1
|
Arner EN, Alzhanova D, Westcott JM, Hinz S, Tiron CE, Blø M, Mai A, Virtakoivu R, Phinney N, Nord S, Aguilera KY, Rizvi A, Toombs JE, Reese TC, Fey V, Micklem D, Gausdal G, Ivaska J, Lorens JB, Brekken RA. AXL-TBK1 driven AKT3 activation promotes metastasis. Sci Signal 2024; 17:eado6057. [PMID: 39689180 DOI: 10.1126/scisignal.ado6057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 11/26/2024] [Indexed: 12/19/2024]
Abstract
The receptor tyrosine kinase AXL promotes tumor progression, metastasis, and therapy resistance through the induction of epithelial-mesenchymal transition (EMT). Here, we found that activation of AXL resulted in the phosphorylation of TANK-binding kinase 1 (TBK1) and the downstream activation of AKT3 and Snail, a transcription factor critical for EMT. Mechanistically, we showed that TBK1 directly bound to and phosphorylated AKT3 in a manner dependent on the multiprotein complex mTORC1. Upon activation, AKT3 interacted with and promoted the nuclear accumulation of Snail, which led to increased EMT as assessed by marker abundance. In human pancreatic ductal adenocarcinoma tissue, nuclear AKT3 colocalized with Snail and correlated with worse clinical outcomes. Primary mouse pancreatic cancer cells deficient in AKT3 showed reduced metastatic spread in vivo, suggesting selective AKT3 inhibition as a potential therapeutic avenue for targeting EMT in aggressive cancers.
Collapse
Affiliation(s)
- Emily N Arner
- Cancer Biology Graduate Program, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Surgery and the Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Dina Alzhanova
- Department of Surgery and the Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jill M Westcott
- Department of Surgery and the Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Stefan Hinz
- Department of Biomedicine and Centre for Cancer Biomarkers, University of Bergen, Bergen, Norway
- BerGenBio ASA, Bergen, Norway
| | - Crina Elena Tiron
- Department of Biomedicine and Centre for Cancer Biomarkers, University of Bergen, Bergen, Norway
- Regional Institute of Oncology, Iasi, Romania
| | | | | | - Reetta Virtakoivu
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland
- Department of Life Technologies, University of Turku, 20520 Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, 20520 Turku, Finland
| | - Natalie Phinney
- Cancer Biology Graduate Program, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Surgery and the Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Silje Nord
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | | | - Ali Rizvi
- Department of Surgery and the Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jason E Toombs
- Department of Surgery and the Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Tanner C Reese
- Cancer Biology Graduate Program, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Vidal Fey
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland
- Department of Life Technologies, University of Turku, 20520 Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, 20520 Turku, Finland
| | | | | | - Johanna Ivaska
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland
- Department of Life Technologies, University of Turku, 20520 Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, 20520 Turku, Finland
| | - James B Lorens
- Department of Biomedicine and Centre for Cancer Biomarkers, University of Bergen, Bergen, Norway
| | - Rolf A Brekken
- Cancer Biology Graduate Program, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Surgery and the Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
2
|
Ma J, Sun F, Li W, Du R, Liu M, Wei Q, Kang B, Yan S, Wang C. SULT2B1: a novel therapeutic target in colorectal cancer via modulation of AKT/PKM2-mediated glycolysis and proliferation. J Transl Med 2024; 22:1093. [PMID: 39623433 PMCID: PMC11613740 DOI: 10.1186/s12967-024-05910-4] [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] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 11/21/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND Sulfotransferase family 2B member 1 (SULT2B1) is involved in regulating cell proliferation, migration and metabolism. However, there is still dispute regarding whether SULT2B1 acts as an oncogene or a suppressor, and the intrinsic mechanisms in modulating tumor progression need to be further elucidated. METHODS This work aims to reveal the relationship among SULT2B1, AKT, PKM2 signaling and glycolytic pathways, and provided a theoretical basis for SULT2B1 as a potential therapeutic target for CRC.Bioinformatics methods, immunohistochemistry (IHC) and immunoblotting assays were performed to analyze the correlation between SULT2B1 and colorectal cancer (CRC). The effect of SULT2B1 on cell proliferation and migration were investigated by several phenotypic experiments in vitro and animal studies. The SULT2B1 interacting proteins were determined by immunofluorescence, immunoprecipitation and GST-pull down assays. Immunoblotting and mCherry-GFP-LC3 assays were performed to analysis autophagy. Chromatin immunoprecipitation (CHIP) assay was utilized to detect the effect of SULT2B1 in regulating transcription. Small molecule agonist/antagonist was used to modify protein activity and therefore analyze the mutual relationships. RESULTS SULT2B1 is a predictive biomarker that is abnormally overexpressed in CRC tissues. Overexpression of SULT2B1 promoted cell proliferation and migration, while its knockout suppressed these processes. Furthermore, SULT2B1 could directly interact with the oncogene AKT and thereby enhance the activity of AKT-mTORC1 signaling. Furthermore, PKM2 was found to bind with SULT2B1, and regulated by SULT2B1 at both transcription and degradation levels. Moreover, blocking glycolysis attenuated the promoting effect of OE-SULT2B1. CONCLUSION SULT2B1 acts as an oncogene in CRC via modulating the AKT/PKM2 axis, therefore making it a promising diagnostic and therapeutic target for CRC.
Collapse
Affiliation(s)
- Jianxing Ma
- Department of General Surgery, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, China
| | - Fengyao Sun
- Precision Medicine Laboratory for Chronic Non-Communicable Diseases of Shandong Province, Institute of Precision Medicine, Jining Medical University, Jining, 272067, China
| | - Wen Li
- Precision Medicine Laboratory for Chronic Non-Communicable Diseases of Shandong Province, Institute of Precision Medicine, Jining Medical University, Jining, 272067, China
| | - Ruihang Du
- Precision Medicine Laboratory for Chronic Non-Communicable Diseases of Shandong Province, Institute of Precision Medicine, Jining Medical University, Jining, 272067, China
| | - Mingchan Liu
- Precision Medicine Laboratory for Chronic Non-Communicable Diseases of Shandong Province, Institute of Precision Medicine, Jining Medical University, Jining, 272067, China
| | - Qiuya Wei
- Department of General Surgery, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, China
| | - Boxiong Kang
- Department of General Surgery, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, China
| | - Siyuan Yan
- Precision Medicine Laboratory for Chronic Non-Communicable Diseases of Shandong Province, Institute of Precision Medicine, Jining Medical University, Jining, 272067, China.
| | - Chen Wang
- Department of General Surgery, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, China.
| |
Collapse
|
3
|
Gelman IH. Metastasis suppressor genes in clinical practice: are they druggable? Cancer Metastasis Rev 2023; 42:1169-1188. [PMID: 37749308 PMCID: PMC11629483 DOI: 10.1007/s10555-023-10135-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 09/01/2023] [Indexed: 09/27/2023]
Abstract
Since the identification of NM23 (now called NME1) as the first metastasis suppressor gene (MSG), a small number of other gene products and non-coding RNAs have been identified that suppress specific parameters of the metastatic cascade, yet which have little or no ability to regulate primary tumor initiation or maintenance. MSG can regulate various pathways or cell biological functions such as those controlling mitogen-activated protein kinase pathway mediators, cell-cell and cell-extracellular matrix protein adhesion, cytoskeletal architecture, G-protein-coupled receptors, apoptosis, and transcriptional complexes. One defining facet of this gene class is that their expression is typically downregulated, not mutated, in metastasis, such that any effective therapeutic intervention would involve their re-expression. This review will address the therapeutic targeting of MSG, once thought to be a daunting task only facilitated by ectopically re-expressing MSG in metastatic cells in vivo. Examples will be cited of attempts to identify actionable oncogenic pathways that might suppress the formation or progression of metastases through the re-expression of specific metastasis suppressors.
Collapse
Affiliation(s)
- Irwin H Gelman
- Department of Cancer Genetics & Genomics, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA.
| |
Collapse
|
4
|
Yang C, Hardy P. The Multifunctional Nature of the MicroRNA/AKT3 Regulatory Axis in Human Cancers. Cells 2023; 12:2594. [PMID: 37998329 PMCID: PMC10670075 DOI: 10.3390/cells12222594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/06/2023] [Accepted: 11/07/2023] [Indexed: 11/25/2023] Open
Abstract
Serine/threonine kinase (AKT) signaling regulates diverse cellular processes and is one of the most important aberrant cell survival mechanisms associated with tumorigenesis, metastasis, and chemoresistance. Targeting AKT has become an effective therapeutic strategy for the treatment of many cancers. AKT3 (PKBγ), the least studied isoform of the AKT family, has emerged as a major contributor to malignancy. AKT3 is frequently overexpressed in human cancers, and many regulatory oncogenic or tumor suppressor small non-coding RNAs (ncRNAs), including microRNAs (miRNAs), have recently been identified to be involved in regulating AKT3 expression. Therefore, a better understanding of regulatory miRNA/AKT3 networks may reveal novel biomarkers for the diagnosis of patients with cancer and may provide invaluable information for developing more effective therapeutic strategies. The aim of this review was to summarize current research progress in the isoform-specific functions of AKT3 in human cancers and the roles of dysregulated miRNA/AKT3 in specific types of human cancers.
Collapse
Affiliation(s)
- Chun Yang
- Research Center of CHU Sainte-Justine, University of Montréal, Montreal, QC H3T 1C5, Canada;
| | - Pierre Hardy
- Research Center of CHU Sainte-Justine, University of Montréal, Montreal, QC H3T 1C5, Canada;
- Department of Pharmacology and Physiology, Department of Pediatrics, University of Montréal, Montreal, QC H3T 1C5, Canada
| |
Collapse
|
5
|
Xio Y, Zhou L, Andl T, Zhang Y. YAP1 controls the N-cadherin-mediated tumor-stroma interaction in melanoma progression. RESEARCH SQUARE 2023:rs.3.rs-2944243. [PMID: 37546745 PMCID: PMC10402251 DOI: 10.21203/rs.3.rs-2944243/v3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Epithelial-to-mesenchymal transition (EMT) is crucial for melanoma cells to escape keratinocyte control, invade underlying dermal tissues, and metastasize to distant organs. The hallmark of EMT is the switch from epithelial cadherin (E-cadherin) to neural cadherin (N-cadherin), allowing melanoma cells to form a homotypic N-cadherin-mediated adhesion with stromal fibroblasts. However, how "cadherin switching" is initiated, maintained, and regulated in melanoma remains unknown. Here, we show that upon Yes-associated protein 1 (YAP1) ablation in cancer-associated fibroblasts (CAFs), the progression of a BRAF-mutant mouse melanoma was significantly suppressed in vivo, and overexpressing YAP1 in CAFs accelerated melanoma growth. CAFs require the YAP1 function to proliferate, migrate, remodel the cytoskeletal machinery and matrix, and promote cancer cell invasion. By RNA-Seq, N-cadherin was identified as a major downstream effector of YAP1 signaling in CAFs. YAP1 silencing led to N-cadherin downregulation in CAFs, which subsequently induced the downregulation of N-cadherin in neighboring melanoma cells. N-cadherin downregulation inhibited the PI3K-AKT signaling pathway in melanoma cells and suppressed melanoma growth in vivo, supporting the role of N-cadherin as an adhesive and signaling molecule in melanoma cells. This finding suggests that YAP1 depletion in CAFs induces the downregulation of p-AKT signaling in melanoma cells through the N-cadherin-mediated interaction between melanoma cells and CAFs. Importantly, our data underscore that CAFs can regulate N-cadherin-mediated interactions with melanoma cells. Thus, disentangling cadherin-mediated cell-cell interactions can potentially disrupt tumor-stroma interactions and reverse the tumor cell invasive phenotype.
Collapse
|
6
|
Schulz B, Schumacher V, Ngezahayo A, Maier-Begandt D, Schadzek N, Wilhelm J, Weidner W, Pilatz A, Fietz D, Kliesch S, Schnepel N, Hambruch N, Rode K, Langeheine M, Brehm R. Analysis of connexin 43, connexin 45 and N-cadherin in the human sertoli cell line FS1 and the human seminoma-like cell line TCam-2 in comparison with human testicular biopsies. BMC Cancer 2023; 23:232. [PMID: 36899312 PMCID: PMC10007848 DOI: 10.1186/s12885-023-10696-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 03/01/2023] [Indexed: 03/12/2023] Open
Abstract
BACKGROUND Germ cell tumors are relatively common in young men. They derive from a non-invasive precursor, called germ cell neoplasia in situ, but the exact pathogenesis is still unknown. Thus, further understanding provides the basis for diagnostics, prognostics and therapy and is therefore paramount. A recently developed cell culture model consisting of human FS1 Sertoli cells and human TCam-2 seminoma-like cells offers new opportunities for research on seminoma. Since junctional proteins within the seminiferous epithelium are involved in cell organization, differentiation and proliferation, they represent interesting candidates for investigations on intercellular adhesion and communication in context with neoplastic progression. METHODS FS1 and TCam-2 cells were characterized regarding gap-junction-related connexin 43 (Cx43) and connexin 45 (Cx45), and adherens-junction-related N-cadherin using microarray, PCR, Western blot, immunocytochemistry and immunofluorescence. Results were compared to human testicular biopsies at different stages of seminoma development via immunohistochemistry to confirm the cell lines' representativeness. Furthermore, dye-transfer measurements were performed to investigate functional cell coupling. RESULTS Cx43, Cx45 and N-cadherin mRNA and protein were generally detectable in both cell lines via qualitative RT-PCR and Western blot. Immunocytochemistry and immunofluorescence revealed a mainly membrane-associated expression of N-cadherin in both cell lines, but gene expression values were higher in FS1 cells. Cx43 expression was also membrane-associated in FS1 cells but barely detectable in TCam-2 cells. Accordingly, a high gene expression value of Cx43 was measured for FS1 and a low value for TCam-2 cells. Cx45 was primary located in the cytoplasm of FS1 and TCam-2 cells and revealed similar low to medium gene expression values in both cell lines. Overall, results were comparable with corresponding biopsies. Additionally, both FS1 and TCam-2 cells showed dye diffusion into neighboring cells. CONCLUSION The junctional proteins Cx43, Cx45 and N-cadherin are expressed in FS1 and TCam-2 cells at mRNA and/or protein level in different amounts and localizations, and cells of both lines are functionally coupled among each other. Concerning the expression of these junctional proteins, FS1 and TCam-2 cells are largely representative for Sertoli and seminoma cells, respectively. Thus, these results provide the basis for further coculture experiments evaluating the role of junctional proteins in context with seminoma progression.
Collapse
Affiliation(s)
- Birte Schulz
- Institute of Anatomy, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany.
| | - Valérie Schumacher
- Department of Urology and Medicine, Boston Children's Hospital, Boston, MA, USA.,Department of Surgery and Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Anaclet Ngezahayo
- Department of Cell Physiology and Biophysics, Institute of Cell Biology and Biophysics, Leibniz University Hannover, Hannover, Germany.,Center for Systems Neuroscience Hannover, University of Veterinary Medicine Hannover Foundation, Hannover, Germany
| | - Daniela Maier-Begandt
- Department of Cell Physiology and Biophysics, Institute of Cell Biology and Biophysics, Leibniz University Hannover, Hannover, Germany
| | - Nadine Schadzek
- Department of Cell Biology, Institute of Cell Biology and Biophysics, Leibniz University Hannover, Hannover, Germany
| | - Jochen Wilhelm
- Institute for Lung Health, Justus Liebig University Giessen, Giessen, Germany.,Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research, Justus Liebig University Giessen, Giessen, Germany.,The Cardiopulmonary Institute, Justus Liebig University Giessen, Giessen, Germany
| | - Wolfgang Weidner
- Department of Urology, Pediatric Urology and Andrology, Justus Liebig University Giessen, Giessen, Germany
| | - Adrian Pilatz
- Department of Urology, Pediatric Urology and Andrology, Justus Liebig University Giessen, Giessen, Germany
| | - Daniela Fietz
- Department of Veterinary Anatomy, Histology and Embryology, Justus Liebig University Giessen, Giessen, Germany
| | - Sabine Kliesch
- Centre of Andrology and Reproductive Medicine, University of Muenster, Muenster, Germany
| | - Nadine Schnepel
- Institute of Anatomy, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Nina Hambruch
- Institute of Anatomy, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Kristina Rode
- Institute of Anatomy, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Marion Langeheine
- Institute of Anatomy, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Ralph Brehm
- Institute of Anatomy, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| |
Collapse
|
7
|
Chi R, Yao C, Chen S, Liu Y, He Y, Zhang J, Ellies LG, Wu X, Zhao Q, Zhou C, Wang Y, Sun H. Elevated BCAA Suppresses the Development and Metastasis of Breast Cancer. Front Oncol 2022; 12:887257. [PMID: 35785192 PMCID: PMC9243538 DOI: 10.3389/fonc.2022.887257] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/16/2022] [Indexed: 11/29/2022] Open
Abstract
Branched-chain amino acids (BCAAs) are the three essential amino acids including leucine, isoleucine, and valine. BCAA metabolism has been linked with the development of a variety of tumors. However, the impact of dietary BCAA intake on breast tumor progression and metastasis remains to be fully explored. Here, we unexpectedly find that the elevated BCAA, either in the genetic model or via increasing dietary intake in mice, suppresses the tumor growth and lung metastasis of breast cancer. The survival analysis shows that BCAA catabolic gene expression is strongly associated with long-term oncological outcomes in patients with breast cancer. In Pp2cm knockout mice in which BCAAs accumulate due to the genetic defect of BCAA catabolism, the breast tumor growth is suppressed. Interestingly, while the cell proliferation and tumor vasculature remain unaffected, more cell death occurs in the tumor in Pp2cm knockout mice, accompanied with increased natural killer (NK) cells. Importantly, increasing BCAA dietary intake suppresses breast tumor growth in mice. On the other hand, there are fewer lung metastases from primary breast tumor in Pp2cm knockout mice and the high BCAA diet-fed mice, suggesting high BCAA also suppresses the lung metastasis of breast cancer. Furthermore, low BCAA diet promotes lung colonization of breast cancer cells in tail vein model. The migration and invasion abilities of breast cancer cells are impaired by high concentration of BCAA in culture medium. The suppressed tumor metastasis and cell migration/invasion abilities by elevated BCAA are accompanied with reduced N-cadherin expression. Together, these data show high BCAA suppresses both tumor growth and metastasis of breast cancer, demonstrating the potential benefits of increasing BCAA dietary intake in the treatment of breast cancer.
Collapse
Affiliation(s)
- Rui Chi
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chengcheng Yao
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Si Chen
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yunxia Liu
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanqi He
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jin Zhang
- Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lesley G. Ellies
- Department of Pathology, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Xuefeng Wu
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qian Zhao
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Cixiang Zhou
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Cixiang Zhou, ; Ying Wang, ; Haipeng Sun,
| | - Ying Wang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Cixiang Zhou, ; Ying Wang, ; Haipeng Sun,
| | - Haipeng Sun
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- NHC Key Laboratory of Hormones and Development, Center for Cardiovascular Diseases, The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
- *Correspondence: Cixiang Zhou, ; Ying Wang, ; Haipeng Sun,
| |
Collapse
|
8
|
Hua H, Zhang H, Chen J, Wang J, Liu J, Jiang Y. Targeting Akt in cancer for precision therapy. J Hematol Oncol 2021; 14:128. [PMID: 34419139 PMCID: PMC8379749 DOI: 10.1186/s13045-021-01137-8] [Citation(s) in RCA: 144] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/03/2021] [Indexed: 02/08/2023] Open
Abstract
Biomarkers-guided precision therapeutics has revolutionized the clinical development and administration of molecular-targeted anticancer agents. Tailored precision cancer therapy exhibits better response rate compared to unselective treatment. Protein kinases have critical roles in cell signaling, metabolism, proliferation, survival and migration. Aberrant activation of protein kinases is critical for tumor growth and progression. Hence, protein kinases are key targets for molecular targeted cancer therapy. The serine/threonine kinase Akt is frequently activated in various types of cancer. Activation of Akt promotes tumor progression and drug resistance. Since the first Akt inhibitor was reported in 2000, many Akt inhibitors have been developed and evaluated in either early or late stage of clinical trials, which take advantage of liquid biopsy and genomic or molecular profiling to realize personalized cancer therapy. Two inhibitors, capivasertib and ipatasertib, are being tested in phase III clinical trials for cancer therapy. Here, we highlight recent progress of Akt signaling pathway, review the up-to-date data from clinical studies of Akt inhibitors and discuss the potential biomarkers that may help personalized treatment of cancer with Akt inhibitors. In addition, we also discuss how Akt may confer the vulnerability of cancer cells to some kinds of anticancer agents.
Collapse
Affiliation(s)
- Hui Hua
- State Key Laboratory of Biotherapy, Laboratory of Stem Cell Biology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Hongying Zhang
- State Key Laboratory of Biotherapy, Laboratory of Oncogene, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jingzhu Chen
- State Key Laboratory of Biotherapy, Laboratory of Oncogene, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jiao Wang
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jieya Liu
- State Key Laboratory of Biotherapy, Laboratory of Oncogene, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yangfu Jiang
- State Key Laboratory of Biotherapy, Laboratory of Oncogene, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
9
|
Hinz N, Baranowsky A, Horn M, Kriegs M, Sibbertsen F, Smit DJ, Clezardin P, Lange T, Schinke T, Jücker M. Knockdown of AKT3 Activates HER2 and DDR Kinases in Bone-Seeking Breast Cancer Cells, Promotes Metastasis In Vivo and Attenuates the TGFβ/CTGF Axis. Cells 2021; 10:cells10020430. [PMID: 33670586 PMCID: PMC7922044 DOI: 10.3390/cells10020430] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/12/2021] [Accepted: 02/16/2021] [Indexed: 12/12/2022] Open
Abstract
Bone metastases frequently occur in breast cancer patients and lack appropriate treatment options. Hence, understanding the molecular mechanisms involved in the multistep process of breast cancer bone metastasis and tumor-induced osteolysis is of paramount interest. The serine/threonine kinase AKT plays a crucial role in breast cancer bone metastasis but the effect of individual AKT isoforms remains unclear. Therefore, AKT isoform-specific knockdowns were generated on the bone-seeking MDA-MB-231 BO subline and the effect on proliferation, migration, invasion, and chemotaxis was analyzed by live-cell imaging. Kinome profiling and Western blot analysis of the TGFβ/CTGF axis were conducted and metastasis was evaluated by intracardiac inoculation of tumor cells into NOD scid gamma (NSG) mice. MDA-MB-231 BO cells exhibited an elevated AKT3 kinase activity in vitro and responded to combined treatment with AKT- and mTOR-inhibitors. Knockdown of AKT3 significantly increased migration, invasion, and chemotaxis in vitro and metastasis to bone but did not significantly enhance osteolysis. Furthermore, knockdown of AKT3 increased the activity and phosphorylation of pro-metastatic HER2 and DDR1/2 but lowered protein levels of CTGF after TGFβ-stimulation, an axis involved in tumor-induced osteolysis. We demonstrated that AKT3 plays a crucial role in bone-seeking breast cancer cells by promoting metastatic potential without facilitating tumor-induced osteolysis.
Collapse
Affiliation(s)
- Nico Hinz
- Center for Experimental Medicine, Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.H.); (F.S.); (D.J.S.)
| | - Anke Baranowsky
- Center for Experimental Medicine, Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (A.B.); (T.S.)
- Department of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Michael Horn
- University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany;
- Mildred Scheel Cancer Career Center Hamburg, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Malte Kriegs
- Department of Radiotherapy & Radiation Oncology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany;
- UCCH Kinomics Core Facility, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Freya Sibbertsen
- Center for Experimental Medicine, Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.H.); (F.S.); (D.J.S.)
| | - Daniel J. Smit
- Center for Experimental Medicine, Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.H.); (F.S.); (D.J.S.)
| | - Philippe Clezardin
- INSERM, Research Unit UMR S1033, LyOS, Faculty of Medicine Lyon-Est, University of Lyon 1, 69372 Lyon, France;
| | - Tobias Lange
- Center for Experimental Medicine, Department of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany;
| | - Thorsten Schinke
- Center for Experimental Medicine, Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (A.B.); (T.S.)
| | - Manfred Jücker
- Center for Experimental Medicine, Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.H.); (F.S.); (D.J.S.)
- Correspondence: ; Tel.: +49-(0)-40-7410-56339
| |
Collapse
|
10
|
Pisano S, Wang X, Garcia-Parra J, Gazze A, Edwards K, Feltracco V, Hu Y, He L, Gonzalez D, Francis LW, Conlan RS, Li C. Nanomicelles potentiate histone deacetylase inhibitor efficacy in vitro. Cancer Nanotechnol 2020. [DOI: 10.1186/s12645-020-00070-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Abstract
Background
Amphiphilic block copolymers used as nanomicelle drug carriers can effectively overcome poor drug solubility and specificity issues. Hence, these platforms have a broad applicability in cancer treatment. In this study, Pluronic F127 was used to fabricate nanomicelles containing the histone deacetylase inhibitor SAHA, which has an epigenetic-driven anti-cancer effect in several tumor types. SAHA-loaded nanomicelles were prepared using a thin-film drying method and characterized for size, surface charge, drug content, and drug release properties. Loaded particles were tested for in vitro activity and their effect on cell cycle and markers of cancer progression.
Results
Following detailed particle characterization, cell proliferation experiments demonstrated that SAHA-loaded nanomicelles more effectively inhibited the growth of HeLa and MCF-7 cell lines compared with free drug formulations. The 30 nm SAHA containing nanoparticles were able to release up to 100% of the encapsulated drug over a 72 h time window. Moreover, gene and protein expression analyses suggested that their cytoreductive effect was achieved through the regulation of p21 and p53 expression. SAHA was also shown to up-regulate E-cadherin expression, potentially influencing tumor migration.
Conclusions
This study highlights the opportunity to exploit pluronic-based nanomicelles for the delivery of compounds that regulate epigenetic processes, thus inhibiting cancer development and progression.
Collapse
|
11
|
Eichberger J, Schulz D, Pscheidl K, Fiedler M, Reichert TE, Bauer RJ, Ettl T. PD-L1 Influences Cell Spreading, Migration and Invasion in Head and Neck Cancer Cells. Int J Mol Sci 2020; 21:ijms21218089. [PMID: 33138288 PMCID: PMC7663567 DOI: 10.3390/ijms21218089] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/21/2020] [Accepted: 10/27/2020] [Indexed: 02/06/2023] Open
Abstract
The programmed cell death protein-1 (PD-1)/programmed cell death ligand-1 (PD-L1) axis blockade has been implemented in advanced-stage tumor therapy for various entities, including head and neck squamous cell carcinoma (HNSCC). Despite a promising tumor response in a subgroup of HNSCC patients, the majority suffer from disease progression. PD-L1 is known to influence several intrinsic mechanisms in cancer cells, such as proliferation, apoptosis, migration and invasion. Here, we modulated PD-L1 expression in three HNSCC cell lines with differential intrinsic PD-L1 expression. In addition to an alteration in the epithelial-to-mesenchymal transition (EMT) marker expression, we observed PD-L1-dependent cell spreading, migration and invasion in a spheroid spreading assay on four different coatings (poly-L-lysine, collagen type I, fibronectin and Matrigel®) and a chemotactic transwell migration/invasion assay. Furthermore, the overexpression of PD-L1 led to increased gene expression and small interfering ribonucleic acid (siRNA) knockdown and decreased gene expression of Rho-GTPases and related proteins in a RT2 Profiler™ PCR Array. Rac1 and Rho-GTPase pulldown assays revealed a change in the activation state concordantly with PD-L1 expression. In summary, our results suggest a major role for PD-L1 in favoring cell motility, including cell spreading, migration and invasion. This is presumably caused by altered N-cadherin expression and changes in the activation states of small Rho-GTPases Rho and Rac1.
Collapse
Affiliation(s)
- Jonas Eichberger
- Department of Oral and Maxillofacial Surgery, University Hospital Regensburg, 9305 Regensburg, Germany; (J.E.); (D.S.); (K.P.); (M.F.); (T.E.R.); (T.E.)
- Department of Oral and Maxillofacial Surgery and Center for Medical Biotechnology, University Hospital Regensburg, 9305 Regensburg, Germany
| | - Daniela Schulz
- Department of Oral and Maxillofacial Surgery, University Hospital Regensburg, 9305 Regensburg, Germany; (J.E.); (D.S.); (K.P.); (M.F.); (T.E.R.); (T.E.)
- Department of Oral and Maxillofacial Surgery and Center for Medical Biotechnology, University Hospital Regensburg, 9305 Regensburg, Germany
| | - Kristian Pscheidl
- Department of Oral and Maxillofacial Surgery, University Hospital Regensburg, 9305 Regensburg, Germany; (J.E.); (D.S.); (K.P.); (M.F.); (T.E.R.); (T.E.)
| | - Mathias Fiedler
- Department of Oral and Maxillofacial Surgery, University Hospital Regensburg, 9305 Regensburg, Germany; (J.E.); (D.S.); (K.P.); (M.F.); (T.E.R.); (T.E.)
- Department of Oral and Maxillofacial Surgery and Center for Medical Biotechnology, University Hospital Regensburg, 9305 Regensburg, Germany
| | - Torsten Eugen Reichert
- Department of Oral and Maxillofacial Surgery, University Hospital Regensburg, 9305 Regensburg, Germany; (J.E.); (D.S.); (K.P.); (M.F.); (T.E.R.); (T.E.)
| | - Richard Josef Bauer
- Department of Oral and Maxillofacial Surgery, University Hospital Regensburg, 9305 Regensburg, Germany; (J.E.); (D.S.); (K.P.); (M.F.); (T.E.R.); (T.E.)
- Department of Oral and Maxillofacial Surgery and Center for Medical Biotechnology, University Hospital Regensburg, 9305 Regensburg, Germany
- Correspondence:
| | - Tobias Ettl
- Department of Oral and Maxillofacial Surgery, University Hospital Regensburg, 9305 Regensburg, Germany; (J.E.); (D.S.); (K.P.); (M.F.); (T.E.R.); (T.E.)
| |
Collapse
|
12
|
Li Y, Lv Z, Zhang S, Wang Z, He L, Tang M, Pu W, Zhao H, Zhang Z, Shi Q, Cai D, Wu M, Hu G, Lui KO, Feng J, Nieto MA, Zhou B. Genetic Fate Mapping of Transient Cell Fate Reveals N-Cadherin Activity and Function in Tumor Metastasis. Dev Cell 2020; 54:593-607.e5. [PMID: 32668208 DOI: 10.1016/j.devcel.2020.06.021] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 04/26/2020] [Accepted: 06/16/2020] [Indexed: 01/06/2023]
Abstract
Genetic lineage tracing unravels cell fate and plasticity in development, tissue homeostasis, and diseases. However, it remains technically challenging to trace temporary or transient cell fate, such as epithelial-to-mesenchymal transition (EMT) in tumor metastasis. Here, we generated a genetic fate-mapping system for temporally seamless tracing of transient cell fate. Highlighting its immediate application, we used it to study EMT gene activity from the local primary tumor to a distant metastatic site in vivo. In a spontaneous breast-to-lung metastasis model, we found that primary tumor cells activated vimentin and N-cadherin in situ, but only N-cadherin was activated and functionally required during metastasis. Tumor cells that have ever expressed N-cadherin constituted the majority of metastases in lungs, and functional deletion of N-cad significantly reduced metastasis. The seamless genetic recording system described here provides an alternative way for understanding transient cell fate and plasticity in biological processes.
Collapse
Affiliation(s)
- Yan Li
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Zan Lv
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Shaohua Zhang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Zhuo Wang
- Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, 201100, Shanghai, China
| | - Lingjuan He
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Muxue Tang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Wenjuan Pu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Huan Zhao
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Zhenqian Zhang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Qihui Shi
- Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, 201100, Shanghai, China
| | - Dongqing Cai
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou 510632, China
| | - Mingfu Wu
- Center for Cardiovascular Sciences, Albany Medical College, Albany, NY 12208, USA
| | - Guohong Hu
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Kathy O Lui
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong SAR 999077, China
| | - Jing Feng
- Laboratory Medicine, Southern Medical University Affiliated Fengxian Hospital, Shanghai 201400, China
| | - M Angela Nieto
- Institute de Neurociencias CSIC-UMH, Avda. Ramon y Cajal s/n, 03550 San Juan de Alicante, Spain
| | - Bin Zhou
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China; Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou 510632, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China.
| |
Collapse
|
13
|
Hinz N, Jücker M. Distinct functions of AKT isoforms in breast cancer: a comprehensive review. Cell Commun Signal 2019; 17:154. [PMID: 31752925 PMCID: PMC6873690 DOI: 10.1186/s12964-019-0450-3] [Citation(s) in RCA: 218] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 10/04/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AKT, also known as protein kinase B, is a key element of the PI3K/AKT signaling pathway. Moreover, AKT regulates the hallmarks of cancer, e.g. tumor growth, survival and invasiveness of tumor cells. After AKT was discovered in the early 1990s, further studies revealed that there are three different AKT isoforms, namely AKT1, AKT2 and AKT3. Despite their high similarity of 80%, the distinct AKT isoforms exert non-redundant, partly even opposing effects under physiological and pathological conditions. Breast cancer as the most common cancer entity in women, frequently shows alterations of the PI3K/AKT signaling. MAIN CONTENT A plethora of studies addressed the impact of AKT isoforms on tumor growth, metastasis and angiogenesis of breast cancer as well as on therapy response and overall survival in patients. Therefore, this review aimed to give a comprehensive overview about the isoform-specific effects of AKT in breast cancer and to summarize known downstream and upstream mechanisms. Taking account of conflicting findings among the studies, the majority of the studies reported a tumor initiating role of AKT1, whereas AKT2 is mainly responsible for tumor progression and metastasis. In detail, AKT1 increases cell proliferation through cell cycle proteins like p21, p27 and cyclin D1 and impairs apoptosis e.g. via p53. On the downside AKT1 decreases migration of breast cancer cells, for instance by regulating TSC2, palladin and EMT-proteins. However, AKT2 promotes migration and invasion most notably through regulation of β-integrins, EMT-proteins and F-actin. Whilst AKT3 is associated with a negative ER-status, findings about the role of AKT3 in regulation of the key properties of breast cancer are sparse. Accordingly, AKT1 is mutated and AKT2 is amplified in some cases of breast cancer and AKT isoforms are associated with overall survival and therapy response in an isoform-specific manner. CONCLUSIONS Although there are several discussed hypotheses how isoform specificity is achieved, the mechanisms behind the isoform-specific effects remain mostly unrevealed. As a consequence, further effort is necessary to achieve deeper insights into an isoform-specific AKT signaling in breast cancer and the mechanism behind it.
Collapse
Affiliation(s)
- Nico Hinz
- Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Manfred Jücker
- Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.
| |
Collapse
|
14
|
Cao ZQ, Wang Z, Leng P. Aberrant N-cadherin expression in cancer. Biomed Pharmacother 2019; 118:109320. [DOI: 10.1016/j.biopha.2019.109320] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 07/26/2019] [Accepted: 07/31/2019] [Indexed: 12/12/2022] Open
|
15
|
Wang Y, Shi L, Li J, Li L, Wang H, Yang H. Long-term cadmium exposure promoted breast cancer cell migration and invasion by up-regulating TGIF. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2019; 175:110-117. [PMID: 30897409 DOI: 10.1016/j.ecoenv.2019.03.046] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 03/09/2019] [Accepted: 03/12/2019] [Indexed: 06/09/2023]
Abstract
Cadmium (Cd) is a known human carcinogen. Previous studies have demonstrated that Cd exposure promoted migration and invasion of breast cancer cells. However, the molecular mechanisms underlying this process have not yet been clearly addressed. The purpose of this study was to investigate whether TG-interacting factor (TGIF) was involved in long-term Cd exposure-induced migration and invasion of breast cancer cells. Human breast cancer cells were continuously exposed to Cd for eight weeks. Western blot and qRT-PCR assays were performed to measure the expression of protein and mRNA. Migration and invasion assays were performed to assess the migratory and invasive ability of human breast cancer cells. Our data indicated that long-term Cd exposure obviously increased the expression of TGIF protein and mRNA in human breast cancer cells. Long-term Cd exposure increased the ability of migration and invasion of human breast cancer cells, which could be inhibited by transfection of small interfering RNA (siRNA) targeting TGIF. We also observed that the long-term Cd exposure-induced up-regulation of MMP2 mRNA expression was modulated by TGIF. In conclusion, our findings suggested that TGIF/MMP2 signaling axis might be involved in malignant progression stimulated by long-term Cd exposure in human breast cancer.
Collapse
Affiliation(s)
- Yadong Wang
- Department of Toxicology, Henan Center for Disease Control and Prevention, Zhengzhou, 450016, China.
| | - Li Shi
- Department of Epidemiology, School of Public Health, Zhengzhou University, Zhengzhou, 450001, China
| | - Jiangmin Li
- Department of Toxicology, Henan Center for Disease Control and Prevention, Zhengzhou, 450016, China
| | - Li Li
- Department of Toxicology, Henan Center for Disease Control and Prevention, Zhengzhou, 450016, China
| | - Haiyu Wang
- Department of Toxicology, Henan Center for Disease Control and Prevention, Zhengzhou, 450016, China
| | - Haiyan Yang
- Department of Epidemiology, School of Public Health, Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
16
|
Basil polysaccharide inhibits hypoxia-induced hepatocellular carcinoma metastasis and progression through suppression of HIF-1α-mediated epithelial-mesenchymal transition. Int J Biol Macromol 2019; 137:32-44. [PMID: 31252022 DOI: 10.1016/j.ijbiomac.2019.06.189] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 05/05/2019] [Accepted: 06/24/2019] [Indexed: 12/24/2022]
Abstract
Invasion and metastasis of cancerous cells affects the treatment and prognosis of hepatocellular carcinoma (HCC). HIF-1α-induced epithelial-mesenchymal transition (EMT) is a critical process associated with cancer metastasis. Basil polysaccharide (BPS), one of the major active ingredients isolated from Basil (Ocimum basilicum L.), has been identified to possess an antitumor activity for HCC. In our current study, BPS was obtained by water extraction and ethanol precipitation method and the characterization was analyzed through ultraviolet absorption spectra and Fourier-transform infrared spectrum. A CoCl2-induced hypoxia model and a HCC cell line-derived xenograft (CDX) model were used to explore the anti-metastasis efficacy and the mechanism that underlies the antitumor activity of BPS. The results showed that hypoxia could facilitate EMT and promote HCC cells migration and/or invasion. Conversely, BPS inhibited the progression and metastasis of tumor, as well as reversed EMT by causing cytoskeletal remodeling under hypoxic conditions. Moreover, BPS alleviated tumor hypoxia by targeting HIF1α, and the mesenchymal markers (β-catenin, N-cadherin and vimentin) were down-regulated, while the epithelial markers (E-cadherin, VMP1 and ZO-1) were up-regulated after BPS treatment under hypoxic conditions. Thus, these results suggested that BPS may be a valuable option for use in clinical treatment of HCC and other malignant tumors.
Collapse
|
17
|
Aras S, Maroun MC, Song Y, Bandyopadhyay S, Stark A, Yang ZQ, Long MP, Grossman LI, Fernández-Madrid F. Mitochondrial autoimmunity and MNRR1 in breast carcinogenesis. BMC Cancer 2019; 19:411. [PMID: 31046734 PMCID: PMC6498478 DOI: 10.1186/s12885-019-5575-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 04/03/2019] [Indexed: 02/07/2023] Open
Abstract
Background Autoantibodies function as markers of tumorigenesis and have been proposed to enhance early detection of malignancies. We recently reported, using immunoscreening of a T7 complementary DNA (cDNA) library of breast cancer (BC) proteins with sera from patients with BC, the presence of autoantibodies targeting several mitochondrial DNA (mtDNA)-encoded subunits of the electron transport chain (ETC) in complexes I, IV, and V. Methods In this study, we have characterized the role of Mitochondrial-Nuclear Retrograde Regulator 1 (MNRR1, also known as CHCHD2), identified on immunoscreening, in breast carcinogenesis. We assessed the protein as well as transcript levels of MNRR1 in BC tissues and in derived cell lines representing tumors of graded aggressiveness. Mitochondrial function was also assayed and correlated with the levels of MNRR1. We studied the invasiveness of BC derived cells and the effect of MNRR1 levels on expression of genes associated with cell proliferation and migration such as Rictor and PGC-1α. Finally, we manipulated levels of MNRR1 to assess its effect on mitochondria and on some properties linked to a metastatic phenotype. Results We identified a nuclear DNA (nDNA)-encoded mitochondrial protein, MNRR1, that was significantly associated with the diagnosis of invasive ductal carcinoma (IDC) of the breast by autoantigen microarray analysis. In focusing on the mechanism of action of MNRR1 we found that its level was nearly twice as high in malignant versus benign breast tissue and up to 18 times as high in BC cell lines compared to MCF10A control cells, suggesting a relationship to aggressive potential. Furthermore, MNRR1 affected levels of multiple genes previously associated with cancer metastasis. Conclusions MNRR1 regulates multiple genes that function in cell migration and cancer metastasis and is higher in cell lines derived from aggressive tumors. Since MNRR1 was identified as an autoantigen in breast carcinogenesis, the present data support our proposal that both mitochondrial autoimmunity and MNRR1 activity in particular are involved in breast carcinogenesis. Virtually all other nuclear encoded genes identified on immunoscreening of invasive BC harbor an MNRR1 binding site in their promoters, thereby placing MNRR1 upstream and potentially making it a novel marker for BC metastasis.
Collapse
Affiliation(s)
- Siddhesh Aras
- Wayne State University School of Medicine, Center for Molecular Medicine and Genetics, 540 E. Canfield Ave, Detroit, MI, 48201, USA
| | - Marie-Claire Maroun
- Department of Internal Medicine, Wayne State University, Detroit, MI, 48201, USA.,Division of Rheumatology, Department of Internal Medicine, Wayne State University, Detroit, MI, 48201, USA
| | - Yeohan Song
- Department of Internal Medicine, Wayne State University, Detroit, MI, 48201, USA
| | | | - Azadeh Stark
- Department of Pathology, Henry Ford Health System, Detroit, MI, 48201, USA
| | - Zeng-Quan Yang
- Department of Oncology and Karmanos Cancer Institute, Wayne State University, Detroit, MI, 48201, USA
| | - Michael P Long
- Department of Pathology, Wayne State University, Detroit, MI, 48201, USA
| | - Lawrence I Grossman
- Wayne State University School of Medicine, Center for Molecular Medicine and Genetics, 540 E. Canfield Ave, Detroit, MI, 48201, USA.
| | - Félix Fernández-Madrid
- Department of Internal Medicine, Wayne State University, Detroit, MI, 48201, USA. .,Division of Rheumatology, Department of Internal Medicine, Wayne State University, Detroit, MI, 48201, USA. .,Wayne State University, University Health Center, 4H, 4201 St. Antoine, Detroit, MI, 48201, USA.
| |
Collapse
|
18
|
Jin W, Li QZ, Zuo YC, Cao YN, Zhang LQ, Hou R, Su WX. Relationship Between DNA Methylation in Key Region and the Differential Expressions of Genes in Human Breast Tumor Tissue. DNA Cell Biol 2018; 38:49-62. [PMID: 30346835 DOI: 10.1089/dna.2018.4276] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Breast cancer has a high mortality rate for females. Aberrant DNA methylation plays a crucial role in the occurrence and progression of breast carcinoma. By comparing DNA methylation differences between tumor breast tissue and normal breast tissue, we calculate and analyze the distributions of the hyper- and hypomethylation sites in different function regions. Results indicate that enhancer regions are often hypomethylated in breast cancer. CpG islands (CGIs) are mainly hypermethylated, while the flanking CGI (shores and shelves) is more easily hypomethylated. The hypomethylation in gene body region is related to the upregulation of gene expression, and the hypomethylation of enhancer regions is closely associated with gene expression upregulation in breast cancer. Some key hypomethylation sites in enhancer regions and key hypermethylation sites in CGIs for regulating key genes are, respectively, found, such as oncogenes ESR1 and ERBB2 and tumor suppressor genes FBLN2, CEBPA, and FAT4. This suggests that the recognizing methylation status of these genes will be useful for the diagnosis of breast cancer.
Collapse
Affiliation(s)
- Wen Jin
- 1 Laboratory of Theoretical Biophysics, School of Physical Science and Technology, Inner Mongolia University , Hohhot, China
| | - Qian-Zhong Li
- 1 Laboratory of Theoretical Biophysics, School of Physical Science and Technology, Inner Mongolia University , Hohhot, China .,2 The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University , Hohhot, China
| | - Yong-Chun Zuo
- 2 The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University , Hohhot, China
| | - Yan-Ni Cao
- 1 Laboratory of Theoretical Biophysics, School of Physical Science and Technology, Inner Mongolia University , Hohhot, China
| | - Lu-Qiang Zhang
- 1 Laboratory of Theoretical Biophysics, School of Physical Science and Technology, Inner Mongolia University , Hohhot, China
| | - Rui Hou
- 1 Laboratory of Theoretical Biophysics, School of Physical Science and Technology, Inner Mongolia University , Hohhot, China
| | - Wen-Xia Su
- 3 College of Science, Inner Mongolia Agricultural University , Hohhot, China
| |
Collapse
|
19
|
[Identification of diagnostic tumour markers and therapeutic targets in testicular tumours]. DER PATHOLOGE 2018; 39:215-220. [PMID: 30206653 DOI: 10.1007/s00292-018-0493-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Today, tumour classification has been expanded due to immunohistochemical and molecular-pathological analyses due to corresponding patterns/profiles of protein and gene expression. The latter analyses often include growth factors and their ligands, intracellular signalling pathways, DNA-binding proteins, and oncogenes and suppressor genes, thus functionally including primarily the regulation of growth including angiogenesis and apoptosis as well as the induction of metastases to adhesion and migration disorders. Based on observations that testicular tumours often show microcalcifications, possibly due to impaired calcium metabolism, we focused on calcium-dependent transmembrane proteins, particularly cadherins, in the search for new tumour markers and therapeutic targets. N‑cadherin is expressed differently in the various subtypes of germ cell tumours and is useful in N‑cadherin-positive germ cell tumours as a novel therapeutic targeting structure, particularly in cisplatin resistance, due to functional analysis. In the tumours of the sex cord stroma beta-catenin and the transcription factor SOX-9 give a clear classification of these tumours. Thus, morphological investigations prove to be pilot experiments to purposefully narrow the spectrum of functionally important proteins and thus to establish promising new differential diagnostic markers or target structures.
Collapse
|
20
|
Zhao X, Ren Y, Cui N, Wang X, Cui Y. Identification of key microRNAs and their targets in exosomes of pancreatic cancer using bioinformatics analysis. Medicine (Baltimore) 2018; 97:e12632. [PMID: 30278585 PMCID: PMC6181532 DOI: 10.1097/md.0000000000012632] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Pancreatic cancer (PC) is one of the most lethal tumors, due to late diagnosis and limited surgical strategies. It has been reported that serum exosomal microRNAs (S-Exo-miRNAs) play a pivotal role as signaling molecules and serve as noninvasive diagnosis methods for PC. The combination of S-Exo-miRNAs with the corresponding target also plays an important role in the tumor microenvironment.Here we investigated S-Exo-miRNAs involved in PC. The gene expression profile was downloaded from the Gene Expression Omnibus (GEO) database. The analysis was carried out using GEO2R. The targets of differentially expressed serum exosomal miRNAs (DE-S-Exo-miRNAs) were predicted by 4 bioinformatic algorithms (miRanda, miRDB, miRWalk, and Targetscan). Further analysis with gene ontology (GO) and Kyoto Encyclopedia of Genomes pathway (KEGG) enrichment analyses were performed with Cytoscape software version 3.4.0. Subsequently, the interaction regulatory network of target genes was performed with the Search Tool for the Retrieval of Interacting Genes (STRING) database (http://www.string-db.org/) and visualized using Cytoscape software.We downloaded the gene expression profile GSE50632, which was based on an Agilent microarray GPL17660 platform containing 4 eligible samples. In total 467 DE-S-Exo-miRNAs were obtained, including 7 overexpressed miRNAs (1.50%), and 460 remaining underexpressed miRNAs (98.50%). The databases miRWalk, miRDB, miRanda, and TargetScan were used to predict their potential targets, which were subsequently submitted to Cytoscape software version 3.4.0 (www.cytoscape.org). Next the functional and pathway enrichment analysis were used for the KEGG pathway and GO categories analysis. The enrichment analysis identified the genes involved in such processes as developmental and negative regulation of multicellular organismal processes, regulation of anatomical structure morphogenesis, regulation of cell death, apoptotic processes and mitogen-activated protein kinase (MAPK) signaling pathway, transforming growth factor - beta (TGF -β) signaling pathway, cyclic adenosine monophosphate (cAMP) signaling pathway, and the phosphatidylinositol-3 kinases/Akt (PI3K-Akt) signaling pathway. Subsequently according to the protein-protein interaction (PPI) network, the top 10 genes were obtained. The enrichment analyses of the genes involved in a significant module revealed that these genes were related to the TGF-β signaling pathway. After reviewing the literature, we identified the apoptosis genes, and their corresponding miRNAs that have a relationship with apoptosis of the tumor.This analysis provides a comprehensive understanding of the roles of S-Exo-miRNAs and the related targets in the development of PC. Additionally, the present study provides promising candidate targets for early diagnosis and therapeutic intervention. However, these predictions require further experimental validation in future studies.
Collapse
Affiliation(s)
- Xin Zhao
- Tianjin Medical University, Tianjin
- Department of Surgery, Tianjin Nankai Hospital, Nankai Clinical School, Tianjin Medical University
| | - Yiming Ren
- Department of Bone and Joint, Tianjin Union Medicine Center, PR China
| | - Naiqiang Cui
- Department of Surgery, Tianjin Nankai Hospital, Nankai Clinical School, Tianjin Medical University
| | - Ximo Wang
- Department of Surgery, Tianjin Nankai Hospital, Nankai Clinical School, Tianjin Medical University
| | - Yunfeng Cui
- Department of Surgery, Tianjin Nankai Hospital, Nankai Clinical School, Tianjin Medical University
| |
Collapse
|
21
|
Hseu YC, Chang GR, Pan JY, Rajendran P, Mathew DC, Li ML, Liao JW, Chen WTL, Yang HL. Antrodia camphorata inhibits epithelial-to-mesenchymal transition by targeting multiple pathways in triple-negative breast cancers. J Cell Physiol 2018; 234:4125-4139. [PMID: 30146779 DOI: 10.1002/jcp.27222] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 07/16/2018] [Indexed: 12/28/2022]
Abstract
Antrodia camphorata (AC) exhibits potential for engendering cell-cycle arrest as well as prompting apoptosis and metastasis inhibition in triple-negative breast cancer (TNBC) cells. We performed the current study to explore the anti-epithelial-to-mesenchymal transition (EMT) properties of fermented AC broth in TNBC cells. Our results illustrated that noncytotoxic concentrations of AC (20-60 μg/ml) reversed the morphological changes (fibroblastic-to-epithelial phenotype) as well as the EMT by upregulating the observed E-cadherin expression. Furthermore, we discovered treatment with AC substantially inhibit the Twist expression in human TNBC (MDA-MB-231) cells as well as in those that were transfected with Twist. In addition, we determined AC to decrease the observed Wnt/β-catenin nuclear translocation through a pathway determined to be dependent on GSK3β. Notably, AC treatment consistently inhibited the EMT by downregulating mesenchymal marker proteins like N-cadherin, vimentin, Snail, ZEB-1, and fibronectin; at that same time upregulating epithelial marker proteins like occludin and ZO-1. Bioluminescence imaging that was executed in vivo demonstrated AC substantially suppressed breast cancer metastasis to the lungs. Notably, we found that western blot analysis confirmed that AC decreased lung metastasis as demonstrated by upregulation of E-cadherin expression in biopsied lung tissue. Together with our results support the anti-EMT activity of AC, indicating AC as having the potential for acting as an anticancer agent for the treatment of human TNBC treatment.
Collapse
Affiliation(s)
- You-Cheng Hseu
- Department of Cosmeceutics, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, Taiwan.,Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan.,Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
| | - Geng-Ruei Chang
- Institute of Biotechnology and Bioinformatics, Asia University, Taichung, Taiwan
| | - Jian-You Pan
- Institute of Biotechnology and Bioinformatics, Asia University, Taichung, Taiwan
| | - Peramaiyan Rajendran
- Department of Cosmeceutics, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, Taiwan
| | - Dony Chacko Mathew
- Department of Cosmeceutics, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, Taiwan
| | - Mei-Ling Li
- Department of Nutrition, Institute of Nutrition, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, Taiwan
| | - Jiunn-Wang Liao
- Graduate Institute of Veterinary Pathology, National Chung Hsing University, Taichung, Taiwan
| | - William Tzu-Liang Chen
- Division of Colorectal Surgery, Department of Surgery, Center of Minimally Invasive Surgery, China Medical University Hospital, China Medical University, Taichung, Taiwan.,College of Medicine, China Medical University, Taichung, Taiwan
| | - Hsin-Ling Yang
- Department of Nutrition, Institute of Nutrition, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, Taiwan
| |
Collapse
|
22
|
Zhang Z, Wen P, Li F, Yao C, Wang T, Liang B, Yang Q, Ma L, He L. HIPK2 inhibits cell metastasis and improves chemosensitivity in esophageal squamous cell carcinoma. Exp Ther Med 2017; 15:1113-1118. [PMID: 29434701 DOI: 10.3892/etm.2017.5468] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 09/22/2017] [Indexed: 12/13/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is one of the most aggressive and lethal malignancies worldwide. At present, the underlying mechanisms of ESCC development and progression are poorly understood. Previous studies have demonstrated that homeodomain-interacting protein kinase-2 (HIPK2) serves an important role in cancer biology, particularly in proliferation and metastasis. However, the role of HIPK2 in ESCC cells remains unclear. In the current study, the expression of HIPK2 in ESCC specimens, adjacent non-cancerous tissues and cell lines was assessed using reverse transcription-quantitative polymerase chain reaction (RT-qPCR). The effects of HIPK2 on cell metastasis, epithelial-mesenchymal transition (EMT) and proliferation were studied using a Transwell assay, RT-qPCR and a Cell Counting Kit-8 assay, respectively. The results indicated that HIPK2 expression was downregulated in ESCC specimens and cell lines, and HIPK2 expression was associated with tumor invasion and lymph node metastasis. Functional studies demonstrated that HIPK2 overexpression inhibited cell metastasis and EMT. Furthermore, HIPK2 overexpression suppressed cell viability during cisplatin treatment. These results suggest that HIPK2 serves an important role in regulating metastasis and the chemosensitivity of ESCC cells, implicating the potential application of HIPK2 in ESCC therapy.
Collapse
Affiliation(s)
- Zhen Zhang
- Department of Clinical Oncology, The First People's Hospital of Nanyang, Nanyang, Henan 473000, P.R. China
| | - Penghai Wen
- Department of Clinical Oncology, The Affiliated Nanshi Hospital of Henan University, Nanyang, Henan 473000, P.R. China
| | - Fangfang Li
- Department of Clinical Oncology, The First People's Hospital of Nanyang, Nanyang, Henan 473000, P.R. China
| | - Chuanshan Yao
- Department of Clinical Oncology, The First People's Hospital of Nanyang, Nanyang, Henan 473000, P.R. China
| | - Tongfu Wang
- Department of Clinical Oncology, The First People's Hospital of Nanyang, Nanyang, Henan 473000, P.R. China
| | - Bing Liang
- Department of Clinical Oncology, The First People's Hospital of Nanyang, Nanyang, Henan 473000, P.R. China
| | - Qingle Yang
- Department of Clinical Oncology, The First People's Hospital of Nanyang, Nanyang, Henan 473000, P.R. China
| | - Lei Ma
- Department of Clinical Oncology, The First People's Hospital of Nanyang, Nanyang, Henan 473000, P.R. China
| | - Limin He
- Department of Clinical Oncology, The First People's Hospital of Nanyang, Nanyang, Henan 473000, P.R. China
| |
Collapse
|
23
|
Suyama K, Yao J, Liang H, Benard O, Loudig OD, Amgalan D, McKimpson WM, Phillips GR, Segall J, Wang Y, Fineberg S, Norton L, Kitsis RN, Hazan RB. An Akt3 Splice Variant Lacking the Serine 472 Phosphorylation Site Promotes Apoptosis and Suppresses Mammary Tumorigenesis. Cancer Res 2017; 78:103-114. [PMID: 29038347 DOI: 10.1158/0008-5472.can-15-1462] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 07/05/2017] [Accepted: 10/10/2017] [Indexed: 12/29/2022]
Abstract
The Akt pathway is a well-known promoter of tumor malignancy. Akt3 is expressed as two alternatively spliced variants, one of which lacks the key regulatory serine 472 phosphorylation site. Whereas the function of full-length Akt3 isoform (Akt3/+S472) is well-characterized, that of Akt3/-S472 isoform remains unknown. Despite being expressed at a substantially lower level than Akt3/+S472 in triple-negative breast cancer cells, specific ablation of Akt3/-S472 enhanced, whereas overexpression, suppressed mammary tumor growth, consistent with a significant association with patient survival duration relative to Akt3/+S472. These effects were due to striking induction of apoptosis, which was mediated by Bim upregulation, leading to conformational activation of Bax and caspase-3 processing. Bim accumulation was caused by marked endocytosis of EGF receptors with concomitant ERK attenuation, which stabilizes BIM. These findings demonstrate an unexpected function of an endogenously expressed Akt isoform in promoting, as opposed to suppressing, apoptosis, underscoring that Akt isoforms may exert dissonant functions in malignancy.Significance: These results illuminate an unexpected function for an endogenously expressed Akt isoform in promoting apoptosis, underscoring the likelihood that different Akt isoforms exert distinct functions in human cancer. Cancer Res; 78(1); 103-14. ©2017 AACR.
Collapse
Affiliation(s)
- Kimita Suyama
- Department of Pathology and Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, New York
| | - Jiahong Yao
- Department of Pathology and Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, New York
| | - Huizhi Liang
- Department of Pathology and Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, New York
| | - Outhiriaradjou Benard
- Department of Pathology and Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, New York
| | - Olivier D Loudig
- Department of Pathology and Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, New York
| | - Dulguun Amgalan
- Department of Medicine and Cell Biology, Wilf Family Cardiovascular Research Institute and Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, New York
| | - Wendy M McKimpson
- Department of Medicine and Cell Biology, Wilf Family Cardiovascular Research Institute and Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, New York
| | - Greg R Phillips
- Department of Biology, College of Staten Island, City University of New York, Staten Island, New York
| | - Jeffrey Segall
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, New York
| | - Yihong Wang
- Department of Pathology, The Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - Susan Fineberg
- Department of Pathology and Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, New York
| | - Larry Norton
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Richard N Kitsis
- Department of Medicine and Cell Biology, Wilf Family Cardiovascular Research Institute and Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, New York
| | - Rachel B Hazan
- Department of Pathology and Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, New York.
| |
Collapse
|
24
|
Chan KK, Matchett KB, Coulter JA, Yuen HF, McCrudden CM, Zhang SD, Irwin GW, Davidson MA, Rülicke T, Schober S, Hengst L, Jaekel H, Platt-Higgins A, Rudland PS, Mills KI, Maxwell P, El-Tanani M, Lappin TR. Erythropoietin drives breast cancer progression by activation of its receptor EPOR. Oncotarget 2017; 8:38251-38263. [PMID: 28418910 PMCID: PMC5503530 DOI: 10.18632/oncotarget.16368] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 02/27/2017] [Indexed: 01/01/2023] Open
Abstract
Breast cancer is a leading cause of cancer-related deaths. Anemia is common in breast cancer patients and can be treated with blood transfusions or with recombinant erythropoietin (EPO) to stimulate red blood cell production. Clinical studies have indicated decreased survival in some groups of cancer patients treated with EPO. Numerous tumor cells express the EPO receptor (EPOR), posing a risk that EPO treatment would enhance tumor growth, but the mechanisms involved in breast tumor progression are poorly understood.Here, we have examined the functional role of the EPO-EPOR axis in pre-clinical models of breast cancer. EPO induced the activation of PI3K/AKT and MAPK pathways in human breast cancer cell lines. EPOR knockdown abrogated human tumor cell growth, induced apoptosis through Bim, reduced invasiveness, and caused downregulation of MYC expression. EPO-induced MYC expression is mediated through the PI3K/AKT and MAPK pathways, and overexpression of MYC partially rescued loss of cell proliferation caused by EPOR downregulation. In a xenotransplantation model, designed to simulate recombinant EPO therapy in breast cancer patients, knockdown of EPOR markedly reduced tumor growth.Thus, our experiments in vitro and in vivo demonstrate that functional EPOR signaling is essential for the tumor-promoting effects of EPO and underline the importance of the EPO-EPOR axis in breast tumor progression.
Collapse
Affiliation(s)
- Ka Kui Chan
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast BT9 7AE, UK
- Department of Pathology, The University of Hong Kong, Hong Kong Special Administrative Region Hong Kong 999077, China
| | - Kyle B. Matchett
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast BT9 7AE, UK
| | | | - Hiu-Fung Yuen
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast BT9 7AE, UK
| | | | - Shu-Dong Zhang
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast BT9 7AE, UK
- Northern Ireland Centre for Stratified Medicine, Biomedical Sciences Research Institute, Ulster University, Londonderry BT47 6SB, UK
| | - Gareth W. Irwin
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast BT9 7AE, UK
| | - Matthew A. Davidson
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast BT9 7AE, UK
| | - Thomas Rülicke
- Institute of Laboratory Animal Science, University of Veterinary Medicine Vienna, Vienna A-1210, Austria
| | - Sophie Schober
- Institute of Laboratory Animal Science, University of Veterinary Medicine Vienna, Vienna A-1210, Austria
| | - Ludger Hengst
- Division of Medical Biochemistry, Biocenter, Innsbruck Medical University, Innsbruck A-6020, Austria
| | - Heidelinde Jaekel
- Division of Medical Biochemistry, Biocenter, Innsbruck Medical University, Innsbruck A-6020, Austria
| | - Angela Platt-Higgins
- Institute of Integrative Biology, University of Liverpool, Liverpool L69 3BX, UK
| | - Philip S. Rudland
- Institute of Integrative Biology, University of Liverpool, Liverpool L69 3BX, UK
| | - Ken I. Mills
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast BT9 7AE, UK
| | - Perry Maxwell
- Northern Ireland Molecular Pathology Laboratory, Belfast Health & Social Care Trust, Queen's University Belfast, Belfast BT9 7AE, UK
| | - Mohamed El-Tanani
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast BT9 7AE, UK
- Institute of Cancer Therapeutics, University of Bradford, Bradford, West Yorkshire BD7 1DP, UK
| | - Terence R. Lappin
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast BT9 7AE, UK
| |
Collapse
|
25
|
Sun H, Liu M, Wu X, Yang C, Zhang Y, Xu Z, Gao K, Wang F. Overexpression of N-cadherin and β-catenin correlates with poor prognosis in patients with nasopharyngeal carcinoma. Oncol Lett 2017; 13:1725-1730. [PMID: 28454316 DOI: 10.3892/ol.2017.5645] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 10/27/2016] [Indexed: 12/20/2022] Open
Abstract
An increasing amount of evidence demonstrates that epithelial-mesenchymal transition (EMT) is important in tumor invasion and metastases. The cell-cell adhesion molecule N-cadherin and the Wnt/β-catenin cascade protein β-catenin are two biomarkers of EMT. The present study aimed to measure the expression levels of N-cadherin and β-catenin in samples from patients with nasopharyngeal carcinoma (NPC) and evaluate their prognostic significance. N-cadherin and β-catenin mRNA was evaluated using reverse transcription-quantitative polymerase chain reaction in 26 NPC tissue samples and 8 nasopharyngeal epithelium samples. Protein expression of N-cadherin and β-catenin was also detected using immunohistochemistry in 128 archival NPC paraffin-embedded specimens. Finally, associations between clinical pathological parameters and prognostic values in NPC were evaluated. The results demonstrated that both the mRNA and protein levels of N-cadherin and β-catenin were significantly increased in NPC tissues compared with the controls. Enhanced expression of N-cadherin and β-catenin protein was strongly correlated with the status of lymph node metastasis and clinical stages in patients with NPC. Notably, high expression of N-cadherin and β-catenin proteins was significantly correlated with lower overall survival (OS) rate in patients with NPC. Finally, multivariate analysis demonstrated that expression of N-cadherin protein and clinical stages were independent prognostic factors for patients with NPC. Therefore, the present study demonstrated that N-cadherin and β-catenin expression may be used as potential prognostic biomarkers for patients with NPC.
Collapse
Affiliation(s)
- Hong Sun
- Department of Otolaryngology, Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Mingyu Liu
- Department of Otolaryngology, Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Xuewen Wu
- Department of Otolaryngology, Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Chunguang Yang
- Department of Otolaryngology, Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Yanni Zhang
- Department of Otolaryngology, Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Zhenhang Xu
- Department of Otolaryngology, Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Kelei Gao
- Department of Otolaryngology, Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Fengjun Wang
- Department of Otolaryngology, Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
26
|
Lin D, Kuang G, Wan J, Zhang X, Li H, Gong X, Li H. Luteolin suppresses the metastasis of triple-negative breast cancer by reversing epithelial-to-mesenchymal transition via downregulation of β-catenin expression. Oncol Rep 2016; 37:895-902. [PMID: 27959422 DOI: 10.3892/or.2016.5311] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 08/23/2016] [Indexed: 11/05/2022] Open
Abstract
The metastasis of breast cancer is associated with dismal prognosis and high mortality due to the lack of effective treatment. Luteolin, a natural flavonoid compound, has been shown to exert antitumor activity in various types of cancers. However, the effects and mechanisms of luteolin on the metastasis of triple-negative breast cancer (TNBC) remain elusive. In the present study, we found that pretreatment of highly metastatic TNBC cell lines with luteolin dose‑dependently inhibited cell migration and invasion, and reversed epithelial-mesenchymal transition (EMT) as determined by altered morphological characteristics, downregulated epithelial markers and upregulated mesenchymal markers, and inhibited EMT-related transcription factors. In an in vivo metastasis experiment using a xenograft model, luteolin markedly inhibited lung metastases of breast cancer and the expression of EMT molecules vimentin and Slug in primary tumor tissues. Notably, luteolin also suppressed the expression of β-catenin mRNA and protein in vitro and in vivo. Furthermore, overexpression of β-catenin by adenoviruses blocked these benefits of luteolin on invasion and metastases of breast cancer. In conclusion, all these results indicated that luteolin effectively suppressed metastases of breast cancer by reversing EMT, which may be mediated by downregulation of β-catenin.
Collapse
Affiliation(s)
- Dan Lin
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Ge Kuang
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Jingyuan Wan
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Xiang Zhang
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Hongzhong Li
- Molecular Oncology and Epigenetics Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Xia Gong
- Department of Anatomy, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Hongyuan Li
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
27
|
Li B, Shi H, Wang F, Hong D, Lv W, Xie X, Cheng X. Expression of E-, P- and N-Cadherin and Its Clinical Significance in Cervical Squamous Cell Carcinoma and Precancerous Lesions. PLoS One 2016; 11:e0155910. [PMID: 27223886 PMCID: PMC4880319 DOI: 10.1371/journal.pone.0155910] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 05/08/2016] [Indexed: 01/08/2023] Open
Abstract
Aberrant expression of classical cadherins has been observed in tumor invasion and metastasis, but its involvement in cervical carcinogenesis and cancer progression is not clear. We investigated E-, P- and N-cadherin expression and its significance in cervical squamous cell carcinoma (SCC) and cervical intraepithelial neoplasia (CIN). This retrospective study enrolled 508 patients admitted to Women's Hospital, School of Medicine, Zhejiang University with cervical lesions between January 2006 and December 2010. Immunochemical staining was performed in 98 samples of normal cervical epithelium (NC), 283 of CIN, and 127 of early-stage SCC. The association of cadherin staining with clinical characteristics and survival of the patients was evaluated by univariate and multivariate analysis. We found gradients of decreasing E-cadherin expression and increasing P-cadherin expression from NC through CIN to SCC. Aberrant E-cadherin and P-cadherin expression were significantly associated with clinical parameters indicating poor prognosis and shorter patient survival. Interestingly, we found very low levels of positive N-cadherin expression in CIN and SCC tissues that were not related to CIN or cancer. Pearson chi-square tests showed that E-cadherin expression in SCC was inversely correlated with P-cadherin expression (E-P switch), and was not correlated with N-cadherin expression. More important, patients with tissues exhibiting an E-P switch in expression had highly aggressive phenotypes and poorer prognosis than those without E-P switch expression. Our findings suggest that E-cadherin and P-cadherin, but not N-cadherin staining, might be useful in diagnosing CIN and for predicting prognosis in patients with early-stage SCC.
Collapse
Affiliation(s)
- Baohua Li
- Department of Gynecologic Oncology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Haiyan Shi
- Department of Pathology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Fenfen Wang
- Department of Gynecologic Oncology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Die Hong
- Department of Gynecologic Oncology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Weiguo Lv
- Department of Gynecologic Oncology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Women’s Reproductive Health Key Laboratory of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xing Xie
- Department of Gynecologic Oncology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Women’s Reproductive Health Key Laboratory of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaodong Cheng
- Department of Gynecologic Oncology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Women’s Reproductive Health Key Laboratory of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- * E-mail:
| |
Collapse
|
28
|
Zuo J, Wen J, Lei M, Wen M, Li S, Lv X, Luo Z, Wen G. Hypoxia promotes the invasion and metastasis of laryngeal cancer cells via EMT. Med Oncol 2016; 33:15. [PMID: 26749588 DOI: 10.1007/s12032-015-0716-6] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 11/27/2015] [Indexed: 12/20/2022]
Abstract
The purpose of this study is to explore the role of hypoxia on the invasion and metastasis of laryngeal carcinoma. The invasion and migration ability of laryngeal cancer SCC10A cell was detected by transwell assay. Western blot was applied to analyze the expression of EMT-related proteins. Fifty-seven samples from postoperative patients with laryngeal cancer were collected to study. Immunohistochemistry was used to examine the expression of GLUT-1 and EMT-related proteins (Vim, E-cad, N-cad) in normal laryngeal squamous epithelial tissue, laryngeal cancer adjacent tissues and laryngeal squamous cell carcinoma tissues. Hypoxia promoted laryngeal cancer cell invasion and migration. Hypoxia also enhanced the expression of GLUT-1, vimentin and N-cad, which exist statistically significant correlation with the clinical staging and lymph node metastases (P < 0.05). The expression of GLUT-1 is positively correlated with Vim and N-cad expression in laryngeal squamous cell carcinoma tissues, but negatively correlated with E-cad expression. The patient survival rate with the positive expression of GLUT-1, Vim and N-cad becomes much shorter compared with those with negative expression of GLUT-1, Vim and N-cad (P < 0.05). Hypoxia promoted laryngeal cancer cell invasion and migration via EMT.
Collapse
Affiliation(s)
- Jianhong Zuo
- Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China. .,Nanhua Hospital, University of South China, Hengyang, 421000, Hunan, People's Republic of China.
| | - Juan Wen
- Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Mingsheng Lei
- Department of Respiratory and Critical Care Medicine, Zhangjiajie City Hospital, Zhangjiajie, 427000, Hunan, People's Republic of China
| | - Meiling Wen
- Nanhua Hospital, University of South China, Hengyang, 421000, Hunan, People's Republic of China
| | - Sai Li
- Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China.,Nanhua Hospital, University of South China, Hengyang, 421000, Hunan, People's Republic of China
| | - Xiu Lv
- Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China.,Nanhua Hospital, University of South China, Hengyang, 421000, Hunan, People's Republic of China
| | - Zhaoyang Luo
- Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China.
| | - Gebo Wen
- Medical School, University of South China, Hengyang, 421001, Hunan, People's Republic of China.
| |
Collapse
|
29
|
Grottke A, Ewald F, Lange T, Nörz D, Herzberger C, Bach J, Grabinski N, Gräser L, Höppner F, Nashan B, Schumacher U, Jücker M. Downregulation of AKT3 Increases Migration and Metastasis in Triple Negative Breast Cancer Cells by Upregulating S100A4. PLoS One 2016; 11:e0146370. [PMID: 26741489 PMCID: PMC4704820 DOI: 10.1371/journal.pone.0146370] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 12/16/2015] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Treatment of breast cancer patients with distant metastases represents one of the biggest challenges in today's gynecological oncology. Therefore, a better understanding of mechanisms promoting the development of metastases is of paramount importance. The serine/threonine kinase AKT was shown to drive cancer progression and metastasis. However, there is emerging data that single AKT isoforms (i.e. AKT1, AKT2 and AKT3) have different or even opposing functions in the regulation of cancer cell migration in vitro, giving rise to the hypothesis that inhibition of distinct AKT isoforms might have undesirable effects on cancer dissemination in vivo. METHODS The triple negative breast cancer cell line MDA-MB-231 was used to investigate the functional roles of AKT in migration and metastasis. AKT single and double knockdown cells were generated using isoform specific shRNAs. Migration was analyzed using live cell imaging, chemotaxis and transwell assays. The metastatic potential of AKT isoform knockdown cells was evaluated in a subcutaneous xenograft mouse model in vivo. RESULTS Depletion of AKT3, but not AKT1 or AKT2, resulted in increased migration in vitro. This effect was even more prominent in AKT2,3 double knockdown cells. Furthermore, combined downregulation of AKT2 and AKT3, as well as AKT1 and AKT3 significantly increased metastasis formation in vivo. Screening for promigratory proteins revealed that downregulation of AKT3 increases the expression of S100A4 protein. In accordance, depletion of S100A4 by siRNA approach reverses the increased migration induced by knockdown of AKT3. CONCLUSIONS We demonstrated that knockdown of AKT3 can increase the metastatic potential of triple negative breast cancer cells. Therefore, our results provide a rationale for the development of AKT isoform specific inhibitors.
Collapse
Affiliation(s)
- Astrid Grottke
- Center for Experimental Medicine, Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Florian Ewald
- Department of Hepatobiliary and Transplant Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Tobias Lange
- Center for Experimental Medicine, Department of Anatomy and Experimental Morphology, University Cancer Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Dominik Nörz
- Center for Experimental Medicine, Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Christiane Herzberger
- Center for Experimental Medicine, Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Johanna Bach
- Center for Experimental Medicine, Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Nicole Grabinski
- Center for Experimental Medicine, Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Lareen Gräser
- Center for Experimental Medicine, Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Frank Höppner
- Center for Experimental Medicine, Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Björn Nashan
- Department of Hepatobiliary and Transplant Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Udo Schumacher
- Center for Experimental Medicine, Department of Anatomy and Experimental Morphology, University Cancer Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Manfred Jücker
- Center for Experimental Medicine, Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
- * E-mail:
| |
Collapse
|
30
|
Tong GH, Tong WW, Qin XS, Lu LP, Liu Y. DBD-F induces apoptosis in gastric cancer-derived cells through suppressing HIF2α expression. Cell Oncol (Dordr) 2015; 38:479-84. [PMID: 26526811 DOI: 10.1007/s13402-015-0253-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/22/2015] [Indexed: 01/29/2023] Open
Abstract
PURPOSE Gastric cancer is the third leading cause of cancer-related death in China. Accumulating evidence indicates that HIF2α may affect the aggressiveness of gastric cancer. It has also been found that HIF2α C-terminal PAS domains can form complexes with inactive benzoxadiazole antagonists. Here, the anti-tumor effect of 4-(N,Ndimethylaminosulphonyl)-7-fluoro-1,2,3-benzoxadiazole (DBD-F) on human gastric cancer cells was examined using both in vitro and in vivo assays. METHODS AND RESULTS We found that DBD-F can induce apoptosis and inhibit the mobility of MKN28 and MKN45 gastric cancer-derived cells in vitro. We also found that DBD-F can suppress tumor growth in established gastric cancer-derived xenograft models in vivo. Finally, we found that DBD-F can inhibit HIF2α expression in gastric cancer-derived cells. CONCLUSIONS From our findings we conclude that DBD-F (i) is cytotoxic to gastric cancer-derived cells and (ii) can induce apoptosis in these cells via the MEK/ERK signaling pathway. In addition, our findings strongly indicate that DBD-F can inhibit HIF2α expression by affecting the phosphorylation status of MEK/ERK in gastric cancer-derived cells.
Collapse
Affiliation(s)
- Guang-Hui Tong
- Department of Laboratory Medicine, ShengJing Affiliated Hospital, China Medical University, Shenyang, 110004, China
| | - Wei-Wei Tong
- Department of Laboratory Medicine, ShengJing Affiliated Hospital, China Medical University, Shenyang, 110004, China
| | - Xiao-Song Qin
- Department of Laboratory Medicine, ShengJing Affiliated Hospital, China Medical University, Shenyang, 110004, China
| | - Li-Ping Lu
- Department of Laboratory Medicine, ShengJing Affiliated Hospital, China Medical University, Shenyang, 110004, China
| | - Yong Liu
- Department of Laboratory Medicine, ShengJing Affiliated Hospital, China Medical University, Shenyang, 110004, China.
| |
Collapse
|
31
|
Ge R, Wang Z, Wu S, Zhuo Y, Otsetov AG, Cai C, Zhong W, Wu CL, Olumi AF. Metformin represses cancer cells via alternate pathways in N-cadherin expressing vs. N-cadherin deficient cells. Oncotarget 2015; 6:28973-87. [PMID: 26359363 PMCID: PMC4745705 DOI: 10.18632/oncotarget.5023] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 08/13/2015] [Indexed: 12/12/2022] Open
Abstract
Metformin has emerged as a potential anticancer agent. Here, we demonstrate that metformin plays an anti-tumor role via repressing N-cadherin, independent of AMPK, in wild-type N-cadherin cancer cells. Ectopic-expression of N-cadherin develops metformin-resistant cancer cells, while suppression of N-cadherin sensitizes cancer to metformin. Manipulation of AMPK expression does not alter sensitivity of cancer to metformin. We show that NF-kappaB is a downstream molecule of N-cadherin and metformin regulates NF-kappaB signaling via suppressing N-cadherin. Moreover, we also suggest that TWIST1 is an upstream molecule of N-cadherin/NF-kappaB signaling and manipulation of TWIST1 expression changes the sensitivity of cancer cells to metformin. In contrast to the cells that express N-cadherin, in N-cadherin deficient cells, metformin plays an anti-tumor role via activation of AMPK. Ectopic expression of N-cadherin makes cancer more resistant to metformin. Therefore, we suggest that metformin's anti-cancer therapeutic effect is mediated through different molecular mechanism in wild-type vs. deficient N-cadherin cancer cells. At last, we selected 49 out of 984 patients' samples with prostatic cancer after radical prostatectomy (selection criteria: Gleason score ≥ 7 and all patients taking metformin) and showed levels of N-cadherin, p65 and AMPK could predict post-surgical recurrence in prostate cancer after treatment of metformin.
Collapse
Affiliation(s)
- Rongbin Ge
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Zongwei Wang
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Shulin Wu
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Yangjia Zhuo
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Aleksandar G. Otsetov
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Chao Cai
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Weide Zhong
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Chin-Lee Wu
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Aria F. Olumi
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
32
|
Wang L, Sang Y, Tang J, Zhang RH, Luo D, Chen M, Deng WG, Kang T. Down-regulation of prostate stem cell antigen (PSCA) by Slug promotes metastasis in nasopharyngeal carcinoma. J Pathol 2015; 237:411-22. [PMID: 26147638 DOI: 10.1002/path.4582] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 06/11/2015] [Accepted: 06/30/2015] [Indexed: 01/27/2023]
Abstract
Distant metastasis and local recurrence are still the major causes for failure of treatment in patients with nasopharyngeal carcinoma (NPC), making it urgent to further elicit the molecular mechanisms of NPC metastasis. Using a gene microarray including transcription factors and known markers for cancer stem cells, prostate stem cell antigen (PSCA) was found to be significantly down-regulated in metastatic NPC in lymph node, compared to its primary tumour, and in NPC cell lines with high metastatic ability compared to those with low metastatic ability. NPC patients with low PSCA expression had a consistently poor metastasis-free survival (p = 0.003). Knockdown and overexpression of PSCA respectively enhanced and impaired the migration and invasion in vitro and the lung metastasis in vivo of NPC cells. Mechanistically, the enhancement of NPC metastasis by knocking down PSCA probably involved epithelial-mesenchymal transition (EMT), by up-regulating N-cadherin and ZEB1/2 and by activating RhoA. The down-regulation of PSCA in NPC cells resulted directly from the binding of Slug to the PSCA promoter. PSCA may be a potential diagnostic marker and therapeutic target for patients with NPC.
Collapse
Affiliation(s)
- Li Wang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Yi Sang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China.,Nanchang Key Laboratory of Cancer Pathogenesis and Translational Research, The Third Affiliated Hospital, Nanchang University, Nanchang, China
| | - Jianjun Tang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Ru-Hua Zhang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Donghua Luo
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Mingyuan Chen
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Wu-Guo Deng
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Tiebang Kang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| |
Collapse
|
33
|
Xu X, Wang W, Kratz K, Fang L, Li Z, Kurtz A, Ma N, Lendlein A. Controlling major cellular processes of human mesenchymal stem cells using microwell structures. Adv Healthc Mater 2014; 3:1991-2003. [PMID: 25313500 DOI: 10.1002/adhm.201400415] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 09/02/2014] [Indexed: 01/17/2023]
Abstract
Directing stem cells towards a desired location and function by utilizing the structural cues of biomaterials is a promising approach for inducing effective tissue regeneration. Here, the cellular response of human adipose-derived mesenchymal stem cells (hADSCs) to structural signals from microstructured substrates comprising arrays of square-shaped or round-shaped microwells is explored as a transitional model between 2D and 3D systems. Microwells with a side length/diameter of 50 μm show advantages over 10 μm and 25 μm microwells for accommodating hADSCs within single microwells rather than in the inter-microwell area. The cell morphologies are three-dimensionally modulated by the microwell structure due to differences in focal adhesion and consequent alterations of the cytoskeleton. In contrast to the substrate with 50 μm round-shaped microwells, the substrate with 50 μm square-shaped microwells promotes the proliferation and osteogenic differentiation potential of hADSCs but reduces the cell migration velocity and distance. Such microwell shape-dependent modulatory effects are highly associated with Rho/ROCK signaling. Following ROCK inhibition, the differences in migration, proliferation, and osteogenesis between cells on different substrates are diminished. These results highlight the possibility to control stem cell functions through the use of structured microwells combined with the manipulation of Rho/ROCK signaling.
Collapse
Affiliation(s)
- Xun Xu
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies; Helmholtz-Zentrum Geesthacht; Kantstraße 55 14513 Teltow Germany
- Institute of Chemistry and Biochemistry; Freie Universität Berlin; Takustraße 3 14195 Berlin Germany
| | - Weiwei Wang
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies; Helmholtz-Zentrum Geesthacht; Kantstraße 55 14513 Teltow Germany
| | - Karl Kratz
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies; Helmholtz-Zentrum Geesthacht; Kantstraße 55 14513 Teltow Germany
- Helmholtz Virtual Institute −Multifunctional Materials in Medicine; Berlin and Teltow; Kantstraße 55 14513 Teltow Germany
| | - Liang Fang
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies; Helmholtz-Zentrum Geesthacht; Kantstraße 55 14513 Teltow Germany
| | - Zhengdong Li
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies; Helmholtz-Zentrum Geesthacht; Kantstraße 55 14513 Teltow Germany
- Institute of Chemistry and Biochemistry; Freie Universität Berlin; Takustraße 3 14195 Berlin Germany
| | - Andreas Kurtz
- Berlin-Brandenburg Center for Regenerative Therapies; Charité - University Medicine Berlin; Augustenburger Platz 1 13353 Berlin Germany
- College of Veterinary Medicine and Research Institute for Veterinary Science; Seoul National University; Gwangk-ro 1 Gwanak-gu Seoul 151-747 Republic of Korea
| | - Nan Ma
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies; Helmholtz-Zentrum Geesthacht; Kantstraße 55 14513 Teltow Germany
- Institute of Chemistry and Biochemistry; Freie Universität Berlin; Takustraße 3 14195 Berlin Germany
- Helmholtz Virtual Institute −Multifunctional Materials in Medicine; Berlin and Teltow; Kantstraße 55 14513 Teltow Germany
| | - Andreas Lendlein
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies; Helmholtz-Zentrum Geesthacht; Kantstraße 55 14513 Teltow Germany
- Institute of Chemistry and Biochemistry; Freie Universität Berlin; Takustraße 3 14195 Berlin Germany
- Helmholtz Virtual Institute −Multifunctional Materials in Medicine; Berlin and Teltow; Kantstraße 55 14513 Teltow Germany
| |
Collapse
|
34
|
Phung TL, Du W, Xue Q, Ayyaswamy S, Gerald D, Antonello Z, Nhek S, Perruzzi CA, Acevedo I, Ramanna-Valmiki R, Rodriguez-Waitkus P, Enayati L, Hochman ML, Lev D, Geeganage S, Benjamin LE. Akt1 and akt3 exert opposing roles in the regulation of vascular tumor growth. Cancer Res 2014; 75:40-50. [PMID: 25388284 DOI: 10.1158/0008-5472.can-13-2961] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Vascular tumors are endothelial cell neoplasms whose mechanisms of tumorigenesis are poorly understood. Moreover, current therapies, particularly those for malignant lesions, have little beneficial effect on clinical outcomes. In this study, we show that endothelial activation of the Akt1 kinase is sufficient to drive de novo tumor formation. Mechanistic investigations uncovered opposing functions for different Akt isoforms in this regulation, where Akt1 promotes and Akt3 inhibits vascular tumor growth. Akt3 exerted negative effects on tumor endothelial cell growth and migration by inhibiting activation of the translation regulatory kinase S6-Kinase (S6K) through modulation of Rictor expression. S6K in turn acted through a negative feedback loop to restrain Akt3 expression. Conversely, S6K signaling was increased in vascular tumor cells where Akt3 was silenced, and the growth of these tumor cells was inhibited by a novel S6K inhibitor. Overall, our findings offer a preclinical proof of concept for the therapeutic utility of treating vascular tumors, such as angiosarcomas, with S6K inhibitors.
Collapse
Affiliation(s)
- Thuy L Phung
- Department of Pathology, Texas Children's Hospital and Baylor College of Medicine, Houston, Texas.
| | - Wa Du
- Department of Pathology, Texas Children's Hospital and Baylor College of Medicine, Houston, Texas
| | - Qi Xue
- Eli Lilly and Company, Indianapolis, Indiana
| | - Sriram Ayyaswamy
- Department of Pathology, Texas Children's Hospital and Baylor College of Medicine, Houston, Texas
| | | | - Zeus Antonello
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Sokha Nhek
- Eli Lilly and Company, Indianapolis, Indiana
| | | | - Isabel Acevedo
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Rajesh Ramanna-Valmiki
- Department of Pathology, Texas Children's Hospital and Baylor College of Medicine, Houston, Texas
| | - Paul Rodriguez-Waitkus
- Department of Pathology, Texas Children's Hospital and Baylor College of Medicine, Houston, Texas
| | - Ladan Enayati
- Department of Pathology, Texas Children's Hospital and Baylor College of Medicine, Houston, Texas
| | - Marcelo L Hochman
- Hemangioma International Treatment Center, Charleston, South Carolina
| | - Dina Lev
- Department of Cancer Biology, MD Anderson Cancer Center, Houston, Texas
| | | | - Laura E Benjamin
- Eli Lilly and Company, Indianapolis, Indiana. Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts.
| |
Collapse
|
35
|
Kim S, Yao J, Suyama K, Qian X, Qian BZ, Bandyopadhyay S, Loudig O, De Leon-Rodriguez C, Zhou ZN, Segall J, Macian F, Norton L, Hazan RB. Slug promotes survival during metastasis through suppression of Puma-mediated apoptosis. Cancer Res 2014; 74:3695-706. [PMID: 24830722 DOI: 10.1158/0008-5472.can-13-2591] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Tumor cells must overcome apoptosis to survive throughout metastatic dissemination and distal organ colonization. Here, we show in the Polyoma Middle T mammary tumor model that N-cadherin (Cdh2) expression causes Slug (Snai2) upregulation, which in turn promotes carcinoma cell survival. Slug was dramatically upregulated in metastases relative to primary tumors. Consistent with a role in metastasis, Slug knockdown in carcinoma cells suppressed lung colonization by decreasing cell survival at metastatic sites, but had no effect on tumor cell invasion or extravasation. In support of this idea, Slug inhibition by shRNA sensitized tumor cells to apoptosis by DNA damage, resulting in caspase-3 and PARP cleavage. The prosurvival effect of Slug was found to be caused by direct repression of the proapoptotic gene, Puma (Bbc3), by Slug. Consistent with a pivotal role for a Slug-Puma axis in metastasis, inhibition of Puma by RNA interference in Slug-knockdown cells rescued lung colonization, whereas Puma overexpression in control tumor cells suppressed lung metastasis. The survival function of the Slug-Puma axis was confirmed in human breast cancer cells, where Slug knockdown increased Puma expression and inhibited lung colonization. This study demonstrates a pivotal role for Slug in carcinoma cell survival, implying that disruption of the Slug-Puma axis may impinge on the survival of metastatic cells.
Collapse
Affiliation(s)
- Seaho Kim
- Authors' Affiliations: Departments of Pathology
| | - Jiahong Yao
- Authors' Affiliations: Departments of Pathology
| | | | - Xia Qian
- Authors' Affiliations: Departments of Pathology
| | | | | | | | | | - Zhen Ni Zhou
- Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx; and
| | - Jeffrey Segall
- Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx; and
| | | | - Larry Norton
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York
| | | |
Collapse
|
36
|
Li H, Yang L, Fu H, Yan J, Wang Y, Guo H, Hao X, Xu X, Jin T, Zhang N. Association between Gαi2 and ELMO1/Dock180 connects chemokine signalling with Rac activation and metastasis. Nat Commun 2013; 4:1706. [PMID: 23591873 PMCID: PMC3644068 DOI: 10.1038/ncomms2680] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Accepted: 02/28/2013] [Indexed: 02/03/2023] Open
Abstract
The chemokine CXCL12 and its G-protein-coupled receptor CXCR4 control the migration, invasiveness and metastasis of breast cancer cells. Binding of CXCL12 to CXCR4 triggers activation of heterotrimeric Gi proteins that regulate actin polymerization and migration. However, the pathways linking chemokine G-protein-coupled receptor/Gi signalling to actin polymerization and cancer cell migration are not known. Here we show that CXCL12 stimulation promotes interaction between Gαi2 and ELMO1. Gi signalling and ELMO1 are both required for CXCL12-mediated actin polymerization, migration and invasion of breast cancer cells. CXCL12 triggers a Gαi2-dependent membrane translocation of ELMO1, which associates with Dock180 to activate small G-proteins Rac1 and Rac2. In vivo, ELMO1 expression is associated with lymph node and distant metastasis, and knocking down ELMO1 impairs metastasis to the lung. Our findings indicate that a chemokine-controlled pathway, consisting of Gαi2, ELMO1/Dock180, Rac1 and Rac2, regulates the actin cytoskeleton during breast cancer metastasis.
Collapse
Affiliation(s)
- Hongyan Li
- Tianjin Medical University Cancer Institute and Hospital and Research Center of Basic Medical Sciences, He Xi District, Tianjin 300060, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
N-cadherin/FGFR promotes metastasis through epithelial-to-mesenchymal transition and stem/progenitor cell-like properties. Oncogene 2013; 33:3411-21. [PMID: 23975425 DOI: 10.1038/onc.2013.310] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Revised: 05/05/2013] [Accepted: 05/20/2013] [Indexed: 12/15/2022]
Abstract
N-cadherin and HER2/neu were found to be co-expressed in invasive breast carcinomas. To test the contribution of N-cadherin and HER2 in mammary tumor metastasis, we targeted N-cadherin expression in the mammary epithelium of the MMTV-Neu mouse. In the context of ErbB2/Neu, N-cadherin stimulated carcinoma cell invasion, proliferation and metastasis. N-cadherin caused fibroblast growth factor receptor (FGFR) upmodulation, resulting in epithelial-to-mesenchymal transition (EMT) and stem/progenitor like properties, involving Snail and Slug upregulation, mammosphere formation and aldehyde dehydrogenase activity. N-cadherin potentiation of the FGFR stimulated extracellular signal regulated kinase (ERK) and protein kinase B (AKT) phosphorylation resulting in differential effects on metastasis. Although ERK inhibition suppressed cyclin D1 expression, cell proliferation and stem/progenitor cell properties, it did not affect invasion or EMT. Conversely, AKT inhibition suppressed invasion through Akt 2 attenuation, and EMT through Snail inhibition, but had no effect on cyclin D1 expression, cell proliferation or mammosphere formation. These findings suggest N-cadherin/FGFR has a pivotal role in promoting metastasis through differential regulation of ERK and AKT, and underscore the potential for targeting the FGFR in advanced ErbB2-amplified breast tumors.
Collapse
|
38
|
Geletu M, Arulanandam R, Chevalier S, Saez B, Larue L, Feracci H, Raptis L. Classical cadherins control survival through the gp130/Stat3 axis. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1833:1947-59. [DOI: 10.1016/j.bbamcr.2013.03.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2013] [Revised: 03/14/2013] [Accepted: 03/18/2013] [Indexed: 01/02/2023]
|
39
|
Agarwal E, Brattain MG, Chowdhury S. Cell survival and metastasis regulation by Akt signaling in colorectal cancer. Cell Signal 2013; 25:1711-9. [PMID: 23603750 PMCID: PMC3686084 DOI: 10.1016/j.cellsig.2013.03.025] [Citation(s) in RCA: 134] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2012] [Revised: 02/23/2013] [Accepted: 03/28/2013] [Indexed: 12/12/2022]
Abstract
Dissemination of cancer cells to distant organ sites is the leading cause of death due to treatment failure in different types of cancer. Mehlen and Puisieux have reviewed the importance of the development of inappropriate cell survival signaling for various steps in the metastatic process and have noted the particular importance of aberrant cell survival to successful colonization at the metastatic site. Therefore, the understanding of mechanisms that govern cell survival fate of these metastatic cells could lead to the understanding of a new paradigm for the control of metastatic potential and could provide the basis for developing novel strategies for the treatment of metastases. Numerous studies have documented the widespread role of Akt in cell survival and metastasis in colorectal cancer, as well as many other types of cancer. Akt acts as a key signaling node that bridges the link between oncogenic receptors to many essential pro-survival cellular functions, and is perhaps the most commonly activated signaling pathway in human cancer. In recent years, Akt2 and Akt3 have emerged as significant contributors to malignancy alongside the well-characterized Akt1 isoform, with distinct non-overlapping functions. This review is aimed at gaining a better understanding of the Akt-driven cell survival mechanisms that contribute to cancer progression and metastasis and the pharmacological inhibitors in clinical trials designed to counter the Akt-driven cell survival responses in cancer.
Collapse
Affiliation(s)
- Ekta Agarwal
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
- Eppley Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Michael G. Brattain
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
- Eppley Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sanjib Chowdhury
- Eppley Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
40
|
Cui Y, Yamada S. N-cadherin dependent collective cell invasion of prostate cancer cells is regulated by the N-terminus of α-catenin. PLoS One 2013; 8:e55069. [PMID: 23359820 PMCID: PMC3554680 DOI: 10.1371/journal.pone.0055069] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Accepted: 12/24/2012] [Indexed: 02/03/2023] Open
Abstract
Cancer cell invasion is the critical first step of metastasis, yet, little is known about how cancer cells invade and initiate metastasis in a complex extracellular matrix. Using a cell line from bone metastasis of prostate cancer (PC3), we analyzed how prostate cancer cells migrate in a physiologically relevant 3D Matrigel. We found that PC3 cells migrated more efficiently as multi-cellular clusters than isolated single cells, suggesting that the presence of cell-cell adhesion improves 3D cell migration. Perturbation of N-cadherin function by transfection of either the N-cadherin cytoplasmic domain or shRNA specific to N-cadherin abolished collective cell migration. Interestingly, PC3 cells do not express α-catenin, an actin binding protein in the cadherin complex. When the full-length α-catenin was re-introduced, the phenotype of PC3 cells reverted back to a more epithelial phenotype with a decreased cell migration rate in 3D Matrigel. Interestingly, we found that the N-terminal half of α-catenin was sufficient to suppress invasive phenotype. Taken together, these data suggest that the formation of N-cadherin junctions promotes 3D cell migration of prostate cancer cells, and this is partly due to an aberrant regulation of the N-cadherin complex in the absence of α-catenin.
Collapse
Affiliation(s)
- Yuanyuan Cui
- Department of Biomedical Engineering, University of California Davis, Davis, California, United States of America
| | - Soichiro Yamada
- Department of Biomedical Engineering, University of California Davis, Davis, California, United States of America
- * E-mail:
| |
Collapse
|
41
|
Radice GL. N-cadherin-mediated adhesion and signaling from development to disease: lessons from mice. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2013; 116:263-89. [PMID: 23481199 PMCID: PMC6047516 DOI: 10.1016/b978-0-12-394311-8.00012-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Of the 20 classical cadherin subtypes identified in mammals, the functions of the two initially identified family members E- (epithelial) and N- (neural) cadherin have been most extensively studied. E- and N-Cadherin have mostly mutually exclusive expression patterns, with E-cadherin expressed primarily in epithelial cells, whereas N-cadherin is found in a variety of cells, including neural, muscle, and mesenchymal cells. N-Cadherin function, in particular, appears to be cell context-dependent, as it can mediate strong cell-cell adhesion in the heart but induces changes in cell behavior in favor of a migratory phenotype in the context of epithelial-mesenchymal transition (EMT). The ability of tumor cells to alter their cadherin expression profile, for example, E- to N-cadherin, is critical for malignant progression. Recent advances in mouse molecular genetics, and specifically tissue-specific knockout and knockin alleles of N-cadherin, have provided some unexpected results. This chapter highlights some of the genetic studies that explored the complex role of N-cadherin in embryonic development and disease.
Collapse
Affiliation(s)
- Glenn L Radice
- Department of Medicine, Center for Translational Medicine, Jefferson Medical College, Philadelphia, Pennsylvania, USA
| |
Collapse
|
42
|
Shih W, Yamada S. N-cadherin as a key regulator of collective cell migration in a 3D environment. Cell Adh Migr 2012; 6:513-7. [PMID: 23076138 PMCID: PMC3547896 DOI: 10.4161/cam.21766] [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] [Indexed: 02/03/2023] Open
Abstract
Cell migration is a critical step of normal developmental processes and disease progression. Often, migrating cells interact and maintain contact with neighboring cells. However, the precise roles of cell-cell adhesion in cell migration have thus far been poorly defined. Often in aggressive cancers, N-cadherin is prominently upregulated, yet, these highly motile cells have limited cell-cell adhesion when plated on a stiff 2D substrate. But, the same cells in a 3D matrix migrate as a multicellular cluster. This new observation suggests that N-cadherin-mediated cell-cell adhesion supports cell interactions between migrating cells in a more physiologically relevant 3D matrix, but not on a 2D substrate. While N-cadherin is an integral part of neural synapses, the ectopic expression of N-cadherin in transformed epithelial cells plays an equally important part in initiating pro-migratory signaling, and providing strong yet flexible cell cohesion essential for persistent cell migration in a 3D matrix. The 3D cell migration analysis for studying cell-to-cell interactions exposes the roles of N-cadherin in multicellular migration, and reveals novel insights into cell migration-dependent normal and pathological processes.
Collapse
Affiliation(s)
- Wenting Shih
- Department of Biomedical Engineering, University of California, Davis, CA USA
| | | |
Collapse
|
43
|
Kashyap MP, Singh AK, Kumar V, Yadav DK, Khan F, Jahan S, Khanna VK, Yadav S, Pant AB. Pkb/Akt1 mediates Wnt/GSK3β/β-catenin signaling-induced apoptosis in human cord blood stem cells exposed to organophosphate pesticide monocrotophos. Stem Cells Dev 2012; 22:224-38. [PMID: 22897592 DOI: 10.1089/scd.2012.0220] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Inhibition mechanisms of protein kinase B (Pkb)/Akt and its consequences on related cell signaling were investigated in human umbilical cord blood stem cells (hUCBSCs) exposed to monocrotophos (MCP, an organophosphate pesticide). In silico data reveal that MCP interacts with kinase and c-terminal regulatory domains of Akt1, resulting into a total docking score of 5.2748 and also forms H-bond between its N-H and Thr-291 residue of Akt1, in addition to possessing several hydrophobic interactions. The main cause of Akt inhibition is considered to be the strong hydrogen bond between N-H and Thr-291, and hydrophobic interactions at Glu-234, and Asp-292 in the vicinity, which is usually occupied by the ribose of ATP, and interaction with residue Phe-161, thus leading to a significant conformational change in that particular portion of the protein. In silico data on Akt inhibition were confirmed by examining the downregulation of phosphorylated (Thr308/Ser493) Akt1 in MCP-exposed hUCBSCs. MCP-mediated altered levels of pAkt downstream targets viz., downregulated pGSK3β (Ser9), unchanged GSK3αβ, and upregulated levels of Bad, P(53), and caspase-9 further confirm the inhibition of pAkt. The cellular fate of such pAkt inhibition was confirmed by increased terminal deoxynucleotide transferase dUTP nick-end labeling positive cells, reduced mitochondrial membrane potential, and the activation of various MAPKs, proapoptotic markers-Bax, and caspases-9/3. Our data demonstrate that Akt1 plays a key role in MCP-induced apoptosis in hUCBSCs. We also identified that such cellular responses of human cord blood stem cells against MCP were due to strong binding and inhibition of kinase and AGC-Kinase-C terminal regulatory domains of Akt1.
Collapse
Affiliation(s)
- Mahendra P Kashyap
- In Vitro Toxicology Laboratory, Indian Institute of Toxicology Research, Council of Scientific & Industrial Research, Lucknow, India
| | | | | | | | | | | | | | | | | |
Collapse
|