51
|
Saindane M, Rallabandi HR, Park KS, Heil A, Nam SE, Yoo YB, Yang JH, Yun IJ. Prognostic Significance of Prostaglandin-Endoperoxide Synthase-2 Expressions in Human Breast Carcinoma: A Multiomic Approach. Cancer Inform 2020; 19:1176935120969696. [PMID: 33223820 PMCID: PMC7656875 DOI: 10.1177/1176935120969696] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 10/03/2020] [Indexed: 01/24/2023] Open
Abstract
Prostaglandin-endoperoxide synthase-2 (PTGS2) plays a pivotal role in inflammation and carcinogenesis in human breast cancer. Our aim of the study is to find the prognostic value of PTGS2 in breast cancer. We conducted a multiomic analysis to determine whether PTGS2 functions as a prognostic biomarker in human breast cancer. We explored PTGS2 mRNA expressions using different public bioinformatics portals. Oncomine, Serial Analysis of Gene Expression (SAGE), GEPIA, ULCAN, PrognoScan database, Kaplan-Meier Plotter, bc-GenExMiner, USC XENA, and Cytoscape/STRING DB were used to identify the prognostic roles of PTGS2 in breast cancer. Based on the clinicopathological analysis, decreased PTGS2 expressions correlated positively with older age, lymph node status, the human epidermal growth factor receptor 2 (HER2) status (P < .0001), estrogen receptor (ER+) expression (P < .0001) Luminal A (P < .0001), and Luminal B (P < .0001). Interestingly, progesterone receptor (PR) (P < .0001) negative showed a high expression of PTGS2. Prostaglandin-endoperoxide synthase-2 was downregulated in breast cancer tissues than in normal tissues. In the PrognoScan database and, Kaplan-Meier Scanner, downregulated expressions of PTGS2 associated with poor overall survival (OS), relapse-free survival (RFS), and distant metastasis-free survival. The methylation levels were significantly higher in the Luminal B subtype. Through oncomine coexpressed gene analysis, we found a positive correlation between PTGS2 and interleukin-6 (IL-6) expression in breast cancer tissues. These results indicate that downregulated expressions of PTGS2 can be used as a promising prognostic biomarker and Luminal B hyper methylation may play an important role in the development of breast cancers. However, to clarify our results, extensive study is required.
Collapse
Affiliation(s)
| | | | - Kyoung Sik Park
- Konkuk University, School of Medicine, Seoul, South Korea.,Research Institute of Medical Science, School of Medicine, Konkuk University, Seoul, South Korea
| | - Alexander Heil
- Institute of Botany and Molecular Genetics, RWTH Aachen University, Aachen, Germany
| | - Sang Eun Nam
- Konkuk University, School of Medicine, Seoul, South Korea.,Research Institute of Medical Science, School of Medicine, Konkuk University, Seoul, South Korea
| | - Young Bum Yoo
- Konkuk University, School of Medicine, Seoul, South Korea.,Research Institute of Medical Science, School of Medicine, Konkuk University, Seoul, South Korea
| | - Jung-Hyun Yang
- Konkuk University, School of Medicine, Seoul, South Korea.,Research Institute of Medical Science, School of Medicine, Konkuk University, Seoul, South Korea
| | - Ik Jin Yun
- Konkuk University, School of Medicine, Seoul, South Korea.,Research Institute of Medical Science, School of Medicine, Konkuk University, Seoul, South Korea
| |
Collapse
|
52
|
Altered Levels of Desaturation and ω-6 Fatty Acids in Breast Cancer Patients' Red Blood Cell Membranes. Metabolites 2020; 10:metabo10110469. [PMID: 33212920 PMCID: PMC7698438 DOI: 10.3390/metabo10110469] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 11/06/2020] [Accepted: 11/12/2020] [Indexed: 02/07/2023] Open
Abstract
Red blood cell (RBC) membrane can reflect fatty acid (FA) contribution from diet and biosynthesis. In cancer, membrane FAs are involved in tumorigenesis and invasiveness, and are indicated as biomarkers to monitor the disease evolution as well as potential targets for therapies and nutritional strategies. The present study provides RBC membrane FA profiles in recently diagnosed breast cancer patients before starting chemotherapy treatment. Patients and controls were recruited, and their dietary habits were collected. FA lipidomic analysis of mature erythrocyte membrane phospholipids in blood samples was performed. Data were adjusted to correct for the effects of diet, body mass index (BMI), and age, revealing that patients showed lower levels of saturated fatty acids (SFA) and higher levels of monounsaturated fatty acid, cis-vaccenic (25%) than controls, with consequent differences in desaturase enzymatic index (∆9 desaturase, -13.1%). In the case of polyunsaturated fatty acids (PUFA), patients had higher values of ω-6 FA (C18:2 (+11.1%); C20:4 (+7.4%)). RBC membrane lipidomic analysis in breast cancer revealed that ω-6 pathways are favored. These results suggest new potential targets for treatments and better nutritional guidelines.
Collapse
|
53
|
Zhou H, Yan Y, Zhang X, Zhao T, Xu J, Han R. Ginseng polysaccharide inhibits MDA-MB-231 cell proliferation by activating the inflammatory response. Exp Ther Med 2020; 20:229. [PMID: 33149784 PMCID: PMC7604739 DOI: 10.3892/etm.2020.9359] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 06/26/2020] [Indexed: 12/16/2022] Open
Abstract
Ginseng polysaccharide (GPS) is known for its efficacy in cancer therapy; however, its regulatory mechanism in breast cancer (BC) remains unclear. To analyze the effect of GPS on BC cell proliferation, cell proliferation rate calculations, western blotting, plasmid transfections, electrophoretic mobility shift assays and chromatin immunoprecipitation assays were performed. GPS treatment in the culture cell medium inhibited cell proliferation in the BC cell line MDA-MB-231. In addition, the E-cadherin level was enhanced while the vimentin level was suppressed following GPS treatment (both P<0.05). Furthermore, the levels of apoptotic markers, including cleaved-Caspase-3 and p53, and inflammatory response markers, including plasminogen activator inhibitor and TNF-α, were induced by GPS treatment in MDA-MB-231 cells (all P<0.05). These results indicated that GPS supplementation activated the inflammatory response and apoptosis in BC cells. GPS treatment activated the phosphorylation levels of c-Jun N-terminal kinase, Akt and NF-κB. In MDA-MB-231 cells, GPS resulted in the accumulation of the NF-κB components p65, p50 and Ikaros family zing finger protein 1 (IKZF1; all, P<0.05). Chromatin immunoprecipitation and electrophoretic mobility shift assays indicated that p65 bound to the IKZF1 promoter. The overexpression of IKZF1 or p65 inhibited MDA-MB-231 cell proliferation (P<0.05), indicating that GPS treatment may inhibit BC cell proliferation by the activation of IKZF1. Taken together, these results suggested that GPS significantly inhibited BC cell proliferation via the control of the biological processes, including the activation of p65-IKZF1 signaling and apoptosis. The data indicated a novel mechanism for further understanding of cancer cell proliferation.
Collapse
Affiliation(s)
- Haoliang Zhou
- Department of Oncology, Wenzhou Central Hospital, Wenzhou, Zhejiang 325000, P.R. China
| | - Yuxiang Yan
- Department of Oncology, Wenzhou Central Hospital, Wenzhou, Zhejiang 325000, P.R. China
| | - Xianbo Zhang
- Department of Oncology, Wenzhou Central Hospital, Wenzhou, Zhejiang 325000, P.R. China
| | - Ting Zhao
- Department of Oncology, Wenzhou Central Hospital, Wenzhou, Zhejiang 325000, P.R. China
| | - Jiangang Xu
- Department of Oncology, Wenzhou Central Hospital, Wenzhou, Zhejiang 325000, P.R. China
| | - Ruokuo Han
- Department of Oncology, Wenzhou Central Hospital, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
54
|
Lappano R, Talia M, Cirillo F, Rigiracciolo DC, Scordamaglia D, Guzzi R, Miglietta AM, De Francesco EM, Belfiore A, Sims AH, Maggiolini M. The IL1β-IL1R signaling is involved in the stimulatory effects triggered by hypoxia in breast cancer cells and cancer-associated fibroblasts (CAFs). JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:153. [PMID: 32778144 PMCID: PMC7418191 DOI: 10.1186/s13046-020-01667-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 08/04/2020] [Indexed: 12/11/2022]
Abstract
Background Hypoxia plays a relevant role in tumor-related inflammation toward the metastatic spread and cancer aggressiveness. The pro-inflammatory cytokine interleukin-1β (IL-β) and its cognate receptor IL1R1 contribute to the initiation and progression of breast cancer determining pro-tumorigenic inflammatory responses. The transcriptional target of the hypoxia inducible factor-1α (HIF-1α) namely the G protein estrogen receptor (GPER) mediates a feedforward loop coupling IL-1β induction by breast cancer-associated fibroblasts (CAFs) to IL1R1 expression by breast cancer cells toward the regulation of target genes and relevant biological responses. Methods In order to ascertain the correlation of IL-β with HIF-1α and further hypoxia-related genes in triple-negative breast cancer (TNBC) patients, a bioinformatics analysis was performed using the information provided by The Invasive Breast Cancer Cohort of The Cancer Genome Atlas (TCGA) project and Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) datasets. Gene expression correlation, statistical analysis and gene set enrichment analysis (GSEA) were carried out with R studio packages. Pathway enrichment analysis was evaluated with Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway. TNBC cells and primary CAFs were used as model system. The molecular mechanisms implicated in the regulation of IL-1β by hypoxia toward a metastatic gene expression profile and invasive properties were assessed performing gene and protein expression studies, PCR arrays, gene silencing and immunofluorescence analysis, co-immunoprecipitation and ChiP assays, ELISA, cell spreading, invasion and spheroid formation. Results We first determined that IL-1β expression correlates with the levels of HIF-1α as well as with a hypoxia-related gene signature in TNBC patients. Next, we demonstrated that hypoxia triggers a functional liaison among HIF-1α, GPER and the IL-1β/IL1R1 signaling toward a metastatic gene signature and a feed-forward loop of IL-1β that leads to proliferative and invasive responses in TNBC cells. Furthermore, we found that the IL-1β released in the conditioned medium of TNBC cells exposed to hypoxic conditions promotes an invasive phenotype of CAFs. Conclusions Our data shed new light on the role of hypoxia in the activation of the IL-1β/IL1R1 signaling, which in turn triggers aggressive features in both TNBC cells and CAFs. Hence, our findings provide novel evidence regarding the mechanisms through which the hypoxic tumor microenvironment may contribute to breast cancer progression and suggest further targets useful in more comprehensive therapeutic strategies.
Collapse
Affiliation(s)
- Rosamaria Lappano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Rende, Italy.
| | - Marianna Talia
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Rende, Italy
| | - Francesca Cirillo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Rende, Italy
| | | | - Domenica Scordamaglia
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Rende, Italy
| | - Rita Guzzi
- Department of Physics, University of Calabria, 87036, Rende, Italy
| | | | - Ernestina Marianna De Francesco
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, 95122, Catania, Italy
| | - Antonino Belfiore
- Endocrinology, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, 95122, Catania, Italy
| | - Andrew H Sims
- MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XR, UK
| | - Marcello Maggiolini
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Rende, Italy.
| |
Collapse
|
55
|
Sherman ME, Vierkant RA, Kaggal S, Hoskin TL, Frost MH, Denison L, Visscher DW, Carter JM, Winham SJ, Jensen MR, Radisky DC, Vachon CM, Degnim AC. Breast Cancer Risk and Use of Nonsteroidal Anti-inflammatory Agents After a Benign Breast Biopsy. Cancer Prev Res (Phila) 2020; 13:967-976. [PMID: 32718942 PMCID: PMC9509660 DOI: 10.1158/1940-6207.capr-20-0178] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/04/2020] [Accepted: 07/21/2020] [Indexed: 11/16/2022]
Abstract
Over one million women in the United States receive biopsy diagnoses of benign breast disease (BBD) each year, which confer a 1.5-4.0-fold increase in breast cancer risk. Studies in the general population suggest that nonsteroidal anti-inflammatory agents (NSAID) lower breast cancer risk; however, associations among women with BBD are unknown. We assessed whether NSAID use among women diagnosed with BBD is associated with lower breast cancer risk. Participants included 3,080 women (mean age = 50.3 ± 13.5 years) in the Mayo BBD surgical biopsy cohort diagnosed between January 1, 1992 and December 31, 2001 who completed breast cancer risk factor questionnaires that assessed NSAID use, and whose biopsies underwent detailed pathology review, masked to outcome. Women were followed from date of BBD biopsy to breast cancer diagnosis (main outcome) or censoring (death, prophylactic mastectomy, reduction mammoplasty, lobular carcinoma in situ or last contact). Median follow-up time was 16.4 ± 6.0 years. Incident breast cancer was diagnosed among 312 women over a median follow-up of 9.9 years. Regular non-aspirin NSAID use was associated with lower breast cancer risk [HR = 0.63; 95% confidence interval (CI) = 0.46-0.85; P = 0.002] with trends of lower risk (highest tertiles of use vs. nonuse) for greater number of years used [HR = 0.55; 95% CI = 0.31-0.97; P trend = 0.003), days used per month (HR = 0.51; 95% CI = 0.33-0.80; P trend = 0.001) and lifetime number of doses taken (HR = 0.53; 95% CI = 0.31-0.89; P trend = 0.003). We conclude that nonaspirin NSAID use is associated with statistically significant lower breast cancer risk after a BBD biopsy, including a dose-response effect, suggesting a potential role for NSAIDs in breast cancer prevention among patients with BBD.
Collapse
Affiliation(s)
- Mark E Sherman
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, Florida.
| | | | | | | | - Marlene H Frost
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota
| | - Lori Denison
- Department of Information Technology, Mayo Clinic, Rochester, Minnesota
| | - Daniel W Visscher
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Jodi M Carter
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | | | | | - Derek C Radisky
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida
| | | | - Amy C Degnim
- Department of Surgery, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
56
|
Yu SM, Han Y, Kim SJ. Simvastatin abolishes nitric oxide- and reactive oxygen species-induced cyclooxygenase-2 expression by blocking the nuclear factor κB pathway in rabbit articular chondrocytes. Cell Biol Int 2020; 44:2153-2162. [PMID: 32706497 DOI: 10.1002/cbin.11424] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 06/28/2020] [Accepted: 07/22/2020] [Indexed: 01/15/2023]
Abstract
Nitric oxide (NO) and reactive oxygen species (ROS) have been shown to be linked with numerous diseases, including osteoarthritis (OA). Our study aimed to examine the effect of simvastatin on NO- or ROS-induced cyclooxygenase-2 (COX-2) expression in OA. Simvastatin has attracted considerable attention since the discovery of its pharmacological effects on different pathogenic processes, including inflammation. Here, we report that simvastatin treatment blocked sodium nitroprusside (SNP)- and interleukin 1 beta (IL-1β)-induced COX-2 production. In addition, simvastatin attenuated SNP-induced NO production and IL-1β-induced ROS generation. Treatment with simvastatin prevented SNP- and IL-1β-induced nuclear factor kappa B (NF-κB) activity. Inhibiting NO production and ROS generation using N-acetylcysteine (NAC) and NG-monomethyl- l-arginine ( l-NMMA), respectively, accelerated the influence of simvastatin on NF-κB activity. In addition, NAC blocked SNP and simvastatin-mediated COX-2 production and NF-κB activity but did not alter IL-1β and simvastatin-mediated COX-2 expression. l-NMMA treatment also abolished IL-1β-mediated COX-2 expression and NF-κB activation, whereas SNP and simvastatin-mediated COX-2 expression were not altered compared with the levels in the SNP and simvastatin-treated cells. Our findings suggested that simvastatin blocks COX-2 expression by inhibiting SNP-induced NO production and IL-1β-induced ROS generation by blocking the NF-κB pathway.
Collapse
Affiliation(s)
- Seon-Mi Yu
- Department of Biological Sciences, College of Natural Sciences, Kongju National University, Gongju, Korea
| | - Yohan Han
- Department of Biological Sciences, College of Natural Sciences, Kongju National University, Gongju, Korea
| | - Song Ja Kim
- Department of Biological Sciences, College of Natural Sciences, Kongju National University, Gongju, Korea
| |
Collapse
|
57
|
Liubomirski Y, Ben-Baruch A. Notch-Inflammation Networks in Regulation of Breast Cancer Progression. Cells 2020; 9:cells9071576. [PMID: 32605277 PMCID: PMC7407628 DOI: 10.3390/cells9071576] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/21/2020] [Accepted: 06/24/2020] [Indexed: 12/20/2022] Open
Abstract
Members of the Notch family and chronic inflammation were each separately demonstrated to have prominent malignancy-supporting roles in breast cancer. Recent investigations indicate that bi-directional interactions that exist between these two pathways promote the malignancy phenotype of breast tumor cells and of their tumor microenvironment. In this review article, we demonstrate the importance of Notch-inflammation interplays in malignancy by describing three key networks that act in breast cancer and their impacts on functions that contribute to disease progression: (1) Cross-talks of the Notch pathway with myeloid cells that are important players in cancer-related inflammation, focusing mainly on macrophages; (2) Cross-talks of the Notch pathway with pro-inflammatory factors, exemplified mainly by Notch interactions with interleukin 6 and its downstream pathways (STAT3); (3) Cross-talks of the Notch pathway with typical inflammatory transcription factors, primarily NF-κB. These three networks enhance tumor-promoting functions in different breast tumor subtypes and act in reciprocal manners, whereby Notch family members activate inflammatory elements and vice versa. These characteristics illustrate the fundamental roles played by Notch-inflammation interactions in elevating breast cancer progression and propose that joint targeting of both pathways together may provide more effective and less toxic treatment approaches in this disease.
Collapse
|
58
|
Bioactive C 17 and C 18 Acetylenic Oxylipins from Terrestrial Plants as Potential Lead Compounds for Anticancer Drug Development. Molecules 2020; 25:molecules25112568. [PMID: 32486470 PMCID: PMC7321150 DOI: 10.3390/molecules25112568] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 05/29/2020] [Accepted: 05/29/2020] [Indexed: 02/06/2023] Open
Abstract
Bioactive C17 and C18 acetylenic oxylipins have shown to contribute to the cytotoxic, anti-inflammatory, and potential anticancer properties of terrestrial plants. These acetylenic oxylipins are widely distributed in plants belonging to the families Apiaceae, Araliaceae, and Asteraceae, and have shown to induce cell cycle arrest and/or apoptosis of cancer cells in vitro and to exert a chemopreventive effect on cancer development in vivo. The triple bond functionality of these oxylipins transform them into highly alkylating compounds being reactive to proteins and other biomolecules. This enables them to induce the formation of anti-inflammatory and cytoprotective phase 2 enzymes via activation of the Keap1–Nrf2 signaling pathway, inhibition of proinflammatory peptides and proteins, and/or induction of endoplasmic reticulum stress, which, to some extent, may explain their chemopreventive effects. In addition, these acetylenic oxylipins have shown to act as ligands for the nuclear receptor PPARγ, which play a central role in growth, differentiation, and apoptosis of cancer cells. Bioactive C17 and C18 acetylenic oxylipins appear, therefore, to constitute a group of promising lead compounds for the development of anticancer drugs. In this review, the cytotoxic, anti-inflammatory and anticancer effects of C17 and C18 acetylenic oxylipins from terrestrial plants are presented and their possible mechanisms of action and structural requirements for optimal cytotoxicity are discussed.
Collapse
|
59
|
de Souza CP, Alves B, Waisberg J, Fonseca F, Carmo ADO, Gehrke F. Detection of COX-2 in liquid biopsy in patients with breast cancer. J Clin Pathol 2020; 73:826-829. [DOI: 10.1136/jclinpath-2020-206576] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 04/14/2020] [Accepted: 04/15/2020] [Indexed: 11/04/2022]
Abstract
AimsTo determine the expression of the cyclooxygenase-2 (COX-2) gene in patients with breast cancer attended at the Centro Universitário Saúde ABC/Faculdade de Medicina do ABC (CUS-ABC/FMABC) outpatient clinic. Breast cancer is the most common cancer in women worldwide. More than two million new cases are reported annually. An overexpression of COX-2 has been observed in many cancers. COX-2 is related to parameters of cancer aggressiveness, including tumour size, positive nodal state and lower survival, and to angiogenesis and resistance to apoptosis.Methods15 mL of peripheral blood was obtained from 34 patients and 21 healthy women. The extracellular RNA of QIAamp RNA was submitted to an RNA sequestration kit for RNA reverse transcriptase. Quantitative real-time PCR was performed using COX-2-specific oligonucleotides and the endogenous Glyceraldehyde-3-Phosphate Dehydrogenase gene.ResultsThe mean remission time was 53 years. The mean progression time was 33 months. The difference observed between the patient and control groups in median COX-2 expression (p<0.001) was significant.ConclusionsPatients with breast cancer showed a higher mean COX-2 expression in peripheral blood samples at diagnosis than the control group. Since this information could prove important in the diagnosis and prognosis of breast cancer, further research is required on larger patient samples.
Collapse
|
60
|
Kuhlman KR, Horn SR, Chiang JJ, Bower JE. Early life adversity exposure and circulating markers of inflammation in children and adolescents: A systematic review and meta-analysis. Brain Behav Immun 2020; 86:30-42. [PMID: 30999015 PMCID: PMC8211400 DOI: 10.1016/j.bbi.2019.04.028] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 12/21/2018] [Accepted: 04/14/2019] [Indexed: 02/06/2023] Open
Abstract
This study provides a comprehensive review of the published research on the association between early life adversity and markers of inflammation in children and adolescents. We conducted a systematic review of the published literature on the association between early life adversity and markers of inflammation in pediatric populations. To date, 27 studies have been published in this area representing a wide range of global populations and diverse methods of which nearly half were prospective, longitudinal studies. Of these 27, only 12 studies shared an inflammatory outcome with 4 or more other studies; 9 for CRP, and 6 for IL-6. The association between early life adversity and both CRP, z = .07 [.04, .10], and IL-6, z = .17 [-.07, .42], were small and only significant for CRP although comparable in magnitude to the effects observed in adult samples. Descriptively, the association between early life adversity and CRP appeared to be stronger in studies conducted in infants and adolescents compared with middle childhood. There was minimal evidence of publication bias for studies measuring CRP, but evidence of publication bias for studies using IL-6. Eight studies have looked at the association between early life adversity and stimulated inflammatory cytokines in vitro, and both the methods and results of these studies were mixed; the majority observed exaggerated production of inflammatory cytokines despite mixed methodological approaches that make comparisons across studies difficult. In summary, the evidence supporting an association between early life adversity and inflammation in pediatric samples is limited so far by the number of studies and their heterogeneous methodological approaches. More research that is grounded in a developmental framework and informed by the complexity of the innate immune system is needed in this area.
Collapse
Affiliation(s)
- Kate R Kuhlman
- Department of Psychology & Social Behavior, University of California Irvine, Irvine, CA 92697, United States; Cousins Center for Psychoneuroimmunology, Semel Institute for Neuroscience & Human Behavior, University of California Los Angeles, Los Angeles, CA 90095, United States.
| | - Sarah R Horn
- Department of Psychology, University of Oregon, Eugene, OR 97403, United States
| | - Jessica J Chiang
- Department of Psychology, Northwestern University, Evanston, IL 60201, United States
| | - Julienne E Bower
- Cousins Center for Psychoneuroimmunology, Semel Institute for Neuroscience & Human Behavior, University of California Los Angeles, Los Angeles, CA 90095, United States; Department of Psychology, University of California Los Angeles, Los Angeles, CA 90095, United States
| |
Collapse
|
61
|
Forcados GE, Sallau AB, Muhammad A, Erukainure OL, James DB. Vitex doniana Leaves Extract Ameliorates Alterations Associated with 7, 12-Dimethyl Benz[a]Anthracene-Induced Mammary Damage in Female Wistar Rats. Nutr Cancer 2020; 73:98-112. [PMID: 32223342 DOI: 10.1080/01635581.2020.1743866] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Vitex doniana leaves are used traditionally in West Africa for the treatment of swellings and cancer. We investigated if Vitex doniana leaves extract could ameliorate 7, 12-dimethylbenz[a]anthracene (DMBA)-induced mammary damage. Female Wistar rats aged 52 ± 2 day were administered 80 mg/kg DMBA. After monitoring for 150 day, rats were administered 0, 50, 100, 200 mg/kg Vitex doniana and 20 mg/kg Tamoxifen for 14 day. Serum estrogen receptor-α, IL-1β and TNF -α levels were determined using ELISA kits. Oxidative stress markers in mammary tissue homogenates were determined using standard spectrophotometric methods. Histopathological examination was done using hematoxylin and eosin staining and cyclooxygenase-2 (COX-2) expression using immunohistochemistry. Liquid chromatography-mass spectrometry was used to determine components present in the extract. Although tumors were not observed, significantly (p < 0.05) lower estrogen receptor-α, malondialdehyde, IL-1β and TNF -α levels, significantly (p < 0.05) higher glutathione and catalase activity, attenuation of malignant epithelial hyperplasia and mild COX-2 expression were observed in rats administered Vitex doniana when compared to DMBA-induced untreated control. Liquid chromatography-mass spectrometry analysis of the V. doniana extract revealed the presence of 4,5-dihydroxy-7-methoxy-6-methylflavone and vanillylamine, which are compounds with reported antioxidant and anti-inflammatory effects. Collectively, treatment with Vitex doniana ameliorated some derangement observed in DMBA-induced rats.
Collapse
Affiliation(s)
| | | | - Aliyu Muhammad
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria, Nigeria
| | - Ochuko Lucky Erukainure
- Nutrition and Toxicology Division, Federal Institute of Industrial Research, Oshodi, Lagos, Nigeria.,Department of Pharmacology, University of the Free State, Bloemfontein, South Africa
| | - Dorcas Bolanle James
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria, Nigeria
| |
Collapse
|
62
|
Watanabe Y, Imanishi Y, Ozawa H, Sakamoto K, Fujii R, Shigetomi S, Habu N, Otsuka K, Sato Y, Sekimizu M, Ito F, Ikari Y, Saito S, Kameyama K, Ogawa K. Selective EP2 and Cox-2 inhibition suppresses cell migration by reversing epithelial-to-mesenchymal transition and Cox-2 overexpression and E-cadherin downregulation are implicated in neck metastasis of hypopharyngeal cancer. Am J Transl Res 2020; 12:1096-1113. [PMID: 32269737 PMCID: PMC7137058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 03/01/2020] [Indexed: 06/11/2023]
Abstract
Cyclooxygenase-2 (Cox-2) has been shown to promote cancer initiation and progression through pleiotropic functions including induction of epithelial-to-mesenchymal transition (EMT) via its predominant product prostaglandin E2 that binds to the cognate receptor EP2. Hence, pharmacological inhibition at the level of EP2 is assumed to be a more selective alternative with less risk to Cox-2 inhibition. However, little is known regarding the anti-cancer effect of an EP2 antagonist on the malignant properties of cancers including hypopharyngeal squamous cell carcinoma (HPSCC). The present study found that both the Cox-2 inhibitor celecoxib and the EP2 antagonist PF-04418948 upregulated CDH-1 expression, restored membranous localization of E-cadherin, and reduced vimentin expression, by downregulating the transcriptional repressors of E-cadherin in BICR6 and FaDu cells. Such Cox-2 or EP2 inhibition-induced EMT reversal led to repressed migration ability in both cells. Immunohistochemical analysis of surgical HPSCC specimens demonstrated an inverse relationship in expression between Cox-2 and E-cadherin both in the context of statistics (P = 0.028) and of reciprocal immunolocalization in situ. Multivariate logistic regression revealed that overexpression of Cox-2 (P < 0.001) and downregulation of E-cadherin (P = 0.016) were both independently predictive of neck metastasis. These results suggest that suppression of cell migration ability via reversing EMT by inhibiting the Cox-2/EP2 signaling may contribute to preventing the development and progression of lymphatic metastasis. Collectively, targeting Cox-2/EP2, especially using EP2 antagonist, can be a promising therapeutic strategy by exerting an anti-metastatic effect via EMT reversal for improving the treatment outcomes of patients with various cancers including HPSCC.
Collapse
Affiliation(s)
- Yoshihiro Watanabe
- Department of Otorhinolaryngology-Head and Neck Surgery, Keio University School of MedicineTokyo, Japan
- Department of Otorhinolaryngology, Tokyo Saiseikai Central HospitalTokyo, Japan
| | - Yorihisa Imanishi
- Department of Otorhinolaryngology-Head and Neck Surgery, Keio University School of MedicineTokyo, Japan
- Department of Otorhinolaryngology-Head and Neck Surgery, Kawasaki Municipal Kawasaki HospitalKawasaki, Japan
| | - Hiroyuki Ozawa
- Department of Otorhinolaryngology-Head and Neck Surgery, Keio University School of MedicineTokyo, Japan
| | - Koji Sakamoto
- Department of Otorhinolaryngology-Head and Neck Surgery, Saiseikai Utsunomiya HospitalUtsunomiya, Japan
| | - Ryoichi Fujii
- Department of Otorhinolaryngology, Saiseikai Yokohamashi Tobu HospitalYokohama, Japan
| | - Seiji Shigetomi
- Department of Otorhinolaryngology, Yokohama Municipal Citizen’s HospitalYokohama, Japan
| | - Noboru Habu
- Department of Otorhinolaryngology, Kyosai Tachikawa HospitalTokyo, Japan
| | - Kuninori Otsuka
- Department of Otorhinolaryngology, Shin-Yurigaoka General HospitalKawasaki, Japan
| | - Yoichiro Sato
- Department of Otorhinolaryngology-Head and Neck Surgery, Kawasaki Municipal Kawasaki HospitalKawasaki, Japan
| | - Mariko Sekimizu
- Department of Otorhinolaryngology-Head and Neck Surgery, Keio University School of MedicineTokyo, Japan
| | - Fumihiro Ito
- Department of Otorhinolaryngology-Head and Neck Surgery, Keio University School of MedicineTokyo, Japan
| | - Yuichi Ikari
- Department of Otorhinolaryngology-Head and Neck Surgery, Keio University School of MedicineTokyo, Japan
| | - Shin Saito
- Department of Otorhinolaryngology-Head and Neck Surgery, Keio University School of MedicineTokyo, Japan
| | - Kaori Kameyama
- Department of Pathology, Keio University School of MedicineTokyo, Japan
| | - Kaoru Ogawa
- Department of Otorhinolaryngology-Head and Neck Surgery, Keio University School of MedicineTokyo, Japan
| |
Collapse
|
63
|
Maskarinec G, Ju D, Shvetsov YB, Horio D, Chan O, Loo LWM, Hernandez BY. Breast tumor tissue inflammation but not lobular involution is associated with survival among breast cancer patients in the Multiethnic Cohort. Cancer Epidemiol 2020; 65:101685. [PMID: 32058311 DOI: 10.1016/j.canep.2020.101685] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 01/29/2020] [Accepted: 02/02/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND This study investigated the association of breast lobular involution status and three inflammatory markers as predictors of survival among breast cancer patients in the Multiethnic Cohort. METHODS Lobular involution was evaluated in tissue sections of normal breast tissue and COX-2, TNF-α, and TGF-β proteins were assessed by immunohistochemistry in tumor microarrays. A summary score added the expression levels of the three markers. Cox regression was applied to estimate hazard ratios (HRs) and 95 % confidence intervals (CI) with age as the time metric and adjustment for factors known to affect mortality. RESULTS Among 254 women (mean age = 61.7 ± 8.7 years) with pathologic blocks and follow-up information, 54 all-cause and 10 breast cancer-specific deaths were identified after a mean follow-up time of 16.0 ± 3.1 years. For 214 participants, an inflammatory score was available and 157 women had information on lobular involution. Lobular involution was not significantly associated with survival. Expression of both COX-2 and TNF-α were significant predictors of lower survival (p = 0.02 and 0.04), while the association for TGF-β was weaker (p = 0.09). When combined into one overall inflammation score, both intermediate (HR = 2.72; 95 % CI 0.90-8.28) and high (HR = 4.21; 95 % CI 1.51-11.8) scores were associated with higher mortality but only the latter was statistically significant. No significant association with breast cancer-specific mortality was detected. CONCLUSIONS These results suggest that strong expression of inflammatory markers in breast tissue predicts a poorer prognosis possibly due to a system-wide state of chronic inflammation.
Collapse
Affiliation(s)
| | - Dan Ju
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | | | - David Horio
- University of Hawaii Cancer Center, Honolulu, HI, United States
| | - Owen Chan
- University of Hawaii Cancer Center, Honolulu, HI, United States
| | - Lenora W M Loo
- University of Hawaii Cancer Center, Honolulu, HI, United States
| | | |
Collapse
|
64
|
Ethyl benzoate bearing pyrrolizine/indolizine moieties: Design, synthesis and biological evaluation of anti-inflammatory and cytotoxic activities. Bioorg Chem 2020; 94:103371. [DOI: 10.1016/j.bioorg.2019.103371] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 10/07/2019] [Accepted: 10/16/2019] [Indexed: 12/19/2022]
|
65
|
Shawky AM, Abourehab MA, Abdalla AN, Gouda AM. Optimization of pyrrolizine-based Schiff bases with 4-thiazolidinone motif: Design, synthesis and investigation of cytotoxicity and anti-inflammatory potency. Eur J Med Chem 2020; 185:111780. [DOI: 10.1016/j.ejmech.2019.111780] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 10/04/2019] [Accepted: 10/09/2019] [Indexed: 12/17/2022]
|
66
|
Wyatt GL, Crump LS, Young CM, Wessells VM, McQueen CM, Wall SW, Gustafson TL, Fan YY, Chapkin RS, Porter WW, Lyons TR. Cross-talk between SIM2s and NFκB regulates cyclooxygenase 2 expression in breast cancer. Breast Cancer Res 2019; 21:131. [PMID: 31783895 PMCID: PMC6884910 DOI: 10.1186/s13058-019-1224-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 11/07/2019] [Indexed: 02/10/2023] Open
Abstract
Background Breast cancer is a leading cause of cancer-related death for women in the USA. Thus, there is an increasing need to investigate novel prognostic markers and therapeutic methods. Inflammation raises challenges in treating and preventing the spread of breast cancer. Specifically, the nuclear factor kappa b (NFκB) pathway contributes to cancer progression by stimulating proliferation and preventing apoptosis. One target gene of this pathway is PTGS2, which encodes for cyclooxygenase 2 (COX-2) and is upregulated in 40% of human breast carcinomas. COX-2 is an enzyme involved in the production of prostaglandins, which mediate inflammation. Here, we investigate the effect of Singleminded-2s (SIM2s), a transcriptional tumor suppressor that is implicated in inhibition of tumor growth and metastasis, in regulating NFκB signaling and COX-2. Methods For in vitro experiments, reporter luciferase assays were utilized in MCF7 cells to investigate promoter activity of NFκB and SIM2. Real-time PCR, immunoblotting, immunohistochemistry, and chromatin immunoprecipitation assays were performed in SUM159 and MCF7 cells. For in vivo experiments, MCF10DCIS.COM cells stably expressing SIM2s-FLAG or shPTGS2 were injected into SCID mice and subsequent tumors harvested for immunostaining and analysis. Results Our results reveal that SIM2 attenuates the activation of NFκB as measured using NFκB-luciferase reporter assay. Furthermore, immunostaining of lysates from breast cancer cells overexpressing SIM2s showed reduction in various NFκB signaling proteins, as well as pAkt, whereas knockdown of SIM2 revealed increases in NFκB signaling proteins and pAkt. Additionally, we show that NFκB signaling can act in a reciprocal manner to decrease expression of SIM2s. Likewise, suppressing NFκB translocation in DCIS.COM cells increased SIM2s expression. We also found that NFκB/p65 represses SIM2 in a dose-dependent manner, and when NFκB is suppressed, the effect on the SIM2 is negated. Additionally, our ChIP analysis confirms that NFκB/p65 binds directly to SIM2 promoter site and that the NFκB sites in the SIM2 promoter are required for NFκB-mediated suppression of SIM2s. Finally, overexpression of SIM2s decreases PTGS2 in vitro, and COX-2 staining in vivo while decreasing PTGS2 and/or COX-2 activity results in re-expression of SIM2. Conclusion Our findings identify a novel role for SIM2s in NFκB signaling and COX-2 expression.
Collapse
Affiliation(s)
- Garhett L Wyatt
- Department of Integrative Biosciences, College of Veterinary Medicine, Texas A&M University, College Station, TX, USA
| | - Lyndsey S Crump
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, USA.,The University of Colorado Cancer Center Young Women's Breast Cancer Translational Program, Aurora, CO, USA
| | - Chloe M Young
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, USA.,The University of Colorado Cancer Center Young Women's Breast Cancer Translational Program, Aurora, CO, USA
| | - Veronica M Wessells
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, USA.,The University of Colorado Cancer Center Young Women's Breast Cancer Translational Program, Aurora, CO, USA
| | - Cole M McQueen
- Department of Integrative Biosciences, College of Veterinary Medicine, Texas A&M University, College Station, TX, USA
| | - Steven W Wall
- Department of Integrative Biosciences, College of Veterinary Medicine, Texas A&M University, College Station, TX, USA
| | - Tanya L Gustafson
- Department of Integrative Biosciences, College of Veterinary Medicine, Texas A&M University, College Station, TX, USA
| | - Yang-Yi Fan
- Department of Nutrition, Texas A&M University, College Station, TX, USA
| | - Robert S Chapkin
- Department of Nutrition, Texas A&M University, College Station, TX, USA
| | - Weston W Porter
- Department of Integrative Biosciences, College of Veterinary Medicine, Texas A&M University, College Station, TX, USA.
| | - Traci R Lyons
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, USA. .,The University of Colorado Cancer Center Young Women's Breast Cancer Translational Program, Aurora, CO, USA.
| |
Collapse
|
67
|
Malecki KMC. Epigenetics and differential effects of aspirin on breast cancer survival: Opportunities for understanding human susceptibility and risk. Cancer 2019; 125:3709-3713. [DOI: 10.1002/cncr.32365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 06/04/2019] [Accepted: 06/05/2019] [Indexed: 11/11/2022]
Affiliation(s)
- Kristen M. C. Malecki
- Department of Population Health Sciences, School of Medicine and Public Health University of Wisconsin Madison Madison Wisconsin
| |
Collapse
|
68
|
Untapped "-omics": the microbial metagenome, estrobolome, and their influence on the development of breast cancer and response to treatment. Breast Cancer Res Treat 2019; 179:287-300. [PMID: 31646389 DOI: 10.1007/s10549-019-05472-w] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 10/10/2019] [Indexed: 02/07/2023]
Abstract
With the advent of next generation sequencing technologies, there is an increasingly complex understanding of the role of gastrointestinal and local breast microbial dysbiosis in breast cancer. In this review, we summarize the current understanding of the microbiome's role in breast carcinogenesis, discussing modifiable risk factors that may affect breast cancer risk by inducing dysbiosis as well as recent sequencing data illustrating breast cancer subtype-specific differences in local breast tissue microbiota. We outline how the 'estrobolome,' the aggregate of estrogen-metabolizing enteric bacterial genes, may affect the risk of developing postmenopausal estrogen receptor-positive breast cancer. We also discuss the microbiome's potent capacity for anticancer therapy activation and deactivation, an important attribute of the gastrointestinal microbiome that has yet to be harnessed clinically.
Collapse
|
69
|
Şenkardeş S, Han Mİ, Kulabaş N, Abbak M, Çevik Ö, Küçükgüzel İ, Küçükgüzel ŞG. Synthesis, molecular docking and evaluation of novel sulfonyl hydrazones as anticancer agents and COX-2 inhibitors. Mol Divers 2019; 24:673-689. [PMID: 31302853 DOI: 10.1007/s11030-019-09974-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 07/04/2019] [Indexed: 10/26/2022]
Abstract
In trying to develop new anticancer agents, a series of sulfonylhydrazones were synthesized. All synthesized compounds were checked for identity and purity using elemental analysis, TLC and HPLC and were characterized by their melting points, FT-IR and NMR spectral data. All synthesized compounds were evaluated for their cytotoxic activity against prostate cancer (PC3), breast cancer (MCF-7) and L929 mouse fibroblast cell lines. Among them, N'-[(2-chloro-3-methoxyphenyl)methylidene]-4-methylbenzenesulfonohydrazide (3k) showed the most potent anticancer activity against both cancer cells with good selectivity (IC50 = 1.38 μM on PC3 with SI = 432.30 and IC50 = 46.09 μM on MCF-7 with SI = 12.94). Further investigation confirmed that 3k displayed morphological alterations in PC3 and MCF-7 cells and promoted apoptosis through down-regulation of the Bcl-2 and upregulation of Bax expression. Additionally, compound 3k was identified as the most potent COX-2 inhibitor (91% inhibition) beside lower COX-1 inhibition. Molecular docking of the tested compounds represented important binding modes which may be responsible for their anticancer activity via inhibition of the COX-2 enzyme. Overall, the lead compound 3k deserves further development as a potential anticancer agent. Sulfonylhydrazones was synthesized and N'-[(2-chloro-3-methoxyphenyl)methylidene]-4- methylbenzenesulfonohydrazide (3k) was identified as the most potent anticancer agent and COX-2 inhibitor. In addition, this compound docked inside the active site of COX-2 succesfully.
Collapse
Affiliation(s)
- Sevil Şenkardeş
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Marmara University, Haydarpaşa, 34668, İstanbul, Turkey.
| | - M İhsan Han
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Erciyes University, Kayseri, Turkey
| | - Necla Kulabaş
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Marmara University, Haydarpaşa, 34668, İstanbul, Turkey
| | - Mürüvvet Abbak
- Scientific Technology Research and Application Centre, Adnan Menderes University, Aydın, Turkey
| | - Özge Çevik
- Department of Biochemistry, School of Medicine, Adnan Menderes University, Aydın, Turkey
| | - İlkay Küçükgüzel
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Marmara University, Haydarpaşa, 34668, İstanbul, Turkey
| | - Ş Güniz Küçükgüzel
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Marmara University, Haydarpaşa, 34668, İstanbul, Turkey
| |
Collapse
|
70
|
Human cytomegalovirus infection is correlated with enhanced cyclooxygenase-2 and 5-lipoxygenase protein expression in breast cancer. J Cancer Res Clin Oncol 2019; 145:2083-2095. [PMID: 31203442 PMCID: PMC6658585 DOI: 10.1007/s00432-019-02946-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 05/27/2019] [Indexed: 01/26/2023]
Abstract
Purpose While enhanced expression of cyclooxygenase-2 (COX-2) and 5-lipoxygenase (5-LO) and their derived metabolites is associated with breast cancer (BC) risk, the precise link between BC carcinogenesis and enhanced inflammatory activity remains to be clarified. Human Cytomegalovirus (HCMV) may induce expression of COX-2 and 5-LO and is frequently found in breast cancer biopsies. Thus, we investigated whether there is an association between HCMV proteins and expression of COX-2 and 5-LO in human BC tissue and BC cell lines. Materials and methods Paraffin embedded biopsies obtained from 49 patients with breast cancer and 26 tissue samples from adjacent, benign breast tissues were retrospectively examined for HCMV-immediate early (IE), HCMV-Late (LA), COX-2, and 5-LO proteins by immunohistochemistry. In vitro, uninfected and HCMV-infected BC cell lines were examined for COX-2 and 5-LO transcripts and proteins by PCR and flow cytometry. Results Extensive expression of COX-2, 5-LO and HCMV-IE proteins were preferentially detected in BC samples. We found a statistically significant concordant correlation between extensive HCMV-IE and COX-2 (P < 0.0001) as well as with HCMV-IE and 5-LO (P = 0.0003) in infiltrating BC. In vitro, HCMV infection induced COX-2 and 5-LO transcripts and COX-2 proteins in MCF-7 cells (P =0.008, P =0.018, respectively). In MDA-MB-231 cells that already had high base line levels of COX-2 expression, HCMV induced both COX-2 and 5-LO proteins but not transcripts. Conclusion Our findings demonstrate a significant correlation between extensive HCMV-IE protein expression and overexpression of COX-2 and 5-LO in human breast cancer. Electronic supplementary material The online version of this article (10.1007/s00432-019-02946-8) contains supplementary material, which is available to authorized users.
Collapse
|
71
|
Esbona K, Yi Y, Saha S, Yu M, Van Doorn RR, Conklin MW, Graham DS, Wisinski KB, Ponik SM, Eliceiri KW, Wilke LG, Keely PJ. The Presence of Cyclooxygenase 2, Tumor-Associated Macrophages, and Collagen Alignment as Prognostic Markers for Invasive Breast Carcinoma Patients. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 188:559-573. [PMID: 29429545 DOI: 10.1016/j.ajpath.2017.10.025] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 10/20/2017] [Accepted: 10/30/2017] [Indexed: 02/08/2023]
Abstract
Inflammation, and the organization of collagen in the breast tumor microenvironment, is an important mediator of breast tumor progression. However, a direct link between markers of inflammation, collagen organization, and patient outcome has yet to be established. A tumor microarray of 371 invasive breast carcinoma biopsy specimens was analyzed for expression of inflammatory markers, including cyclooxygenase 2 (COX-2), macrophages, and several collagen features in the tumor nest (TN) or the tumor-associated stroma (TS). The tumor microarray cohort included females, aged 18 to 80 years, with a median follow-up of 8.4 years. High expression of COX-2 (TN), CD68 (TS), and CD163 (TN and TS) predicted worse patient overall survival (OS). This notion was strengthened by the finding from the multivariate analysis that high numbers of CD163+ macrophages in the TS is an independent prognostic factor. Overall collagen deposition was associated with high stromal expression of COX-2 and CD163; however, total collagen deposition was not a predictor for OS. Conversely, local collagen density, alignment and perpendicular alignment to the tumor boundary (tumor-associated collagen signature-3) were predictors of OS. These results suggest that in invasive carcinoma, the localization of inflammatory cells and aligned collagen orientation predict poor patient survival. Additional clinical studies may help validate whether therapy with selective COX-2 inhibitors alters expression of CD68 and CD163 inflammatory markers.
Collapse
Affiliation(s)
- Karla Esbona
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin; Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin.
| | - Yanyao Yi
- Department of Statistics, University of Wisconsin-Madison, Madison, Wisconsin; Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin
| | - Sandeep Saha
- Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin
| | - Menggang Yu
- Department of Biostatistics and Medical Informatics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin
| | - Rachel R Van Doorn
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin
| | - Matthew W Conklin
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin; Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin
| | - Douglas S Graham
- Department of Information Technology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Kari B Wisinski
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin
| | - Suzanne M Ponik
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin; Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin
| | - Kevin W Eliceiri
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin; Laboratory for Optical and Computational Instrumentation, University of Wisconsin-Madison, Madison, Wisconsin
| | - Lee G Wilke
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin; Department of Surgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin
| | - Patricia J Keely
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin; Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin
| |
Collapse
|
72
|
The role of omega 3 fatty acids in suppressing muscle protein catabolism: A possible therapeutic strategy to reverse cancer cachexia? J Funct Foods 2019. [DOI: 10.1016/j.jff.2018.12.033] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
73
|
Greish K, Nehoff H, Bahman F, Pritchard T, Taurin S. Raloxifene nano-micelles effect on triple-negative breast cancer is mediated through estrogen receptor-β and epidermal growth factor receptor. J Drug Target 2019; 27:903-916. [PMID: 30615483 DOI: 10.1080/1061186x.2019.1566341] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer that differs in progression, recurrence, and prognosis from other forms of breast cancer. The heterogeneity of TNBC has remained a challenge as no targeted therapy is currently available. Previously, we and others have demonstrated that raloxifene, a selective oestrogen receptor modulator, was also acting independently of the oestrogen receptor-α. However, raloxifene is characterised by a low bioavailability in vivo. Thus, we encapsulated raloxifene into a styrene-maleic acid (SMA) micelle to improve its pharmacokinetics. The micellar raloxifene had higher cytotoxicity when compared to the free formulation, promoted a higher cellular uptake and affected critical signalling pathways. Furthermore, SMA-raloxifene reduced TNBC tumour growth more efficiently than free raloxifene. Finally, we showed that this effect was partially mediated through oestrogen receptor-β. In conclusion, we have provided new insight into the role of raloxifene nanoformulation in improving the management of TNBC.
Collapse
Affiliation(s)
- Khaled Greish
- a Department of Molecular Medicine, and Nanomedicine Unit , College of Medicine and Medical Sciences, Princess Al-Jawhara Center for Molecular Medicine and Inherited Disorders, Arabian Gulf University , Manama , Kingdom of Bahrain.,b Department of Oncology , Suez Canal University , Ismailia , Egypt
| | - Hayley Nehoff
- c Department of Pharmacology and Toxicology , University of Otago , Dunedin , New Zealand
| | - Fatemah Bahman
- a Department of Molecular Medicine, and Nanomedicine Unit , College of Medicine and Medical Sciences, Princess Al-Jawhara Center for Molecular Medicine and Inherited Disorders, Arabian Gulf University , Manama , Kingdom of Bahrain
| | - Tara Pritchard
- d Malaghan Institute of Medical Research , Wellington , New Zealand
| | - Sebastien Taurin
- a Department of Molecular Medicine, and Nanomedicine Unit , College of Medicine and Medical Sciences, Princess Al-Jawhara Center for Molecular Medicine and Inherited Disorders, Arabian Gulf University , Manama , Kingdom of Bahrain
| |
Collapse
|
74
|
Yun SJ, Lee B, Komori K, Lee MJ, Lee BG, Kim K, Park S. Regulation of TIM-3 expression in a human T cell line by tumor-conditioned media and cyclic AMP-dependent signaling. Mol Immunol 2018; 105:224-232. [PMID: 30554083 DOI: 10.1016/j.molimm.2018.12.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 11/01/2018] [Accepted: 12/08/2018] [Indexed: 12/17/2022]
Abstract
T cell immunoglobulin and mucin domain-3 (TIM-3) expression increases in exhausted T cells, which inhibits T cell function. TIM-3 expression is supposedly up-regulated in tumor-bearing individuals via chronic antigenic stimulation of T cells. Considering the immunosuppressive nature of the tumor microenvironment, we investigated whether tumor-secreted molecules might enhance TIM-3 expression in Jurkat T cells. We observed that TIM-3 expression was increased by the activation of prostaglandin (PG) E2 and cyclic AMP (cAMP) signaling pathways. Adenylate cyclase activation led to protein kinase A (PKA)-dependent upregulation of the TIM-3 minimal promoter region and of upstream conserved non-coding sequences. TIM-3 expression in Jurkat T cells was increased by the exposure to breast tumor cell-conditioned media partially through the interaction between PGE2 and its receptor, EP4. Our results propose that tumor-secreted molecules such as PGE2, which activates PKA and EPAC, may regulate TIM-3 expression in T cells.
Collapse
Affiliation(s)
- Su Jin Yun
- Department of Microbiology and Ajou University School of Medicine, Youngtongku Worldcupro 206, Suwon, 16499, Republic of Korea; Department of Biomedical Sciences (Graduate Program of Molecular Medicine), Ajou University Graduate School of Medicine, Suwon, 16499, Republic of Korea
| | - Bokyoung Lee
- Department of Microbiology and Ajou University School of Medicine, Youngtongku Worldcupro 206, Suwon, 16499, Republic of Korea; Department of Biomedical Sciences (Graduate Program of Molecular Medicine), Ajou University Graduate School of Medicine, Suwon, 16499, Republic of Korea
| | - Kuniharu Komori
- Department of Microbiology and Ajou University School of Medicine, Youngtongku Worldcupro 206, Suwon, 16499, Republic of Korea; Department of Biomedical Sciences (Graduate Program of Molecular Medicine), Ajou University Graduate School of Medicine, Suwon, 16499, Republic of Korea
| | - Mi Jin Lee
- Department of Microbiology and Ajou University School of Medicine, Youngtongku Worldcupro 206, Suwon, 16499, Republic of Korea
| | - Byoung Gill Lee
- Department of Microbiology and Ajou University School of Medicine, Youngtongku Worldcupro 206, Suwon, 16499, Republic of Korea; Department of Biomedical Sciences (Graduate Program of Molecular Medicine), Ajou University Graduate School of Medicine, Suwon, 16499, Republic of Korea
| | - Kyongmin Kim
- Department of Microbiology and Ajou University School of Medicine, Youngtongku Worldcupro 206, Suwon, 16499, Republic of Korea; Department of Biomedical Sciences (Graduate Program of Molecular Medicine), Ajou University Graduate School of Medicine, Suwon, 16499, Republic of Korea
| | - Sun Park
- Department of Microbiology and Ajou University School of Medicine, Youngtongku Worldcupro 206, Suwon, 16499, Republic of Korea; Department of Biomedical Sciences (Graduate Program of Molecular Medicine), Ajou University Graduate School of Medicine, Suwon, 16499, Republic of Korea.
| |
Collapse
|
75
|
Peng Y, Wang Y, Tang N, Sun D, Lan Y, Yu Z, Zhao X, Feng L, Zhang B, Jin L, Yu F, Ma X, Lv C. Andrographolide inhibits breast cancer through suppressing COX-2 expression and angiogenesis via inactivation of p300 signaling and VEGF pathway. J Exp Clin Cancer Res 2018; 37:248. [PMID: 30314513 PMCID: PMC6186120 DOI: 10.1186/s13046-018-0926-9] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 10/02/2018] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Andrographolide (Andro), a diterpenoid lactone, has been used for treatment of various cancers with less adverse effects. However, the underlying mechanisms regarding its anti-tumor mechanism still remain unclear. METHODS Cell viability and proliferation were measured by CCK8 and CFSE dilution assay. The localization of p50/p65 or cytochrome c was determined using confocal immunofluorescence. Streptavidin-agarose pulldown or ChIP assays were used to detect the binding of multiple transactivators to COX-2 promoter. The promoter activity was examined by a dual-Luciferase reporter assay. The functions of Andro on COX-2-mediated angiogenesis were also investigated using human HUVEC cells through tube formation and spheroids sprouting assay. The in vivo anti-tumor efficacy of Andro was analyzed in xenografts nude mice. RESULTS The results indicated that Andro could significantly inhibit the proliferation of human breast cancers, and suppress COX-2 expression at both protein and mRNA levels. Furthermore, Andro could dose-dependently inhibit COX-2-mediated angiogenesis in human endothelial cells. We have also found that Andro significantly promoted the activation of cytochrome c and activated caspase-dependent apoptotic signaling pathway. Our further explorations demonstrated that Andro inhibited the binding of the transactivators CREB2, C-Fos and NF-κB and blocked the recruitment of coactivator p300 to COX-2 promoter. Moreover, Andro could effectively inhibit the activity of p300 histone acetyltransferase (HAT), thereby attenuating the p300-mediated acetylation of NF-κB. Besides, Andro could also dramatically inhibit the migration, invasion and tubulogenesis of HUVECs in vitro. In addition, Andro also exhibited effective anti-tumor efficacy as well as angiogenesis inhibition in vivo. CONCLUSION In current study, we explore the potential effects of Andro in suppressing breast cancer growth and tumor angiogenesis, as well as the precise mechanisms. This work demonstrated the potential anti-cancer effects of Andro, indicating that Andro could inhibit COX-2 expression through attenuating p300 HAT activity and suppress angiogenesis via VEGF pathway, and thereby could be developed as an antitumor agent for the treatment of breast cancer.
Collapse
Affiliation(s)
- Yulin Peng
- Institute of Integrative Medicine, College of Pharmacy, College of Basic Medical Science, Dalian Medical University, Dalian, 116044 China
| | - Yan Wang
- Institute of Integrative Medicine, College of Pharmacy, College of Basic Medical Science, Dalian Medical University, Dalian, 116044 China
| | - Ning Tang
- Institute of Integrative Medicine, College of Pharmacy, College of Basic Medical Science, Dalian Medical University, Dalian, 116044 China
- Department of Integrative Medicine, Liaoning University of Traditional Chinese Medicine Xinglin College, Shenyang, 110167 China
| | - Dongdong Sun
- Institute of Integrative Medicine, College of Pharmacy, College of Basic Medical Science, Dalian Medical University, Dalian, 116044 China
| | - Yulong Lan
- Institute of Integrative Medicine, College of Pharmacy, College of Basic Medical Science, Dalian Medical University, Dalian, 116044 China
| | - Zhenlong Yu
- Institute of Integrative Medicine, College of Pharmacy, College of Basic Medical Science, Dalian Medical University, Dalian, 116044 China
- Emergency Department, The Second Affiliated Hospital of Hainan Medical University, Haikou, 571199 China
| | - Xinyu Zhao
- Institute of Integrative Medicine, College of Pharmacy, College of Basic Medical Science, Dalian Medical University, Dalian, 116044 China
| | - Lei Feng
- Institute of Integrative Medicine, College of Pharmacy, College of Basic Medical Science, Dalian Medical University, Dalian, 116044 China
- Emergency Department, The Second Affiliated Hospital of Hainan Medical University, Haikou, 571199 China
| | - Baojing Zhang
- Institute of Integrative Medicine, College of Pharmacy, College of Basic Medical Science, Dalian Medical University, Dalian, 116044 China
| | - Lingling Jin
- Institute of Integrative Medicine, College of Pharmacy, College of Basic Medical Science, Dalian Medical University, Dalian, 116044 China
| | - Fabiao Yu
- Emergency Department, The Second Affiliated Hospital of Hainan Medical University, Haikou, 571199 China
| | - Xiaochi Ma
- Institute of Integrative Medicine, College of Pharmacy, College of Basic Medical Science, Dalian Medical University, Dalian, 116044 China
- Emergency Department, The Second Affiliated Hospital of Hainan Medical University, Haikou, 571199 China
| | - Chuanzhu Lv
- Emergency Department, The Second Affiliated Hospital of Hainan Medical University, Haikou, 571199 China
| |
Collapse
|
76
|
Substrate-inactivated cyclooxygenase-2 is disposed of by exosomes through the ER-Golgi pathway. Biochem J 2018; 475:3141-3151. [PMID: 30217900 DOI: 10.1042/bcj20180530] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 09/06/2018] [Accepted: 09/13/2018] [Indexed: 01/16/2023]
Abstract
Catalysis of arachidonic acid (AA) by cyclooxygenase-2 (COX-2) gives rise to a single product that serves as a precursor for all prostaglandins, which are central mediators of inflammation. Rapid up-regulation of COX-2 expression in response to pro-inflammatory stimuli is a well-characterized means of generating the large pool of prostaglandins necessary for inflammation. However, an efficient inflammatory process must also terminate rapidly and thus requires cessation of COX-2 enzymatic activity and removal of excess protein from the cell. Previous studies showed that COX-2 that has not been exposed to AA ('naive') degrades in the cellular proteasome. However, continuous exposure to AA induces suicide inactivation of COX-2 and its elimination no longer occurs in neither the proteasomal nor lysosomal machineries. In the present study, we show that either overexpressed or endogenously induced COX-2 is secreted via exosomes through the endoplasmic reticulum-Golgi pathway. We further find that excretion of COX-2 is significantly enhanced by prolonged exposure to AA. Genetic or chemical inhibition of COX-2 enzymatic activity has no effect on its secretion in the absence of substrate, but prevents the additional activity-dependent secretion. Finally, transfer of COX-2 to target cells only occurs in the absence of AA stimulation. Together, these results suggest that exosomal secretion of AA-activated COX-2 constitutes a means to remove damaged inactive COX-2 from the cell.
Collapse
|
77
|
Kennedy BM, Harris RE. Cyclooxygenase and lipoxygenase gene expression in the inflammogenesis of breast cancer. Inflammopharmacology 2018; 26:10.1007/s10787-018-0489-6. [PMID: 29736687 DOI: 10.1007/s10787-018-0489-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 04/22/2018] [Indexed: 02/06/2023]
Abstract
We examined the expression of major inflammatory genes, cyclooxygenase-1 and 2 (COX1, COX2) and arachidonate 5-lipoxygenase (ALOX5) in 1090 tumor samples of invasive breast cancer from The Cancer Genome Atlas (TCGA). Mean cyclooxygenase expression (COX1 + COX2) ranked in the upper 99th percentile of all 20,531 genes and surprisingly, the mean expression of COX1 was more than tenfold higher than COX2. Highly significant correlations were observed between COX2 with eight tumor-promoting genes (EGR2, IL6, RGS2, B3GNT5, SGK1, SLC2A3, SFRP1 and ETS2) and between ALOX5 and ten tumor promoter genes (CD33, MYOF1, NLRP1, GAB3, CD4, IFR8, CYTH4, BTK, FGR, CD37). Expression of CYP19A1 (aromatase) was significantly correlated with COX2, but only in tumors positive for ER, PR and HER2. Tumor-promoting genes correlated with the expression of COX1, COX2, and ALOX5 are known to effectively increase mitogenesis, mutagenesis, angiogenesis, cell survival, immunosuppression and metastasis in the pathogenesis of breast cancer.
Collapse
Affiliation(s)
- Brian M Kennedy
- Colleges of Public Health and Medicine, The Ohio State University Comprehensive Cancer Center, The Ohio State University, 1841 Neil Avenue (306 Cunz Hall), Columbus, OH, 43210-1351, USA
| | - Randall E Harris
- Colleges of Public Health and Medicine, The Ohio State University Comprehensive Cancer Center, The Ohio State University, 1841 Neil Avenue (306 Cunz Hall), Columbus, OH, 43210-1351, USA.
| |
Collapse
|
78
|
Gérard C, Brown KA. Obesity and breast cancer - Role of estrogens and the molecular underpinnings of aromatase regulation in breast adipose tissue. Mol Cell Endocrinol 2018; 466:15-30. [PMID: 28919302 DOI: 10.1016/j.mce.2017.09.014] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Revised: 09/12/2017] [Accepted: 09/13/2017] [Indexed: 12/15/2022]
Abstract
One in eight women will develop breast cancer over their lifetime making it the most common female cancer. The cause of breast cancer is multifactorial and includes hormonal, genetic and environmental cues. Obesity is now an accepted risk factor for breast cancer in postmenopausal women, particularly for the hormone-dependent subtype of breast cancer. Obesity, which is characterized by an excess accumulation of body fat, is at the origin of chronic inflammation of white adipose tissue and is associated with dramatic changes in the biology of adipocytes leading to their dysfunction. Inflammatory factors found in the breast of obese women considerably impact estrogen signaling, mainly by driving changes in aromatase expression the enzyme responsible for estrogen production, and therefore promote tumor formation and progression. There is thus a strong link between adipose inflammation and estrogen biosynthesis and their signaling pathways converge in obese patients. This review describes how obesity-related factors can affect the risk of hormone-dependent breast cancer, highlighting the different molecular mechanisms and metabolic pathways involved in aromatase regulation, estrogen production and breast malignancy in the context of obesity.
Collapse
Affiliation(s)
- Céline Gérard
- Metabolism & Cancer Laboratory, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Kristy A Brown
- Metabolism & Cancer Laboratory, Hudson Institute of Medical Research, Clayton, VIC, Australia; Department of Physiology, Monash University, Clayton, VIC, Australia; Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
79
|
Wang YX, Zhang ZY, Wang JQ, Qian XL, Cui J. HPV16 E7 increases COX-2 expression and promotes the proliferation of breast cancer. Oncol Lett 2018; 16:317-325. [PMID: 29928417 PMCID: PMC6006466 DOI: 10.3892/ol.2018.8624] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 03/09/2018] [Indexed: 12/15/2022] Open
Abstract
Breast cancer remains the leading cause of mortality worldwide. Human papilloma virus 16 (HPV16) may serve a function in the pathogenesis and development of breast cancer. However, the detection rate of HPV16 in breast carcinoma may vary by region. In the present study, the expression of HPV16 E7 in paraffin-embedded tissues from patients with breast cancer from North China was detected. Additionally, the molecular mechanisms underlying the function of HPV16 E7 in the proliferation of breast cancer cells were examined. The results demonstrated that the DNA of HPV16 E7 was detected in 30.5% of the samples, and that HPV16 E7 promoted the proliferation of breast cancer cells in vitro and in vivo. Additionally, HPV16 E7-mediated proliferation of breast cancer cells was suppressed in response to treatment with cyclooxygenase-2 (COX-2)-specific small interfering RNA and celecoxib. The results of the present study revealed that HPV16 E7 may promote the proliferation of breast cancer cells by upregulating COX-2, suggesting that COX-2 may be a potential therapeutic target for HPV16 E7-mediated progression of breast cancer.
Collapse
Affiliation(s)
- Yong-Xia Wang
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Zhe-Ying Zhang
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Jian-Qiang Wang
- Department of Pathology, Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Xin-Lai Qian
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Jing Cui
- Department of Pathology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| |
Collapse
|
80
|
Gautam S, Rawat AK, Sammi SR, Roy S, Singh M, Devi U, Yadav RK, Singh L, Rawat JK, Ansari MN, Saeedan AS, Kumar D, Pandey R, Kaithwas G. DuCLOX-2/5 Inhibition Attenuates Inflammatory Response and Induces Mitochondrial Apoptosis for Mammary Gland Chemoprevention. Front Pharmacol 2018; 9:314. [PMID: 29681851 PMCID: PMC5897656 DOI: 10.3389/fphar.2018.00314] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 03/19/2018] [Indexed: 12/11/2022] Open
Abstract
The present study is a pursuit to define implications of dual cyclooxygenase-2 (COX-2) and 5-lipoxygenase (5-LOX) (DuCLOX-2/5) inhibition on various aspects of cancer augmentation and chemoprevention. The monotherapy and combination therapy of zaltoprofen (COX-2 inhibitor) and zileuton (5-LOX inhibitor) were validated for their effect against methyl nitrosourea (MNU) induced mammary gland carcinoma in albino wistar rats. The combination therapy demarcated significant effect upon the cellular proliferation as evidenced through decreased in alveolar bud count and restoration of the histopathological architecture when compared to toxic control. DuCLOX-2/5 inhibition also upregulated levels of caspase-3 and caspase-8, and restored oxidative stress markers (GSH, TBARs, protein carbonyl, SOD and catalase). The immunoblotting and qRT-PCR studies revealed the participation of the mitochondrial mediated death apoptosis pathway along with favorable regulation of COX-2, 5-LOX. Aforementioned combination restored the metabolic changes to normal when scrutinized through 1H NMR studies. Henceforth, the DuCLOX-2/5 inhibition was recorded to import significant anticancer effects in comparison to either of the individual treatments.
Collapse
Affiliation(s)
- Swetlana Gautam
- Department of Pharmaceutical Sciences, School of Biosciences and Biotechnology, Babasaheb Bhimrao Ambedkar University (A Central University), Lucknow, India
| | - Atul K Rawat
- Center for Biomedical Research, Sanjay Gandhi Post Graduate Institute of Medical Sciences Campus, Lucknow, India
| | - Shreesh R Sammi
- Department of Microbial Technology and Nematology, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
| | - Subhadeep Roy
- Department of Pharmaceutical Sciences, School of Biosciences and Biotechnology, Babasaheb Bhimrao Ambedkar University (A Central University), Lucknow, India
| | - Manjari Singh
- Department of Pharmaceutical Sciences, School of Biosciences and Biotechnology, Babasaheb Bhimrao Ambedkar University (A Central University), Lucknow, India
| | - Uma Devi
- Department of Pharmaceutical Sciences, Faculty of Health and Medical Sciences, Sam Higginbottom Institute of Agricultural Sciences and Technology, Allahabad, India
| | - Rajnish K Yadav
- Department of Pharmaceutical Sciences, School of Biosciences and Biotechnology, Babasaheb Bhimrao Ambedkar University (A Central University), Lucknow, India
| | - Lakhveer Singh
- Department of Pharmaceutical Sciences, School of Biosciences and Biotechnology, Babasaheb Bhimrao Ambedkar University (A Central University), Lucknow, India
| | - Jitendra K Rawat
- Department of Pharmaceutical Sciences, School of Biosciences and Biotechnology, Babasaheb Bhimrao Ambedkar University (A Central University), Lucknow, India
| | - Mohd N Ansari
- Department of Pharmacology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Abdulaziz S Saeedan
- Department of Pharmacology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Dinesh Kumar
- Department of Microbial Technology and Nematology, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
| | - Rakesh Pandey
- Department of Microbial Technology and Nematology, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
| | - Gaurav Kaithwas
- Department of Pharmaceutical Sciences, School of Biosciences and Biotechnology, Babasaheb Bhimrao Ambedkar University (A Central University), Lucknow, India
| |
Collapse
|
81
|
Alessandra-Perini J, Perini JA, Rodrigues-Baptista KC, de Moura RS, Junior AP, dos Santos TA, Souza PJC, Nasciutti LE, Machado DE. Euterpe oleracea extract inhibits tumorigenesis effect of the chemical carcinogen DMBA in breast experimental cancer. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 18:116. [PMID: 29609579 PMCID: PMC5879811 DOI: 10.1186/s12906-018-2183-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 03/21/2018] [Indexed: 12/27/2022]
Abstract
Background Among the processes involved in the breast tumor microenvironment, angiogenesis and inflammation play a central role, and the main factors of these processes are the vascular endothelial growth factor (VEGF), cyclooxygenase 2 (COX-2) and macrophages. Recently, the extract of Euterpe oleracea (açaí), a fruit that is widely found in the Amazon region, already showed antitumorigenic effects in vitro in human breast cancer cell lines. The present study aimed to investigate the effect of açaí on breast cancer using a chemically DMBA (7,12-dimethylbenzanthracene) experimental model. Methods One day after initiation of treatment with açaí, mammary carcinogenesis was induced in female Wistar rats using a subcutaneous injection of 25 mg/kg of DMBA in the mammary gland. Forty rats were randomized into two groups: treated with 200 mg/kg of either açaí extract or vehicle, via gastric tube for 16 consecutive weeks. After treatment, the tumor was collected for macroscopic, histological and immunohistochemical (VEGF, vascular endothelial growth factor receptor 2 -VEGFR-2, COX-2 and matrix metalloproteinase -MMP-9) analyses; peritoneal fluid was subjected to flow cytometry (F4–80/MAC-2+) and ELISA immunoassay (VEGF, prostaglandin E2 -PGE2 and interleukin-10 -IL-10). Heart, liver and kidney samples were collected for histological analysis. Results After 16 weeks of induction, the mammary carcinoma was confirmed by macroscopic and histological evaluation. Survival analysis indicates that açaí increased the survival (P = .0002, long-rank test) and reduced the deaths number (P = .0036, Chi-square test). Açaí treatment decreased the number of inflammatory cells and macrophage positive cells (Mac-2 + F4–80+), as well as promoting a reduction in immunostaining of VEGF, VEGFR-2 and COX-2. The açaí group also exhibited lower concentrations of PGE2, VEGF and IL-10 compared to the control. The histopathological results of the liver and kidneys showed protective effect of açaí, since in the control group, there was an increase in fibrosis, atypical cells and hemorrhagic microenvironment. Conclusion The results of this study demonstrated the antiangiogenic and anti-inflammatory potential of açaí, like due to the decreases of the number of activated macrophages, resulting in the inhibition of DMBA carcinogenicity in breast cancer. Electronic supplementary material The online version of this article (10.1186/s12906-018-2183-z) contains supplementary material, which is available to authorized users.
Collapse
|
82
|
Desai SJ, Prickril B, Rasooly A. Mechanisms of Phytonutrient Modulation of Cyclooxygenase-2 (COX-2) and Inflammation Related to Cancer. Nutr Cancer 2018; 70:350-375. [PMID: 29578814 DOI: 10.1080/01635581.2018.1446091] [Citation(s) in RCA: 138] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The link between chronic inflammation and cancer involves cytokines and mediators of inflammatory pathways. Cyclooxygenase-2 (COX-2), a key enzyme in fatty acid metabolism, is upregulated during both inflammation and cancer. COX-2 is induced by pro-inflammatory cytokines at the site of inflammation and enhanced COX-2-induced synthesis of prostaglandins stimulates cancer cell proliferation, promotes angiogenesis, inhibits apoptosis, and increases metastatic potential. As a result, COX-2 inhibitors are a subject of intense research interest toward potential clinical applications. Epidemiological studies highlight the potential benefits of diets rich in phytonutrients for cancer prevention. Plants contain numerous phytonutrient secondary metabolites shown to modulate COX-2. Studies have shown that these metabolites, some of which are used in traditional medicine, can reduce inflammation and carcinogenesis. This review describes the molecular mechanisms by which phytonutrients modulate inflammation, including studies of carotenoids, phenolic compounds, and fatty acids targeting various inflammation-related molecules and pathways associated with cancer. Examples of pathways include those of COX-2, mitogen-activated protein kinase kinase kinase, mitogen-activated protein kinase, pro-inflammatory cytokines, and transcription factors like nuclear factor kappa B. Such phytonutrient modulation of COX-2 and inflammation continue to be explored for applications in the prevention and treatment of cancer.
Collapse
Affiliation(s)
- Shreena J Desai
- a Office of Cancer Complementary and Alternative Medicine , National Cancer Institute , Rockville , Maryland , USA
| | - Ben Prickril
- a Office of Cancer Complementary and Alternative Medicine , National Cancer Institute , Rockville , Maryland , USA
| | - Avraham Rasooly
- a Office of Cancer Complementary and Alternative Medicine , National Cancer Institute , Rockville , Maryland , USA
| |
Collapse
|
83
|
Freitas-Alves DR, Vieira-Monteiro HDA, Piranda DN, Sobral-Leite M, da Silva TSL, Bergmann A, Valença SS, Perini JA, Vianna-Jorge R. PTGS2 polymorphism rs689466 favors breast cancer recurrence in obese patients. Endocr Relat Cancer 2018; 25:351-365. [PMID: 29321183 DOI: 10.1530/erc-17-0374] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 01/10/2018] [Indexed: 11/08/2022]
Abstract
Breast cancer is the leading cancer among women, and its increasing incidence is a challenge worldwide. Estrogen exposure is the main risk factor, but obesity among postmenopausal women has been shown to favor disease onset and progression. The link between obesity and mammary carcinogenesis involves elevated estrogen production and proinflammatory stimuli within the adipose tissue, with activation of the cyclooxygenase-2 pathway. Here, we evaluate the impact of the four most common cyclooxygenase-2 gene polymorphisms (rs689465, rs689466, rs20417 and rs20417), in combination with obesity, on the risk of breast cancer progression in a cohort of Brazilian breast cancer patients (N = 1038). Disease-free survival was evaluated using Kaplan-Meier curves, with multivariate Cox proportional hazards regression models for calculation of adjusted hazard ratios (HRadj). Obesity did not affect disease progression, whereas rs689466 variant genotypes increased the recurrence risk among obese patients (HRadj = 2.5; 95% CI = 1.4-4.3), either for luminal (HRadj = 2.2; 95% CI = 1.1-4.2) or HER2-like and triple-negative tumors (HRadj = 3.2; 95% CI = 1.2-8.5). Likewise, the haplotype *4, which contains variant rs689466, was associated with shorter disease-free survival among obese patients (HRadj = 3.3; 95% CI = 1.8-6.0), either in luminal (HRadj = 3.5; 95% CI = 1.6-7.3) or HER2-like and triple-negative (HRadj = 3.1; 95% CI = 1.1-8.9) tumors. Such deleterious impact of variant rs689466 on disease-free survival of obese breast cancer patients was restricted to postmenopausal women. In conclusion, cyclooxygenase-2 genotyping may add to the prognostic evaluation of obese breast cancer patients.
Collapse
Affiliation(s)
- Daniely Regina Freitas-Alves
- Coordenação de PesquisaInstituto Nacional do Câncer, Rio de Janeiro, Rio de Janeiro, Brasil
- Programa de Pós-Graduação em Saúde Pública e Meio AmbienteEscola Nacional de Saúde Pública, FIOCRUZ, Rio de Janeiro, Rio de Janeiro, Brasil
- Instituto de Ciências BiomédicasUniversidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brasil
| | - Hayra de Andrade Vieira-Monteiro
- Coordenação de PesquisaInstituto Nacional do Câncer, Rio de Janeiro, Rio de Janeiro, Brasil
- Programa de Pós-Graduação em Saúde Pública e Meio AmbienteEscola Nacional de Saúde Pública, FIOCRUZ, Rio de Janeiro, Rio de Janeiro, Brasil
| | - Diogo Nascimento Piranda
- Coordenação de PesquisaInstituto Nacional do Câncer, Rio de Janeiro, Rio de Janeiro, Brasil
- Instituto de Ciências BiomédicasUniversidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brasil
| | - Marcelo Sobral-Leite
- Coordenação de PesquisaInstituto Nacional do Câncer, Rio de Janeiro, Rio de Janeiro, Brasil
- Division of Molecular PathologyThe Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Taiana Sousa Lopes da Silva
- Coordenação de PesquisaInstituto Nacional do Câncer, Rio de Janeiro, Rio de Janeiro, Brasil
- Instituto de Ciências BiomédicasUniversidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brasil
- Programa de Pós-Graduação em Biologia Molecular e CelularInstituto Biomédico, Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brasil
| | - Anke Bergmann
- Coordenação de PesquisaInstituto Nacional do Câncer, Rio de Janeiro, Rio de Janeiro, Brasil
| | - Samuel Santos Valença
- Instituto de Ciências BiomédicasUniversidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brasil
| | - Jamila Alessandra Perini
- Programa de Pós-Graduação em Saúde Pública e Meio AmbienteEscola Nacional de Saúde Pública, FIOCRUZ, Rio de Janeiro, Rio de Janeiro, Brasil
- Laboratório de Pesquisa de Ciências FarmacêuticasUnidade de Farmácia, Centro Universitário Estadual da Zona Oeste, Rio de Janeiro, Rio de Janeiro, Brasil
| | - Rosane Vianna-Jorge
- Coordenação de PesquisaInstituto Nacional do Câncer, Rio de Janeiro, Rio de Janeiro, Brasil
- Programa de Pós-Graduação em Saúde Pública e Meio AmbienteEscola Nacional de Saúde Pública, FIOCRUZ, Rio de Janeiro, Rio de Janeiro, Brasil
- Instituto de Ciências BiomédicasUniversidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brasil
| |
Collapse
|
84
|
Raposo TP, Arias-Pulido H, Chaher N, Fiering SN, Argyle DJ, Prada J, Pires I, Queiroga FL. Comparative aspects of canine and human inflammatory breast cancer. Semin Oncol 2018. [PMID: 29526258 DOI: 10.1053/j.seminoncol.2017.10.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Inflammatory breast cancer (IBC) in humans is the most aggressive form of mammary gland cancer and shares clinical, pathologic, and molecular patterns of disease with canine inflammatory mammary carcinoma (CIMC). Despite the use of multimodal therapeutic approaches, including targeted therapies, the prognosis for IBC/CIMC remains poor. The aim of this review is to critically analyze IBC and CIMC in terms of biology and clinical features. While rodent cancer models have formed the basis of our understanding of cancer biology, the translation of this knowledge into improved outcomes has been limited. However, it is possible that a comparative "one health" approach to research, using a natural canine model of the disease, may help advance our knowledge on the biology of the disease. This will translate into better clinical outcomes for both species. We propose that CIMC has the potential to be a useful model for developing and testing novel therapies for IBC. Further, this strategy could significantly improve and accelerate the design and establishment of new clinical trials to identify novel and improved therapies for this devastating disease in a more predictable way.
Collapse
Affiliation(s)
- Teresa P Raposo
- Division of Cancer and Stem Cells, Faculty of Medicine, University of Nottingham, United Kingdom
| | - Hugo Arias-Pulido
- Department of Microbiology and Immunology and Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire 03756, USA
| | - Nabila Chaher
- Department of Pathology, Centre Pierre et Marie Curie, 1, Avenue Battendier, Place May 1st, Algiers, Algeria
| | - Steven N Fiering
- Department of Microbiology and Immunology and Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire 03756, USA
| | - David J Argyle
- The Royal (Dick) School of Veterinary Studies and Roslin Institute, Easter Bush Campus, Midlothian, University of Edinburgh, United Kingdom
| | - Justina Prada
- Departament of Veterinary Sciences, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal; Animal and Veterinary research Centre (CECAV), University of Trás-os-Montes and Alto Douro, Vila Real, Portugal
| | - Isabel Pires
- Departament of Veterinary Sciences, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal; Animal and Veterinary research Centre (CECAV), University of Trás-os-Montes and Alto Douro, Vila Real, Portugal
| | - Felisbina Luísa Queiroga
- Departament of Veterinary Sciences, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal; Center for the Study of Animal Sciences, CECA-ICETA, University of Porto, Porto, Portugal; Center for Research and Technology of Agro-Environment and Biological Sciences, University of Trás-os-Montes and Alto Douro, Vila Real, Portugal.
| |
Collapse
|
85
|
Li N, Wang Q, Zhu T, Qiao L, Zhang F, Mi R, Wang B, Chen L, Gu J, Lu Y, Zheng Q. In vitro functional characterization of prostaglandin-endoperoxide synthase 2 during chondrocyte hypertrophic differentiation. Oncotarget 2017; 7:36280-36292. [PMID: 27121205 PMCID: PMC5095000 DOI: 10.18632/oncotarget.8889] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 04/04/2016] [Indexed: 01/02/2023] Open
Abstract
Cyclooxygenase 2 (Cox-2) has been implicated an essential role during bone repair, but the mechanisms remain elusive. Bone repair healing is known to include processes similar to endochondral ossification. In this study, we investigated the in vitro effect of Cox-2 on Col10a1 expression and chondrocyte hypertrophy, two critical components of endochondral ossification. Using quantitative RT-PCR, we detected increased mRNA levels of Cox-2 and Col10a1 in hypertrophic MCT cells, while cells treated with Cox-2 inhibitor, NS398, showed decreased mRNA and protein levels of Cox-2 and Col10a1. Increased Cox-2 also correlated with significantly upregulated Col10a1 in hypertrophic ATDC5 cells, whereas inhibition of Cox-2 significantly decreased Col10a1 expression. We further generated a Cox-2-expressing ATDC5 stable cell line. Compared with the controls, Cox-2 over-expression significantly increased Col10a1 as early as day 7 of continuous culturing, but not at days 14 and 21. Enhanced Alp staining was also observed in day 7 stable cell line. Correspondingly, we detected significantly increased levels of Runx2, Alp, Bcl-2, Bax, Col1a1, Osterix, and Bsp in day 7 stable line. Most of these genes have been associated with chondrocyte maturation and apoptosis. Together, our results support that Cox-2 promotes Col10a1 expression and chondrocyte hypertrophy in vitro, possibly through upregulation of Runx2 and other relevant transcription factors.
Collapse
Affiliation(s)
- Na Li
- Department of Hematological Laboratory Science, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Qian Wang
- Department of Hematological Laboratory Science, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Ting Zhu
- Department of Hematological Laboratory Science, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Longwei Qiao
- Center for Reproduction and Genetics, Suzhou Hospital Affiliated to Nanjing Medical University, Suzhou, Jiangsu, 215002, China
| | - Fei Zhang
- Department of Hematological Laboratory Science, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Rui Mi
- Department of Hematological Laboratory Science, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Bo Wang
- Department of Hematological Laboratory Science, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Lin Chen
- State Key Laboratory of Trauma, Burns and Combined Injury, Center of Bone Metabolism and Repair, Trauma Center, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Junxia Gu
- Department of Hematological Laboratory Science, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Yaojuan Lu
- Department of Hematological Laboratory Science, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Qiping Zheng
- Department of Hematological Laboratory Science, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| |
Collapse
|
86
|
Zanoaga O, Jurj A, Raduly L, Cojocneanu-Petric R, Fuentes-Mattei E, Wu O, Braicu C, Gherman CD, Berindan-Neagoe I. Implications of dietary ω-3 and ω-6 polyunsaturated fatty acids in breast cancer. Exp Ther Med 2017; 15:1167-1176. [PMID: 29434704 PMCID: PMC5776638 DOI: 10.3892/etm.2017.5515] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 07/20/2017] [Indexed: 12/20/2022] Open
Abstract
Breast cancer represents one of the most common forms of cancer in women worldwide, with an increase in the number of newly diagnosed patients in the last decade. The role of fatty acids, particularly of a diet rich in ω-3 and ω-6 polyunsaturated fatty acids (PUFAs), in breast cancer development is not fully understood and remains controversial due to their complex mechanism of action. However, a large number of animal models and cell culture studies have demonstrated that high levels of ω-3 PUFAs have an inhibitory role in the development and progression of breast cancer, compared to ω-6 PUFAs. The present review focused on recent studies regarding the correlation between dietary PUFAs and breast cancer development, and aimed to emphasize the main molecular mechanisms involved in the modification of cell membrane structure and function, modulation of signal transduction pathways, gene expression regulation, and antiangiogenic and antimetastatic effects. Furthermore, the anticancer role of ω-3 PUFAs through the modulation of microRNA expression levels was also reviewed.
Collapse
Affiliation(s)
- Oana Zanoaga
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Ancuta Jurj
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Lajos Raduly
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania.,Department of Physiopathology, University of Agricultural Science and Veterinary Medicine, 400372 Cluj-Napoca, Romania
| | - Roxana Cojocneanu-Petric
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Enrique Fuentes-Mattei
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Oscar Wu
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.,Texas Tech University Honors College, McClellan Hall, Lubbock, TX 79409, USA
| | - Cornelia Braicu
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Claudia Diana Gherman
- Surgical Clinic II Hospital, 400006 Cluj-Napoca, Romania.,Department of Surgery, Iuliu Haţieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania.,MEDFUTURE-Research Center for Advanced Medicine, University of Medicine and Pharmacy Iuliu-Hatieganu, 400349 Cluj-Napoca, Romania.,Department of Functional Genomics, Proteomics and Experimental Pathology, Prof Dr Ion Chiricuta Oncology Institute, 400015 Cluj-Napoca, Romania
| |
Collapse
|
87
|
HPV16 E6 Promotes Breast Cancer Proliferation via Upregulation of COX-2 Expression. BIOMED RESEARCH INTERNATIONAL 2017; 2017:2948467. [PMID: 29250535 PMCID: PMC5700552 DOI: 10.1155/2017/2948467] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 06/10/2017] [Accepted: 07/03/2017] [Indexed: 01/10/2023]
Abstract
Background. Breast cancer remains the leading cause of cancer-related mortality worldwide. It has been indicated that human papillomaviruses 16 (HPV16) might participate in the pathogenesis and development of breast cancer. However, the detected rate of HPV16 varies with region. We will investigate HPV16 E6 expression in North China and explore the effects and mechanism of HPV16 E6 on breast cancer proliferation in this study. Methods. The expressions of HPV16 E6 and COX-2 in paraffin-embedded tissues of the invasive ductal breast cancer were detected by qPCR and IHC. The effects of HPV16 E6 on breast cancer proliferation were determined by function studies. The mechanism of HPV16 E6 in promoting breast cancer proliferation was explored by Western blot and Dual-Luciferase Reporter Assay. Results. HPV16 E6 was positive in 28% invasive ductal breast carcinoma in North China; HPV16 E6 promoted breast cancer proliferation. Inhibition of COX-2 by siCOX-2 or Celecoxib attenuated the proliferation of breast cancer cells with HPV16 E6 expression; and the upregulation of COX-2 could be suppressed by the inhibition of NF-κB activity. Conclusion. HPV16 E6 promotes breast cancer proliferation by activation of NF-κB signaling pathway and increase of COX-2 expression. COX-2 will be a potential target for HPV16 E6-associated breast cancer.
Collapse
|
88
|
Zhang X, Harbeck N, Jeschke U, Doisneau-Sixou S. Influence of vitamin D signaling on hormone receptor status and HER2 expression in breast cancer. J Cancer Res Clin Oncol 2017; 143:1107-1122. [PMID: 28025696 DOI: 10.1007/s00432-016-2325-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 12/16/2016] [Indexed: 12/14/2022]
Abstract
PURPOSE Breast cancer is a significant global public health issue. It is the leading cause of death among women around the world, with an incidence increasing annually. In recent years, there has been more and more information in the literature regarding a protective role of vitamin D in cancer. Increasingly preclinical and clinical studies suggest that vitamin D optimal levels can reduce the risk of breast cancer development and regulate cancer-related pathways. METHOD In this review, we focus on the importance of vitamin D in breast cancers, discussing especially the influence of vitamin D signaling on estrogen receptor and human epidermal growth factor receptor 2 (HER2), two major biomarkers of breast cancer today. CONCLUSION We discuss the possibility of actual and future targeted therapeutic approaches for vitamin D signaling in breast cancer.
Collapse
Affiliation(s)
- Xi Zhang
- Brustzentrum der Universität München, Klinik und Poliklinik für Frauenheilkunde und Geburtshilfe, Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, Maistraße 11, 80337, Munich, Germany
| | - Nadia Harbeck
- Brustzentrum der Universität München, Klinik und Poliklinik für Frauenheilkunde und Geburtshilfe, Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, Maistraße 11, 80337, Munich, Germany
| | - Udo Jeschke
- Brustzentrum der Universität München, Klinik und Poliklinik für Frauenheilkunde und Geburtshilfe, Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, Maistraße 11, 80337, Munich, Germany
| | - Sophie Doisneau-Sixou
- Brustzentrum der Universität München, Klinik und Poliklinik für Frauenheilkunde und Geburtshilfe, Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, Maistraße 11, 80337, Munich, Germany.
- Faculté des Sciences Pharmaceutiques, Université Paul Sabatier Toulouse III, 31062, Toulouse Cedex 09, France.
| |
Collapse
|
89
|
Anti-Inflammatory Mechanism Involved in Pomegranate-Mediated Prevention of Breast Cancer: the Role of NF-κB and Nrf2 Signaling Pathways. Nutrients 2017; 9:nu9050436. [PMID: 28452959 PMCID: PMC5452166 DOI: 10.3390/nu9050436] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 04/05/2017] [Indexed: 12/14/2022] Open
Abstract
Pomegranate (Punica granatum L.), a nutrient-rich unique fruit, has been used for centuries for the prevention and treatment of various inflammation-driven diseases. Based on our previous study, a characterized pomegranate emulsion (PE) exhibited a striking inhibition of dimethylbenz(a)anthracene (DMBA)-initiated rat mammary tumorigenesis via antiproliferative and apoptosis-inducing mechanisms. The objective of the present work is to investigate the anti-inflammatory mechanism of action of PE during DMBA rat mammary carcinogenesis by evaluating the expression of cyclooxygenase-2 (COX-2), heat shock protein 90 (HSP90), nuclear factor-κB (NF-κB) and nuclear factor erythroid 2p45 (NF-E2)-related factor 2 (Nrf2). Mammary tumor samples were harvested from our previous chemopreventive study in which PE (0.2–5.0 g/kg) was found to reduce mammary tumorigenesis in a dose-dependent manner. The expressions of COX-2, HSP90, NF-κB, inhibitory κBα (IκBα) and Nrf2 were detected by immunohistochemical techniques. PE decreased the expression of COX-2 and HSP90, prevented the degradation of IκBα, hindered the translocation of NF-κB from cytosol to nucleus and increased the expression and nuclear translocation of Nrf2 during DMBA-induced mammary tumorigenesis. These findings, together with our previous results, indicate that PE-mediated prevention of DMBA-evoked mammary carcinogenesis may involve anti-inflammatory mechanisms through concurrent but differential regulation of two interrelated molecular pathways, namely NF-κB and Nrf2 signaling.
Collapse
|
90
|
Kanda Y, Osaki M, Okada F. Chemopreventive Strategies for Inflammation-Related Carcinogenesis: Current Status and Future Direction. Int J Mol Sci 2017; 18:E867. [PMID: 28422073 PMCID: PMC5412448 DOI: 10.3390/ijms18040867] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 04/14/2017] [Accepted: 04/17/2017] [Indexed: 02/07/2023] Open
Abstract
A sustained and chronically-inflamed environment is characterized by the presence of heterogeneous inflammatory cellular components, including neutrophils, macrophages, lymphocytes and fibroblasts. These infiltrated cells produce growth stimulating mediators (inflammatory cytokines and growth factors), chemotactic factors (chemokines) and genotoxic substances (reactive oxygen species and nitrogen oxide) and induce DNA damage and methylation. Therefore, chronic inflammation serves as an intrinsic niche for carcinogenesis and tumor progression. In this article, we summarize the up-to-date findings regarding definitive/possible causes and mechanisms of inflammation-related carcinogenesis derived from experimental and clinical studies. We also propose 10 strategies, as well as candidate agents for the prevention of inflammation-related carcinogenesis.
Collapse
Affiliation(s)
- Yusuke Kanda
- Division of Pathological Biochemistry, Tottori University Faculty of Medicine, Yonago, Tottori 683-8503, Japan.
| | - Mitsuhiko Osaki
- Division of Pathological Biochemistry, Tottori University Faculty of Medicine, Yonago, Tottori 683-8503, Japan.
- Chromosome Engineering Research Center, Tottori University, Yonago, Tottori 683-8503, Japan.
| | - Futoshi Okada
- Division of Pathological Biochemistry, Tottori University Faculty of Medicine, Yonago, Tottori 683-8503, Japan.
- Chromosome Engineering Research Center, Tottori University, Yonago, Tottori 683-8503, Japan.
| |
Collapse
|
91
|
Gao S, Sun Y, Liu X, Zhang D, Yang X. EpCAM and COX-2 expression are positively correlated in human breast cancer. Mol Med Rep 2017; 15:3755-3760. [DOI: 10.3892/mmr.2017.6447] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 02/09/2017] [Indexed: 11/06/2022] Open
|
92
|
Feng J, Cao Z, Wang L, Wan Y, Peng N, Wang Q, Chen X, Zhou Y, Zhu Y. Inducible GBP5 Mediates the Antiviral Response via Interferon-Related Pathways during Influenza A Virus Infection. J Innate Immun 2017; 9:419-435. [PMID: 28376501 DOI: 10.1159/000460294] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 02/08/2017] [Indexed: 12/18/2022] Open
Abstract
Guanylate binding protein (GBP) 5 belongs to the GBP family, which is involved in important cellular processes, including signal transduction, translation, vesicle trafficking, and exocytosis. Structurally, GBPs display a high degree of homology and share highly conserved GTP-binding or hydrolysis domains. GBP5 was reported to be a critical cellular factor in inflammasome assembly. However, little is known about its role in the host antiviral innate immune response. In this study, we found that GBP5 expression was significantly elevated in influenza patients and influenza A virus-infected A549 human lung epithelial cells. The overexpression of GBP5 inhibited virus replication by enhancing the expression of virus-induced interferon (IFN) and IFN-related effectors. Knockdown of GBP5 had the opposite effect. Moreover, GBP5 enhanced endogenous IFN expression by interacting with the NF-κB-essential modulator complex and stimulating NF-κB signaling. Additionally, the expression of proinflammatory factors, such as IL-6, IL-8, tumor necrosis factor-α, cyclooxygenase-2, and inducible nitric oxide synthase, was also activated by GBP5. Taken together, our results reveal that GBP5 inhibited virus replication through the activation of IFN signaling and proinflammatory factors.
Collapse
Affiliation(s)
- Jian Feng
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | | | | | | | | | | | | | | | | |
Collapse
|
93
|
Mon NN, Senga T, Ito S. Interleukin-1β activates focal adhesion kinase and Src to induce matrix metalloproteinase-9 production and invasion of MCF-7 breast cancer cells. Oncol Lett 2016; 13:955-960. [PMID: 28356984 DOI: 10.3892/ol.2016.5521] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 10/14/2016] [Indexed: 11/06/2022] Open
Abstract
Interleukin-1β (IL-1b) is a pleiotropic cytokine that is important in tumor progression and invasion. Matrix metalloproteinase-9 (MMP-9), which is a secreted matrix-degrading enzyme, is one of the key regulators of tumor invasion and metastasis. The current report indicated that IL-1b promotes MMP-9 production and cell invasion in non-metastatic MCF-7 breast cancer cells. IL-1b activated focal adhesion kinase (FAK) and proto-oncogene tyrosine-protein kinase Src (Src). Moreover, inhibiting the Src/FAK pathway reduced the IL-1b-induced production of MMP-9 and cell invasion. To investigate the functional role of FAK in MMP-9 production cell lines expressing mutant FAK in FAK knock-out mouse fibroblasts were generated. In wild-type FAK-expressing cells, MMP-9 production was induced by IL-1b stimulation. By contrast, IL-1b-induced MMP-9 production was abrogated in FAK knock-out, FAK Y397F, FAK Y925F, and kinase dead mutant-expressing cells. Therefore the results of the current study indicate that FAK and Src kinases are activated by IL-1b and play a critical role in MMP-9 production and tumor cell invasion.
Collapse
Affiliation(s)
- Naing Naing Mon
- Division of Cancer Biology, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya 466-8550, Japan
| | - Takeshi Senga
- Division of Cancer Biology, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya 466-8550, Japan
| | - Satoko Ito
- Division of Cancer Biology, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya 466-8550, Japan
| |
Collapse
|
94
|
Kim S, Campbell J, Yoo W, Taylor JA, Sandler DP. Systemic Levels of Estrogens and PGE 2 Synthesis in Relation to Postmenopausal Breast Cancer Risk. Cancer Epidemiol Biomarkers Prev 2016; 26:383-388. [PMID: 27864342 DOI: 10.1158/1055-9965.epi-16-0556] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 09/23/2016] [Accepted: 10/18/2016] [Indexed: 12/14/2022] Open
Abstract
Background: Prostaglandin E2 (PGE2) induces aromatase expression in adipose tissue, leading to increased estrogen production that may promote the development and progression of breast cancer. However, few studies have simultaneously investigated systemic levels of PGE2 and estrogen in relation to postmenopausal breast cancer risk.Methods: Here, we determined urinary estrogen metabolites (EM) using mass spectrometry in a case-cohort study (295 incident breast cancer cases and 294 subcohort members), and using linear regression estimated the effect of urinary levels of a major PGE2 metabolite (PGE-M) on EMs. HRs for the risk of developing breast cancer in relation to PGE-M and EMs were compared between Cox regression models with and without mutual adjustment.Results: PGE-M was a significant predictor of estrone (E1), but not estradiol (E2) levels in multivariable analysis. Elevated E2 levels were associated with an increased risk of developing breast cancer [HRQ5vs.Q1, 1.54; 95% confidence interval (CI), 1.01-2.35], and this association remained unchanged after adjustment for PGE-M (HRQ5vs.Q1, 1.52; 95% CI, 0.99-2.33). Similarly, elevated levels of PGE-M were associated with increased risk of developing breast cancer (HRQ4vs.Q1, 2.01; 95% CI, 1.01-4.29), and this association was only nominally changed after consideration of E1 or E2 levels.Conclusions: Urinary levels of PGE-M and estrogens were independently associated with future risk of developing breast cancer among these postmenopausal women.Impact: Increased breast cancer risk associated with PGE-M might not be fully explained by the estrogens-breast cancer association alone but also by additional effects related to inflammation. Cancer Epidemiol Biomarkers Prev; 26(3); 383-8. ©2016 AACR.
Collapse
Affiliation(s)
- Sangmi Kim
- Medical College of Georgia, Department of Medicine, Section of Hematology/Oncology, Augusta University Georgia Cancer Center, Augusta, Georgia.
| | - Jeff Campbell
- Medical College of Georgia, Department of Medicine, Section of Hematology/Oncology, Augusta University Georgia Cancer Center, Augusta, Georgia
| | - Wonsuk Yoo
- Institute of Public and Preventive Health - Data Coordinating Center, Augusta University, Augusta, Georgia
| | - Jack A Taylor
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Dale P Sandler
- Epidemiology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| |
Collapse
|
95
|
Singh S, Awasthi M, Pandey VP, Dwivedi UN. Plant derived anti-cancerous secondary metabolites as multipronged inhibitor of COX, Topo, and aromatase: molecular modeling and dynamics simulation analyses. J Biomol Struct Dyn 2016; 35:3082-3097. [DOI: 10.1080/07391102.2016.1241720] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Swati Singh
- Bioinformatics Infrastructure Facility, Center of Excellence in Bioinformatics, Department of Biochemistry, University of Lucknow, Lucknow 226007, Uttar Pradesh, India
| | - Manika Awasthi
- Bioinformatics Infrastructure Facility, Center of Excellence in Bioinformatics, Department of Biochemistry, University of Lucknow, Lucknow 226007, Uttar Pradesh, India
| | - Veda P. Pandey
- Bioinformatics Infrastructure Facility, Center of Excellence in Bioinformatics, Department of Biochemistry, University of Lucknow, Lucknow 226007, Uttar Pradesh, India
| | - Upendra N. Dwivedi
- Bioinformatics Infrastructure Facility, Center of Excellence in Bioinformatics, Department of Biochemistry, University of Lucknow, Lucknow 226007, Uttar Pradesh, India
| |
Collapse
|
96
|
Cyclooxygenase-2: A Role in Cancer Stem Cell Survival and Repopulation of Cancer Cells during Therapy. Stem Cells Int 2016; 2016:2048731. [PMID: 27882058 PMCID: PMC5108861 DOI: 10.1155/2016/2048731] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 09/16/2016] [Accepted: 09/26/2016] [Indexed: 12/13/2022] Open
Abstract
Cyclooxygenase-2 (COX-2) is an inducible form of the enzyme that catalyses the synthesis of prostanoids, including prostaglandin E2 (PGE2), a major mediator of inflammation and angiogenesis. COX-2 is overexpressed in cancer cells and is associated with progressive tumour growth, as well as resistance of cancer cells to conventional chemotherapy and radiotherapy. These therapies are often delivered in multiple doses, which are spaced out to allow the recovery of normal tissues between treatments. However, surviving cancer cells also proliferate during treatment intervals, leading to repopulation of the tumour and limiting the effectiveness of the treatment. Tumour cell repopulation is a major cause of treatment failure. The central dogma is that conventional chemotherapy and radiotherapy selects resistant cancer cells that are able to reinitiate tumour growth. However, there is compelling evidence of an active proliferative response, driven by increased COX-2 expression and downstream PGE2 release, which contribute to the repopulation of tumours and poor patient outcome. In this review, we will examine the evidence for a role of COX-2 in cancer stem cell biology and as a mediator of tumour repopulation that can be molecularly targeted to overcome resistance to therapy.
Collapse
|
97
|
Dietz BM, Hajirahimkhan A, Dunlap TL, Bolton JL. Botanicals and Their Bioactive Phytochemicals for Women's Health. Pharmacol Rev 2016; 68:1026-1073. [PMID: 27677719 PMCID: PMC5050441 DOI: 10.1124/pr.115.010843] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Botanical dietary supplements are increasingly popular for women's health, particularly for older women. The specific botanicals women take vary as a function of age. Younger women will use botanicals for urinary tract infections, especially Vaccinium macrocarpon (cranberry), where there is evidence for efficacy. Botanical dietary supplements for premenstrual syndrome (PMS) are less commonly used, and rigorous clinical trials have not been done. Some examples include Vitex agnus-castus (chasteberry), Angelica sinensis (dong quai), Viburnum opulus/prunifolium (cramp bark and black haw), and Zingiber officinale (ginger). Pregnant women have also used ginger for relief from nausea. Natural galactagogues for lactating women include Trigonella foenum-graecum (fenugreek) and Silybum marianum (milk thistle); however, rigorous safety and efficacy studies are lacking. Older women suffering menopausal symptoms are increasingly likely to use botanicals, especially since the Women's Health Initiative showed an increased risk for breast cancer associated with traditional hormone therapy. Serotonergic mechanisms similar to antidepressants have been proposed for Actaea/Cimicifuga racemosa (black cohosh) and Valeriana officinalis (valerian). Plant extracts with estrogenic activities for menopausal symptom relief include Glycine max (soy), Trifolium pratense (red clover), Pueraria lobata (kudzu), Humulus lupulus (hops), Glycyrrhiza species (licorice), Rheum rhaponticum (rhubarb), Vitex agnus-castus (chasteberry), Linum usitatissimum (flaxseed), Epimedium species (herba Epimedii, horny goat weed), and Medicago sativa (alfalfa). Some of the estrogenic botanicals have also been shown to have protective effects against osteoporosis. Several of these botanicals could have additional breast cancer preventive effects linked to hormonal, chemical, inflammatory, and/or epigenetic pathways. Finally, although botanicals are perceived as natural safe remedies, it is important for women and their healthcare providers to realize that they have not been rigorously tested for potential toxic effects and/or drug/botanical interactions. Understanding the mechanism of action of these supplements used for women's health will ultimately lead to standardized botanical products with higher efficacy, safety, and chemopreventive properties.
Collapse
Affiliation(s)
- Birgit M Dietz
- University of Illinois at Chicago/National Institutes of Health Center for Botanical Dietary Supplements, Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois
| | - Atieh Hajirahimkhan
- University of Illinois at Chicago/National Institutes of Health Center for Botanical Dietary Supplements, Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois
| | - Tareisha L Dunlap
- University of Illinois at Chicago/National Institutes of Health Center for Botanical Dietary Supplements, Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois
| | - Judy L Bolton
- University of Illinois at Chicago/National Institutes of Health Center for Botanical Dietary Supplements, Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
98
|
Li W, Yue W, Wang H, Lai B, Yang X, Zhang C, Wang Y, Gu M. Cyclooxygenase-2 is associated with malignant phenotypes in human lung cancer. Oncol Lett 2016; 12:3836-3844. [PMID: 27895738 PMCID: PMC5104181 DOI: 10.3892/ol.2016.5207] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 07/12/2016] [Indexed: 12/27/2022] Open
Abstract
The objective of the present study was to investigate whether cyclooxygenase-2 (COX-2) is associated with malignancy, and to investigate its molecular mechanisms in human lung cancer tumor malignancy. The present study used RNA interference (RNAi) methodology and celecoxib, a COX-2 inhibitor, to investigate the effect of COX-2 knockdown on the proliferation and invasion abilities of lung cancer cells and the molecular mechanisms involved. Human lung adenocarcinoma A549-si10 and LTEP-A2 cells transfected with a specific small interfering RNA (A549-si10 and LTEP-A2-si10, respectively) grew more slowly compared with parental cell lines and cells transfected with pU6. The colony formation of A549-si10 and LTEP-A2-si10 cells was also reduced. In addition, A549-si10 and LTEP-A2-si10 cells were characterized by decreased metastatic and invasive abilities. The proliferation and invasive potential of parental A549 and LTEP-A2 cells was inhibited following treatment with celecoxib. In vivo, a COX-2 knockdown resulted in a decrease of proliferation and reduction of vascular endothelial growth factor (VEGF), matrix metalloproteinase-2 (MMP-2) and endothelial growth factor receptor (EGFR) expression in A549 xenografts. In conclusion, the present study revealed that COX-2 plays a extremely important role in tumor growth, infiltration and metastasis via the regulation of VEGF, MMP-2 and EGRF expression. Therefore, COX-2 is a potential therapeutic target for lung cancer.
Collapse
Affiliation(s)
- Weiying Li
- Department of Cellular & Molecular Biology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing 101149, P.R. China
| | - Wentao Yue
- Department of Cellular & Molecular Biology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing 101149, P.R. China
| | - Hui Wang
- Department of Cellular & Molecular Biology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing 101149, P.R. China
| | - Baitang Lai
- Department of Cellular & Molecular Biology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing 101149, P.R. China
| | - Xuehui Yang
- Department of Cellular & Molecular Biology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing 101149, P.R. China
| | - Chunyan Zhang
- Department of Cellular & Molecular Biology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing 101149, P.R. China
| | - Yue Wang
- Department of Cellular & Molecular Biology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing 101149, P.R. China
| | - Meng Gu
- Department of Cellular & Molecular Biology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing 101149, P.R. China
| |
Collapse
|
99
|
Kumar M, Dhatwalia SK, Dhawan DK. Role of angiogenic factors of herbal origin in regulation of molecular pathways that control tumor angiogenesis. Tumour Biol 2016; 37:14341-14354. [DOI: 10.1007/s13277-016-5330-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 09/06/2016] [Indexed: 12/19/2022] Open
|
100
|
Filipenko I, Schwalm S, Reali L, Pfeilschifter J, Fabbro D, Huwiler A, Zangemeister-Wittke U. Upregulation of the S1P 3 receptor in metastatic breast cancer cells increases migration and invasion by induction of PGE 2 and EP 2/EP 4 activation. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:1840-1851. [PMID: 27616330 DOI: 10.1016/j.bbalip.2016.09.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 09/02/2016] [Accepted: 09/07/2016] [Indexed: 12/20/2022]
Abstract
Breast cancer is one of the most common and devastating malignancies among women worldwide. Recent evidence suggests that malignant progression is also driven by processes involving the sphingolipid molecule sphingosine 1-phosphate (S1P) and its binding to cognate receptor subtypes on the cell surface. To investigate the effect of this interaction on the metastatic phenotype, we used the breast cancer cell line MDA-MB-231 and the sublines 4175 and 1833 derived from lung and bone metastases in nude mice, respectively. In both metastatic cell lines expression of the S1P3 receptor was strongly upregulated compared to the parental cells and correlated with higher S1P-induced intracellular calcium ([Ca2+]i), higher cyclooxygenase (COX)-2 and microsomal prostaglandin (PG) E2 synthase expression, and consequently with increased PGE2 synthesis. PGE2 synthesis was decreased by antagonists and siRNA against S1P3 and S1P2. Moreover, in parental MDA-MB-231 cells overexpression of S1P3 by cDNA transfection also increased PGE2 synthesis, but only after treatment with the DNA methyltransferase inhibitor 5-aza-2-deoxycytidine, indicating reversible silencing of the COX-2 promoter. Functionally, the metastatic sublines showed enhanced migration and Matrigel invasion in adapted Boyden chamber assays, which further increased by S1P stimulation. This response was abrogated by either S1P3 antagonism, COX-2 inhibition or PGE2 receptor 2 (EP2) and 4 (EP4) antagonism, but not by S1P2 antagonism. Our data demonstrate that in breast cancer cells overexpression of S1P3 and its activation by S1P has pro-inflammatory and pro-metastatic potential by inducing COX-2 expression and PGE2 signaling via EP2 and EP4.
Collapse
Affiliation(s)
- Iuliia Filipenko
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, CH-3011 Bern, Switzerland
| | - Stephanie Schwalm
- Pharmazentrum Frankfurt/ZAFES, Klinikum der Goethe Universität Frankfurt am Main, Theodor Stern Kai 7, D-60590 Frankfurt am Main, Germany
| | - Luca Reali
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, CH-3011 Bern, Switzerland
| | - Josef Pfeilschifter
- Pharmazentrum Frankfurt/ZAFES, Klinikum der Goethe Universität Frankfurt am Main, Theodor Stern Kai 7, D-60590 Frankfurt am Main, Germany
| | - Doriano Fabbro
- PIQUR Therapeutics AG, Hochbergstrasse 60C, CH-4057 Basel, Switzerland
| | - Andrea Huwiler
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, CH-3011 Bern, Switzerland.
| | - Uwe Zangemeister-Wittke
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, CH-3011 Bern, Switzerland.
| |
Collapse
|