151
|
SOX9 Regulates Cancer Stem-Like Properties and Metastatic Potential of Single-Walled Carbon Nanotube-Exposed Cells. Sci Rep 2017; 7:11653. [PMID: 28912540 PMCID: PMC5599589 DOI: 10.1038/s41598-017-12037-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 09/01/2017] [Indexed: 12/16/2022] Open
Abstract
Engineered nanomaterials hold great promise for the future development of innovative products but their adverse health effects are a major concern. Recent studies have indicated that certain nanomaterials, including carbon nanotubes (CNTs), may be carcinogenic. However, the underlying mechanisms behind their potential malignant properties remain unclear. In this study, we linked SOX9, a stem cell associated transcription factor, to the neoplastic-like properties of human lung epithelial cells chronically exposed to a low-dose of single-walled carbon nanotubes (SWCNTs). We found that SOX9 is upregulated in SWCNT-exposed cells, which is consistent with their abilities to induce tumor formation and metastasis in vivo. We therefore hypothesized that SOX9 overexpression may be responsible for the neoplastic-like phenotype observed in our model. Indeed, SOX9 knockdown inhibited anchorage-independent cell growth in vitro and lung colonization in vivo in a mouse xenograft model. SOX9 depletion also suppressed the formation of cancer stem-like cells (CSCs), as determined by tumor sphere formation and aldehyde dehydrogenase (ALDH) activity (Aldefluor) assays. Furthermore, SOX9 knockdown suppressed tumor metastasis and the expression of the stem cell marker ALDH1A1. Taken together, our findings provide a mechanistic insight into SWCNT-induced carcinogenesis and the role of SOX9 in CSC regulation and metastasis.
Collapse
|
152
|
Seguin L, Camargo MF, Wettersten HI, Kato S, Desgrosellier JS, von Schalscha T, Elliott KC, Cosset E, Lesperance J, Weis SM, Cheresh DA. Galectin-3, a Druggable Vulnerability for KRAS-Addicted Cancers. Cancer Discov 2017; 7:1464-1479. [PMID: 28893801 DOI: 10.1158/2159-8290.cd-17-0539] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 08/17/2017] [Accepted: 09/05/2017] [Indexed: 01/12/2023]
Abstract
Identifying the molecular basis for cancer cell dependence on oncogenes such as KRAS can provide new opportunities to target these addictions. Here, we identify a novel role for the carbohydrate-binding protein galectin-3 as a lynchpin for KRAS dependence. By directly binding to the cell surface receptor integrin αvβ3, galectin-3 gives rise to KRAS addiction by enabling multiple functions of KRAS in anchorage-independent cells, including formation of macropinosomes that facilitate nutrient uptake and ability to maintain redox balance. Disrupting αvβ3/galectin-3 binding with a clinically active drug prevents their association with mutant KRAS, thereby suppressing macropinocytosis while increasing reactive oxygen species to eradicate αvβ3-expressing KRAS-mutant lung and pancreatic cancer patient-derived xenografts and spontaneous tumors in mice. Our work reveals galectin-3 as a druggable target for KRAS-addicted lung and pancreas cancers, and indicates integrin αvβ3 as a biomarker to identify susceptible tumors.Significance: There is a significant unmet need for therapies targeting KRAS-mutant cancers. Here, we identify integrin αvβ3 as a biomarker to identify mutant KRAS-addicted tumors that are highly sensitive to inhibition of galectin-3, a glycoprotein that binds to integrin αvβ3 to promote KRAS-mediated activation of AKT. Cancer Discov; 7(12); 1464-79. ©2017 AACR.This article is highlighted in the In This Issue feature, p. 1355.
Collapse
Affiliation(s)
- Laetitia Seguin
- Department of Pathology, Moores UCSD Cancer Center, and Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, California.
| | - Maria F Camargo
- Department of Pathology, Moores UCSD Cancer Center, and Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, California
| | - Hiromi I Wettersten
- Department of Pathology, Moores UCSD Cancer Center, and Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, California
| | - Shumei Kato
- School of Medicine, Division of Hematology/Oncology, University of California, San Diego, La Jolla, California
| | - Jay S Desgrosellier
- Department of Pathology, Moores UCSD Cancer Center, and Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, California
| | - Tami von Schalscha
- Department of Pathology, Moores UCSD Cancer Center, and Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, California
| | - Kathryn C Elliott
- Department of Pathology, Moores UCSD Cancer Center, and Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, California
| | - Erika Cosset
- Department of Pathology, Moores UCSD Cancer Center, and Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, California
| | - Jacqueline Lesperance
- Department of Pathology, Moores UCSD Cancer Center, and Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, California
| | - Sara M Weis
- Department of Pathology, Moores UCSD Cancer Center, and Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, California
| | - David A Cheresh
- Department of Pathology, Moores UCSD Cancer Center, and Sanford Consortium for Regenerative Medicine, University of California, San Diego, La Jolla, California.
| |
Collapse
|
153
|
Chen QY, Costa M. A comprehensive review of metal-induced cellular transformation studies. Toxicol Appl Pharmacol 2017; 331:33-40. [DOI: 10.1016/j.taap.2017.05.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 04/30/2017] [Accepted: 05/05/2017] [Indexed: 01/07/2023]
|
154
|
Anderson M, Marayati R, Moffitt R, Yeh JJ. Hexokinase 2 promotes tumor growth and metastasis by regulating lactate production in pancreatic cancer. Oncotarget 2017; 8:56081-56094. [PMID: 28915575 PMCID: PMC5593546 DOI: 10.18632/oncotarget.9760] [Citation(s) in RCA: 140] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 05/02/2016] [Indexed: 12/12/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a KRAS-driven cancer with a high incidence of metastasis and an overall poor prognosis. Previous work in a genetically engineered mouse model of PDAC showed glucose metabolism to be important for maintaining tumor growth. Multiple glycolytic enzymes, including hexokinase 2 (HK2), were upregulated in primary PDAC patient tumors, supporting a role for glycolysis in promoting human disease. HK2 was most highly expressed in PDAC metastases, suggesting a link between HK2 and aggressive tumor biology. In support of this we found HK2 expression to be associated with shorter overall survival in PDAC patients undergoing curative surgery. Transient and stable knockdown of HK2 in primary PDAC cell lines decreased lactate production, anchorage independent growth (AIG) and invasion through a reconstituted matrix. Conversely, stable overexpression of HK2 increased lactate production, cell proliferation, AIG and invasion. Pharmacologic inhibition of lactate production reduced the HK2-driven increase in invasion while addition of extracellular lactate enhanced invasion, together providing a link between glycolytic activity and metastatic potential. Stable knockdown of HK2 decreased primary tumor growth in cell line xenografts and decreased incidence of lung metastasis after tail vein injection. Gene expression analysis of tumors with decreased HK2 expression showed alterations in VEGF-A signaling, a pathway important for angiogenesis and metastasis, consistent with a requirement of HK2 in promoting metastasis. Overall our data provides strong evidence for the role of HK2 in promoting PDAC disease progression, suggesting that direct inhibition of HK2 may be a promising approach in the clinic.
Collapse
Affiliation(s)
- Marybeth Anderson
- Curriculum in Genetics & Molecular Biology, The University of North Carolina, Chapel Hill, NC
- Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, NC
| | - Raoud Marayati
- Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, NC
| | - Richard Moffitt
- Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, NC
| | - Jen Jen Yeh
- Curriculum in Genetics & Molecular Biology, The University of North Carolina, Chapel Hill, NC
- Lineberger Comprehensive Cancer Center, The University of North Carolina, Chapel Hill, NC
- Departments of Surgery and Pharmacology, The University of North Carolina, Chapel Hill, NC
| |
Collapse
|
155
|
Manandhar S, Kim CG, Lee SH, Kang SH, Basnet N, Lee YM. Exostosin 1 regulates cancer cell stemness in doxorubicin-resistant breast cancer cells. Oncotarget 2017; 8:70521-70537. [PMID: 29050299 PMCID: PMC5642574 DOI: 10.18632/oncotarget.19737] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 06/26/2017] [Indexed: 12/18/2022] Open
Abstract
Cancer stem cells (CSCs) are associated with cancer recurrence following radio/chemotherapy owing to their high resistance to therapeutic intervention. In this study, we investigated the role of exostoxin 1 (EXT1), an endoplasmic reticulum (ER)-residing type II transmembrane glycoprotein, in cancer cell stemness. DNA microarray analysis revealed that doxorubicin-resistant MCF7/ADR cells have high levels of EXT1 expression compared to its parental cell line, MCF7. These cells showed significantly higher populations of CSCs and larger populations of aldehyde dehydrogenase (ALDH+) and CD44+/CD24-cells, as compared to MCF7 cells. siRNA-mediated knockdown of EXT1 in MCF7/ADR cells significantly reduced cancer stem cell markers, populations of ALDH+and CD44+/CD24- cells, mRNA and protein expression for CD44, and mammosphere number. Furthermore, epithelial mesenchymal transition (EMT) markers and migratory behavior were also repressed with reduced EXT1. In an in vitro soft agar colony formation assay, EXT1 knockdown by short hairpin RNA (shRNA) reduced the colony formation ability of these cells. Based on these results, we suggest that EXT1 could be a promising novel target to overcome cancer cell stemness in anthracycline-based therapeutic resistance.
Collapse
Affiliation(s)
- Sarala Manandhar
- BK21 Plus Multi-Omics Based Creative Drug Research Training Team (22A20154413076), National Basic Research Laboratory of Vascular Homeostasis Regulation, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu 41566, South Korea
| | - Chang-Gu Kim
- BK21 Plus Multi-Omics Based Creative Drug Research Training Team (22A20154413076), National Basic Research Laboratory of Vascular Homeostasis Regulation, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu 41566, South Korea
| | - Sun-Hee Lee
- BK21 Plus Multi-Omics Based Creative Drug Research Training Team (22A20154413076), National Basic Research Laboratory of Vascular Homeostasis Regulation, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu 41566, South Korea
| | - Soo Hyun Kang
- BK21 Plus Multi-Omics Based Creative Drug Research Training Team (22A20154413076), National Basic Research Laboratory of Vascular Homeostasis Regulation, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu 41566, South Korea
| | - Nikita Basnet
- BK21 Plus Multi-Omics Based Creative Drug Research Training Team (22A20154413076), National Basic Research Laboratory of Vascular Homeostasis Regulation, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu 41566, South Korea
| | - You Mie Lee
- BK21 Plus Multi-Omics Based Creative Drug Research Training Team (22A20154413076), National Basic Research Laboratory of Vascular Homeostasis Regulation, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu 41566, South Korea
| |
Collapse
|
156
|
Espindola-Netto JM, Chini CCS, Tarragó M, Wang E, Dutta S, Pal K, Mukhopadhyay D, Sola-Penna M, Chini EN. Preclinical efficacy of the novel competitive NAMPT inhibitor STF-118804 in pancreatic cancer. Oncotarget 2017; 8:85054-85067. [PMID: 29156703 PMCID: PMC5689593 DOI: 10.18632/oncotarget.18841] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 06/05/2017] [Indexed: 11/25/2022] Open
Abstract
NAD salvage is one of the pathways used to generate NAD in mammals. Nicotinamide phosphoribosyltransferase (NAMPT), the rate-limiting enzyme in this pathway, uses nicotinamide (NAM) to generate nicotinamide mononucleotide (NMN). NMN is one of the main precursors of NAD synthesis in cells. Our previous study showed the importance of NAMPT in maintaining NAD levels in pancreatic ductal adenocarcinoma cells (PDAC), and that the NAMPT inhibitor FK866 decreased pancreatic cancer growth. We now tested the effect of STF-118804, a new highly specific NAMPT inhibitor, in models of pancreatic ductal adenocarcinoma. STF-118804 reduced viability and growth of different PDAC lines, as well as the formation of colonies in soft agar. In addition, STF-118804 decreased glucose uptake, lactate excretion, and ATP levels, resulting in metabolic collapse. STF-118804 treatment activated AMPK and inhibited of mTOR pathways in these cells. This effect was significantly potentiated by pharmacological AMPK activation and mTOR inhibition. Exogenous NMN blocked both the activation of the AMPK pathway and the decrease in cell viability. Panc-1 cells expressing GFP-luciferase were orthotopically implanted on mice pancreas to test the in vivo effectiveness of STF-118804. Both STF-118804 and FK866 reduced tumor size after 21 days of treatment. Combinations of STF-118804 with chemotherapeutic agents such as paclitaxel, gemcitabine, and etoposide showed an additive effect in decreasing cell viability and growth. In conclusion, our preclinical study shows that the NAMPT inhibitor STF-118804 reduced the growth of PDAC in vitro and in vivo and had an additive effect in combination with main current chemotherapeutic drugs.
Collapse
Affiliation(s)
- Jair Machado Espindola-Netto
- Laboratory of Signal Transduction and Molecular Nutrition, Department of Anesthesiology, Mayo Clinic College of Medicine, Rochester, MN, U.S.A.,Laboratório de Enzimologia e Controle do Metabolismo (LabECoM), Departamento de Biotecnologia Farmacêutica (BioTecFar), Faculdade de Farmacia, Centro de Ciencias da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Claudia C S Chini
- Laboratory of Signal Transduction and Molecular Nutrition, Department of Anesthesiology, Mayo Clinic College of Medicine, Rochester, MN, U.S.A
| | - Mariana Tarragó
- Laboratory of Signal Transduction and Molecular Nutrition, Department of Anesthesiology, Mayo Clinic College of Medicine, Rochester, MN, U.S.A
| | - Enfeng Wang
- Department of Biochemistry and Molecular Biology, College of Medicine, Mayo Clinic, Rochester, MN, U.S.A
| | - Shamit Dutta
- Department of Biochemistry and Molecular Biology, College of Medicine, Mayo Clinic, Rochester, MN, U.S.A
| | - Krishnendu Pal
- Department of Biochemistry and Molecular Biology, College of Medicine, Mayo Clinic, Rochester, MN, U.S.A
| | - Debabrata Mukhopadhyay
- Department of Biochemistry and Molecular Biology, College of Medicine, Mayo Clinic, Rochester, MN, U.S.A
| | - Mauro Sola-Penna
- Laboratório de Enzimologia e Controle do Metabolismo (LabECoM), Departamento de Biotecnologia Farmacêutica (BioTecFar), Faculdade de Farmacia, Centro de Ciencias da Saúde, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Eduardo N Chini
- Laboratory of Signal Transduction and Molecular Nutrition, Department of Anesthesiology, Mayo Clinic College of Medicine, Rochester, MN, U.S.A
| |
Collapse
|
157
|
Shirnekhi HK, Kelley EP, DeLuca JG, Herman JA. Spindle assembly checkpoint signaling and sister chromatid cohesion are disrupted by HPV E6-mediated transformation. Mol Biol Cell 2017; 28:2035-2041. [PMID: 28539402 PMCID: PMC5509418 DOI: 10.1091/mbc.e16-12-0853] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 05/18/2017] [Accepted: 05/18/2017] [Indexed: 12/20/2022] Open
Abstract
Aneuploidy, a condition that results from unequal partitioning of chromosomes during mitosis, is a hallmark of many cancers, including those caused by human papillomaviruses (HPVs). E6 and E7 are the primary transforming proteins in HPV that drive tumor progression. In this study, we stably expressed E6 and E7 in noncancerous RPE1 cells and analyzed the specific mitotic defects that contribute to aneuploidy in each cell line. We find that E6 expression results in multiple chromosomes associated with one or both spindle poles, causing a significant mitotic delay. In most cells, the misaligned chromosomes eventually migrated to the spindle equator, leading to mitotic exit. In some cells, however, mitotic exit occurred in the presence of pole-associated chromosomes. We determined that this premature mitotic exit is due to defects in spindle assembly checkpoint (SAC) signaling, such that cells are unable to maintain a prolonged mitotic arrest in the presence of unaligned chromosomes. This SAC defect is caused in part by a loss of kinetochore-associated Mad2 in E6-expressing cells. Our results demonstrate that E6-expressing cells exhibit previously unappreciated mitotic defects that likely contribute to HPV-mediated cancer progression.
Collapse
Affiliation(s)
- Hazheen K Shirnekhi
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523
| | - Erin P Kelley
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523
| | - Jennifer G DeLuca
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523 )
| | - Jacob A Herman
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523 )
| |
Collapse
|
158
|
A tumor suppressor role for C/EBPα in solid tumors: more than fat and blood. Oncogene 2017; 36:5221-5230. [PMID: 28504718 DOI: 10.1038/onc.2017.151] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 03/29/2017] [Accepted: 04/10/2017] [Indexed: 12/12/2022]
Abstract
The transcription factor CCAAT/enhancer-binding protein alpha (C/EBPα) plays a critical role during embryogenesis and is thereafter required for homeostatic glucose metabolism, adipogenesis and myeloid development. Its ability to regulate the expression of lineage-specific genes and induce growth arrest contributes to the terminal differentiation of several cell types, including hepatocytes, adipocytes and granulocytes. CEBPA loss of-function mutations contribute to the development of ~10% of acute myeloid leukemia (AML), stablishing a tumor suppressor role for C/EBPα. Deregulation of C/EBPα expression has also been reported in a variety of additional human neoplasias, including liver, breast and lung cancer. However, functional CEBPA mutations have not been found in solid tumors, suggesting that abrogation of C/EBPα function in non-hematopoietic tissues is regulated by alternative mechanisms. Here we review the function of C/EBPα in solid tumors and focus on the molecular mechanisms underlying its tumor suppressive role.
Collapse
|
159
|
Bizjak M, Malavašič P, Dolinar K, Pohar J, Pirkmajer S, Pavlin M. Combined treatment with Metformin and 2-deoxy glucose induces detachment of viable MDA-MB-231 breast cancer cells in vitro. Sci Rep 2017; 7:1761. [PMID: 28496098 PMCID: PMC5431940 DOI: 10.1038/s41598-017-01801-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 04/04/2017] [Indexed: 12/13/2022] Open
Abstract
Triple naegative breast cancer has an increased rate of distant metastasis and consequently poor prognosis. To metastasize, breast cancer cells must detach from the main tumour mass and resist anoikis, a programmed cell death induced by lack of cell-extracellular matrix communication. Although cancer cells must detach to metastasize in vivo, the viability of floating cancer cells in vitro is rarely investigated. Here we show that co-treatment of anoikis-resistant MDA-MB-231 cells with metformin and 2-deoxy-D-glucose (2-DG) increased the percentage of floating cells, of which about 95% were viable. Floating cells resumed their proliferation once they were reseeded in the pharmacological compound-free medium. Similar effects on detachment were observed on anoikis-prone MCF-7 cells. Co-treatment of MDA-MB-231 cells with metformin and 2-DG induced a strong activation of AMP-activated protein kinase (AMPK), which was reduced by AMPK inhibitor compound C that prevented detachment of MDA-MB-231 cells. However, direct AMPK activators A-769662 and AICAR did not have any major effect on the percentage of floating MDA-MB-231 cells, indicating that AMPK activation is necessary but not sufficient for triggering detachment of cancer cells. Our results demonstrate that separate analysis of floating and attached cancer cells might be important for evaluation of anti-cancer agents.
Collapse
Affiliation(s)
- Maruša Bizjak
- Group for nano and biotechnological applications, Faculty of Electrical Engineering, University of Ljubljana, Ljubljana, Slovenia
| | - Petra Malavašič
- Group for nano and biotechnological applications, Faculty of Electrical Engineering, University of Ljubljana, Ljubljana, Slovenia
| | - Klemen Dolinar
- Group for nano and biotechnological applications, Faculty of Electrical Engineering, University of Ljubljana, Ljubljana, Slovenia.,Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Jelka Pohar
- Department of Synthetic Biology and Immunology, National institute of Chemistry, Ljubljana, Slovenia.,Centre of Excellence EN-FIST, Ljubljana, Slovenia
| | - Sergej Pirkmajer
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Mojca Pavlin
- Group for nano and biotechnological applications, Faculty of Electrical Engineering, University of Ljubljana, Ljubljana, Slovenia. .,Institute of Biophysics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia.
| |
Collapse
|
160
|
Lakshminarasimhan R, Andreu-Vieyra C, Lawrenson K, Duymich CE, Gayther SA, Liang G, Jones PA. Down-regulation of ARID1A is sufficient to initiate neoplastic transformation along with epigenetic reprogramming in non-tumorigenic endometriotic cells. Cancer Lett 2017; 401:11-19. [PMID: 28483516 DOI: 10.1016/j.canlet.2017.04.040] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 04/25/2017] [Accepted: 04/26/2017] [Indexed: 12/28/2022]
Abstract
The chromatin remodeler AT-Rich Interactive Domain 1A (ARID1A) is frequently mutated in ovarian clear cell carcinoma (OCCC) and endometriosis precursor lesions. Here, we show that knocking down ARID1A in an immortalized endometriosis cell line is sufficient to induce phenotypic changes indicative of neoplastic transformation as evidenced by higher efficiency of anchorage-independent growth, increased propensity to adhere to collagen, and greater capacity to invade basement membrane extract in vitro. ARID1A knockdown is associated with expression dysregulation of 99 target genes, and many of these expression changes are also observed in primary OCCC tissues. Further, pathway analysis indicates these genes fall within networks highly relevant to tumorigenesis including integrin and paxillin pathways. We demonstrate that the down-regulation of ARID1A does not markedly alter global chromatin accessibility or DNA methylation but unexpectedly, we find strong increases in the active H3K27ac mark in promoter regions and decreases of H3K27ac at potential enhancers. Taken together, these data provide evidence that ARID1A mutation can be an early stage event in the oncogenic transformation of endometriosis cells giving rise to OCCC.
Collapse
Affiliation(s)
- Ranjani Lakshminarasimhan
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Claudia Andreu-Vieyra
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Department of Urology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Kate Lawrenson
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, USA; Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Suite 290W, Los Angeles, CA, USA
| | - Christopher E Duymich
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Simon A Gayther
- Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Suite 290W, Los Angeles, CA, USA; Center for Bioinformatics and Functional Genomics, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Gangning Liang
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Department of Urology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| | - Peter A Jones
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Department of Urology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Department of Biochemistry & Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Van Andel Research Institute, Grand Rapids, MI 49503, USA.
| |
Collapse
|
161
|
Deficiency of CCN5/WISP-2-Driven Program in breast cancer Promotes Cancer Epithelial cells to mesenchymal stem cells and Breast Cancer growth. Sci Rep 2017; 7:1220. [PMID: 28450698 PMCID: PMC5430628 DOI: 10.1038/s41598-017-00916-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 03/16/2017] [Indexed: 12/31/2022] Open
Abstract
Breast cancer progression and relapse is conceivably due to tumor initiating cells (TICs)/cancer stem cells. EMT (epithelial-mesenchymal-transition)-signaling regulates TICs’ turnover. However, the mechanisms associated with this episode are unclear. We show that, in triple-negative-breast cancer (TNBC) cells enriched with TICs, CCN5 significantly blocks cellular growth via apoptosis, reversing EMT-signaling and impairing mammosphere formation, thereby blocking the tumor-forming ability and invasive capacity of these cells. To corroborate these findings, we isolated tumor-initiating side populations (SP) and non-side population (NSP or main population) from MCF-7 cell line, and evaluated the impact of CCN5 on these subpopulations. CCN5 was overexpressed in the NSP but downregulated in the SP. Characteristically, NSP cells are ER-α positive and epithelial type with little tumorigenic potency, while SP cells are very similar to triple-negative ones that do not express ER-α- and Her-2 and are highly tumorigenic in xenograft models. The overexpression of CCN5 in SP results in EMT reversion, ER-α upregulation and delays in tumor growth in xenograft models. We reasoned that CCN5 distinguishes SP and NSP and could reprogram SP to NSP transition, thereby delaying tumor growth in the xenograft model. Collectively, we reveal how CCN5-signaling underlies the driving force to prevent TNBC growth and progression.
Collapse
|
162
|
Lai WT, Li YJ, Wu SB, Yang CN, Wu TS, Wei YH, Deng YT. Connective tissue growth factor decreases mitochondrial metabolism through ubiquitin-mediated degradation of mitochondrial transcription factor A in oral squamous cell carcinoma. J Formos Med Assoc 2017; 117:212-219. [PMID: 28438434 DOI: 10.1016/j.jfma.2017.04.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 04/06/2017] [Accepted: 04/07/2017] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND/PURPOSE Deregulation of metabolic pathways is one of the hallmarks of cancer progression. Connective tissue growth factor (CTGF/CCN2) acts as a tumor suppressor in oral squamous cell carcinoma (OSCC). However, the role of CTGF in modulating cancer metabolism is still unclear. METHODS OSCC cells stably overexpressing CTGF (SAS/CTGF) and shRNA against CTGF (TW2.6/shCTGF) were established. Oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) were examined by the Seahorse XF24 analyzer. The expression of CTGF and mitochondrial biogenesis related genes was measured by real-time polymerase chain reaction or Western blot analysis. RESULTS CTGF decreased OCR, ECAR, adenosine triphosphate (ATP) generation, mitochondrial DNA (mtDNA), and mitochondrial transcription factor A (mtTFA) protein expression in OSCC cells. Overexpression of mtTFA restored CTGF-decreased OCR, ECAR, mtDNA copy number, migration and invasion of SAS/CTGF cells. Immunoprecipitation assay showed a higher level of ubiquitinated mtTFA protein after CTGF treatment. MG132, an inhibitor of proteasomal degradation, reversed the effect of CTGF on mtTFA protein expression in SAS cells. CONCLUSION CTGF can decrease glycolysis, mitochondrial oxidative phosphorylation, ATP generation, and mtDNA copy number by increasing mtTFA protein degradation through ubiquitin proteasome pathway and in turn reduces migration and invasion of OSCC cells. Therefore, CTGF may be developed as a potential additive therapeutic drug for oral cancer in the near future.
Collapse
Affiliation(s)
- Wei-Ting Lai
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan
| | - Yue-Ju Li
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan
| | - Shi-Bei Wu
- Institute of Biotechnology in Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Cheng-Ning Yang
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan
| | - Tai-Sheng Wu
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan
| | - Yau-Huei Wei
- Institute of Biotechnology in Medicine, National Yang-Ming University, Taipei, Taiwan; Department of Medicine, Mackay Medical College, Taiwan
| | - Yi-Ting Deng
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan; Department of Dentistry, National Taiwan University Hospital, Hsin-Chu Branch, Hsin-Chu, Taiwan.
| |
Collapse
|
163
|
Basset C, Bonnet-Magnaval F, Navarro MGJ, Touriol C, Courtade M, Prats H, Garmy-Susini B, Lacazette E. Api5 a new cofactor of estrogen receptor alpha involved in breast cancer outcome. Oncotarget 2017; 8:52511-52526. [PMID: 28881748 PMCID: PMC5581047 DOI: 10.18632/oncotarget.17281] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 03/10/2017] [Indexed: 01/31/2023] Open
Abstract
Api5 (Apoptosis inhibitor 5) is an anti-apoptotic factor that confers resistance to genotoxic stress in human cancer. Api5 is also expressed in endothelial cells and participates to the Estrogen Receptor α (ERα) signaling to promote cell migration. In this study, we found an over expression of Api5 in human breast cancer. Given that we show that high expression of Api5 in breast cancer patients is associated with shorter recurrence free survival, we investigated the relationship between ERα and Api5 at the molecular level. We found that Api5 Nuclear Receptor box (NR box) drives a direct interaction with the C domain of ERα. Furthermore, Api5 participates to gene transcription activation of ERα target genes upon estrogen treatment. Besides, Api5 expression favors tumorigenicity and migration and is necessary for tumor growth in vivo in mice xenografted model of breast cancer cell line. These finding suggest that Api5 is a new cofactor of ERα that functionally participates to the tumorigenic phenotype of breast cancer cells. In ERα breast cancer patients, Api5 overexpression is associated with poor survival, and may be used as a predictive marker of breast cancer recurrence free survival.
Collapse
Affiliation(s)
- Céline Basset
- U1037-CRCT, INSERM, Université Toulouse, F-31037, Toulouse, France.,Laboratoire d'Histologie-Embryologie, Faculté de Médecine Rangueil, F-31062, Toulouse, France
| | | | | | | | - Monique Courtade
- U1037-CRCT, INSERM, Université Toulouse, F-31037, Toulouse, France.,Laboratoire d'Histologie-Embryologie, Faculté de Médecine Rangueil, F-31062, Toulouse, France
| | - Hervé Prats
- U1037-CRCT, INSERM, Université Toulouse, F-31037, Toulouse, France
| | | | - Eric Lacazette
- UMR 1048-I2MC, INSERM, Université Toulouse, F-31432, Toulouse, France
| |
Collapse
|
164
|
Pereira LP, Silva P, Duarte M, Rodrigues L, Duarte CMM, Albuquerque C, Serra AT. Targeting Colorectal Cancer Proliferation, Stemness and Metastatic Potential Using Brassicaceae Extracts Enriched in Isothiocyanates: A 3D Cell Model-Based Study. Nutrients 2017; 9:nu9040368. [PMID: 28394276 PMCID: PMC5409707 DOI: 10.3390/nu9040368] [Citation(s) in RCA: 43] [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: 03/01/2017] [Revised: 03/23/2017] [Accepted: 04/01/2017] [Indexed: 01/28/2023] Open
Abstract
Colorectal cancer (CRC) recurrence is often attributable to circulating tumor cells and/or cancer stem cells (CSCs) that resist to conventional therapies and foster tumor progression. Isothiocyanates (ITCs) derived from Brassicaceae vegetables have demonstrated anticancer effects in CRC, however little is known about their effect in CSCs and tumor initiation properties. Here we examined the effect of ITCs-enriched Brassicaceae extracts derived from watercress and broccoli in cell proliferation, CSC phenotype and metastasis using a previously developed three-dimensional HT29 cell model with CSC-like traits. Both extracts were phytochemically characterized and their antiproliferative effect in HT29 monolayers was explored. Next, we performed cell proliferation assays and flow cytometry analysis in HT29 spheroids treated with watercress and broccoli extracts and respective main ITCs, phenethyl isothiocyanate (PEITC) and sulforaphane (SFN). Soft agar assays and relative quantitative expression analysis of stemness markers and Wnt/β-catenin signaling players were performed to evaluate the effect of these phytochemicals in stemness and metastasis. Our results showed that both Brassicaceae extracts and ITCs exert antiproliferative effects in HT29 spheroids, arresting cell cycle at G₂/M, possibly due to ITC-induced DNA damage. Colony formation and expression of LGR5 and CD133 cancer stemness markers were significantly reduced. Only watercress extract and PEITC decreased ALDH1 activity in a dose-dependent manner, as well as β-catenin expression. Our research provides new insights on CRC therapy using ITC-enriched Brassicaceae extracts, specially watercress extract, to target CSCs and circulating tumor cells by impairing cell proliferation, ALDH1-mediated chemo-resistance, anoikis evasion, self-renewal and metastatic potential.
Collapse
Affiliation(s)
- Lucília P Pereira
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal.
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa (ITQB NOVA), 2780-157 Oeiras, Portugal.
| | - Patrícia Silva
- Unidade de Investigação em Patobiologia Molecular (UIPM), Instituto Português de Oncologia de Lisboa Francisco Gentil, E.P.E (IPOLFG, EPE), 1099-023 Lisboa, Portugal.
| | - Marlene Duarte
- Unidade de Investigação em Patobiologia Molecular (UIPM), Instituto Português de Oncologia de Lisboa Francisco Gentil, E.P.E (IPOLFG, EPE), 1099-023 Lisboa, Portugal.
| | - Liliana Rodrigues
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal.
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa (ITQB NOVA), 2780-157 Oeiras, Portugal.
| | - Catarina M M Duarte
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal.
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa (ITQB NOVA), 2780-157 Oeiras, Portugal.
| | - Cristina Albuquerque
- Unidade de Investigação em Patobiologia Molecular (UIPM), Instituto Português de Oncologia de Lisboa Francisco Gentil, E.P.E (IPOLFG, EPE), 1099-023 Lisboa, Portugal.
| | - Ana Teresa Serra
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2780-901 Oeiras, Portugal.
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa (ITQB NOVA), 2780-157 Oeiras, Portugal.
| |
Collapse
|
165
|
Zhou D, Springer MZ, Xu D, Liu D, Hudmon A, Macleod KF, Meroueh SO. Small molecules inhibit STAT3 activation, autophagy, and cancer cell anchorage-independent growth. Bioorg Med Chem 2017; 25:2995-3005. [PMID: 28438385 DOI: 10.1016/j.bmc.2017.03.048] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 03/18/2017] [Accepted: 03/21/2017] [Indexed: 12/25/2022]
Abstract
Triple-negative breast cancers (TNBCs) lack the signature targets of other breast tumors, such as HER2, estrogen receptor, and progesterone receptor. These aggressive basal-like tumors are driven by a complex array of signaling pathways that are activated by multiple driver mutations. Here we report the discovery of 6 (KIN-281), a small molecule that inhibits multiple kinases including maternal leucine zipper kinase (MELK) and the non-receptor tyrosine kinase bone marrow X-linked (BMX) with single-digit micromolar IC50s. Several derivatives of 6 were synthesized to gain insight into the binding mode of the compound to the ATP binding pocket. Compound 6 was tested for its effect on anchorage-dependent and independent growth of MDA-MB-231 and MDA-MB-468 breast cancer cells. The effect of 6 on BMX prompted us to evaluate its effect on STAT3 phosphorylation and DNA binding. The compound's inhibition of cell growth led to measurements of survivin, Bcl-XL, p21WAF1/CIP1, and cyclin A2 levels. Finally, LC3B-II levels were quantified following treatment of cells with 6 to determine whether the compound affected autophagy, a process that is known to be activated by STAT3. Compound 6 provides a starting point for the development of small molecules with polypharmacology that can suppress TNBC growth and metastasis.
Collapse
Affiliation(s)
- Donghui Zhou
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, United States
| | - Maya Z Springer
- The Ben May Department for Cancer Research, University of Chicago, United States
| | - David Xu
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, United States; Department of BioHealth Informatics, Indiana University School of Informatics and Computing, United States
| | - Degang Liu
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, United States
| | - Andy Hudmon
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, United States
| | - Kay F Macleod
- The Ben May Department for Cancer Research, University of Chicago, United States
| | - Samy O Meroueh
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, United States; Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, United States.
| |
Collapse
|
166
|
Christenson JL, Butterfield KT, Spoelstra NS, Norris JD, Josan JS, Pollock JA, McDonnell DP, Katzenellenbogen BS, Katzenellenbogen JA, Richer JK. MMTV-PyMT and Derived Met-1 Mouse Mammary Tumor Cells as Models for Studying the Role of the Androgen Receptor in Triple-Negative Breast Cancer Progression. Discov Oncol 2017; 8:69-77. [PMID: 28194662 DOI: 10.1007/s12672-017-0285-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 01/27/2017] [Indexed: 12/19/2022] Open
Abstract
Triple-negative breast cancer (TNBC) has a faster rate of metastasis compared to other breast cancer subtypes, and no effective targeted therapies are currently FDA-approved. Recent data indicate that the androgen receptor (AR) promotes tumor survival and may serve as a potential therapeutic target in TNBC. Studies of AR in disease progression and the systemic effects of anti-androgens have been hindered by the lack of an AR-positive (AR+) immunocompetent preclinical model. In this study, we identified the transgenic MMTV-PyMT (mouse mammary tumor virus-polyoma middle tumor-antigen) mouse mammary gland carcinoma model of breast cancer and Met-1 cells derived from this model as tools to study the role of AR in breast cancer progression. AR protein expression was examined in late-stage primary tumors and lung metastases from MMTV-PyMT mice as well as in Met-1 cells by immunohistochemistry (IHC). Sensitivity of Met-1 cells to the AR agonist dihydrotestosterone (DHT) and anti-androgen therapy was examined using cell viability, migration/invasion, and anchorage-independent growth assays. Late-stage primary tumors and lung metastases from MMTV-PyMT mice and Met-1 cells expressed abundant nuclear AR protein, while negative for estrogen and progesterone receptors. Met-1 sensitivity to DHT and AR antagonists demonstrated a reliance on AR for survival, and AR antagonists inhibited invasion and anchorage-independent growth. These data suggest that the MMTV-PyMT model and Met-1 cells may serve as valuable tools for mechanistic studies of the role of AR in disease progression and how anti-androgens affect the tumor microenvironment.
Collapse
Affiliation(s)
- Jessica L Christenson
- Department of Pathology, University of Colorado, Anschutz Medical Campus, 12800 E. 19th Ave., Aurora, CO, 80045, USA
| | - Kiel T Butterfield
- Department of Pathology, University of Colorado, Anschutz Medical Campus, 12800 E. 19th Ave., Aurora, CO, 80045, USA
| | - Nicole S Spoelstra
- Department of Pathology, University of Colorado, Anschutz Medical Campus, 12800 E. 19th Ave., Aurora, CO, 80045, USA
| | - John D Norris
- Department of Pharmacology and Cancer Biology, Duke University, 450 Research Drive, Durham, NC, 27708, USA
| | - Jatinder S Josan
- Department of Chemistry, Virginia Tech University, 900 West Campus Drive, Blacksburg, VA, 24061, USA
| | - Julie A Pollock
- Department of Chemistry, University of Richmond, 28 Westhampton Way, Richmond, VA, 23173, USA
| | - Donald P McDonnell
- Department of Pharmacology and Cancer Biology, Duke University, 450 Research Drive, Durham, NC, 27708, USA
| | - Benita S Katzenellenbogen
- Department of Molecular and Integrative Physiology, University of Illinois, 407 South Goodwin Avenue, Urbana, IL, 61801, USA
| | - John A Katzenellenbogen
- Department of Chemistry, University of Illinois, 600 South Mathews Avenue, Urbana, IL, 61801, USA
| | - Jennifer K Richer
- Department of Pathology, University of Colorado, Anschutz Medical Campus, 12800 E. 19th Ave., Aurora, CO, 80045, USA.
| |
Collapse
|
167
|
Ho SM, Rao R, To S, Schoch E, Tarapore P. Bisphenol A and its analogues disrupt centrosome cycle and microtubule dynamics in prostate cancer. Endocr Relat Cancer 2017; 24:83-96. [PMID: 27998958 PMCID: PMC5226663 DOI: 10.1530/erc-16-0175] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 12/19/2016] [Indexed: 12/11/2022]
Abstract
Humans are increasingly exposed to structural analogues of bisphenol A (BPA), as BPA is being replaced by these compounds in BPA-free consumer products. We have previously shown that chronic and developmental exposure to BPA is associated with increased prostate cancer (PCa) risk in human and animal models. Here, we examine whether exposure of PCa cells (LNCaP, C4-2) to low-dose BPA and its structural analogues (BPS, BPF, BPAF, TBBPA, DMBPA and TMBPA) affects centrosome amplification (CA), a hallmark of cancer initiation and progression. We found that exposure to BPA, BPS, DMBPA and TBBPA, in descending order, increased the number of cells with CA, in a non-monotonic dose-response manner. Furthermore, cells treated with BPA and their analogues initiated centrosome duplication at 8 h after release from serum starvation, significantly earlier in G-1 phase than control cells. This response was attended by earlier release of nucleophosmin from unduplicated centrosomes. BPA-exposed cells exhibited increased expression of cyclin-dependent kinase CDK6 and decreased expression of CDK inhibitors (p21Waf1/CIP1 and p27KIP1). Using specific antagonists for estrogen/androgen receptors, CA in the presence of BPA or its analogues was likely to be mediated via ESR1 signaling. Change in microtubule dynamics was observed on exposure to these analogues, which, for BPA, was accompanied by increased expression of centrosome-associated protein CEP350 Similar to BPA, chronic treatment of cells with DMBPA, but not other analogues, resulted in the enhancement of anchorage-independent growth. We thus conclude that selected BPA analogues, similar to BPA, disrupt centrosome function and microtubule organization, with DMBPA displaying the broadest spectrum of cancer-promoting effects.
Collapse
Affiliation(s)
- Shuk-Mei Ho
- Department of Environmental HealthUniversity of Cincinnati Medical Center, Cincinnati, Ohio, USA
- Center for Environmental GeneticsUniversity of Cincinnati Medical Center, Cincinnati, Ohio, USA
- Cincinnati Cancer CenterCincinnati, Ohio, USA
- Cincinnati Veteran Affairs Hospital Medical CenterCincinnati, Ohio, USA
| | - Rahul Rao
- Department of Environmental HealthUniversity of Cincinnati Medical Center, Cincinnati, Ohio, USA
| | - Sarah To
- Department of Environmental HealthUniversity of Cincinnati Medical Center, Cincinnati, Ohio, USA
- Center for Cancer ResearchHudson Institute of Medical Research, Clayton, Victoria, Australia
- Monash UniversityClayton, Victoria, Australia
| | - Emma Schoch
- Department of Environmental HealthUniversity of Cincinnati Medical Center, Cincinnati, Ohio, USA
| | - Pheruza Tarapore
- Department of Environmental HealthUniversity of Cincinnati Medical Center, Cincinnati, Ohio, USA
- Center for Environmental GeneticsUniversity of Cincinnati Medical Center, Cincinnati, Ohio, USA
- Cincinnati Cancer CenterCincinnati, Ohio, USA
| |
Collapse
|
168
|
Saha S, Ghosh M, Dutta SK. The dual-hit metabolic modulator LDCA synergistically potentiates doxorubicin to selectively combat cancer-associated hallmarks. RSC Adv 2017. [DOI: 10.1039/c7ra08625c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Dual-hit metabolic modulator LDCA synergistically potentiates doxorubicin to counter melanoma progression.
Collapse
Affiliation(s)
- Suchandrima Saha
- Drug Development Diagnostics and Biotechnology
- CSIR-Indian Institute of Chemical Biology (CSIR-IICB)
- Kolkata-700032
- India
| | - Monisankar Ghosh
- Drug Development Diagnostics and Biotechnology
- CSIR-Indian Institute of Chemical Biology (CSIR-IICB)
- Kolkata-700032
- India
| | - Samir Kumar Dutta
- Drug Development Diagnostics and Biotechnology
- CSIR-Indian Institute of Chemical Biology (CSIR-IICB)
- Kolkata-700032
- India
| |
Collapse
|
169
|
Efficient Transformation of Primary Human Mesenchymal Stromal Cells by Adenovirus Early Region 1 Oncogenes. J Virol 2016; 91:JVI.01782-16. [PMID: 27795433 DOI: 10.1128/jvi.01782-16] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 10/15/2016] [Indexed: 11/20/2022] Open
Abstract
Previous observations that human amniotic fluid cells (AFC) can be transformed by human adenovirus type 5 (HAdV-5) E1A/E1B oncogenes prompted us to identify the target cells in the AFC population that are susceptible to transformation. Our results demonstrate that one cell type corresponding to mesenchymal stem/stroma cells (hMSCs) can be reproducibly transformed by HAdV-5 E1A/E1B oncogenes as efficiently as primary rodent cultures. HAdV-5 E1-transformed hMSCs exhibit all properties commonly associated with a high grade of oncogenic transformation, including enhanced cell proliferation, anchorage-independent growth, increased growth rate, and high telomerase activity as well as numerical and structural chromosomal aberrations. These data confirm previous work showing that HAdV preferentially transforms cells of mesenchymal origin in rodents. More importantly, they demonstrate for the first time that human cells with stem cell characteristics can be completely transformed by HAdV oncogenes in tissue culture with high efficiency. Our findings strongly support the hypothesis that undifferentiated progenitor cells or cells with stem cell-like properties are highly susceptible targets for HAdV-mediated cell transformation and suggest that virus-associated tumors in humans may originate, at least in part, from infections of these cell types. We expect that primary hMSCs will replace the primary rodent cultures in HAdV viral transformation studies and are confident that these investigations will continue to uncover general principles of viral oncogenesis that can be extended to human DNA tumor viruses as well. IMPORTANCE It is generally believed that transformation of primary human cells with HAdV-5 E1 oncogenes is very inefficient. However, a few cell lines have been successfully transformed with HAdV-5 E1A and E1B, indicating that there is a certain cell type which is susceptible to HAdV-mediated transformation. Interestingly, all those cell lines have been derived from human embryonic tissue, albeit the exact cell type is not known yet. We show for the first time the successful transformation of primary human mesenchymal stromal cells (hMSCs) by HAdV-5 E1A and E1B. Further, we show upon HAdV-5 E1A and E1B expression that these primary progenitor cells exhibit features of tumor cells and can no longer be differentiated into the adipogenic, chondrogenic, or osteogenic lineage. Hence, primary hMSCs represent a robust and novel model system to elucidate the underlying molecular mechanisms of adenovirus-mediated transformation of multipotent human progenitor cells.
Collapse
|
170
|
Zhang RY, Yu ZH, Zeng L, Zhang S, Bai Y, Miao J, Chen L, Xie J, Zhang ZY. SHP2 phosphatase as a novel therapeutic target for melanoma treatment. Oncotarget 2016; 7:73817-73829. [PMID: 27650545 PMCID: PMC5342016 DOI: 10.18632/oncotarget.12074] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Accepted: 09/02/2016] [Indexed: 12/23/2022] Open
Abstract
Melanoma ranks among the most aggressive and deadly human cancers. Although a number of targeted therapies are available, they are effective only in a subset of patients and the emergence of drug resistance often reduces durable responses. Thus there is an urgent need to identify new therapeutic targets and develop more potent pharmacological agents for melanoma treatment. Herein we report that SHP2 levels are frequently elevated in melanoma, and high SHP2 expression is significantly associated with more metastatic phenotype and poorer prognosis. We show that SHP2 promotes melanoma cell viability, motility, and anchorage-independent growth, through activation of both ERK1/2 and AKT signaling pathways. We demonstrate that SHP2 inhibitor 11a-1 effectively blocks SHP2-mediated ERK1/2 and AKT activation and attenuates melanoma cell viability, migration and colony formation. Most importantly, SHP2 inhibitor 11a-1 suppresses xenografted melanoma tumor growth, as a result of reduced tumor cell proliferation and enhanced tumor cell apoptosis. Taken together, our data reveal SHP2 as a novel target for melanoma and suggest SHP2 inhibitors as potential novel therapeutic agents for melanoma treatment.
Collapse
Affiliation(s)
- Ruo-Yu Zhang
- Department of Medicinal Chemistry and Molecular Pharmacology, Center for Cancer Research, and Institute for Drug Discovery, Purdue University, West Lafayette, IN 47907, USA
| | - Zhi-Hong Yu
- Department of Medicinal Chemistry and Molecular Pharmacology, Center for Cancer Research, and Institute for Drug Discovery, Purdue University, West Lafayette, IN 47907, USA
| | - Lifan Zeng
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Sheng Zhang
- Department of Medicinal Chemistry and Molecular Pharmacology, Center for Cancer Research, and Institute for Drug Discovery, Purdue University, West Lafayette, IN 47907, USA
| | - Yunpeng Bai
- Department of Medicinal Chemistry and Molecular Pharmacology, Center for Cancer Research, and Institute for Drug Discovery, Purdue University, West Lafayette, IN 47907, USA
| | - Jinmin Miao
- Department of Medicinal Chemistry and Molecular Pharmacology, Center for Cancer Research, and Institute for Drug Discovery, Purdue University, West Lafayette, IN 47907, USA
| | - Lan Chen
- Department of Medicinal Chemistry and Molecular Pharmacology, Center for Cancer Research, and Institute for Drug Discovery, Purdue University, West Lafayette, IN 47907, USA
| | - Jingwu Xie
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Zhong-Yin Zhang
- Department of Medicinal Chemistry and Molecular Pharmacology, Center for Cancer Research, and Institute for Drug Discovery, Purdue University, West Lafayette, IN 47907, USA
- Department of Chemistry, Center for Cancer Research, and Institute for Drug Discovery, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
171
|
Piao MY, Cao HL, He NN, Xu MQ, Dong WX, Wang WQ, Wang BM, Zhou B. Potential role of TRIM3 as a novel tumour suppressor in colorectal cancer (CRC) development. Scand J Gastroenterol 2016; 51:572-82. [PMID: 26691157 DOI: 10.3109/00365521.2015.1124285] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVE Colorectal cancer (CRC) is the third leading cause of cancer-related mortality in the United States. Recent cancer genome-sequencing efforts and complementary functional studies have led to the identification of a collection of candidate 'driver' genes involved in CRC tumorigenesis. Tripartite motif (TRIM3) is recently identified as a tumour suppressor in glioblastoma but this tumour-suppressive function has not been investigated in CRC. MATERIAL AND METHODS In this study, we investigated the potential role of TRIM3 as a tumour suppressor in CRC development by manipulating the expression of TRIM3 in two authentic CRC cell lines, HCT116 and DLD1, followed by various functional assays, including cell proliferation, colony formation, scratch wound healing, soft agar, and invasion assays. Xenograft experiment was performed to examine in vivo tumour-suppressive properties of TRIM3. RESULTS Small-interfering RNA (siRNA) mediated knockdown of TRIM3 conferred growth advantage in CRC cells. In contrast, overexpression of TRIM3 affected cell survival, cell migration, anchorage independent growth and invasive potential in CRC cells. In addition, TRIM3 was found to be down-regulated in human colon cancer tissues compared with matched normal colon tissues. Overexpression of TRIM3 significantly inhibited tumour growth in vivo using xenograft mouse models. Mechanistic investigation revealed that TRIM3 can regulate p53 protein level through its stabilisation. CONCLUSIONS TRIM3 functions as a tumour suppressor in CRC progression. This tumour-suppressive function is exerted partially through regulation of p53 protein. Therefore, this protein may represent a novel therapeutic target for prevention or intervention of CRC.
Collapse
Affiliation(s)
- Mei-Yu Piao
- a Department of Gastroenterology and Hepatology , General Hospital, Tianjin Medical University , Heping District , Tianjin , China
| | - Hai-Long Cao
- a Department of Gastroenterology and Hepatology , General Hospital, Tianjin Medical University , Heping District , Tianjin , China
| | - Na-Na He
- a Department of Gastroenterology and Hepatology , General Hospital, Tianjin Medical University , Heping District , Tianjin , China
| | - Meng-Que Xu
- a Department of Gastroenterology and Hepatology , General Hospital, Tianjin Medical University , Heping District , Tianjin , China
| | - Wen-Xiao Dong
- a Department of Gastroenterology and Hepatology , General Hospital, Tianjin Medical University , Heping District , Tianjin , China
| | - Wei-Qiang Wang
- a Department of Gastroenterology and Hepatology , General Hospital, Tianjin Medical University , Heping District , Tianjin , China
| | - Bang-Mao Wang
- a Department of Gastroenterology and Hepatology , General Hospital, Tianjin Medical University , Heping District , Tianjin , China
| | - Bing Zhou
- b Department of Gastroenterology , Tanggu Traditional Chinese Medicine Hospital of Tianjin Binhai New Area , Tanggu Binhai New Area , Tianjin , China
| |
Collapse
|
172
|
Thomas C, Ji Y, Lodhi N, Kotova E, Pinnola AD, Golovine K, Makhov P, Pechenkina K, Kolenko V, Tulin AV. Non-NAD-Like poly(ADP-Ribose) Polymerase-1 Inhibitors effectively Eliminate Cancer in vivo. EBioMedicine 2016; 13:90-98. [PMID: 27727003 PMCID: PMC5264309 DOI: 10.1016/j.ebiom.2016.10.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 10/03/2016] [Accepted: 10/03/2016] [Indexed: 01/29/2023] Open
Abstract
The clinical potential of PARP-1 inhibitors has been recognized >10years ago, prompting intensive research on their pharmacological application in several branches of medicine, particularly in oncology. However, natural or acquired resistance of tumors to known PARP-1 inhibitors poses a serious problem for their clinical implementation. Present study aims to reignite clinical interest to PARP-1 inhibitors by introducing a new method of identifying highly potent inhibitors and presenting the largest known collection of structurally diverse inhibitors. The majority of PARP-1 inhibitors known to date have been developed as NAD competitors. NAD is utilized by many enzymes other than PARP-1, resulting in a trade-off trap between their specificity and efficacy. To circumvent this problem, we have developed a new strategy to blindly screen a small molecule library for PARP-1 inhibitors by targeting a highly specific rout of its activation. Based on this screen, we present a collection of PARP-1 inhibitors and provide their structural classification. In addition to compounds that show structural similarity to NAD or known PARP-1 inhibitors, the screen identified structurally new non-NAD-like inhibitors that block PARP-1 activity in cancer cells with greater efficacy and potency than classical PARP-1 inhibitors currently used in clinic. These non-NAD-like PARP-1 inhibitors are effective against several types of human cancer xenografts, including kidney, prostate, and breast tumors in vivo. Our pre-clinical testing of these inhibitors using laboratory animals has established a strong foundation for advancing the new inhibitors to clinical trials.
Collapse
Affiliation(s)
- Colin Thomas
- Fox Chase Cancer Center, Philadelphia, PA, United States
| | - Yingbiao Ji
- Fox Chase Cancer Center, Philadelphia, PA, United States
| | - Niraj Lodhi
- Fox Chase Cancer Center, Philadelphia, PA, United States
| | - Elena Kotova
- Fox Chase Cancer Center, Philadelphia, PA, United States
| | | | | | - Peter Makhov
- Fox Chase Cancer Center, Philadelphia, PA, United States
| | | | | | - Alexei V Tulin
- Fox Chase Cancer Center, Philadelphia, PA, United States.
| |
Collapse
|
173
|
Liu X, Liao W, Yuan Q, Ou Y, Huang J. TTK activates Akt and promotes proliferation and migration of hepatocellular carcinoma cells. Oncotarget 2016; 6:34309-20. [PMID: 26418879 PMCID: PMC4741454 DOI: 10.18632/oncotarget.5295] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2015] [Accepted: 09/07/2015] [Indexed: 12/15/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most malignant cancers with poor clinical outcome. The protein kinase human monopolar spindle 1 (hMps1/TTK) gene expression is significantly increased in HCCs. However, its contributions to hepatocarcinogenesis remain unclear. In this study, we found that TTK was overexpressed in 77.63% (118/152) HCC specimens. Elevated TTK expression positively correlated with large tumor size and presence of the portal vein tumor thrombus (PVTT). Demethylation in its promoter increased TTK expression in HCC. In vitro assays revealed that TTK not only promoted cell proliferation and anchorage-independent growth, but also cell migration. Subsequent investigations revealed that TTK activated Akt/mTOR pathway in a p53 dependent manner. We also found that TTK specific kinase inhibitor AZ3146 could decrease HCC cell growth. In conclusion, TTK contributes to HCC tumorigenesis via promoting cell proliferation and migration. It may serve as a novel biomarker and a potential target in HCC cancer therapy.
Collapse
Affiliation(s)
- Xing Liu
- National Engineering Center for Biochip at Shanghai, Shanghai, China.,Shanghai-MOST Key Laboratory for Disease and Health Genomics, Chinese National Human Genome Center at Shanghai, Shanghai, China
| | - Weijia Liao
- Hepatology Institute of Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, China
| | - Qing Yuan
- National Engineering Center for Biochip at Shanghai, Shanghai, China
| | - Ying Ou
- National Engineering Center for Biochip at Shanghai, Shanghai, China
| | - Jian Huang
- National Engineering Center for Biochip at Shanghai, Shanghai, China.,Shanghai-MOST Key Laboratory for Disease and Health Genomics, Chinese National Human Genome Center at Shanghai, Shanghai, China.,Key Laboratory of Systems Biomedicine (Ministry of Education) and Collaborative Innovation Center of Systems Biomedicine, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
174
|
Amith SR, Wilkinson JM, Fliegel L. KR-33028, a potent inhibitor of the Na +/H + exchanger NHE1, suppresses metastatic potential of triple-negative breast cancer cells. Biochem Pharmacol 2016; 118:31-39. [PMID: 27521504 DOI: 10.1016/j.bcp.2016.08.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 08/09/2016] [Indexed: 10/21/2022]
Abstract
Hyper-activation of the Na+/H+ exchanger NHE1 occurs at the onset of oncogenic transformation and plays a critical role in breast cancer carcinogenesis. Dysregulation of NHE1 activity results in intracellular alkalinization and the acidification of the extracellular tumor microenvironment that promotes metastasis. Hence, the use of chemical inhibitors of NHE1 as chemotherapeutic agents is an alluring prospect. We previously demonstrated that two structurally different NHE1 inhibitors, EMD87580 [(2-methyl-4,5-di-(methylsulfonyl)-benzoyl)-guanidine], and HMA [5-(N,N-hexamethylene)-amiloride], were effective as co-adjuvants to potentiate paclitaxel-mediated cytotoxic chemotherapy in triple-negative breast cancer (TNBC) cells. Both these drugs, however, had reduced or minimal anti-cancer effects when used alone. Here, we tested KR-33028 (4-cyano (benzo[b]thiophene-2-carbonyl)guanidine), a potent and selective inhibitor of NHE1, to determine its efficacy in inhibition of metastatic potential of TNBC cells. In highly invasive MDA-MB-231, moderately invasive MDA-MB-468, and lowly invasive Hs578T TNBC cells, KR-33028 considerably reduced rates of cell migration and anchorage-independent colony growth. Invasion of MDA-MB-231 and MDA-MB-468 cells through extracellular matrix was also dramatically decreased in response to KR-33028. We further tested the effect of KR-33028 on MDA-MB-231 cells lacking NHE1 expression (231koNHE1); no differences were observed between untreated control and KR-33028-treated 231koNHE1 cells. Taken together, our results highlight the in vitro efficacy of KR-33028-mediated NHE1 inhibition on limiting cellular functions that are predictive of metastasis in vivo. We suggest that targeting NHE1 in the development of novel chemotherapeutics could be highly effective in combatting triple-negative breast cancer and that KR-33028 is potentially useful in prevention of metastasis.
Collapse
Affiliation(s)
- Schammim Ray Amith
- Department of Biochemistry, University of Alberta, Edmonton, Alberta T6G 2H7, Canada.
| | - Jodi Marie Wilkinson
- Department of Biochemistry, University of Alberta, Edmonton, Alberta T6G 2H7, Canada.
| | - Larry Fliegel
- Department of Biochemistry, University of Alberta, Edmonton, Alberta T6G 2H7, Canada.
| |
Collapse
|
175
|
Guanidine Alkaloids from the Marine Sponge Monanchora pulchra Show Cytotoxic Properties and Prevent EGF-Induced Neoplastic Transformation in Vitro. Mar Drugs 2016; 14:md14070133. [PMID: 27428983 PMCID: PMC4962023 DOI: 10.3390/md14070133] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 07/05/2016] [Accepted: 07/08/2016] [Indexed: 12/13/2022] Open
Abstract
Guanidine alkaloids from sponges Monanchora spp. represent diverse bioactive compounds, however, the mechanisms underlying bioactivity are very poorly understood. Here, we report results of studies on cytotoxic action, the ability to inhibit EGF-induced neoplastic transformation, and the effects on MAPK/AP-1 signaling of eight rare guanidine alkaloids, recently isolated from the marine sponge Monanchora pulchra, namely: monanchocidin A (1), monanchocidin B (2), monanchomycalin C (3), ptilomycalin A (4), monanchomycalin B (5), normonanchocidin D (6), urupocidin A (7), and pulchranin A (8). All of the compounds induced cell cycle arrest (apart from 8) and programmed death of cancer cells. Ptilomycalin A-like compounds 1–6 activated JNK1/2 and ERK1/2, following AP-1 activation and caused p53-independent programmed cell death. Compound 7 induced p53-independent cell death without activation of AP-1 or caspase-3/7, and the observed JNK1/2 activation did not contribute to the cytotoxic effect of the compound. Alkaloid 8 induced JNK1/2 (but not ERK1/2) activation leading to p53-independent cell death and strong suppression of AP-1 activity. Alkaloids 1–4, 7, and 8 were able to inhibit the EGF-induced neoplastic transformation of JB6 P+ Cl41 cells. Our results suggest that investigated guanidine marine alkaloids hold potential to eliminate human cancer cells and prevent cancer cell formation and spreading.
Collapse
|
176
|
Troilo A, Benson EK, Esposito D, Garibsingh RAA, Reddy EP, Mungamuri SK, Aaronson SA. Angiomotin stabilization by tankyrase inhibitors antagonizes constitutive TEAD-dependent transcription and proliferation of human tumor cells with Hippo pathway core component mutations. Oncotarget 2016; 7:28765-82. [PMID: 27144834 PMCID: PMC5045355 DOI: 10.18632/oncotarget.9117] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 03/26/2016] [Indexed: 12/29/2022] Open
Abstract
The evolutionarily conserved Hippo inhibitory pathway plays critical roles in tissue homeostasis and organ size control, while mutations affecting certain core components contribute to tumorigenesis. Here we demonstrate that proliferation of Hippo pathway mutant human tumor cells exhibiting high constitutive TEAD transcriptional activity was markedly inhibited by dominant negative TEAD4, which did not inhibit the growth of Hippo wild-type cells with low levels of regulatable TEAD-mediated transcription. The tankyrase inhibitor, XAV939, identified in a screen for inhibitors of TEAD transcriptional activity, phenocopied these effects independently of its other known functions by stabilizing angiomotin and sequestering YAP in the cytosol. We also identified one intrinsically XAV939 resistant Hippo mutant tumor line exhibiting lower and less durable angiomotin stabilization. Thus, angiomotin stabilization provides a new mechanism for targeting tumors with mutations in Hippo pathway core components as well as a biomarker for sensitivity to such therapy.
Collapse
Affiliation(s)
- Albino Troilo
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Erica K. Benson
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Davide Esposito
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - E. Premkumar Reddy
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sathish Kumar Mungamuri
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Stuart A. Aaronson
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
177
|
Katoh D, Nishizuka M, Osada S, Imagawa M. FAD104, a Regulator of Adipogenesis and Osteogenesis, Interacts with the C-Terminal Region of STAT3 and Represses Malignant Transformation of Melanoma Cells. Biol Pharm Bull 2016; 39:849-55. [PMID: 26948083 DOI: 10.1248/bpb.b15-01026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Anchorage-independent growth is one of the defining characteristics of cancer cells. Many oncogenes and tumor suppressor genes are involved in regulating this type of growth. Factor for adipocyte differentiation 104 gene (fad104) is a regulator of adipogenesis and osteogenesis. Previously, we reported that fad104 suppressed metastasis as well as invasion of melanoma cells. However, it is unclear whether fad104 is involved in malignant transformation, which is associated with metastasis. In this study, we revealed that fad104 negatively regulated the colony forming activity of melanoma cells. The presence of the N-terminal region of FAD104 was required for the regulation of malignant transformation of melanoma cells. In addition, the deletion mutant of FAD104 that contained the N-terminal region and transmembrane domain interacted with signal transducer and activator of transcription 3 (STAT3) and suppressed STAT3 activity. However, the deletion mutant of FAD104 lacking the N-terminal region did not influence the interaction with STAT3 or suppress the STAT3 activity. Moreover, FAD104 interacted with the C-terminal region of STAT3. In summary, we demonstrated that fad104 suppressed anchorage-independent growth of melanoma cells, and that the N-terminal region of FAD104 is essential for inhibiting malignant transformation and STAT3 activity.
Collapse
Affiliation(s)
- Daiki Katoh
- Department of Molecular Biology, Graduate School of Pharmaceutical Sciences, Nagoya City University
| | | | | | | |
Collapse
|
178
|
Vales G, Rubio L, Marcos R. Genotoxic and cell-transformation effects of multi-walled carbon nanotubes (MWCNT) following in vitro sub-chronic exposures. JOURNAL OF HAZARDOUS MATERIALS 2016; 306:193-202. [PMID: 26736170 DOI: 10.1016/j.jhazmat.2015.12.021] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 11/23/2015] [Accepted: 12/14/2015] [Indexed: 06/05/2023]
Abstract
BEAS-2B cells were sub-chronically exposed (up to 4 weeks) to low doses of multi-walled carbon nanotubes (MWCNT, NM403). Genotoxic effects were evaluated using the comet and the micronucleus (MN) assays at three different time-points. The expression of different interleukins (IL) such as IL-1B, IL-6 and IL-8, as well as HO-1 as stress marker, was assessed after 3 weeks treatments. As a hallmark biomarker of cell-transforming ability we used the soft-agar assay, which detects anchorage-independent cell growth. Our results show high levels of intracellular reactive oxygen species (ROS) associated to MWCNT exposure. Nevertheless, an important proportion of these ROS levels seems to be associated to solubilized metals contaminants present in NM403, more than to the internalized MWCNT. No primary DNA damage was obtained in the Comet assay although significant levels of chromosome damage were detected using the micronucleus assay. A significant decrease in the expression of the studied cytokines was observed and significant increases in the number of induced colonies were obtained when the ability of induce anchorage-independent growth was determined. These results show that chromosome damage and reducing inflammatory signalling correlated with an increase in attachment-independent growth associated with sub-chronic MWCNT exposure.
Collapse
Affiliation(s)
- Gerard Vales
- Grup de Mutagènesi, Departament de Genètica i de Microbiologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Laura Rubio
- Grup de Mutagènesi, Departament de Genètica i de Microbiologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Ricard Marcos
- Grup de Mutagènesi, Departament de Genètica i de Microbiologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Spain; CIBER Epidemiología y Salud Pública, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
179
|
Saha S, Ghosh M, Dutta SK. Role of metabolic modulator Bet-CA in altering mitochondrial hyperpolarization to suppress cancer associated angiogenesis and metastasis. Sci Rep 2016; 6:23552. [PMID: 27003027 PMCID: PMC4802328 DOI: 10.1038/srep23552] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 03/07/2016] [Indexed: 01/05/2023] Open
Abstract
Solid tumors characteristically reflect a metabolic switching from glucose oxidation to glycolysis that plays a fundamental role in angiogenesis and metastasis to facilitate aggressive tumor outcomes. Hyperpolarized mitochondrial membrane potential is a manifestation of malignant cells that compromise the intrinsic pathways of apoptosis and confer a suitable niche to promote the cancer associated hallmark traits. We have previously reported that co-drug Bet-CA selectively targets cancer cells by inducing metabolic catastrophe without a manifest in toxicity. Here we report that the same molecule at a relatively lower concentration deregulates the cardinal phenotypes associated with angiogenesis and metastasis. In mice syngeneic 4T1 breast cancer model, Bet-CA exhibited effective abrogation of angiogenesis and concomitantly obliterated lung metastasis consistent with altered mitochondrial bioenergetics. Furthermore, Bet-CA significantly lowered vascular endothelial growth factor (VEGF) levels and obviated matrix metalloproteases (MMP-2/9) production directly to the criterion where abrogation of autocrine VEGF/VEGFR2 signalling loop was documented. In vitro studies anticipatedly documented the role of Bet-CA in inhibiting actin remodeling, lamellipodia formation and cell membrane ruffling to constitutively suppress cell motility and invasion. Results comprehensively postulate that Bet-CA, a mitochondria targeting metabolic modulator may serve as an excellent candidate for combating angiogenesis and metastasis.
Collapse
Affiliation(s)
- Suchandrima Saha
- Drug Development Diagnostic and Biotechnology Division, CSIR- Indian Institute of Chemical Biology (CSIR-IICB), 4, Raja S.C. Mullick Road, Kolkata-700032, West Bengal, India
| | - Monisankar Ghosh
- Drug Development Diagnostic and Biotechnology Division, CSIR- Indian Institute of Chemical Biology (CSIR-IICB), 4, Raja S.C. Mullick Road, Kolkata-700032, West Bengal, India
| | - Samir Kumar Dutta
- Drug Development Diagnostic and Biotechnology Division, CSIR- Indian Institute of Chemical Biology (CSIR-IICB), 4, Raja S.C. Mullick Road, Kolkata-700032, West Bengal, India
| |
Collapse
|
180
|
Ghosh M, Saha S, Dutta SK. 'Dual hit' metabolic modulator LDCA selectively kills cancer cells by efficient competitive inhibition of LDH-A. Chem Commun (Camb) 2016; 52:2401-4. [PMID: 26732434 DOI: 10.1039/c5cc09903j] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Herein, we synthesize and elucidate the potential of a novel 'dual hit' molecule, LDCA, to constitutively block lactate dehydrogenase isoform-A (LDH-A) to selectively subvert apoptosis and rigorously attenuate breast tumor progression in a mouse model, comprehensively delineating the therapeutic prospectus of LDCA in the field of cancer metabolics.
Collapse
Affiliation(s)
- Monisankar Ghosh
- Division of Drug Development, Diagnostics and Biotechnology, CSIR- Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Kolkata-700032, West Bengal, India.
| | - Suchandrima Saha
- Division of Drug Development, Diagnostics and Biotechnology, CSIR- Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Kolkata-700032, West Bengal, India.
| | - Samir Kumar Dutta
- Division of Drug Development, Diagnostics and Biotechnology, CSIR- Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Kolkata-700032, West Bengal, India.
| |
Collapse
|
181
|
Ghosh M, Saha S, Dutta SK. Synergism of metabolic modulators Bet-CA and LDCA: a rational combinatorial approach to selectively combat cancer associated hallmark traits. RSC Adv 2016. [DOI: 10.1039/c6ra14366k] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The strategic combination of two metabolic modulators, Bet-CA and LDCA in a calculated ratio selectively and synergistically inhibits the hallmark traits of cancer by altering the highly resilient mitochondrial bioenergetics.
Collapse
Affiliation(s)
- Monisankar Ghosh
- Drug Development Diagnostic and Biotechnology Division
- CSIR – Indian Institute of Chemical Biology (CSIR-IICB)
- Kolkata-700032
- India
| | - Suchandrima Saha
- Drug Development Diagnostic and Biotechnology Division
- CSIR – Indian Institute of Chemical Biology (CSIR-IICB)
- Kolkata-700032
- India
| | - Samir Kumar Dutta
- Drug Development Diagnostic and Biotechnology Division
- CSIR – Indian Institute of Chemical Biology (CSIR-IICB)
- Kolkata-700032
- India
| |
Collapse
|
182
|
Madajewski B, Boatman MA, Chakrabarti G, Boothman DA, Bey EA. Depleting Tumor-NQO1 Potentiates Anoikis and Inhibits Growth of NSCLC. Mol Cancer Res 2015; 14:14-25. [PMID: 26553038 DOI: 10.1158/1541-7786.mcr-15-0207-t] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 11/01/2015] [Indexed: 12/20/2022]
Abstract
UNLABELLED The fundamental role that NAD(P)H/quinone oxidoreductase 1 (NQO1) plays, in normal cells, as a cytoprotective enzyme guarding against stress induced by reactive oxygen species (ROS) is well documented. However, what is not known is whether the observed overexpression of NQO1 in neoplastic cells contributes to their survival. The current study discovered that depleting NQO1 expression in A549 and H292 lung adenocarcinoma cells caused an increase in ROS formation, inhibited anchorage-independent growth, increased anoikis sensitization, and decreased three-dimensional tumor spheroid invasion. These in vivo data further implicate tumor-NQO1 expression in a protumor survival role, because its depletion suppressed cell proliferation and decreased lung tumor xenograft growth. Finally, these data reveal an exploitable link between tumor-NQO1 expression and the survival of lung tumors because NQO1 depletion significantly decreased the percentage of ALDH((high)) cancer cells within the tumor population. IMPLICATIONS Loss of tumor-NQO1 expression inhibits tumor growth and suggests that novel therapeutics directed at tumor-NQO1 may have clinical benefit.
Collapse
Affiliation(s)
- Brian Madajewski
- Department of Pharmaceutical Sciences, Mary Babb Randolph Cancer Center, West Virginia University, Morgantown, West Virginia 26506
| | - Michael A Boatman
- Department of Pharmaceutical Sciences, Mary Babb Randolph Cancer Center, West Virginia University, Morgantown, West Virginia 26506
| | - Gaurab Chakrabarti
- Department of Pharmacology, Laboratory of Molecular Cell Stress Responses, Program in Cell Stress and Cancer Nanomedicine, Simmons Cancer Center, UT Southwestern Medical Center at Dallas, TX 75390-8807
| | - David A Boothman
- Department of Pharmacology, Laboratory of Molecular Cell Stress Responses, Program in Cell Stress and Cancer Nanomedicine, Simmons Cancer Center, UT Southwestern Medical Center at Dallas, TX 75390-8807
| | - Erik A Bey
- Department of Pharmaceutical Sciences, Mary Babb Randolph Cancer Center, West Virginia University, Morgantown, West Virginia 26506
| |
Collapse
|
183
|
Löffek S, Ullrich N, Görgens A, Murke F, Eilebrecht M, Menne C, Giebel B, Schadendorf D, Singer BB, Helfrich I. CEACAM1-4L Promotes Anchorage-Independent Growth in Melanoma. Front Oncol 2015; 5:234. [PMID: 26539411 PMCID: PMC4609850 DOI: 10.3389/fonc.2015.00234] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 10/05/2015] [Indexed: 11/13/2022] Open
Abstract
Widespread metastasis is the leading course of death in many types of cancer, including malignant melanoma. The process of metastasis can be divided into a number of complex cell biological events, collectively termed the “invasion-metastasis cascade.” Previous reports have characterized the capability of anchorage-independent growth of cancer cells in vitro as a key characteristic of highly aggressive tumor cells, particularly with respect to metastatic potential. Biological heterogeneity as well as drastic alterations in cell adhesion of disseminated cancer cells support escape mechanisms for metastases to overcome conventional therapies. Here, we show that exclusively the carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) splice variant CEACAM1-4L supports an anchorage-independent signature in malignant melanoma. These results highlight important variant-specific modulatory functions of CEACAM1 for metastatic spread in patients suffering malignant melanoma.
Collapse
Affiliation(s)
- Stefanie Löffek
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University of Duisburg-Essen , Essen , Germany ; German Cancer Consortium (DKTK), Medical Faculty, University of Duisburg-Essen , Essen , Germany
| | - Nico Ullrich
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University of Duisburg-Essen , Essen , Germany ; German Cancer Consortium (DKTK), Medical Faculty, University of Duisburg-Essen , Essen , Germany
| | - André Görgens
- Institute for Transfusion Medicine, Medical Faculty, University of Duisburg-Essen , Essen , Germany
| | - Florian Murke
- Institute for Transfusion Medicine, Medical Faculty, University of Duisburg-Essen , Essen , Germany
| | - Mara Eilebrecht
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University of Duisburg-Essen , Essen , Germany ; German Cancer Consortium (DKTK), Medical Faculty, University of Duisburg-Essen , Essen , Germany
| | - Christopher Menne
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University of Duisburg-Essen , Essen , Germany ; German Cancer Consortium (DKTK), Medical Faculty, University of Duisburg-Essen , Essen , Germany
| | - Bernd Giebel
- Institute for Transfusion Medicine, Medical Faculty, University of Duisburg-Essen , Essen , Germany
| | - Dirk Schadendorf
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University of Duisburg-Essen , Essen , Germany ; German Cancer Consortium (DKTK), Medical Faculty, University of Duisburg-Essen , Essen , Germany
| | - Bernhard B Singer
- Institute of Anatomy, Medical Faculty, University of Duisburg-Essen , Essen , Germany
| | - Iris Helfrich
- Skin Cancer Unit of the Dermatology Department, Medical Faculty, West German Cancer Center, University of Duisburg-Essen , Essen , Germany ; German Cancer Consortium (DKTK), Medical Faculty, University of Duisburg-Essen , Essen , Germany
| |
Collapse
|
184
|
Cockbill LMR, Murk K, Love S, Hanley JG. Protein interacting with C kinase 1 suppresses invasion and anchorage-independent growth of astrocytic tumor cells. Mol Biol Cell 2015; 26:4552-61. [PMID: 26466675 PMCID: PMC4678014 DOI: 10.1091/mbc.e15-05-0270] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 10/07/2015] [Indexed: 12/11/2022] Open
Abstract
Astrocytic tumors are the most common form of primary brain tumor. Astrocytic tumor cells infiltrate the surrounding CNS tissue, allowing them to evade removal upon surgical resection of the primary tumor. Dynamic changes to the actin cytoskeleton are crucial to cancer cell invasion, but the specific mechanisms that underlie the particularly invasive phenotype of astrocytic tumor cells are unclear. Protein interacting with C kinase 1 (PICK1) is a PDZ and BAR domain-containing protein that inhibits actin-related protein 2/3 (Arp2/3)-dependent actin polymerization and is involved in regulating the trafficking of a number of cell-surface receptors. Here we report that, in contrast to other cancers, PICK1 expression is down-regulated in grade IV astrocytic tumor cell lines and also in clinical cases of the disease in which grade IV tumors have progressed from lower-grade tumors. Exogenous expression of PICK1 in the grade IV astrocytic cell line U251 reduces their capacity for anchorage-independent growth, two-dimensional migration, and invasion through a three-dimensional matrix, strongly suggesting that low PICK1 expression plays an important role in astrocytic tumorigenesis. We propose that PICK1 negatively regulates neoplastic infiltration of astrocytic tumors and that manipulation of PICK1 is an attractive possibility for therapeutic intervention.
Collapse
Affiliation(s)
- Louisa M R Cockbill
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, United Kingdom
| | - Kai Murk
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, United Kingdom
| | - Seth Love
- School of Clinical Sciences, University of Bristol, Bristol BS10 5NB, United Kingdom
| | - Jonathan G Hanley
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, United Kingdom
| |
Collapse
|
185
|
In vitro study on the safety of near infrared laser therapy in its potential application as postmastectomy lymphedema treatment. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2015; 151:285-96. [PMID: 26355716 DOI: 10.1016/j.jphotobiol.2015.08.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 07/29/2015] [Accepted: 08/11/2015] [Indexed: 12/18/2022]
Abstract
Clinical studies demonstrated the effectiveness of laser therapy in the management of postmastectomy lymphedema, a discomforting disease that can arise after surgery/radiotherapy and gets progressively worse and chronic. However, safety issues restrict the possibility to treat cancer patients with laser therapy, since the effects of laser radiation on cancer cell behavior are not completely known and the possibility of activating postmastectomy residual cancer cells must be considered. This paper reports the results of an in vitro study aimed to investigate the effect of a class IV, dual-wavelength (808 nm and 905 nm), NIR laser system on the behavior of two human breast adenocarcinoma cell lines (namely, MCF7 and MDA-MB361 cell lines), using human dermal fibroblasts as normal control. Cell viability, proliferation, apoptosis, cell cycle and ability to form colonies were analyzed in order to perform a cell-based safety testing of the laser treatment in view of its potential application in the management of postmastectomy lymphedema. The results showed that, limited to the laser source, treatment conditions and experimental models used, laser radiation did not significantly affect the behavior of human breast adenocarcinoma cells, including their clonogenic efficiency. Although these results do not show any significant laser-induced modification of cancer cell behavior, further studies are needed to assess the possibility of safely applying NIR laser therapy for the management of postmastectomy lymphedema.
Collapse
|
186
|
Al-Shammari AM, Alshami MA, Umran MA, Almukhtar AA, Yaseen NY, Raad K, Hussien AA. Establishment and characterization of a receptor-negative, hormone-nonresponsive breast cancer cell line from an Iraqi patient. BREAST CANCER (DOVE MEDICAL PRESS) 2015; 7:223-30. [PMID: 26300657 PMCID: PMC4536763 DOI: 10.2147/bctt.s74509] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
A new breast cancer cell line (AMJ13) has been established from an Iraqi breast cancer patient. It is considered unique because it is the first for an Iraqi population, and is expected to be a useful tool in breast cancer research. The AMJ13 cell line was established from the primary tumor of a 70-year-old Iraqi woman with a histological diagnosis of infiltrating ductal carcinoma. The cells were morphologically characterized by light and scanning electron microscopy, and found to be elongated multipolar epithelial-like cells with a population doubling time of 22 hours. The anchorage-independent growth ability test showed that the cells were able to grow in semisolid agarose, confirming their transformed nature. Cytogenetic study of these cells showed chromosomal aberrations with many structural and numerical abnormalities, producing chromosomes of unknown origin called marker chromosomes. Immunocytochemistry showed that the estrogen receptor and the progesterone receptor were not expressed, and a weak positive result was found for HER2/neu gene expression. AMJ13 cells were positive for BRCA1 and BRCA2, as well as for vimentin. This cell line should be useful when testing new therapies for breast cancer in the Middle East.
Collapse
Affiliation(s)
- Ahmed Majeed Al-Shammari
- Experimental Therapy Department, Iraqi Center for Cancer and Medical Genetic Research, Mustansiriya University, Baghdad, Iraq
| | - Mortadha A Alshami
- Biotechnology Department, Collage of Science, Baghdad University, Baghdad, Iraq
| | | | - Asmaa Amer Almukhtar
- Medical Genetics Department, Iraqi Center for Cancer and Medical Genetic Research, Mustansiriya University, Baghdad, Iraq
| | - Nahi Y Yaseen
- Experimental Therapy Department, Iraqi Center for Cancer and Medical Genetic Research, Mustansiriya University, Baghdad, Iraq
| | - Khansaa Raad
- Experimental Therapy Department, Iraqi Center for Cancer and Medical Genetic Research, Mustansiriya University, Baghdad, Iraq
| | - Ayman A Hussien
- Experimental Therapy Department, Iraqi Center for Cancer and Medical Genetic Research, Mustansiriya University, Baghdad, Iraq
| |
Collapse
|
187
|
Kamada M, Mitsui Y, Matsuo T, Takahashi T. Reversible transformation and de-differentiation of human cells derived from induced pluripotent stem cell teratomas. Hum Cell 2015; 29:1-9. [PMID: 26069211 PMCID: PMC4705144 DOI: 10.1007/s13577-015-0119-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 05/26/2015] [Indexed: 11/29/2022]
Abstract
We first aimed to generate transformed cell lines from a human induced pluripotent stem cell (hiPSC)-teratoma, and then examined the tumorigenic risks of the differentiated cells from hiPSC explant, because hiPSC-derivatives give rise to tumors in immune-deficient mice when transplanted. The colonies isolated from sparse cultures of hiPSC-teratoma cells expressed NANOG and OCT3/4 strongly, and telomerase reverse transcriptase (TERT) weakly. However, soft agar assay demonstrated that only one of them generated colonies in the gel, though hiPSCs, hTERT-transfected immortal cells, and its oncogene-transfected cells did not form any colonies. Furthermore, none of colonies isolated from the soft agar gel on primary culture (passage 0) of teratoma cells, expressed NANOG and OCT3/4 in the expanded cultures. The second soft agar assay on the colony-derived cells was unexpectedly negative. The cumulative growth curve, telomere shortening, and senescence-associated β-galactosidase (SA β-gal) staining confirmed the mortality of these cells, suggesting their reversible transformation. By using medium for embryonic stem cell (ESC medium) after MCDB 131 (MCDB) medium, the differentiated culture cells derived from hiPSC-teratoma converted into the cells expressing undifferentiated marker proteins, which lost afterwords even in ESC medium with feeder SNL76/7. The reversibility of transformation and de-differentiation suggest that tumorigenic risks of differentiated cells arise when they are exposed to suitable niches in vivo. Thus, removal of only the undifferentiated cells from iPSC-derivatives before transplantation does not solve the problem. Elucidation of mechanisms of reversibility and control of epigenetic changes is discussed as a safety bottleneck for hiPSC therapy.
Collapse
Affiliation(s)
- Mizuna Kamada
- Laboratory of Physiological Chemistry, Faculty of Pharmaceutical Sciences at Kagawa, Tokushima Bunri University, 1314-1 Shido, Sanuki, Kagawa, 769-2193, Japan
| | - Youji Mitsui
- Laboratory of Physiological Chemistry, Faculty of Pharmaceutical Sciences at Kagawa, Tokushima Bunri University, 1314-1 Shido, Sanuki, Kagawa, 769-2193, Japan. .,Foundation for Advancement of International Science, Department of Research Development, Ibaraki, 305-0821, Japan.
| | - Taira Matsuo
- Laboratory of Physiological Chemistry, Faculty of Pharmaceutical Sciences at Kagawa, Tokushima Bunri University, 1314-1 Shido, Sanuki, Kagawa, 769-2193, Japan
| | - Tomoko Takahashi
- Laboratory of Physiological Chemistry, Faculty of Pharmaceutical Sciences at Kagawa, Tokushima Bunri University, 1314-1 Shido, Sanuki, Kagawa, 769-2193, Japan.
| |
Collapse
|
188
|
Thapa N, Choi S, Tan X, Wise T, Anderson RA. Phosphatidylinositol Phosphate 5-Kinase Iγ and Phosphoinositide 3-Kinase/Akt Signaling Couple to Promote Oncogenic Growth. J Biol Chem 2015; 290:18843-54. [PMID: 26070568 DOI: 10.1074/jbc.m114.596742] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Indexed: 11/06/2022] Open
Abstract
The assembly of signaling complexes at the plasma membrane is required for the initiation and propagation of cellular signaling upon cell activation. The class I PI3K and the serine/threonine-specific protein kinase Akt signaling pathways (PI3K/Akt) are often activated in tumors. These pathways are initiated by the generation of phosphatidylinositol 3,4,5-triphosphate (PIP3) by PI3K-mediated phosphorylation of phosphatidylinositol 4,5-biphosphate (PIP2), synthesized by phosphatidylinositol 4-phosphate 5-kinase (PIPKI) enzymes. The mechanism of how tumor cells recruit and organize the PIP2-synthesizing enzymes with PI3K in the plasma membrane for activation of PI3K/Akt signaling is not defined. Here, we demonstrated a role for the phosphatidylinositol 4-phosphate 5-kinase Iγ (PIPKIγ) in PI3K/Akt signaling. PIPKIγ is overexpressed in triple-negative breast cancers. Loss of PIPKIγ or its focal adhesion-targeting variant, PIPKIγi2, impaired PI3K/Akt activation upon stimulation with growth factors or extracellular matrix proteins in different tumor cells. PIPKIγi2 assembles into a complex containing Src and PI3K; Src was required for the recruitment of PI3K enzyme into the complex. PIPKIγi2 interaction with Src and its lipid kinase activity were required for promoting PI3K/Akt signaling. These results define a mechanism by which PIPKIγi2 and PI3K are integrated into a complex regulated by Src, resulting in the spatial generation of PIP2, which is the substrate PI3K required for PIP3 generation and subsequent Akt activation. This study elucidates the mechanism by which PIP2-generating enzyme controls Akt activation upstream of a PI3K enzyme. This pathway may represent a signaling nexus required for the survival and growth of metastasizing and circulating tumor cells in vivo.
Collapse
Affiliation(s)
- Narendra Thapa
- From the Molecular and Cellular Pharmacology Program, University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin 53706
| | - Suyong Choi
- From the Molecular and Cellular Pharmacology Program, University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin 53706
| | - Xiaojun Tan
- From the Molecular and Cellular Pharmacology Program, University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin 53706
| | - Thomas Wise
- From the Molecular and Cellular Pharmacology Program, University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin 53706
| | - Richard A Anderson
- From the Molecular and Cellular Pharmacology Program, University of Wisconsin, School of Medicine and Public Health, Madison, Wisconsin 53706
| |
Collapse
|
189
|
Ansong E, Ying Q, Ekoue DN, Deaton R, Hall AR, Kajdacsy-Balla A, Yang W, Gann PH, Diamond AM. Evidence that selenium binding protein 1 is a tumor suppressor in prostate cancer. PLoS One 2015; 10:e0127295. [PMID: 25993660 PMCID: PMC4436248 DOI: 10.1371/journal.pone.0127295] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Accepted: 04/13/2015] [Indexed: 12/25/2022] Open
Abstract
Selenium-Binding Protein 1 (SBP1, SELENBP1, hSP56) is a selenium-associated protein shown to be at lower levels in tumors, and its lower levels are frequently predictive of a poor clinical outcome. Distinguishing indolent from aggressive prostate cancer is a major challenge in disease management. Associations between SBP1 levels, tumor grade, and disease recurrence following prostatectomy were investigated by duplex immunofluorescence imaging using a tissue microarray containing tissue from 202 prostate cancer patients who experienced biochemical (PSA) recurrence after prostatectomy and 202 matched control patients whose cancer did not recur. Samples were matched by age, ethnicity, pathological stage and Gleason grade, and images were quantified using the Vectra multispectral imaging system. Fluorescent labels were targeted for SBP1 and cytokeratins 8/18 to restrict scoring to tumor cells, and cell-by-cell quantification of SBP1 in the nucleus and cytoplasm was performed. Nuclear SBP1 levels and the nuclear to cytoplasm ratio were inversely associated with tumor grade using linear regression analysis. Following classification of samples into quartiles based on the SBP1 levels among controls, tumors in the lowest quartile were more than twice as likely to recur compared to those in any other quartile. Inducible ectopic SBP1 expression reduced the ability of HCT-116 human tumor cells to grow in soft agar, a measure of transformation, without affecting proliferation. Cells expressing SBP1 also demonstrated a robust induction in the phosphorylation of the p53 tumor suppressor at serine 15. These data indicate that loss of SBP1 may play an independent contributing role in prostate cancer progression and its levels might be useful in distinguishing indolent from aggressive disease.
Collapse
Affiliation(s)
- Emmanuel Ansong
- Department of Pathology, University of Illinois at Chicago, Chicago, Illinois, United States of America
- * E-mail:
| | - Qi Ying
- Department of Pathology, University of Illinois at Chicago, Chicago, Illinois, United States of America
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, China
- Department of Pathology, Xinxiang Medical University, Xinxiang, China
| | - Dede N. Ekoue
- Department of Pathology, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Ryan Deaton
- Department of Pathology, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Andrew R. Hall
- University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Andre Kajdacsy-Balla
- Department of Pathology, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Wancai Yang
- Department of Pathology, University of Illinois at Chicago, Chicago, Illinois, United States of America
- Department of Pathology, Xinxiang Medical University, Xinxiang, China
| | - Peter H. Gann
- Department of Pathology, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Alan M. Diamond
- Department of Pathology, University of Illinois at Chicago, Chicago, Illinois, United States of America
| |
Collapse
|
190
|
Boeckx C, Blockx L, de Beeck KO, Limame R, Camp GV, Peeters M, Vermorken JB, Specenier P, Wouters A, Baay M, Lardon F. Establishment and characterization of cetuximab resistant head and neck squamous cell carcinoma cell lines: focus on the contribution of the AP-1 transcription factor. Am J Cancer Res 2015; 5:1921-1938. [PMID: 26269754 PMCID: PMC4529614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2015] [Accepted: 05/10/2015] [Indexed: 06/04/2023] Open
Abstract
BACKGROUND After an initial response to EGFR targeted therapy, secondary resistance almost invariably ensues, thereby limiting the clinical benefit of the drug. Hence, it has been recognized that the successful implementation of targeted therapy in the treatment of HNSCC cancer is very much dependent on predictive biomarkers for patient selection. METHODS We generated an in vitro model of acquired cetuximab resistance by chronically exposing three HNSCC cell lines to increasing cetuximab doses. Gene expression profiles of sensitive parental cells and resistant daughter cells were compared using microarray analysis. Growth inhibitory experiments were performed with an HB-EGF antibody and the MMP inhibitor, both in combination with cetuximab. Characteristics of EMT were analyzed using migration and invasion assays, immunofluorescent vimentin staining and qRT-PCR for several genes involved in this process. The function of the transcription factor AP-1 was investigated using qRT-PCR for several genes upregulated or downregulated in cetuximab resistant cells. Furthermore, anchorage-independent growth was investigated using the soft agar assay. RESULTS Gene expression profiling shows that cetuximab resistant cells upregulate several genes, including interleukin 8, the EGFR ligand HB-EGF and the metalloproteinase ADAM19. Cytotoxicity experiments with neutralizing HB-EGF antibody could not induce any growth inhibition, whereas an MMP inhibitor inhibited cell growth in cetuximab resistant cells. However, no synergetic effects combined with cetuximab could be observed. Cetuximab resistant cells showed traits of EMT, as witnessed by increased migratory potential, increased invasive potential, increased vimentine expression and increased expression of several genes involved in EMT. Furthermore, expression of upregulated genes could be repressed by the treatment with apigenin. The cetuximab resistant LICR-HN2 R10.3 cells tend to behave differently in cell culture, forming spheres. Therefore, soft agar assay was performed and showed more and larger colonies when challenged with cetuximab compared to PBS challenged cells. CONCLUSIONS In summary, our results indicate that increased expression of the ligand HB-EGF could contribute to resistance towards cetuximab in our cetuximab resistant HNSCC cells. Furthermore, several genes upregulated or downregulated in cetuximab resistant cells are under control of the AP-1 transcription factor. However, more studies are warranted to further unravel the role of AP-1 in cetuximab resistance.
Collapse
Affiliation(s)
- Carolien Boeckx
- Center for Oncological Research (CORE) Antwerp, Laboratory of Cancer Research and Clinical Oncology, University of AntwerpBelgium
| | - Lina Blockx
- Center for Oncological Research (CORE) Antwerp, Laboratory of Cancer Research and Clinical Oncology, University of AntwerpBelgium
| | - Ken Op de Beeck
- Center for Oncological Research (CORE) Antwerp, Laboratory of Cancer Research and Clinical Oncology, University of AntwerpBelgium
- Center for Medical Genetics, Department of Biomedical Sciences, University of AntwerpBelgium
| | - Ridha Limame
- Center for Oncological Research (CORE) Antwerp, Laboratory of Cancer Research and Clinical Oncology, University of AntwerpBelgium
| | - Guy Van Camp
- Center for Medical Genetics, Department of Biomedical Sciences, University of AntwerpBelgium
| | - Marc Peeters
- Center for Oncological Research (CORE) Antwerp, Laboratory of Cancer Research and Clinical Oncology, University of AntwerpBelgium
- Department of Medical Oncology, Antwerp University HospitalBelgium
| | - Jan B Vermorken
- Center for Oncological Research (CORE) Antwerp, Laboratory of Cancer Research and Clinical Oncology, University of AntwerpBelgium
- Department of Medical Oncology, Antwerp University HospitalBelgium
| | - Pol Specenier
- Center for Oncological Research (CORE) Antwerp, Laboratory of Cancer Research and Clinical Oncology, University of AntwerpBelgium
- Department of Medical Oncology, Antwerp University HospitalBelgium
| | - An Wouters
- Center for Oncological Research (CORE) Antwerp, Laboratory of Cancer Research and Clinical Oncology, University of AntwerpBelgium
| | - Marc Baay
- Center for Oncological Research (CORE) Antwerp, Laboratory of Cancer Research and Clinical Oncology, University of AntwerpBelgium
| | - Filip Lardon
- Center for Oncological Research (CORE) Antwerp, Laboratory of Cancer Research and Clinical Oncology, University of AntwerpBelgium
| |
Collapse
|
191
|
Ricciardelli C, Lokman NA, Cheruvu S, Tan IA, Ween MP, Pyragius CE, Ruszkiewicz A, Hoffmann P, Oehler MK. Transketolase is upregulated in metastatic peritoneal implants and promotes ovarian cancer cell proliferation. Clin Exp Metastasis 2015; 32:441-55. [PMID: 25895698 DOI: 10.1007/s10585-015-9718-1] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Accepted: 04/07/2015] [Indexed: 12/14/2022]
Abstract
Ovarian cancer, the most lethal gynaecological cancer, is characterised by the shedding of epithelial cells from the ovarian surface, followed by metastasis and implantation onto the peritoneal surfaces of abdominal organs. Our proteomic studies investigating the interactions between peritoneal (LP-9) and ovarian cancer (OVCAR-5) cells found transketolase (TKT) to be regulated in the co-culture system. This study characterized TKT expression in advanced stage (III/IV) serous ovarian cancers (n = 125 primary and n = 54 peritoneal metastases), normal ovaries (n = 6) and benign serous cystadenomas (n = 10) by immunohistochemistry. In addition, we also evaluated the function of TKT in ovarian cancer cells in vitro. Nuclear TKT was present in all primary serous ovarian cancer tissues examined (median 82.0 %, range 16.5-100 %) and was significantly increased in peritoneal metastases compared with matching primary cancers (P = 0.01, Wilcoxon Rank test). Kaplan-Meier survival and Cox regression analyses showed that high nuclear TKT positivity in peritoneal metastases (>94 %) was significantly associated with reduced overall survival (P = 0.006) and a 2.8 fold increased risk of ovarian cancer death (95 % CI 1.29-5.90, P = 0.009). Knockdown of TKT by siRNAs significantly reduced SKOV-3 cell proliferation but had no effect on their motility or invasion. Oxythiamine, an inhibitor of TKT activity, significantly inhibited the proliferation of four ovarian cancer cell lines (OV-90, SKOV-3, OVCAR-3 and OVCAR-5) and primary serous ovarian cancer cells isolated from patient ascites. In conclusion, these findings indicate that TKT plays an important role in the proliferation of metastatic ovarian cancer cells and could be used as novel therapeutic target for advanced disease.
Collapse
Affiliation(s)
- Carmela Ricciardelli
- Discipline of Obstetrics and Gynaecology, School of Paediatrics and Reproductive Health, Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia,
| | | | | | | | | | | | | | | | | |
Collapse
|
192
|
An J, Guo Y, Wang T, Pantuck AJ, Rettig MB. Pomegranate extract inhibits EMT in clear cell renal cell carcinoma in a NF-κB and JNK dependent manner. Asian J Urol 2015; 2:38-45. [PMID: 29264118 PMCID: PMC5730684 DOI: 10.1016/j.ajur.2015.04.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 08/19/2014] [Accepted: 08/29/2014] [Indexed: 01/06/2023] Open
Abstract
Objective Clear cell renal cell carcinoma (ccRCC) is the most common subtype of renal cell carcinoma (RCC) and is characterized by biallelic inactivation of the von Hippel-Lindau (VHL) tumor suppressor gene. One effect of VHL inactivation is hypoxia inducible factor alpha (HIFα)-independent constitutive activation of nuclear factor kappa B (NF-κB) and c-jun N-terminal kinase (JNK). Both NF-κB and JNK drive ccRCC growth and epithelial to mesenchymal transition (EMT). The purpose of this study was to determine the biochemical effects of pomegranate juice extracts (PE) on RCC cell lines. Methods The pre-clinical effects of PE on NF-κB, JNK, and the EMT phenotype were assayed, including its effect on proliferation, anchorage-independent growth, and invasion of pVHL-deficient RCCs. Results PE inhibits the NF-κB and JNK pathways and consequently inhibits the EMT phenotype of pVHL-deficient ccRCCs. The effects of PE are concentration-dependent and affect not only biochemical markers of EMT (i.e., cadherin expression) but also functional manifestations of EMT, such as invasion. These effects are manifested within days of exposure to PE when diluted 2000-fold. Highly dilute concentrations of PE (106 dilution), which do not impact these pathways in the short term, were found to have NF-κB and JNK inhibitory effects and ability to reverse the EMT phenotype following prolonged exposure. Conclusion These findings suggest that PE may mediate inhibition growth of pVHL-deficient ccRCCs and raises the possibility of its use as a dietary adjunct to managing patients with active surveillance for small, localized, incidentally identified renal tumors so as to avoid more invasive procedures such as nephrectomy.
Collapse
Affiliation(s)
- Jiabin An
- Department of Medicine, Division of Hematology-Oncology, VA Greater Los Angeles Healthcare System, Los Angeles, CA, 90073, USA
| | - Yanchuan Guo
- Technical Institute of Physics and Chemistry, Chinese Academy of Science, Beijing, 100190, China
| | - Ting Wang
- Department of Medicine, Division of Hematology-Oncology, VA Greater Los Angeles Healthcare System, Los Angeles, CA, 90073, USA
| | - Allan J Pantuck
- Department of Urology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Matthew B Rettig
- Department of Medicine, Division of Hematology-Oncology, VA Greater Los Angeles Healthcare System, Los Angeles, CA, 90073, USA.,Department of Urology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA.,Department of Medicine, Division of Hematology-Oncology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| |
Collapse
|
193
|
Briehl MM. Oxygen in human health from life to death--An approach to teaching redox biology and signaling to graduate and medical students. Redox Biol 2015; 5:124-139. [PMID: 25912168 PMCID: PMC4412967 DOI: 10.1016/j.redox.2015.04.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 04/08/2015] [Indexed: 02/07/2023] Open
Abstract
In the absence of oxygen human life is measured in minutes. In the presence of oxygen, normal metabolism generates reactive species (ROS) that have the potential to cause cell injury contributing to human aging and disease. Between these extremes, organisms have developed means for sensing oxygen and ROS and regulating their cellular processes in response. Redox signaling contributes to the control of cell proliferation and death. Aberrant redox signaling underlies many human diseases. The attributes acquired by altered redox homeostasis in cancer cells illustrate this particularly well. This teaching review and the accompanying illustrations provide an introduction to redox biology and signaling aimed at instructors of graduate and medical students. The ability to sense oxygen and respond to oxidative stress is ancient. Chemical and kinetic properties of ROS are key to understanding redox signaling. Redox signaling participates in normal control of cell proliferation and death. Aberrant redox signaling contributes to the hallmarks of cancer. Novel redox-based chemotherapeutics are being developed.
Collapse
Affiliation(s)
- Margaret M Briehl
- Department of Pathology, University of Arizona, PO Box 24-5043, Tucson, AZ 85724-5043, USA.
| |
Collapse
|
194
|
Foley JM, Scholten DJ, Monks NR, Cherba D, Monsma DJ, Davidson P, Dylewski D, Dykema K, Winn ME, Steensma MR. Anoikis-resistant subpopulations of human osteosarcoma display significant chemoresistance and are sensitive to targeted epigenetic therapies predicted by expression profiling. J Transl Med 2015; 13:110. [PMID: 25889105 PMCID: PMC4419490 DOI: 10.1186/s12967-015-0466-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 03/17/2015] [Indexed: 01/27/2023] Open
Abstract
Background Osteosarcoma (OS) is the most common type of solid bone cancer, with latent metastasis being a typical mode of disease progression and a major contributor to poor prognosis. For this to occur, cells must resist anoikis and be able to recapitulate tumorigenesis in a foreign microenvironment. Finding novel approaches to treat osteosarcoma and target those cell subpopulations that possess the ability to resist anoikis and contribute to metastatic disease is imperative. Here we investigate anchorage-independent (AI) cell growth as a model to better characterize anoikis resistance in human osteosarcoma while using an expression profiling approach to identify and test targetable signaling pathways. Methods Established human OS cell lines and patient-derived human OS cell isolates were subjected to growth in either adherent or AI conditions using Ultra-Low Attachment plates in identical media conditions. Growth rate was assessed using cell doubling times and chemoresistance was assessed by determining cell viability in response to a serial dilution of either doxorubicin or cisplatin. Gene expression differences were examined using quantitative reverse-transcription PCR and microarray with principal component and pathway analysis. In-vivo OS xenografts were generated by either subcutaneous or intratibial injection of adherent or AI human OS cells into athymic nude mice. Statistical significance was determined using student’s t-tests with significance set at α = 0.05. Results We show that AI growth results in a global gene expression profile change accompanied by significant chemoresistance (up to 75 fold, p < 0.05). AI cells demonstrate alteration of key mediators of mesenchymal differentiation (β-catenin, Runx2), stemness (Sox2), proliferation (c-myc, Akt), and epigenetic regulation (HDAC class 1). AI cells were equally tumorigenic as their adherent counterparts, but showed a significantly decreased rate of growth in-vitro and in-vivo (p < 0.05). Treatment with the pan-histone deacetylase inhibitor vorinostat and the DNA methyltransferase inhibitor 5-azacytidine mitigated AI growth, while 5-azacytidine sensitized anoikis-resistant cells to doxorubicin (p < 0.05). Conclusions These data demonstrate remarkable plasticity in anoikis-resistant human osteosarcoma subpopulations accompanied by a rapid development of chemoresistance and altered growth rates mirroring the early stages of latent metastasis. Targeting epigenetic regulation of this process may be a viable therapeutic strategy. Electronic supplementary material The online version of this article (doi:10.1186/s12967-015-0466-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jessica M Foley
- Helen DeVos Children's Hospital, Spectrum Health System, Grand Rapids, MI, USA.
| | - Donald J Scholten
- Van Andel Research Institute, Grand Rapids, MI, USA. .,Michigan State University College of Human Medicine, Grand Rapids, MI, USA.
| | - Noel R Monks
- Van Andel Research Institute, Grand Rapids, MI, USA.
| | - David Cherba
- Van Andel Research Institute, Grand Rapids, MI, USA.
| | | | | | | | - Karl Dykema
- Van Andel Research Institute, Grand Rapids, MI, USA.
| | - Mary E Winn
- Van Andel Research Institute, Grand Rapids, MI, USA.
| | - Matthew R Steensma
- Helen DeVos Children's Hospital, Spectrum Health System, Grand Rapids, MI, USA. .,Van Andel Research Institute, Grand Rapids, MI, USA. .,Michigan State University College of Human Medicine, Grand Rapids, MI, USA.
| |
Collapse
|
195
|
Nasti TH, Cochran JB, Tsuruta Y, Yusuf N, McKay KM, Athar M, Timares L, Elmets CA. A murine model for the development of melanocytic nevi and their progression to melanoma. Mol Carcinog 2015; 55:646-58. [PMID: 25788145 PMCID: PMC4575238 DOI: 10.1002/mc.22310] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 01/22/2015] [Accepted: 02/06/2015] [Indexed: 01/25/2023]
Abstract
Acquired melanocytic nevi are commonly found in sun exposed and unexposed human skin, but the potential for their transformation into invasive melanoma is not clear. Therefore, a mouse model of nevus initiation and progression was developed in C3H/HeN mice using a modified chemical carcinogenesis protocol. Nevi develop due to DNA damage initiated by dimethylbenz(a) anthracene (DMBA) followed by chronic promotion with 12‐O‐tetradecanoyl‐phorbol‐13‐acetate (TPA). Dysplastic pigmented skin lesions appeared in 7–9 wk with 100% penetrance. Nests of melanocytic cells appeared in a subset of skin draining lymph nodes (dLN) by 25 wk, but not in age matched controls. Immunohistochemistry, real‐time PCR, and flow cytometric analyses confirmed their melanocytic origin. Transformed cells were present in a subset of nevi and dLNs, which exhibited anchorage‐independent growth, tumor development, and metastasis in nude mice. Approximately 50% of the cell lines contained H‐Ras mutations and lost tumor suppressor p16Ink4a expression. While most studies of melanoma focus on tumor progression in transgenic mouse models where the mutations are present from birth, our model permits investigation of acquired mutations at the earliest stages of nevus initiation and promotion of nevus cell transformation. This robust nevus/melanoma model may prove useful for identifying genetic loci associated with nevus formation, novel oncogenic pathways, tumor targets for immune‐prevention, screening therapeutics, and elucidating mechanisms of immune surveillance and immune evasion. © 2015 The Authors. Molecular Carcinogenesis, published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Tahseen H Nasti
- The Department of Dermatology, The University of Alabama at Birmingham School of Medicine, Birmingham, Alabama
| | - J Barry Cochran
- The Department of Dermatology, The University of Alabama at Birmingham School of Medicine, Birmingham, Alabama
| | - Yuko Tsuruta
- The Department of Dermatology, The University of Alabama at Birmingham School of Medicine, Birmingham, Alabama.,The Skin Diseases Research Center, and The University of Alabama at Birmingham School of Medicine, Birmingham, Alabama
| | - Nabiha Yusuf
- The Department of Dermatology, The University of Alabama at Birmingham School of Medicine, Birmingham, Alabama.,The Skin Diseases Research Center, and The University of Alabama at Birmingham School of Medicine, Birmingham, Alabama.,The Birmingham VA Medical Center, Birmingham, Alabama
| | - Kristopher M McKay
- The Department of Dermatology, The University of Alabama at Birmingham School of Medicine, Birmingham, Alabama
| | - Mohammad Athar
- The Department of Dermatology, The University of Alabama at Birmingham School of Medicine, Birmingham, Alabama.,The Skin Diseases Research Center, and The University of Alabama at Birmingham School of Medicine, Birmingham, Alabama
| | - Laura Timares
- The Department of Dermatology, The University of Alabama at Birmingham School of Medicine, Birmingham, Alabama.,The Skin Diseases Research Center, and The University of Alabama at Birmingham School of Medicine, Birmingham, Alabama.,The Birmingham VA Medical Center, Birmingham, Alabama
| | - Craig A Elmets
- The Department of Dermatology, The University of Alabama at Birmingham School of Medicine, Birmingham, Alabama.,The Skin Diseases Research Center, and The University of Alabama at Birmingham School of Medicine, Birmingham, Alabama.,The Birmingham VA Medical Center, Birmingham, Alabama
| |
Collapse
|
196
|
Drewe J, Bucher KA, Zahner C. A systematic review of non-hormonal treatments of vasomotor symptoms in climacteric and cancer patients. SPRINGERPLUS 2015; 4:65. [PMID: 25713759 PMCID: PMC4331402 DOI: 10.1186/s40064-015-0808-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 01/09/2015] [Indexed: 12/03/2022]
Abstract
The cardinal climacteric symptoms of hot flushes and night sweats affect 24-93% of all women during the physiological transition from reproductive to post-reproductive life. Though efficacious, hormonal therapy and partial oestrogenic compounds are linked to a significant increase in breast cancer. Non-hormonal treatments are thus greatly appreciated. This systematic review of published hormonal and non-hormonal treatments for climacteric, and breast and prostate cancer-associated hot flushes, examines clinical efficacy and therapy-related cancer risk modulation. A PubMed search included literature up to June 19, 2014 without limits for initial dates or language, with the search terms, (hot flush* OR hot flash*) AND (clinical trial* OR clinical stud*) AND (randomi* OR observational) NOT review). Retrieved references identified further papers. The focus was on hot flushes; other symptoms (night sweats, irritability, etc.) were not specifically screened. Included were some 610 clinical studies where a measured effect of the intervention, intensity and severity were documented, and where patients received treatment of pharmaceutical quality. Only 147 of these references described studies with alternative non-hormonal treatments in post-menopausal women and in breast and prostate cancer survivors; these results are presented in Additional file 1. The most effective hot flush treatment is oestrogenic hormones, or a combination of oestrogen and progestins, though benefits are partially outweighed by a significantly increased risk for breast cancer development. This review illustrates that certain non-hormonal treatments, including selective serotonin reuptake inhibitors, gabapentin/pregabalin, and Cimicifuga racemosa extracts, show a positive risk-benefit ratio. Key pointsSeveral non-hormonal alternatives to hormonal therapy have been established and registered for the treatment of vasomotor climacteric symptoms in peri- and post-menopausal women. There are indications that non-hormonal treatments are useful alternatives in patients with a history of breast and prostate cancer. However, confirmation by larger clinical trials is required.
Collapse
Affiliation(s)
- Juergen Drewe
- Max Zeller AG, Seeblickstr. 4, 8590 Romanshorn, Switzerland
| | | | | |
Collapse
|
197
|
Guo Y, Shu L, Zhang C, Su ZY, Kong ANT. Curcumin inhibits anchorage-independent growth of HT29 human colon cancer cells by targeting epigenetic restoration of the tumor suppressor gene DLEC1. Biochem Pharmacol 2015; 94:69-78. [PMID: 25640947 DOI: 10.1016/j.bcp.2015.01.009] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 01/13/2015] [Accepted: 01/21/2015] [Indexed: 01/25/2023]
Abstract
Colorectal cancer remains the most prevalent malignancy in humans. The impact of epigenetic alterations on the development of this complex disease is now being recognized. The dynamic and reversible nature of epigenetic modifications makes them a promising target in colorectal cancer chemoprevention and treatment. Curcumin (CUR), the major component in Curcuma longa, has been shown as a potent chemopreventive phytochemical that modulates various signaling pathways. Deleted in lung and esophageal cancer 1 (DLEC1) is a tumor suppressor gene with reduced transcriptional activity and promoter hypermethylation in various cancers, including colorectal cancer. In the present study, we aimed to investigate the inhibitory role of DLEC1 in anchorage-independent growth of the human colorectal adenocarcinoma HT29 cells and epigenetic regulation by CUR. Specifically, we found that CUR treatment inhibited colony formation of HT29 cells, whereas stable knockdown of DLEC1 using lentiviral short hairpin RNA vector increased cell proliferation and colony formation. Knockdown of DLEC1 in HT29 cells attenuated the ability of CUR to inhibit anchorage-independent growth. Methylation-specific polymerase chain reaction (MSP), bisulfite genomic sequencing, and methylated DNA immunoprecipitation revealed that CUR decreased CpG methylation of the DLEC1 promoter in HT29 cells after 5 days of treatment, corresponding to increased mRNA expression of DLEC1. Furthermore, CUR decreased the protein expression of DNA methyltransferases and subtypes of histone deacetylases (HDAC4, 5, 6, and 8). Taken together, our results suggest that the inhibitory effect of CUR on anchorage-independent growth of HT29 cells could, at least in part, involve the epigenetic demethylation and up-regulation of DLEC1.
Collapse
Affiliation(s)
- Yue Guo
- Graduate Program in Pharmaceutical Sciences, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA; Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Room 228, 160 Frelinghuysen Road, Piscataway, NJ 08854, USA
| | - Limin Shu
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Room 228, 160 Frelinghuysen Road, Piscataway, NJ 08854, USA
| | - Chengyue Zhang
- Graduate Program in Pharmaceutical Sciences, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA; Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Room 228, 160 Frelinghuysen Road, Piscataway, NJ 08854, USA
| | - Zheng-Yuan Su
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Room 228, 160 Frelinghuysen Road, Piscataway, NJ 08854, USA
| | - Ah-Ng Tony Kong
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Room 228, 160 Frelinghuysen Road, Piscataway, NJ 08854, USA.
| |
Collapse
|
198
|
Abstract
PARP-1 is a nuclear enzyme regulating transcription, chromatin restructuring, and DNA repair. PARP-1 is activated by interaction with NAD+, DNA, and core histones. Each route of PARP-1 activation leads to somewhat different outcomes. PARP-1 interactions with core histones control PARP-1 functions during transcriptional activation in euchromatin. DNA-dependent regulation of PARP-1 determines its localization in heterochromatin and PARP-1-dependent silencing. Here we address the biological significance of DNA-dependent PARP-1 regulation in vitro and in vivo. We report that minor grove binding ligands (MGBLs) specifically target PARP-1 interaction with DNA, and, hence, the DNA-dependent pathway of PARP-1 activation. By obstructing its interaction with DNA molecules, MGBLs block PARP-1 activity in vitro and in vivo, as we demonstrate using Drosophila, as well as human cancer-derived cells. We also demonstrate synergistic inhibition of PARP-1, combining MGBLs with conventional NAD+-dependent inhibitors in human cancer cells. These results suggest that combining different classes of PARP-1 inhibitors can precisely modulate PARP-1 activity in living cells, thus holding promise for new avenues of cancer treatment.
Collapse
|
199
|
An J, Guo Y, Wang T, Pantuck AJ, Rettig MB. WITHDRAWN: Pomegranate extract inhibits EMT in clear cell renal cell carcinoma in a NF-κB and JNK dependent manner. Asian J Urol 2014. [DOI: 10.1016/j.ajur.2014.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
200
|
Wang C, Tseng T, Jhang Y, Tseng J, Hsieh C, Wu WG, Lee S. Loss of cell invasiveness through PKC-mediated syndecan-1 downregulation in melanoma cells under anchorage independency. Exp Dermatol 2014; 23:843-9. [DOI: 10.1111/exd.12550] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/15/2014] [Indexed: 12/26/2022]
Affiliation(s)
- ChiaChen Wang
- School of Medicine; FuJen Catholic University; New Taipei City Taiwan
- Department of Dermatology; Cardinal Tien Hospital; New Taipei City Taiwan
| | - TingTing Tseng
- School of Medicine; FuJen Catholic University; New Taipei City Taiwan
| | - Yaoyun Jhang
- School of Medicine; FuJen Catholic University; New Taipei City Taiwan
| | - JenChih Tseng
- School of Medicine; FuJen Catholic University; New Taipei City Taiwan
| | - ChiaoHui Hsieh
- School of Medicine; FuJen Catholic University; New Taipei City Taiwan
| | - Wen-guey Wu
- Department of Life Sciences and Institute of Bioinformatics and Structural Biology; National TsingHua University; Hsinchu Taiwan
| | - ShaoChen Lee
- School of Medicine; FuJen Catholic University; New Taipei City Taiwan
| |
Collapse
|