from the translation of the variant ERBB2 mRNA under the transcriptional control of a distal promoter that is actively used in breast cancer cells. We show that both membrane ERBB2a and nuclear ERBB2b isoforms are prevalently expressed in breast cancer cell lines and carcinoma samples. The ERBB2b isoform, which is translated from mRNA variant 2, can directly translocate into the nucleus due to the lack of the signal peptide which is required for an intermediate membrane location. Small interfering RNA-mediated gene silencing showed that ERBB2b can repress ERBB2a expression, encoded by variant 1, whereas ERBB2a activates ERBB2b. Nuclear ERBB2 binding to its own promoter was revealed by chromatin immunoprecipitation assay. Altogether, our results provide new insights into the origin and function of nuclear ERBB2 where it can participate at the same time in a positive or a negative feedback autoregulatory loop, dependent on which of its promoters this bona fide transcription factor is acting. They also provide a new understanding for the resistance to therapies targeting the membrane-anchored ERBB2 in breast cancer.
Collapse
Affiliation(s)
- Guoqiang Hua
- INSERM UMR1090 TAGC, Aix-Marseille University, Marseille, France
- INSERM UMR1260, RNM, FMTS, Strasbourg, France
- Faculté de Chirurgie Dentaire de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Aurélie Bergon
- INSERM UMR1090 TAGC, Aix-Marseille University, Marseille, France
| | - Pierre Cauchy
- INSERM UMR1090 TAGC, Aix-Marseille University, Marseille, France
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg im Breisgau, Germany
| | | | - François Bertucci
- Laboratoire d'Oncologie Prédictive, CRCM, CNRS UMR 7258, INSERM U1068, Institut Paoli-Calmettes, Aix-Marseille University, Marseille, France
| | - Daniel Birnbaum
- Laboratoire d'Oncologie Prédictive, CRCM, CNRS UMR 7258, INSERM U1068, Institut Paoli-Calmettes, Aix-Marseille University, Marseille, France
| | - Nadia Benkirane-Jessel
- INSERM UMR1260, RNM, FMTS, Strasbourg, France
- Faculté de Chirurgie Dentaire de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Jean Imbert
- INSERM UMR1090 TAGC, Aix-Marseille University, Marseille, France
| |
Collapse
|
7
|
Canonical ErbB-2 isoform and ErbB-2 variant c located in the nucleus drive triple negative breast cancer growth. Oncogene 2020; 39:6245-6262. [PMID: 32843720 DOI: 10.1038/s41388-020-01430-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 08/07/2020] [Accepted: 08/11/2020] [Indexed: 02/06/2023]
Abstract
Triple negative breast cancer (TNBC) refers to tumors that do not express clinically significant levels of estrogen and progesterone receptors, and lack membrane overexpression or gene amplification of ErbB-2/HER2, a receptor tyrosine kinase. Transcriptome and proteome heterogeneity of TNBC poses a major challenge to precision medicine. Clinical biomarkers and targeted therapies for this disease remain elusive, so chemotherapy has been the standard of care for early and metastatic TNBC. Our present findings placed ErbB-2 in an unanticipated scenario: the nucleus of TNBC (NErbB-2). Our study on ErbB-2 alternative splicing events, using a PCR-sequencing approach combined with an RNA interference strategy, revealed that TNBC cells express either the canonical (wild-type) ErbB-2, encoded by transcript variant 1, or the non-canonical ErbB-2 isoform c, encoded by alternative variant 3 (RefSeq), or both. These ErbB-2 isoforms function in the nucleus as transcription factors. Evicting both from the nucleus or silencing isoform c only, blocks TN cell and tumor growth. This reveals not only NErbB-2 canonical and alternative isoforms role as targets of therapy in TNBC, but also isoform c dominant oncogenic potential. Furthermore, we validated our findings in the clinic and observed that NErbB-2 correlates with poor prognosis in primary TN tumors, disclosing NErbB-2 as a novel biomarker for TNBC. Our discoveries challenge the present scenario of drug development for personalized BC medicine that focuses on wild-type RefSeq proteins, which conserve the canonical domains and are located in their classical cellular compartments.
Collapse
|
8
|
Nuclear PDCD4 Expression Defines a Subset of Luminal B-Like Breast Cancers with Good Prognosis. Discov Oncol 2020; 11:218-239. [PMID: 32632815 DOI: 10.1007/s12672-020-00392-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 06/19/2020] [Indexed: 02/07/2023] Open
Abstract
The hormone receptor-positive (estrogen and/or progesterone receptor (PR)-positive) and HER2-negative breast cancer (BC) subtype is a biologically heterogeneous entity that includes luminal A-like (LumA-like) and luminal B-like (LumB-like) subtypes. Decreased PR levels is a distinctive biological feature of LumB-like tumors. These tumors also show reduced sensitivity to endocrine therapies and poorer prognosis than LumA-like tumors. Identification of biomarkers to accurately predict disease relapse in these subtypes is crucial in order to select effective therapies. We identified the tumor suppressor PDCD4 (programmed cell death 4), located in the nucleus (NPDCD4), as an independent prognostic factor of good clinical outcome in LumA-like and LumB-like subtypes. NPDCD4-positive LumB-like tumors presented overall and disease-free survival rates comparable to those of NPDCD4-positive LumA-like tumors, indicating that NPDCD4 improves the outcome of LumB-like patients. In contrast, NPDCD4 loss increased the risk of disease recurrence and death in LumB-like compared with LumA-like tumors. This, along with our results showing that LumB-like tumors present lower NPDCD4 positivity than LumA-like tumors, suggests that NPDCD4 loss contributes to endocrine therapy resistance in LumB-like BCs. We also revealed that PR induces PDCD4 transcription in LumB-like BC, providing a mechanistic explanation to the low PDCD4 levels in LumB-like BCs lacking PR. Finally, PDCD4 silencing enhanced BC cell survival in a patient-derived explant model of LumB-like disease. Our discoveries highlight NPDCD4 as a novel biomarker in LumA- and LumB-like subtypes, which could be included in the panel of immunohistochemical markers used in the clinic to accurately predict the prognosis of LumB-like tumors.
Collapse
|
9
|
Cenciarini ME, Proietti CJ. Molecular mechanisms underlying progesterone receptor action in breast cancer: Insights into cell proliferation and stem cell regulation. Steroids 2019; 152:108503. [PMID: 31562879 DOI: 10.1016/j.steroids.2019.108503] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 09/13/2019] [Accepted: 09/19/2019] [Indexed: 02/07/2023]
Abstract
The ovarian steroid hormone progesterone and its nuclear receptor, the Progesterone Receptor (PR), play an essential role in the regulation of cell proliferation and differentiation in the mammary gland. In addition, experimental and clinical evidence demonstrate their critical role in controlling mammary gland tumorigenesis and breast cancer development. When bound to its ligand, the main action of PR is as a transcription factor, which regulates the expression of target genes networks. PR also activates signal transduction pathways through a rapid or non-genomic mechanism in breast cancer cells, an event that is fully integrated with its genomic effects. This review summarizes the molecular mechanisms of the ligand-activated PR actions that drive epithelial cell proliferation and the regulation of the stem cell population in the normal breast and in breast cancer.
Collapse
Affiliation(s)
- Mauro E Cenciarini
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Vuelta de Obligado 2490, Buenos Aires C1428ADN, Argentina
| | - Cecilia J Proietti
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Vuelta de Obligado 2490, Buenos Aires C1428ADN, Argentina.
| |
Collapse
|
10
|
Schimmack S, Yang Y, Felix K, Herbst M, Li Y, Schenk M, Bergmann F, Hackert T, Strobel O. C-reactive protein (CRP) promotes malignant properties in pancreatic neuroendocrine neoplasms. Endocr Connect 2019; 8:1007-1019. [PMID: 31234146 PMCID: PMC6652262 DOI: 10.1530/ec-19-0132] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 06/24/2019] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Elevated pre-operative C-reactive protein (CRP) serum values have been reported to be associated with poor overall survival for patients with pancreatic neuroendocrine neoplasms (pNEN). The aim of this study was to identify mechanisms linking CRP to poor prognosis in pNEN. METHODS The malignant properties of pNENs were investigated using the human pNEN cell-lines BON1 and QGP1 exposed to CRP or IL-6. Analyses were performed by ELISA, Western blot, flow cytometry and immunocytochemistry as well as invasion and proliferation assays. To compare cytokine profiles and CRP levels, 76 serum samples of pNEN patients were analyzed using Luminex technology. In parallel, the expression of CRP and growth signaling pathway proteins was assessed on cell lines and paraffin-embedded primary pNEN. RESULTS In BON1 and QGP1 cells, inflammation (exposure to IL-6) significantly upregulated CRP expression and secretion as well as migratory properties. CRP stimulation of BON1 cells increased IL-6 secretion and invasion. This was accompanied by activation/phosphorylation of the ERK, AKT and/or STAT3 pathways. Although known CRP receptors - CD16, CD32 and CD64 - were not detected on BON1 cells, CRP uptake of pNEN cells was shown after CRP exposure. In patients, increased pre-operative CRP levels (≥5 mg/L) were associated with significantly higher serum levels of IL-6 and G-CSF, as well as with an increased CRP expression and ERK/AKT/STAT3 phosphorylation in pNEN tissue. CONCLUSION The malignant properties of pNEN cells can be stimulated by CRP and IL-6 promoting ERK/AKT/STAT pathways activation as well as invasion, thus linking systemic inflammation and poor prognosis.
Collapse
Affiliation(s)
- Simon Schimmack
- European Pancreas Center, Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Yongchao Yang
- European Pancreas Center, Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
- Department of Burns and Plastic Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Klaus Felix
- European Pancreas Center, Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Markus Herbst
- European Pancreas Center, Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Yixiong Li
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Miriam Schenk
- European Pancreas Center, Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Frank Bergmann
- Institute of Pathology, Heidelberg University, Heidelberg, Germany
| | - Thilo Hackert
- European Pancreas Center, Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Oliver Strobel
- European Pancreas Center, Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
11
|
Zhao Z, Li L, Du P, Ma L, Zhang W, Zheng L, Lan B, Zhang B, Ma F, Xu B, Zhan Q, Song Y. Transcriptional Downregulation of miR-4306 serves as a New Therapeutic Target for Triple Negative Breast Cancer. Theranostics 2019; 9:1401-1416. [PMID: 30867840 PMCID: PMC6401504 DOI: 10.7150/thno.30701] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 12/23/2018] [Indexed: 12/31/2022] Open
Abstract
Rationale: Triple-negative breast cancer (TNBC) is characterized by the absence of estrogen receptor alpha (ER-α), human epidermal growth factor receptor 2 (HER2) and progesterone receptor (PR) expression, but the effect of lacking the three factors on TNBC is unclear. Whether loss of the three factors contributes to deregulate genes that participate in the progress of TNBC remains unknown. Methods: We performed microRNA arrays and comprehensive analysis to screen for miRNAs that are transcriptionally regulated by ER-α, HER2 and PR. Functional assays and molecular mechanism studies were used to investigate the role of miR-4306 in TNBC. An orthotopic mouse model of TNBC was used to evaluate the therapeutic potential of a cholesterol-conjugated miR-4306 mimic. Results: We found that miR-4306 is transcriptionally regulated by ER-α, HER2 and PR, and the downregulation of miR-4306 in TNBC is caused by the loss of ER-α, HER2 and PR. Clinically, low miR-4306 expression is strongly associated with lymph node metastasis and poor survival for TNBC. Upregulation of miR-4306 greatly suppresses TNBC cell proliferation, migration and invasion and abrogates angiogenesis and lymphangiogenesis in vitro. According to in vivo models, miR-4306 overexpression considerably inhibits TNBC growth, lung metastasis, angiogenesis and lymph node metastasis. Mechanistic analyses indicate that miR-4306 directly targets SIX1/Cdc42/VEGFA to inactivate the signaling pathways mediated by SIX1/Cdc42/VEGFA. Finally, the orthotopic mouse model of TNBC reveals that miR-4306 mimic can be used for TNBC treatment in combination with cisplatin. Conclusions: Our findings suggest that miR-4306 acts as a tumor suppressor in TNBC and is a potential therapeutic target for TNBC treatment.
Collapse
Affiliation(s)
- Zitong Zhao
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Lin Li
- BGI-Shenzhen, Shenzhen, Guangdong 518083, China
| | - Peina Du
- BGI-Shenzhen, Shenzhen, Guangdong 518083, China
| | - Liying Ma
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Weimin Zhang
- Laboratory of Molecular Oncology, Peking University Cancer Hospital, Beijing 100142, China
| | - Leilei Zheng
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Bo Lan
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Bailin Zhang
- Department of Breast Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Fei Ma
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Bo Xu
- Breast Cancer Center and the Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Qimin Zhan
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
- Laboratory of Molecular Oncology, Peking University Cancer Hospital, Beijing 100142, China
| | - Yongmei Song
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| |
Collapse
|
12
|
Cordo Russo RI, Chervo MF, Madera S, Charreau EH, Elizalde PV. Nuclear ErbB-2: a Novel Therapeutic Target in ErbB-2-Positive Breast Cancer? Discov Oncol 2019; 10:64-70. [PMID: 30656558 DOI: 10.1007/s12672-018-0356-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 12/12/2018] [Indexed: 12/20/2022] Open
Abstract
Membrane overexpression of ErbB-2 (MErbB-2), a member of the ErbB family of receptor tyrosine kinases, occurs in 15-20% of breast cancers (BC) and constitutes a therapeutic target in this BC subtype (ErbB-2-positive). Although MErbB-2-targeted therapies have significantly improved patients' clinical outcome, resistance to available drugs is still a major issue in the clinic. Lack of accurate biomarkers for predicting responses to anti-ErbB-2 drugs at the time of diagnosis is also an important unresolved issue. Hence, a better understanding of the ErbB-2 signaling pathway constitutes a critical task in the battle against BC. In its canonical mechanism of action, MErbB-2 activates downstream signaling pathways, which transduce its proliferative effects in BC. The dogma of ErbB-2 mechanism of action has been challenged by the demonstration that MErbB-2 migrates to the nucleus, where it acts as a transcriptional regulator. Accumulating findings demonstrate that nuclear ErbB-2 (NErbB-2) is involved in BC growth and metastasis. Emerging evidence also reveal a role of NErbB-2 in the response to available anti-MErbB-2 agents. Here, we will review NErbB-2 function in BC and will particularly discuss the role of NErbB-2 as a novel target for therapy in ErbB-2-positive BC.
Collapse
Affiliation(s)
- Rosalía I Cordo Russo
- Laboratory of Molecular Mechanisms of Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Vuelta de Obligado 2490, C1428ADN, Buenos Aires, Argentina.
| | - María F Chervo
- Laboratory of Molecular Mechanisms of Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Vuelta de Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | - Santiago Madera
- Laboratory of Molecular Mechanisms of Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Vuelta de Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | - Eduardo H Charreau
- Laboratory of Molecular Mechanisms of Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Vuelta de Obligado 2490, C1428ADN, Buenos Aires, Argentina
| | - Patricia V Elizalde
- Laboratory of Molecular Mechanisms of Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Vuelta de Obligado 2490, C1428ADN, Buenos Aires, Argentina.
| |
Collapse
|
13
|
Radenkovic S, Konjevic G, Gavrilovic D, Stojanovic-Rundic S, Plesinac-Karapandzic V, Stevanovic P, Jurisic V. pSTAT3 expression associated with survival and mammographic density of breast cancer patients. Pathol Res Pract 2018; 215:366-372. [PMID: 30598340 DOI: 10.1016/j.prp.2018.12.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 12/01/2018] [Accepted: 12/24/2018] [Indexed: 01/03/2023]
Abstract
BACKGROUND Constitutive activation of STAT3 have been shown in several tumor types including breast cancer. We investigate STAT3 expresion as possible molecular marker for breast cancer early detection, as well as prognostic factor for determination of tumor agressiveness. METHODS In this study we measure p(Y705)STAT3 expression in tumor and adjacent tissue of breast cancer patients by Western blot. For relapse-free survival (RFS) and overall survival (OS) we used Log-Rank test. RESULTS We show that average expression of p (Y705) STAT3 in tumor tissue is higher compared to adjacent tissue. Moreover, we found that patients with HER2 positive receptors had significantly higher pSTAT3 expression compared to HER2 negative patients. We showed that patients with high mammographic density had significantly higher tumor expression of pSTAT3 compared to patients with low mammographic density. Also, we show that pSTAT3 expression correlates with longer RFS in the entire group of patients, as well as in the group of ER positive, in lymph node positive and in older group of breast cancer patients (with age over 50). Furthermore, in the entire group of patients, in ER positive, in lymph node positive and in older group of patient, high expression of pSTAT3 showed a better survival than low expression of pSTAT3. CONCLUSION Considering that the expression of pSTAT3 is associated with longer RFS and survival, it can be used as prognostic tools for determination of group of breast cancer patients with low-risk.
Collapse
Affiliation(s)
- Sandra Radenkovic
- Institute of Oncology and Radiology of Serbia, Department of Radiation Oncology and Diagnostics, Belgrade, Serbia
| | - Gordana Konjevic
- Institute of Oncology and Radiology of Serbia, Department of Radiation Oncology and Diagnostics, Belgrade, Serbia; Institute of Oncology and Radiology of Serbia, Department of Experimental Oncology, Serbia
| | - Dusica Gavrilovic
- Institute of Oncology and Radiology of Serbia, Department of Radiation Oncology and Diagnostics, Belgrade, Serbia
| | | | | | | | - Vladimir Jurisic
- Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia.
| |
Collapse
|
14
|
Yang L, Yu X, Yang Y. Autotaxin upregulated by STAT3 activation contributes to invasion in pancreatic neuroendocrine neoplasms. Endocr Connect 2018; 7:1299-1307. [PMID: 30352421 PMCID: PMC6240148 DOI: 10.1530/ec-18-0356] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 10/10/2018] [Indexed: 12/14/2022]
Abstract
Although the upregulation of autotaxin (ATX) is associated with many solid tumours, its role in pancreatic neuroendocrine neoplasms (pNEN) has not been well elucidated. The expression of ATX in pNEN tissues and pNEN cell line BON1 was analysed by Western blot, PCR and immunocytochemistry upon exposure to interleukin-6 (IL-6). Additionally, pNEN cell line BON1 was transfected with siRNAs against ATX or signal transducer and activator of transcription 3 (STAT3) and assessed by in vitro invasion assays. The following results were obtained. The expression of ATX in pNEN tissues was significantly increased compared with that in normal pancreatic tissues. High ATX expression was strongly correlated with tumour grade, lymph node metastasis and tumour-node-metastasis stage. Furthermore, ATX downregulation notably inhibited the metastatic capacity of pNEN cells, whereas STAT3 knockdown was found to downregulate the expression of ATX. ATX expression was upregulated in BON1 cells upon stimulation with IL-6, and this was accompanied by activation/phosphorylation of STAT3. Western blot analysis of human pNEN tissue extracts confirmed increased ATX expression and STAT3 phosphorylation with elevated expression levels of IL-6. In conclusion, ATX is upregulated in pNEN and is correlated with the metastatic capacity of pNEN cells, potentially via interaction with STAT3 activation.
Collapse
Affiliation(s)
- Linfei Yang
- Center for Medical Experiments, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xiao Yu
- Department of General Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yongchao Yang
- Department of Burns and Plastic Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
- European Pancreas Center, Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
- Correspondence should be addressed to Y Yang:
| |
Collapse
|
15
|
Yang Y, Yang L, Li Y. Neuropilin-1 (NRP-1) upregulated by IL-6/STAT3 signaling contributes to invasion in pancreatic neuroendocrine neoplasms. Hum Pathol 2018; 81:192-200. [PMID: 30420046 DOI: 10.1016/j.humpath.2018.06.030] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 06/19/2018] [Accepted: 06/22/2018] [Indexed: 12/18/2022]
Abstract
Although the upregulation of Neuropilin-1 (NRP-1) is associated with many solid tumors, its role in pancreatic neuroendocrine neoplasms (pNEN) has not been well elucidated. The aim of this study was to investigate the role of NRP-1 in improving treatment and determining the prognosis of pNEN. In this study, the expression of NRP-1 in pNEN tissue samples and pNEN cell line BON1 was analyzed by Western blot, polymerase chain reaction (PCR) and immunocytochemistry upon exposure to interleukin-6 (IL-6). Additionally, pNEN cell line BON1 was transfected with small interfering RNAs against NRP-1 or signal transducer and activator of transcription 3 (STAT3) and assessed by in vitro invasion assays. The expression of NRP-1 in pNEN tissues was markedly increased compared with adjacent normal pancreatic tissues. High NRP-1 expression was strongly correlated with tumor grades (P = .026), lymph node metastasis (P = .025), and tumor-node-metastasis stages (P = .012). Furthermore, NRP-1 downregulation notably inhibited the metastatic capacity of pNEN cells, and STAT3 knockdown was found to downregulate the expression of NRP-1. BON1 cells upregulated NRP-1 expression upon stimulation with IL-6. This was accompanied by activation/phosphorylation of the AKT and STAT3 signaling pathways. Western blot of extracts of human pNENs confirmed increased NRP-1 expression, as well as AKT/STAT3 phosphorylation in tissue of pNENs with elevated expression levels of IL-6. In conclusion, our findings suggest that NRP-1 is upregulated in pNEN and is correlated with the metastatic capacity of pNEN cells, potentially via interaction with the IL-6/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Yongchao Yang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Linfei Yang
- Center for Medical Experiments, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Yixiong Li
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha 410008, China.
| |
Collapse
|
16
|
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
|
17
|
Kim DH, Park JE, Chae IG, Park G, Lee S, Chun KS. Isoliquiritigenin inhibits the proliferation of human renal carcinoma Caki cells through the ROS-mediated regulation of the Jak2/STAT3 pathway. Oncol Rep 2017; 38:575-583. [PMID: 28560439 DOI: 10.3892/or.2017.5677] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 04/24/2017] [Indexed: 11/06/2022] Open
Abstract
Isoliquiritigenin (ISL) is a flavonoid with chalcone structure that has been noted in licorice and shallot, which are generally used in traditional Chinese medicine. ISL has demonstrated various pharmacological effects including antioxidant, anti-inflammatory and antitumor activity. However, the molecular mechanisms underlying the anticancer effects of ISL remain poorly understood. The present study revealed that ISL significantly decreased viability and induced apoptosis in human renal carcinoma Caki cells. The ISL-induced apoptosis was associated with the cleavage of caspase-9, -7 and -3, and that of PARP. Moreover, ISL increased the expression of pro-apoptotic protein Bax and diminished the expression of anti-apoptotic protein Bcl-2, and Bcl-xl, thereby increasing cytochrome c release. Treatment of cells with ISL also induced the expression of p53 through downregulation of murine double minute 2 (Mdm2). Furthermore, ISL generated reactive oxygen species (ROS), and pretreatment with ROS scavenger N-acetyl cysteine (NAC) and NADPH oxidase inhibitor diphenyleneiodonium abrogated the ISL-induced apoptosis. One of the key oncogenic signaling pathways is mediated through signal transducer and activator of transcription 3 (STAT3), which promotes abnormal cell proliferation. Incubation of cells with ISL markedly diminished phosphorylation and DNA binding activity of STAT3, and reduced expression of STAT3 responsive gene products, such as cyclin D1 and D2. ISL also attenuated constitutive phosphorylation of upstream kinase, Janus-activated kinase 2 (Jak2). Pretreatment with NAC abrogated the inhibitory effect of ISL on activation of STAT3 and blocked the cleavage of caspase-9, -7 and -3, and that of PARP in Caki cells. Taken together, the present study provides the first report that ISL induces apoptosis in Caki cells via generation of ROS, which causes induction of p53 and inhibition of the STAT3 signaling pathway.
Collapse
Affiliation(s)
- Do-Hee Kim
- College of Pharmacy, Seoul National University, Seoul 151-742, Republic of Korea
| | - Ji Eun Park
- College of Pharmacy, Keimyung University, Daegu 704-701, Republic of Korea
| | - In Gyeong Chae
- College of Pharmacy, Keimyung University, Daegu 704-701, Republic of Korea
| | - Geumi Park
- College of Pharmacy, Keimyung University, Daegu 704-701, Republic of Korea
| | - Sooyeun Lee
- College of Pharmacy, Keimyung University, Daegu 704-701, Republic of Korea
| | - Kyung-Soo Chun
- College of Pharmacy, Keimyung University, Daegu 704-701, Republic of Korea
| |
Collapse
|
18
|
Timmermans-Sprang EPM, Gracanin A, Mol JA. Molecular Signaling of Progesterone, Growth Hormone, Wnt, and HER in Mammary Glands of Dogs, Rodents, and Humans: New Treatment Target Identification. Front Vet Sci 2017; 4:53. [PMID: 28451590 PMCID: PMC5389977 DOI: 10.3389/fvets.2017.00053] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 03/28/2017] [Indexed: 12/21/2022] Open
Abstract
Mammary tumors are the most common form of neoplasia in the bitch. Female dogs are protected when they are spayed before the first estrus cycle, but this effect readily disappears and is already absent when dogs are spayed after the second heat. As the ovaries are removed during spaying, ovarian steroids are assumed to play an essential role in tumor development. The sensitivity toward tumor development is already present during early life, which may be caused by early mutations in stem cells during the first estrus cycles. Later on in life, tumors arise that are mostly steroid-receptor positive, although a small subset of tumors overexpressing human epidermal growth factor 2 (HER2) and some lacking estrogen receptor, progesterone receptor (PR), and HER2 (triple negative) are present, as is the situation in humans. Progesterone (P4), acting through PR, is the major steroid involved in outgrowth of mammary tissue. PRs are expressed in two forms, the progesterone receptor A (PRA) and progesterone receptor B (PRB) isoforms derived from splice variants from a single gene. The dog and the whole family of canids have only a functional PRA isoform, whereas the PRB isoform, if expressed at all, is devoid of intrinsic biological activity. In human breast cancer, overexpression of the PRA isoform is related to more aggressive carcinomas making the dog a unique model to study PRA-related mammary cancer. Administration of P4 to adult dogs results in local mammary expression of growth hormone (GH) and wing less-type mouse mammary tumor virus integration site family 4 (Wnt4). Both proteins play a role in activation of mammary stem cells. In this review, we summarize what is known on P4, GH, and Wnt signaling in canine mammary cancer, how the family of HER receptors could interact with this signaling, and what this means for comparative and translational oncological aspects of human breast cancer development.
Collapse
Affiliation(s)
| | - Ana Gracanin
- Department of Clinical Sciences of Companion Animals, Utrecht University, Utrecht, Netherlands
| | - Jan A Mol
- Department of Clinical Sciences of Companion Animals, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
19
|
Elizalde PV, Cordo Russo RI, Chervo MF, Schillaci R. ErbB-2 nuclear function in breast cancer growth, metastasis and resistance to therapy. Endocr Relat Cancer 2016; 23:T243-T257. [PMID: 27765799 DOI: 10.1530/erc-16-0360] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 10/06/2016] [Indexed: 12/17/2022]
Abstract
Approximately 15-20% of breast cancers (BC) show either membrane overexpression of ErbB-2 (MErbB-2), a member of the ErbBs family of receptor tyrosine kinases, or ERBB2 gene amplification. Until the development of MErbB-2-targeted therapies, this BC subtype, called ErbB-2-positive, was associated with increased metastatic potential and poor prognosis. Although these therapies have significantly improved overall survival and cure rates, resistance to available drugs is still a major clinical issue. In its classical mechanism, MErbB-2 activates downstream signaling cascades, which transduce its effects in BC. The fact that ErbB-2 is also present in the nucleus of BC cells was discovered over twenty years ago. Also, compelling evidence revealed a non-canonical function of nuclear ErbB-2 as a transcriptional regulator. As a deeper understanding of nuclear ErbB-2 actions would be crucial to the disclosure of its role as a biomarker and a target of therapy in BC, we will here review its function in BC, in particular, its role in growth, metastatic spreading and response to currently available MErbB-2-positive BC therapies.
Collapse
Affiliation(s)
- Patricia V Elizalde
- Laboratory of Molecular Mechanisms of CarcinogenesisInstituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
| | - Rosalía I Cordo Russo
- Laboratory of Molecular Mechanisms of CarcinogenesisInstituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
| | - Maria F Chervo
- Laboratory of Molecular Mechanisms of CarcinogenesisInstituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
| | - Roxana Schillaci
- Laboratory of Molecular Mechanisms of CarcinogenesisInstituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
| |
Collapse
|
20
|
Grimm SL, Hartig SM, Edwards DP. Progesterone Receptor Signaling Mechanisms. J Mol Biol 2016; 428:3831-49. [PMID: 27380738 DOI: 10.1016/j.jmb.2016.06.020] [Citation(s) in RCA: 138] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 06/25/2016] [Accepted: 06/27/2016] [Indexed: 12/27/2022]
Abstract
Progesterone receptor (PR) is a master regulator in female reproductive tissues that controls developmental processes and proliferation and differentiation during the reproductive cycle and pregnancy. PR also plays a role in progression of endocrine-dependent breast cancer. As a member of the nuclear receptor family of ligand-dependent transcription factors, the main action of PR is to regulate networks of target gene expression in response to binding its cognate steroid hormone, progesterone. This paper summarizes recent advances in understanding the structure-function properties of the receptor protein and the tissue/cell-type-specific PR signaling pathways that contribute to the biological actions of progesterone in the normal breast and in breast cancer.
Collapse
Affiliation(s)
- Sandra L Grimm
- Department of Molecular and Cellular Biology, Baylor College of Medicine,Houston, TX 77030, USA
| | - Sean M Hartig
- Department of Molecular and Cellular Biology, Baylor College of Medicine,Houston, TX 77030, USA
| | - Dean P Edwards
- Department of Molecular and Cellular Biology, Baylor College of Medicine,Houston, TX 77030, USA.
| |
Collapse
|
21
|
Venturutti L, Romero LV, Urtreger AJ, Chervo MF, Cordo Russo RI, Mercogliano MF, Inurrigarro G, Pereyra MG, Proietti CJ, Izzo F, Díaz Flaqué MC, Sundblad V, Roa JC, Guzmán P, Bal de Kier Joffé ED, Charreau EH, Schillaci R, Elizalde PV. Stat3 regulates ErbB-2 expression and co-opts ErbB-2 nuclear function to induce miR-21 expression, PDCD4 downregulation and breast cancer metastasis. Oncogene 2016; 35:2208-22. [PMID: 26212010 DOI: 10.1038/onc.2015.281] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 06/15/2015] [Accepted: 06/18/2015] [Indexed: 02/07/2023]
Abstract
Membrane overexpression of the receptor tyrosine kinase ErbB-2 (MErbB-2) accounts for a clinically aggressive breast cancer (BC) subtype (ErbB-2-positive) with increased incidence of metastases. We and others demonstrated that nuclear ErbB-2 (NErbB-2) also plays a key role in BC and is a poor prognostic factor in ErbB-2-positive tumors. The signal transducer and activator of transcription 3 (Stat3), another player in BC, has been recognized as a downstream mediator of MErbB-2 action in BC metastasis. Here, we revealed an unanticipated novel direction of the ErbB-2 and Stat3 interaction underlying BC metastasis. We found that Stat3 binds to its response elements (GAS) at the ErbB-2 promoter to upregulate ErbB-2 transcription in metastatic, ErbB-2-positive BC. We validated these results in several BC subtypes displaying metastatic and non-metastatic ability, highlighting Stat3 general role as upstream regulator of ErbB-2 expression in BC. Moreover, we showed that Stat3 co-opts NErbB-2 function by recruiting ErbB-2 as its coactivator at the GAS sites in the promoter of microRNA-21 (miR-21), a metastasis-promoting microRNA (miRNA). Using an ErbB-2 nuclear localization domain mutant and a constitutively activated ErbB-2 variant, we found that NErbB-2 role as a Stat3 coactivator and also its direct role as transcription factor upregulate miR-21 in BC. This reveals a novel function of NErbB-2 as a regulator of miRNAs expression. Increased levels of miR-21, in turn, downregulate the expression of the metastasis-suppressor protein programmed cell death 4 (PDCD4), a validated miR-21 target. Using an in vivo model of metastatic ErbB-2-postive BC, in which we silenced Stat3 and reconstituted ErbB-2 or miR-21 expression, we showed that both are downstream mediators of Stat3-driven metastasis. Supporting the clinical relevance of our results, we found an inverse correlation between ErbB-2/Stat3 nuclear co-expression and PDCD4 expression in ErbB-2-positive primary invasive BCs. Our findings identify Stat3 and NErbB-2 as novel therapeutic targets to inhibit ErbB-2-positive BC metastasis.
Collapse
Affiliation(s)
- L Venturutti
- Laboratory of Molecular Mechanisms of Carcinogenesis, Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
| | - L V Romero
- Laboratory of Molecular Mechanisms of Carcinogenesis, Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
| | - A J Urtreger
- Research Area, Institute of Oncology 'Angel H. Roffo', University of Buenos Aires, Buenos Aires, Argentina
| | - M F Chervo
- Laboratory of Molecular Mechanisms of Carcinogenesis, Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
| | - R I Cordo Russo
- Laboratory of Molecular Mechanisms of Carcinogenesis, Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
| | - M F Mercogliano
- Laboratory of Molecular Mechanisms of Carcinogenesis, Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
| | - G Inurrigarro
- Servicio de Patología, Sanatorio Mater Dei, Buenos Aires, Argentina
| | - M G Pereyra
- Laboratory of Molecular Mechanisms of Carcinogenesis, Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
| | - C J Proietti
- Laboratory of Molecular Mechanisms of Carcinogenesis, Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
| | - F Izzo
- Laboratory of Molecular Mechanisms of Carcinogenesis, Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
| | - M C Díaz Flaqué
- Laboratory of Molecular Mechanisms of Carcinogenesis, Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
| | - V Sundblad
- Laboratory of Molecular Mechanisms of Carcinogenesis, Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
| | - J C Roa
- Departamento de Anatomía Patológica (BIOREN), Universidad de La Frontera, Temuco, Chile
- Departamento de Anatomía Patológica, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago de Chile, Chile
- Advanced Center for Chronic Diseases (ACCDIS), Pontificia Universidad Católica de Chile, Santiago de Chile, Chile
| | - P Guzmán
- Departamento de Anatomía Patológica (BIOREN), Universidad de La Frontera, Temuco, Chile
| | - E D Bal de Kier Joffé
- Research Area, Institute of Oncology 'Angel H. Roffo', University of Buenos Aires, Buenos Aires, Argentina
| | - E H Charreau
- Laboratory of Molecular Mechanisms of Carcinogenesis, Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
| | - R Schillaci
- Laboratory of Molecular Mechanisms of Carcinogenesis, Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
| | - P V Elizalde
- Laboratory of Molecular Mechanisms of Carcinogenesis, Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
| |
Collapse
|
22
|
Scheving LA, Zhang X, Stevenson MC, Weintraub MA, Abbasi A, Clarke AM, Threadgill DW, Russell WE. Loss of hepatocyte ERBB3 but not EGFR impairs hepatocarcinogenesis. Am J Physiol Gastrointest Liver Physiol 2015; 309:G942-54. [PMID: 26492920 PMCID: PMC4683301 DOI: 10.1152/ajpgi.00089.2015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 10/18/2015] [Indexed: 01/31/2023]
Abstract
Epidermal growth factor receptor (EGFR) and ERBB3 have been implicated in hepatocellular carcinogenesis (HCC). However, it is not known whether altering the activity of either EGFR or ERBB3 affects HCC development. We now show that Egfr(Dsk5) mutant mice, which have a gain-of-function allele that increases basal EGFR kinase activity, develop spontaneous HCC by 10 mo of age. Their tumors show increased activation of EGFR, ERBB2, and ERBB3 as well as AKT and ERK1,2. Hepatocyte-specific models of EGFR and ERBB3 gene ablation were generated to evaluate how the loss of these genes affected tumor progression. Loss of either receptor tyrosine kinase did not alter liver development or regenerative liver growth following carbon tetrachloride injection. However, using a well-characterized model of HCC in which N-nitrosodiethylamine is injected into 14-day-old mice, we discovered that loss of hepatocellular ERBB3 but not EGFR, which occurred after tumor initiation, retarded liver tumor formation and cell proliferation. We found no evidence that this was due to increased apoptosis or diminished phosphatidylinositol-3-kinase activity in the ERBB3-null cells. However, the relative amount of phospho-STAT3 was diminished in tumors derived from these mice, suggesting that ERBB3 may promote HCC through STAT3 activation.
Collapse
Affiliation(s)
- Lawrence A. Scheving
- 1Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee;
| | - Xiuqi Zhang
- 1Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee;
| | - Mary C. Stevenson
- 1Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee;
| | - Michael A. Weintraub
- 1Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee;
| | - Annam Abbasi
- 1Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee;
| | - Andrea M. Clarke
- 1Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee;
| | - David W. Threadgill
- 6Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas; and ,7Department of Molecular and Cellular Medicine, Texas A&M University, College Station, Texas
| | - William E. Russell
- 1Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee; ,5Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee;
| |
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
|
Kamińska K, Czarnecka AM, Escudier B, Lian F, Szczylik C. Interleukin-6 as an emerging regulator of renal cell cancer. Urol Oncol 2015; 33:476-85. [PMID: 26296264 DOI: 10.1016/j.urolonc.2015.07.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 07/10/2015] [Accepted: 07/11/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND Our knowledge on the molecular basis of kidney cancer metastasisis still relatively low. About 25-30% of patients suffering from clear cell renal cell carcinoma (ccRCC)present metastatic disease at the time of primary diagnosis. Only 10% of patients diagnosed with stage IV disease survive 5 years and 20-50% of patients diagnosed with localized tumor develop metastases within 3 years. High mortality of patients with this cancer is associated with a large potential for metastasis and resistance to oncologic treatments such as chemo- and radiotherapy. Literature data based on studies conducted on other types of cancers suggest that in metastatic ccRCC, the complex of interleukin-6 (IL-6) and its soluble receptor (sIL-6R; complex IL-6/sIL-6R) and the signal transduction pathway (gp130/STAT3) might play a key role in this process. PURPOSE Therefore, in this review we focus on the role of IL-6 and its signaling pathways as a factor for development and spread of RCC. Analyzing the molecular basis of cancer spreading will enable the development of prognostic tests, evaluate individual predisposition for metastasis, and produce drugs that target metastases. As the development of effective systemic treatments evolve from advancements in molecular biology, continued studies directed at understanding the genetic and molecular complexities of this disease are critical to improve RCC treatment options.
Collapse
Affiliation(s)
| | - Anna M Czarnecka
- Department of Oncology, Military Institute of Medicine, Warsaw, Poland
| | - Bernard Escudier
- Medical Oncology Department, Institut Gustave Roussy, Villejuif, France
| | - Fei Lian
- Emory University School of Medicine, Atlanta, GA
| | - Cezary Szczylik
- Department of Oncology, Military Institute of Medicine, Warsaw, Poland
| |
Collapse
|
25
|
Cordo Russo RI, Béguelin W, Díaz Flaqué MC, Proietti CJ, Venturutti L, Galigniana N, Tkach M, Guzmán P, Roa JC, O'Brien NA, Charreau EH, Schillaci R, Elizalde PV. Targeting ErbB-2 nuclear localization and function inhibits breast cancer growth and overcomes trastuzumab resistance. Oncogene 2015; 34:3413-28. [PMID: 25174405 DOI: 10.1038/onc.2014.272] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 07/08/2014] [Accepted: 07/19/2014] [Indexed: 12/11/2022]
Abstract
Membrane overexpression of ErbB-2/HER2 receptor tyrosine kinase (membrane ErbB-2 (MErbB-2)) has a critical role in breast cancer (BC). We and others have also shown the role of nuclear ErbB-2 (NErbB-2) in BC, whose presence we identified as a poor prognostic factor in MErbB-2-positive tumors. Current anti-ErbB-2 therapies, as with the antibody trastuzumab (Ttzm), target only MErbB-2. Here, we found that blockade of NErbB-2 action abrogates growth of BC cells, sensitive and resistant to Ttzm, in a scenario in which ErbB-2, ErbB-3 and Akt are phosphorylated, and ErbB-2/ErbB-3 dimers are formed. Also, inhibition of NErbB-2 presence suppresses growth of a preclinical BC model resistant to Ttzm. We showed that at the cyclin D1 promoter, ErbB-2 assembles a transcriptional complex with Stat3 (signal transducer and activator of transcription 3) and ErbB-3, another member of the ErbB family, which reveals the first nuclear function of ErbB-2/ErbB-3 dimer. We identified NErbB-2 as the major proliferation driver in Ttzm-resistant BC, and demonstrated that Ttzm inability to disrupt the Stat3/ErbB-2/ErbB-3 complex underlies its failure to inhibit growth. Furthermore, our results in the clinic revealed that nuclear interaction between ErbB-2 and Stat3 correlates with poor overall survival in primary breast tumors. Our findings challenge the paradigm of anti-ErbB-2 drug design and highlight NErbB-2 as a novel target to overcome Ttzm resistance.
Collapse
MESH Headings
- Active Transport, Cell Nucleus/drug effects
- Animals
- Antibodies, Monoclonal, Humanized/therapeutic use
- Breast Neoplasms/drug therapy
- Breast Neoplasms/pathology
- Cell Nucleus/drug effects
- Cell Nucleus/metabolism
- Cell Proliferation/drug effects
- Cell Proliferation/genetics
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Drug Synergism
- Female
- Genes, Dominant/physiology
- Humans
- Mice, Inbred BALB C
- Mice, Nude
- Molecular Targeted Therapy/methods
- Mutant Proteins/pharmacology
- Mutant Proteins/therapeutic use
- Protein Isoforms/pharmacology
- Protein Isoforms/therapeutic use
- Protein Transport/drug effects
- Receptor, ErbB-2/antagonists & inhibitors
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/metabolism
- Receptor, ErbB-2/physiology
- Trastuzumab
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- R I Cordo Russo
- Laboratory of Molecular Mechanisms of Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - W Béguelin
- Laboratory of Molecular Mechanisms of Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - M C Díaz Flaqué
- Laboratory of Molecular Mechanisms of Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - C J Proietti
- Laboratory of Molecular Mechanisms of Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - L Venturutti
- Laboratory of Molecular Mechanisms of Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - N Galigniana
- Laboratory of Molecular Mechanisms of Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - M Tkach
- Laboratory of Molecular Mechanisms of Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - P Guzmán
- Departamento de Anatomía Patológica (BIOREN), Universidad de La Frontera, Temuco, Chile
| | - J C Roa
- Departamento de Anatomía Patológica (BIOREN), Universidad de La Frontera, Temuco, Chile
| | - N A O'Brien
- Department of Medicine, Division of Hematology/Oncology, Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - E H Charreau
- Laboratory of Molecular Mechanisms of Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - R Schillaci
- Laboratory of Molecular Mechanisms of Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | - P V Elizalde
- Laboratory of Molecular Mechanisms of Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| |
Collapse
|
26
|
Bennesch MA, Picard D. Minireview: Tipping the balance: ligand-independent activation of steroid receptors. Mol Endocrinol 2015; 29:349-63. [PMID: 25625619 DOI: 10.1210/me.2014-1315] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Steroid receptors are prototypical ligand-dependent transcription factors and a textbook example for allosteric regulation. According to this canonical model, binding of cognate steroid is an absolute requirement for transcriptional activation. Remarkably, the simple one ligand-one receptor model could not be farther from the truth. Steroid receptors, notably the sex steroid receptors, can receive multiple inputs. Activation of steroid receptors by other signals, working through their own signaling pathways, in the absence of the cognate steroids, represents the most extreme form of signaling cross talk. Compared with cognate steroids, ligand-independent activation pathways produce similar but not identical outputs. Here we review the phenomena and discuss what is known about the underlying molecular mechanisms and the biological significance. We hypothesize that steroid receptors may have evolved to be trigger happy. In addition to their cognate steroids, many posttranslational modifications and interactors, modulated by other signals, may be able to tip the balance.
Collapse
Affiliation(s)
- Marcela A Bennesch
- Département de Biologie Cellulaire, Université de Genève, Sciences III, CH-1211 Genève 4, Switzerland
| | | |
Collapse
|
27
|
Izzo F, Mercogliano F, Venturutti L, Tkach M, Inurrigarro G, Schillaci R, Cerchietti L, Elizalde PV, Proietti CJ. Progesterone receptor activation downregulates GATA3 by transcriptional repression and increased protein turnover promoting breast tumor growth. Breast Cancer Res 2014; 16:491. [PMID: 25479686 PMCID: PMC4303201 DOI: 10.1186/s13058-014-0491-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 11/28/2014] [Indexed: 11/10/2022] Open
Abstract
Introduction The transcription factor GATA3 is involved in mammary gland development and is crucial for the maintenance of the differentiated status of luminal epithelial cells. The role of GATA3 in breast cancer as a tumor suppressor has been established, although insights into the mechanism of GATA3 expression loss are still required. Methods Chromatin immunoprecipitation assays were conducted to study progestin modulation of recruitment of transcription factors to GATA3 promoter. We performed western blot and reverse RT-qPCR experiments to explore progestin regulation of GATA3 protein and mRNA expression respectively. Confocal microscopy and in vitro phosphorylation studies were conducted to examine progestin capacity to induce GATA3 serine phosphorylation in its 308 residue. GATA3 participation in progestin-induced breast cancer growth was addressed in in vitro proliferation and in vivo tumor growth experiments. Results In this study, we demonstrate that progestin-activated progesterone receptor (PR) reduces GATA3 expression through regulation at the transcriptional and post-translational levels in breast cancer cells. In the former mechanism, the histone methyltransferase enhancer of zeste homolog 2 is co-recruited with activated PR to a putative progesterone response element in the GATA3 proximal promoter, increasing H3K27me3 levels and inducing chromatin compaction, resulting in decreased GATA3 mRNA levels. This transcriptional regulation is coupled with increased GATA3 protein turnover through progestin-induced GATA3 phosphorylation at serine 308 followed by 26S proteasome-mediated degradation. Both molecular mechanisms converge to accomplish decreased GATA3 expression levels in breast cancer cells upon PR activation. In addition, we demonstrated that decreased GATA3 levels are required for progestin-induced upregulation of cyclin A2, which mediates the G1 to S phase transition of the cell cycle and was reported to be associated with poor prognosis in breast cancer. Finally, we showed that downregulation of GATA3 is required for progestin stimulation of both in vitro cell proliferation and in vivo tumor growth. Conclusions In the present study, we reveal that progestin-induced PR activation leads to loss of GATA3 expression in breast cancer cells through transcriptional and post-translational regulation. Importantly, we demonstrate that GATA3 downregulation is required for progestin-induced upregulation of cyclin A2 and for progestin-induced in vitro and in vivo breast cancer cell growth. Electronic supplementary material The online version of this article (doi:10.1186/s13058-014-0491-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Franco Izzo
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Vuelta de Obligado 2490, Buenos Aires, 1428 ADN, Argentina.
| | - Florencia Mercogliano
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Vuelta de Obligado 2490, Buenos Aires, 1428 ADN, Argentina.
| | - Leandro Venturutti
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Vuelta de Obligado 2490, Buenos Aires, 1428 ADN, Argentina.
| | - Mercedes Tkach
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Vuelta de Obligado 2490, Buenos Aires, 1428 ADN, Argentina.
| | | | - Roxana Schillaci
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Vuelta de Obligado 2490, Buenos Aires, 1428 ADN, Argentina.
| | | | - Patricia V Elizalde
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Vuelta de Obligado 2490, Buenos Aires, 1428 ADN, Argentina.
| | - Cecilia J Proietti
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Vuelta de Obligado 2490, Buenos Aires, 1428 ADN, Argentina.
| |
Collapse
|
28
|
Daniel AR, Gaviglio AL, Knutson TP, Ostrander JH, D'Assoro AB, Ravindranathan P, Peng Y, Raj GV, Yee D, Lange CA. Progesterone receptor-B enhances estrogen responsiveness of breast cancer cells via scaffolding PELP1- and estrogen receptor-containing transcription complexes. Oncogene 2014; 34:506-15. [PMID: 24469035 PMCID: PMC4112172 DOI: 10.1038/onc.2013.579] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Revised: 11/26/2013] [Accepted: 12/10/2013] [Indexed: 12/16/2022]
Abstract
Progesterone and estrogen are important drivers of breast cancer proliferation. Herein, we probed ER-alpha and PR cross-talk in breast cancer models. Stable expression of PR-B in PR-low/ER+ MCF7 cells increased cellular sensitivity to estradiol and IGF1, as measured in growth assays performed in the absence of exogenous progestin; similar results were obtained in PR-null/ER+ T47D cells stably expressing PR-B. Genome-wide microarray analyses revealed that unliganded PR-B induced robust expression of a subset of estradiol-responsive ER-target genes, including CathepsinD (CTSD). Estradiol-treated MCF7 cells stably expressing PR-B exhibited enhanced ER Ser167 phosphorylation and recruitment of ER, PR, and the proline, glutamate and leucine rich protein 1 (PELP1) to an estrogen response element (ERE) in the CTSD distal promoter; this complex co-immunoprecipitated with IGF1R in whole cell lysates. Importantly, ER/PR/PELP1 complexes were also detected in human breast cancer samples. Inhibition of IGF1R or PI3K blocked PR-B-dependent CTSD mRNA upregulation in response to estradiol. Similarly, inhibition of IGF1R or PR significantly reduced ER recruitment to the CTSD promoter. Stable knockdown of endogenous PR or onapristone treatment of multiple unmodified breast cancer cell lines blocked estradiol-mediated CTSD induction, inhibited growth in soft agar, and partially restored tamoxifen-sensitivity of resistant cells. Further, combination treatment of breast cancer cells with both onapristone and IGF1R tyrosine kinase inhibitor AEW541 was more effective than either agent alone. In summary, unliganded PR-B enhanced proliferative responses to estradiol and IGF1 via scaffolding of ERalpha/PELP1/IGF1R-containing complexes. Our data provide a strong rationale for targeting PR in combination with ER and IGF1R in patients with luminal breast cancer.
Collapse
Affiliation(s)
- A R Daniel
- Departments of Medicine and Pharmacology, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - A L Gaviglio
- Departments of Medicine and Pharmacology, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - T P Knutson
- Departments of Medicine and Pharmacology, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - J H Ostrander
- Department of Medicine, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - A B D'Assoro
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - P Ravindranathan
- Department of Urology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Y Peng
- Department of Pathology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - G V Raj
- Department of Urology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - D Yee
- Departments of Medicine and Pharmacology, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - C A Lange
- Departments of Medicine and Pharmacology, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
29
|
Díaz Flaqué MC, Galigniana NM, Béguelin W, Vicario R, Proietti CJ, Russo RC, Rivas MA, Tkach M, Guzmán P, Roa JC, Maronna E, Pineda V, Muñoz S, Mercogliano MF, Charreau EH, Yankilevich P, Schillaci R, Elizalde PV. Progesterone receptor assembly of a transcriptional complex along with activator protein 1, signal transducer and activator of transcription 3 and ErbB-2 governs breast cancer growth and predicts response to endocrine therapy. Breast Cancer Res 2013; 15:R118. [PMID: 24345432 PMCID: PMC3978912 DOI: 10.1186/bcr3587] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Accepted: 12/09/2013] [Indexed: 12/28/2022] Open
Abstract
INTRODUCTION The role of the progesterone receptor (PR) in breast cancer remains a major clinical challenge. Although PR induces mammary tumor growth, its presence in breast tumors is a marker of good prognosis. We investigated coordinated PR rapid and nonclassical transcriptional effects governing breast cancer growth and endocrine therapy resistance. METHODS We used breast cancer cell lines expressing wild-type and mutant PRs, cells sensitive and resistant to endocrine therapy, a variety of molecular and cellular biology approaches, in vitro proliferation studies and preclinical models to explore PR regulation of cyclin D1 expression, tumor growth, and response to endocrine therapy. We investigated the clinical significance of activator protein 1 (AP-1) and PR interaction in a cohort of 99 PR-positive breast tumors by an immunofluorescence protocol we developed. The prognostic value of AP-1/PR nuclear colocalization in overall survival (OS) was evaluated using Kaplan-Meier method, and Cox model was used to explore said colocalization as an independent prognostic factor for OS. RESULTS We demonstrated that at the cyclin D1 promoter and through coordinated rapid and transcriptional effects, progestin induces the assembly of a transcriptional complex among AP-1, Stat3, PR, and ErbB-2 which functions as an enhanceosome to drive breast cancer growth. Our studies in a cohort of human breast tumors identified PR and AP-1 nuclear interaction as a marker of good prognosis and better OS in patients treated with tamoxifen (Tam), an anti-estrogen receptor therapy. Rationale for this finding was provided by our demonstration that Tam inhibits rapid and genomic PR effects, rendering breast cancer cells sensitive to its antiproliferative effects. CONCLUSIONS We here provided novel insight into the paradox of PR action as well as new tools to identify the subgroup of ER+/PR + patients unlikely to respond to ER-targeted therapies.
Collapse
Affiliation(s)
- María C Díaz Flaqué
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Obligado 2490, Buenos Aires 1428, Argentina
| | - Natalia M Galigniana
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Obligado 2490, Buenos Aires 1428, Argentina
| | - Wendy Béguelin
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Obligado 2490, Buenos Aires 1428, Argentina
| | - Rocío Vicario
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Obligado 2490, Buenos Aires 1428, Argentina
| | - Cecilia J Proietti
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Obligado 2490, Buenos Aires 1428, Argentina
| | - Rosalía Cordo Russo
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Obligado 2490, Buenos Aires 1428, Argentina
| | - Martín A Rivas
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Obligado 2490, Buenos Aires 1428, Argentina
| | - Mercedes Tkach
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Obligado 2490, Buenos Aires 1428, Argentina
| | | | - Juan C Roa
- Universidad de La Frontera, Temuco, Chile
| | - Esteban Maronna
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Obligado 2490, Buenos Aires 1428, Argentina
- Sanatorio Mater Dei, Buenos Aires, Argentina
| | | | | | | | - Eduardo H Charreau
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Obligado 2490, Buenos Aires 1428, Argentina
| | - Patricio Yankilevich
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA), CONICET - Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - Roxana Schillaci
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Obligado 2490, Buenos Aires 1428, Argentina
| | - Patricia V Elizalde
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Obligado 2490, Buenos Aires 1428, Argentina
| |
Collapse
|
30
|
Koperek O, Aumayr K, Schindl M, Werba G, Soleiman A, Schoppmann S, Sahora K, Birner P. Phosphorylation of STAT3 correlates with HER2 status, but not with survival in pancreatic ductal adenocarcinoma. APMIS 2013; 122:476-81. [PMID: 24164699 DOI: 10.1111/apm.12194] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Accepted: 07/30/2013] [Indexed: 12/16/2022]
Abstract
Activation of signal-transcriptional factor signal transducer and activator of transcription 3 (STAT3) is associated with more aggressive behaviour in a variety of human malignancies. As selective STAT3 inhibitors exist, this protein might represent a novel therapeutic target. Although STAT3 seems to play an important role in progression of pancreatic ductal carcinoma (PDAC), only few data on this subject exist. The aim of our study was the investigation of STAT3 activation and its correlation with its possible regulator HER2. Expression of tyrosine-705 phosphorylated STAT3 (pSTAT3) was determined immunohistochemically in 79 PDACs. HER2 status assessed by immunohistochemistry and double colour silver in situ hybridization was available from a previous study. PSTAT3 expression was seen in 33 (41.8%) patients. Six patients were scored as HER2 positive having strong correlation with pSTAT3 expression (p = 0.004, Fisher's exact test). No association of pSTAT3 expression with patients' age, tumour staging and grading, perineural invasion of tumour cells or survival time was seen. pSTAT3 is frequently expressed in PDAC. Nevertheless, its immediate clinical relevance seems to be low. However, further research needs to determine whether STAT3 status in PDAC is predictive for the response to novel targeting therapies.
Collapse
Affiliation(s)
- Oskar Koperek
- Department of Pathology, Medical University of Vienna, Vienna
| | | | | | | | | | | | | | | |
Collapse
|
31
|
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
|
32
|
Tkach M, Rosemblit C, Rivas MA, Proietti CJ, Díaz Flaqué MC, Mercogliano MF, Beguelin W, Maronna E, Guzmán P, Gercovich FG, Deza EG, Elizalde PV, Schillaci R. p42/p44 MAPK-mediated Stat3Ser727 phosphorylation is required for progestin-induced full activation of Stat3 and breast cancer growth. Endocr Relat Cancer 2013; 20:197-212. [PMID: 23329648 DOI: 10.1530/erc-12-0194] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Stat3 is a signaling node for multiple oncogenic pathways and is therefore frequently active in breast cancer. As experimental and clinical evidence reveals that progestins are key players in controlling mammary gland tumorigenesis, we studied Stat3 participation in this event. We have previously shown that progestins induce Stat3Tyr705 phosphorylation and its transcriptional activation in breast cancer cells. In this study, we demonstrate that progestins also induce Stat3 phosphorylation at Ser727 residue, which occurs via activation of c-Src/p42/p44 MAPK pathways in murine progestin-dependent C4HD cells and in T-47D cells. Expression of a Stat3S727A vector, which carries a serine-to-alanine substitution at codon 727, shows that Stat3Ser727 phosphorylation is required for full transcriptional activation of cyclin D1 gene expression by progestins and for in vivo Stat3 recruitment on cyclin D1 promoter. Transfection of Stat3S727A in murine and human breast cancer cells abolished progestin-induced in vitro and in vivo growth. Moreover, we found a positive correlation between progesterone receptor expression and nuclear localization of Stat3Ser727 phosphorylation in breast cancer biopsies. These data highlight Stat3 phosphorylation in Ser727 residue as a nongenomic action by progestins, necessary to promote breast cancer growth.
Collapse
Affiliation(s)
- Mercedes Tkach
- Laboratory of Molecular Mechanisms of Carcinogenesis, Instituto de Biología y Medicina Experimental, CONICET, Vuelta de Obligado 2490, Buenos Aires C1428ADN, Argentina
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Cruz GI, Martínez ME, Natarajan L, Wertheim BC, Gago-Dominguez M, Bondy M, Daneri-Navarro A, Meza-Montenegro MM, Gutierrez-Millan LE, Brewster A, Schedin P, Komenaka IK, Castelao JE, Carracedo A, Redondo CM, Thompson PA. Hypothesized role of pregnancy hormones on HER2+ breast tumor development. Breast Cancer Res Treat 2013; 137:237-46. [PMID: 23135573 PMCID: PMC4054812 DOI: 10.1007/s10549-012-2313-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2012] [Accepted: 10/24/2012] [Indexed: 12/25/2022]
Abstract
Breast cancer incidence rates have declined among older but not younger women; the latter are more likely to be diagnosed with breast cancers carrying a poor prognosis. Epidemiological evidence supports an increase in breast cancer incidence following pregnancy with risk elevated as much as 10 years post-partum. We investigated the association between years since last full-term pregnancy at the time of diagnosis (≤10 or >10 years) and breast tumor subtype in a case series of premenopausal Hispanic women (n = 627). Participants were recruited in the United States, Mexico, and Spain. Cases with known estrogen receptor (ER), progesterone receptor (PR), and HER2 status, with one or more full-term pregnancies ≥1 year prior to diagnosis were eligible for this analysis. Cases were classified into three tumor subtypes according to hormone receptor (HR+ = ER+ and/or PR+; HR- = ER- and PR-) expression and HER2 status: HR+/HER2-, HER2+ (regardless of HR), and triple negative breast cancer. Case-only odds ratios (ORs) and 95 % confidence intervals (CIs) were calculated for HER2+ tumors in reference to HR+/HER2- tumors. Participants were pooled in a mixed-effects logistic regression model with years since pregnancy as a fixed effect and study site as a random effect. When compared to HR+/HER2- cases, women with HER2+ tumors were more likely be diagnosed in the post-partum period of ≤10 years (OR = 1.68; 95 % CI, 1.12-2.52). The effect was present across all source populations and independent of the HR status of the HER2+ tumor. Adjusting for age at diagnosis (≤45 or >45 years) did not materially alter our results (OR = 1.78; 95 % CI, 1.08-2.93). These findings support the novel hypothesis that factors associated with the post-partum breast, possibly hormonal, are involved in the development of HER2+ tumors.
Collapse
Affiliation(s)
- Giovanna I. Cruz
- School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - María Elena Martínez
- Moores UCSD Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Loki Natarajan
- Moores UCSD Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | | | - Manuela Gago-Dominguez
- Genomic Medicine Group, Galician Foundation of Genomic Medicine, Complejo Hospitalario Universitario de Santiago, SERGAS, IDIS, Santiago de Compostela, Spain
| | - Melissa Bondy
- Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | | | | | | | - Abenaa Brewster
- Department of Clinical Cancer Prevention, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Pepper Schedin
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Denver, CO, USA
| | - Ian K. Komenaka
- Maricopa Medical Center, Department of Surgery, Phoenix, AZ, USA
| | - J. Esteban Castelao
- Oncology and Genetics Unit, Complejo Hospitalario Universitario de Vigo, Genomic Medicine Group, SERGAS, Vigo, Spain
| | - Angel Carracedo
- Genomic Medicine Group, Galician Foundation of Genomic Medicine, Complejo Hospitalario Universitario de Santiago, SERGAS, IDIS, Santiago de Compostela, Spain
| | - Carmen M. Redondo
- Oncology and Genetics Unit, Complejo Hospitalario Universitario de Vigo, Genomic Medicine Group, SERGAS, Vigo, Spain
| | - Patricia A. Thompson
- Department of Cellular and Molecular Medicine, College of Medicine, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
34
|
Cork DMW, Lennard TWJ, Tyson-Capper AJ. Progesterone receptor (PR) variants exist in breast cancer cells characterised as PR negative. Tumour Biol 2012; 33:2329-40. [PMID: 22956396 DOI: 10.1007/s13277-012-0495-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Accepted: 08/16/2012] [Indexed: 01/18/2023] Open
Abstract
Progesterone receptor (PR) expression is measured in breast cancer by immunohistochemistry using N-terminally targeted antibodies and serves as a biomarker for endocrine therapeutic decisions. Extensive PR alternative splicing has been reported which may generate truncated PR variant proteins which are not detected by current breast cancer screening or may alter the function of proteins detected in screening. However, the existence of such truncated PR variants remains controversial. We have characterised PR protein expression in breast cancer cell lines using commercial PR antibodies targeting different epitopes. Truncated PR proteins are detected in reportedly PR negative MDA-MB-231 cells using a C-terminally targeted antibody. Antibody specificity was confirmed by immunoblotting following siRNA knockdown of PR expression. We have further demonstrated that alternatively spliced PR mRNA is present in MDA-MB-231 cells and in reportedly PR-negative breast tumour tissue which could encode the truncated PR proteins detected by the C-terminal antibody. The potential function of PR variant proteins present in MDA-MB-231 cells was also assessed, indicating the ability of these PR variants to bind progesterone, interact with a nuclear PR co-factor and bind DNA. These findings suggest that alternative splicing may generate functional truncated PR variant proteins which are not detected by breast cancer screening using N-terminally targeted antibodies leading to misclassification as PR negative.
Collapse
Affiliation(s)
- David M W Cork
- Reproductive and Vascular Biology Research Group, Institute of Cellular Medicine, Medical School, Newcastle University, 3rd Floor William Leech Building, Framlington Place, Newcastle-upon-Tyne NE2 4HH, UK
| | | | | |
Collapse
|
35
|
Tanos T, Rojo L, Echeverria P, Brisken C. ER and PR signaling nodes during mammary gland development. Breast Cancer Res 2012; 14:210. [PMID: 22809143 PMCID: PMC3680919 DOI: 10.1186/bcr3166] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The ovarian hormones estrogen and progesterone orchestrate postnatal mammary gland development and are implicated in breast cancer. Most of our understanding of the molecular mechanisms of estrogen receptor (ER) and progesterone receptor (PR) signaling stems from in vitro studies with hormone receptor-positive cell lines. They have shown that ER and PR regulate gene transcription either by binding to DNA response elements directly or via other transcription factors and recruiting co-regulators. In addition they cross-talk with other signaling pathways through nongenomic mechanisms. Mouse genetics combined with tissue recombination techniques have provided insights about the action of these two hormones in vivo. It has emerged that hormones act on a subset of mammary epithelial cells and relegate biological functions to paracrine factors. With regards to hormonal signaling in breast carcinomas, global gene expression analyses have led to the identification of gene expression signatures that are characteristic of ERα-positive tumors that have stipulated functional studies of hitherto poorly understood transcription factors. Here, we highlight what has been learned about ER and PR signaling nodes in these different systems and attempt to lay out in which way the insights may converge.
Collapse
|
36
|
Hagan CR, Daniel AR, Dressing GE, Lange CA. Role of phosphorylation in progesterone receptor signaling and specificity. Mol Cell Endocrinol 2012; 357:43-9. [PMID: 21945472 PMCID: PMC3265648 DOI: 10.1016/j.mce.2011.09.017] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2011] [Revised: 08/08/2011] [Accepted: 09/11/2011] [Indexed: 10/17/2022]
Abstract
Progesterone receptors (PR), in concert with peptide growth factor-initiated signaling pathways, initiate massive expansion of the epithelial cell compartment associated with the process of alveologenesis in the developing mammary gland. PR-dependent signaling events also contribute to inappropriate proliferation observed in breast cancer. Notably, PR-B isoform-specific cross talk with growth factor-driven pathways is required for the proliferative actions of progesterone. Indeed, PRs act as heavily phosphorylated transcription factor "sensors" for mitogenic protein kinases that are often elevated and/or constitutively activated in invasive breast cancers. In addition, phospho-PR-target genes frequently include the components of mitogenic signaling pathways, revealing a mechanism for feed-forward signaling that confers increased responsiveness of, PR +mammary epithelial cells to these same mitogenic stimuli. Understanding the mechanisms and isoform selectivity of PR/kinase interactions may yield further insight into targeting altered signaling networks in breast and other hormonally responsive cancers (i.e. lung, uterine and ovarian) in the clinic. This review focuses on PR phosphorylation by mitogenic protein kinases and mechanisms of PR-target gene selection that lead to increased cell proliferation.
Collapse
Affiliation(s)
- Christy R Hagan
- University of Minnesota, Departments of Medicine and Pharmacology, Division of Hematology, Oncology, and Transplantation, Women's Cancer Program, Masonic Cancer Center, Minneapolis, MN 55455, United States
| | | | | | | |
Collapse
|
37
|
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
|
38
|
Phosphorylation of signal transducer and activator of transcription 3 (STAT3) correlates with Her-2 status, carbonic anhydrase 9 expression and prognosis in esophageal cancer. Clin Exp Metastasis 2012; 29:615-24. [DOI: 10.1007/s10585-012-9475-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2012] [Accepted: 03/25/2012] [Indexed: 12/12/2022]
|
39
|
Influence of conformationally restricted pyrimidines on the activity of 10–23 DNAzymes. Bioorg Med Chem 2012; 20:2581-6. [DOI: 10.1016/j.bmc.2012.02.047] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2011] [Revised: 02/14/2012] [Accepted: 02/20/2012] [Indexed: 01/05/2023]
|
40
|
Schillaci R, Guzmán P, Cayrol F, Beguelin W, Díaz Flaqué MC, Proietti CJ, Pineda V, Palazzi J, Frahm I, Charreau EH, Maronna E, Roa JC, Elizalde PV. Clinical relevance of ErbB-2/HER2 nuclear expression in breast cancer. BMC Cancer 2012; 12:74. [PMID: 22356700 PMCID: PMC3342900 DOI: 10.1186/1471-2407-12-74] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Accepted: 02/22/2012] [Indexed: 11/25/2022] Open
Abstract
Background The biological relevance of nuclear ErbB-2/HER2 (NuclErbB-2) presence in breast tumors remains unexplored. In this study we assessed the clinical significance of ErbB-2 nuclear localization in primary invasive breast cancer. The reporting recommendations for tumor marker prognostic studies (REMARK) guidelines were used as reference. Methods Tissue microarrays from a cohort of 273 primary invasive breast carcinomas from women living in Chile, a Latin American country, were examined for membrane (MembErbB-2) and NuclErbB-2 expression by an immunofluorescence (IF) protocol we developed. ErbB-2 expression was also evaluated by immunohistochemistry (IHC) with a series of antibodies. Correlation between NuclErbB-2 and MembErbB-2, and between NuclErbB-2 and clinicopathological characteristics of tumors was studied. The prognostic value of NuclErbB-2 in overall survival (OS) was evaluated using Kaplan-Meier method, and Cox model was used to explore NuclErbB-2 as independent prognostic factor for OS. Results The IF protocol we developed showed significantly higher sensitivity for detection of NuclErbB-2 than IHC procedures, while its specificity and sensitivity to detect MembErbB-2 were comparable to those of IHC procedures. We found 33.6% NuclErbB-2 positivity, 14.2% MembErbB-2 overexpression by IF, and 13.0% MembErbB-2 prevalence by IHC in our cohort. We identified NuclErbB-2 positivity as a significant independent predictor of worse OS in patients with MembErbB-2 overexpression. NuclErbB-2 was also a biomarker of lower OS in tumors that overexpress MembErbB-2 and lack steroid hormone receptors. Conclusions We revealed a novel role for NuclErbB-2 as an independent prognostic factor of poor clinical outcome in MembErbB-2-positive breast tumors. Our work indicates that patients presenting NuclErbB-2 may need new therapeutic strategies involving specific blockage of ErbB-2 nuclear migration.
Collapse
Affiliation(s)
- Roxana Schillaci
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Buenos Aires, Argentina
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Durfort T, Tkach M, Meschaninova MI, Rivas MA, Elizalde PV, Venyaminova AG, Schillaci R, François JC. Small interfering RNA targeted to IGF-IR delays tumor growth and induces proinflammatory cytokines in a mouse breast cancer model. PLoS One 2012; 7:e29213. [PMID: 22235273 PMCID: PMC3250415 DOI: 10.1371/journal.pone.0029213] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2011] [Accepted: 11/22/2011] [Indexed: 12/30/2022] Open
Abstract
Insulin-like growth factor I (IGF-I) and its type I receptor (IGF-IR) play significant roles in tumorigenesis and in immune response. Here, we wanted to know whether an RNA interference approach targeted to IGF-IR could be used for specific antitumor immunostimulation in a breast cancer model. For that, we evaluated short interfering RNA (siRNAs) for inhibition of in vivo tumor growth and immunological stimulation in immunocompetent mice. We designed 2′-O-methyl-modified siRNAs to inhibit expression of IGF-IR in two murine breast cancer cell lines (EMT6, C4HD). Cell transfection of IGF-IR siRNAs decreased proliferation, diminished phosphorylation of downstream signaling pathway proteins, AKT and ERK, and caused a G0/G1 cell cycle block. The IGF-IR silencing also induced secretion of two proinflammatory cytokines, TNF- α and IFN-γ. When we transfected C4HD cells with siRNAs targeting IGF-IR, mammary tumor growth was strongly delayed in syngenic mice. Histology of developing tumors in mice grafted with IGF-IR siRNA treated C4HD cells revealed a low mitotic index, and infiltration of lymphocytes and polymorphonuclear neutrophils, suggesting activation of an antitumor immune response. When we used C4HD cells treated with siRNA as an immunogen, we observed an increase in delayed-type hypersensitivity and the presence of cytotoxic splenocytes against wild-type C4HD cells, indicative of evolving immune response. Our findings show that silencing IGF-IR using synthetic siRNA bearing 2′-O-methyl nucleotides may offer a new clinical approach for treatment of mammary tumors expressing IGF-IR. Interestingly, our work also suggests that crosstalk between IGF-I axis and antitumor immune response can mobilize proinflammatory cytokines.
Collapse
Affiliation(s)
- Tiphanie Durfort
- Institut National de la Santé et de la Recherche Médicale (INSERM) U565, Paris, France
- Centre National de la Recherche, Scientifique, UMR 7196; Muséum National d'Histoire Naturelle, Paris, France
| | - Mercedes Tkach
- Instituto de Biología y Medicina Experimental (IBYME), Consejo de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Mariya I. Meschaninova
- Institute of Chemical Biology and Fundamental Medicine - Siberian Division of Russian Academy of Sciences (SB-RAS), Novosibirsk, Russia
| | - Martín A. Rivas
- Instituto de Biología y Medicina Experimental (IBYME), Consejo de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Patricia V. Elizalde
- Instituto de Biología y Medicina Experimental (IBYME), Consejo de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Alya G. Venyaminova
- Institute of Chemical Biology and Fundamental Medicine - Siberian Division of Russian Academy of Sciences (SB-RAS), Novosibirsk, Russia
| | - Roxana Schillaci
- Instituto de Biología y Medicina Experimental (IBYME), Consejo de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Jean-Christophe François
- Institut National de la Santé et de la Recherche Médicale (INSERM) U565, Paris, France
- Centre National de la Recherche, Scientifique, UMR 7196; Muséum National d'Histoire Naturelle, Paris, France
- * E-mail:
| |
Collapse
|
42
|
Fiszman GL, Jasnis MA. Molecular Mechanisms of Trastuzumab Resistance in HER2 Overexpressing Breast Cancer. Int J Breast Cancer 2011; 2011:352182. [PMID: 22295219 PMCID: PMC3262573 DOI: 10.4061/2011/352182] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2011] [Accepted: 07/01/2011] [Indexed: 01/24/2023] Open
Abstract
The epidermal growth factor receptor 2 (HER2) is a tyrosine kinase overexpressed in nearly 20% to 25% of invasive breast cancers. Trastuzumab is a humanized monoclonal antibody that targets HER2. The majority of patients with metastatic breast cancer initially respond to trastuzumab, however, within 1 year of treatment disease progresses. Several molecular mechanisms have been described as contributing to the development of trastuzumab resistance. They could be grouped as impaired access of trastuzumab to HER2, upregulation of HER2 downstream signaling pathways, signaling of alternative pathways, and impaired immune antitumor mechanisms. However, since many of them have overlapping effects, it would be of great clinical impact to identify the principal signaling pathways involved in drug resistance. Significant efforts are being applied to find other therapeutic modalities besides trastuzumab treatment to be used alone or in combination with current modalities.
Collapse
Affiliation(s)
- Gabriel L Fiszman
- Immunobiology Department, Institute of Oncology A. H. Roffo, University of Buenos Aires, Avenida San Martín 5481, CP1417 DTB Buenos Aires, Argentina
| | | |
Collapse
|
43
|
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
|
44
|
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
|
45
|
Tang JZ, Kong XJ, Banerjee A, Muniraj N, Pandey V, Steiner M, Perry JK, Zhu T, Liu DX, Lobie PE. STAT3alpha is oncogenic for endometrial carcinoma cells and mediates the oncogenic effects of autocrine human growth hormone. Endocrinology 2010; 151:4133-45. [PMID: 20668024 DOI: 10.1210/en.2010-0273] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
We herein demonstrate an oncogenic role for signal transducer and activator of transcription (STAT)-3alpha (the full length STAT3 isoform), which also mediates autocrine human GH (hGH)-stimulated oncogenicity, in human endometrial carcinoma (EC) cells. Autocrine hGH stimulated Y705 phosphorylation of STAT3 and STAT3-mediated transcriptional activity in a SRC and Janus-2 Kinase dependent manner in human EC cell lines. Forced expression of a constitutively active variant of STAT3alpha increased proliferation, anchorage-independent, three-dimensional (3D) Matrigel, and xenograft growth and promoted epithelial-mesenchymal transition, migration, and invasion of EC cells. Conversely, the oncogenic capacity of EC cells was significantly impaired by treatment with JSI-124, an inhibitor of STAT3 phosphorylation and activity, small interfering RNA-mediated depletion of STAT3alpha, or a dominant-negative variant of STAT3alpha. Furthermore, the enhanced EC cell oncogenicity stimulated by autocrine hGH, was also abrogated by functional inhibition or small interfering RNA-mediated depletion of STAT3alpha. STAT3alpha may therefore be a common mediator of oncogenic signaling pathways stimulating progression of EC.
Collapse
Affiliation(s)
- Jian-Zhong Tang
- Liggins Institute, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Barbieri I, Quaglino E, Maritano D, Pannellini T, Riera L, Cavallo F, Forni G, Musiani P, Chiarle R, Poli V. Stat3 is required for anchorage-independent growth and metastasis but not for mammary tumor development downstream of the ErbB-2 oncogene. Mol Carcinog 2010; 49:114-20. [PMID: 20027636 DOI: 10.1002/mc.20605] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The oncogenic transcription factor Stat3 is constitutively active in a high percentage of human tumors including mammary adenocarcinomas and is reported to participate in the ErbB-2 oncogene signaling. In order to assess the role of signal transducer and activator of transcription 3 (Stat3) in mammary tumorigenesis downstream of ErbB-2, we generated mice expressing the activated rat ErbB-2 (neu) but lacking Stat3 in the mammary epithelium. Stat3 is apparently not required for neu-driven mammary tumorigenesis as tumors developed similarly in both Stat3-sufficient and Stat3-deficient glands. However, short hairpin RNA (shRNA)-mediated Stat3 silencing in a neu-overexpressing tumor-derived cell line completely abolished both neu-driven anchorage-independent growth and lung metastasis. Our data suggest that Stat3 might be a useful therapeutic target in breast tumors showing amplification and/or overexpression of the ErbB-2 oncogene, which normally display aggressive, metastatic behavior.
Collapse
Affiliation(s)
- Isaia Barbieri
- Molecular Biotechnology Center (MBC), University of Turin, 10126 Turin, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
Steroid receptors (SRs) are hormone-activated transcription factors important for a wide variety of cellular functions. Post-translational modifications of SRs, including phosphorylation, ubiquitination, acetylation, and sumoylation regulate their expression and function. The remarkable number of phosphorylation sites in these receptors and the wide variety of kinases shown to modulate phosphorylation influence the integration between cell-signaling pathways and SR action. These phosphorylation sites have been identified in all of the functional domains with the majority being located within the amino-terminal portions of the receptors. The regulation of function is receptor specific, site specific, and often dependent on the cellular context. Numerous roles for site-specific phosphorylation have been elucidated including sensitivity of hormone response, DNA binding, expression, stability, subcellular localization, dimerization, and protein-protein interactions that can determine the regulation of specific target genes. This review summarizes the current knowledge regarding receptor site-specific phosphorylation and regulation of function. As functional assays become more sophisticated, it is likely that additional roles for phosphorylation in receptor function will be identified.
Collapse
Affiliation(s)
- Robert D Ward
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | | |
Collapse
|
48
|
Rivas MA, Tkach M, Beguelin W, Proietti CJ, Rosemblit C, Charreau EH, Elizalde PV, Schillaci R. Transactivation of ErbB-2 induced by tumor necrosis factor alpha promotes NF-kappaB activation and breast cancer cell proliferation. Breast Cancer Res Treat 2009; 122:111-24. [PMID: 19760502 DOI: 10.1007/s10549-009-0546-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2009] [Accepted: 09/03/2009] [Indexed: 01/06/2023]
Abstract
Tumor necrosis factor alpha (TNFalpha) is a pleiotropic cytokine which, acting locally, induces tumor growth. Accumulating evidence, including our findings, showed that TNFalpha is mitogenic in breast cancer cells in vitro and in vivo. In the present study, we explored TNFalpha involvement on highly aggressive ErbB-2-overexpressing breast cancer cells. We found that TNFalpha induces ErbB-2 phosphorylation in mouse breast cancer C4HD cells and in the human breast cancer cell lines SK-BR-3 and BT-474. ErbB-2 phosphorylation at Tyr877 residue was mediated by TNFalpha-induced c-Src activation. Moreover, TNFalpha promoted ErbB-2/ErbB-3 heterocomplex formation, Akt activation and NF-kappaB transcriptional activation. Inhibition of ErbB-2 by addition of AG825, an epidermal growth factor receptor/ErbB-2-tyrosine kinase inhibitor, or knockdown of ErbB-2 by RNA interference strategy, blocked TNFalpha-induced NF-kappaB activation and proliferation. However, the humanized monoclonal antibody anti-ErbB-2 Herceptin could not inhibit TNFalpha ability to promote breast cancer growth. Interestingly, our work disclosed that TNFalpha is able to transactivate ErbB-2 and use it as an obligatory downstream signaling molecule in the generation of mitogenic signals. As TNFalpha has been shown to be present in the tumor microenvironment of a significant proportion of human infiltrating breast cancers, our findings would have clinical implication in ErbB-2-positive breast cancer treatment.
Collapse
Affiliation(s)
- Martín A Rivas
- Laboratory of Molecular Mechanisms of Carcinogenesis, Instituto de Biología y Medicina Experimental, CONICET, Vuelta de Obligado 2490, Buenos Aires C1428ADN, Argentina
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Guo C, Yang G, Khun K, Kong X, Levy D, Lee P, Melamed J. Activation of Stat3 in renal tumors. Am J Transl Res 2009; 1:283-290. [PMID: 19956438 PMCID: PMC2776322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2009] [Accepted: 02/25/2009] [Indexed: 05/28/2023]
Abstract
Signal transducer and activator of transcription 3 (Stat3) plays a vital role in signal transduction pathways that mediate transformation and inhibit apoptosis. Oncogenic Stat3 is persistently activated in several human cancers and transformed cell lines. Previous studies indicate activation of Stat3 in renal cell carcinoma (RCC). However, the detailed characterization of the Stat3 expression pattern in different histologic types of RCC is lacking. We have analyzed the immunoprofile of activated or phosphorylated Stat3 (pStat3) in a tissue microarray of renal tumors of different histologic types, including 42 cases of conventional clear cell type, 24 chromophobe, and 7 papillary, 15 oncocytoma, 7 urothelial carcinoma and 21 normal kidney tissues using an anti-pStat3 antibody (recognizes only activated STAT3). pStat3 nuclear staining was observed in 25 of 42 conventional clear cell RCC (59.5 %), 8 of 24 chromophobe RCC (33.3%), 4 of 7 papillary RCC (57.1%). In the other tumor groups, 4 of 15 oncocytomas (26.7%) and 6 of 7 urothelial carcinomas (85.7%) showed positive nuclear staining. Weak nuclear immunoreactivity for pStat3 was seen in 4 of 21 cases of non-neoplastic kidney tissue (19.0%). The extent of Stat3 activation as determined by nuclear expression of its phosphorylated form is increased in histologic types of renal tumors with greater malignant potential, specifically conventional clear cell RCC, papillary RCC and urothelial carcinoma, only slightly increased in chromophobe RCC, and not increased in oncocytoma. These results suggest a role of Stat3 activation in different types of renal neoplasia, possibly serving as a prognostic marker or therapeutic target.
Collapse
Affiliation(s)
- Charles Guo
- Department of Pathology, New York University School of Medicine New York, NY
| | | | | | | | | | | | | |
Collapse
|