1
|
Itakura H, Hata T, Okuzaki D, Takeda K, Iso K, Qian Y, Morimoto Y, Adachi T, Hirose H, Yokoyama Y, Ogino T, Miyoshi N, Takahashi H, Uemura M, Mizushima T, Hinoi T, Mori M, Doki Y, Eguchi H, Yamamoto H. Tumor-suppressive role of the musculoaponeurotic fibrosarcoma gene in colorectal cancer. iScience 2023; 26:106478. [PMID: 37091240 PMCID: PMC10119606 DOI: 10.1016/j.isci.2023.106478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 12/21/2022] [Accepted: 03/19/2023] [Indexed: 04/25/2023] Open
Abstract
Somatic cell reprogramming using the microRNAs miR-200c, miR-302s, and miR-369s leads to increased expression of cyclin-dependent kinase inhibitors in human colorectal cancer (CRC) cells and suppressed tumor growth. Here, we investigated whether these microRNAs inhibit colorectal tumorigenesis in CPC;Apc mice, which are prone to colon and rectal polyps. Repeated administration of microRNAs inhibited polyp formation. Microarray analysis indicated that c-MAF, which reportedly shows oncogene-like behavior in multiple myeloma and T cell lymphoma, decreased in tumor samples but increased in microRNA-treated normal mucosa. Immunohistochemistry identified downregulation of c-MAF as an early tumorigenesis event in CRC, with low c-MAF expression associated with poor prognosis. Of note, c-MAF expression and p53 protein levels were inversely correlated in CRC samples. c-MAF knockout led to enhanced tumor formation in azoxymethane/dextran sodium sulfate-treated mice, with activation of cancer-promoting genes. c-MAF may play a tumor-suppressive role in CRC development.
Collapse
Affiliation(s)
- Hiroaki Itakura
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Yamadaoka 2-2, Suita, Osaka 565-0871, Japan
| | - Tsuyoshi Hata
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Yamadaoka 2-2, Suita, Osaka 565-0871, Japan
| | - Daisuke Okuzaki
- Genome Information Research Centre, Research Institute for Microbial Diseases, Osaka University, Yamadaoka 3-1, Suita, Osaka 565-0871, Japan
- Laboratory of Human Immunology (Single Cell Genomics), WPI Immunology Research Center, Osaka University, Yamadaoka 3-1, Suita, Osaka 565-0871, Japan
| | - Koki Takeda
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Yamadaoka 2-2, Suita, Osaka 565-0871, Japan
| | - Kenji Iso
- Department of Molecular Pathology, Division of Health Sciences, Graduate School of Medicine, Osaka University, Yamadaoka 1-7, Suita, Osaka 565-0871, Japan
| | - Yamin Qian
- Department of Molecular Pathology, Division of Health Sciences, Graduate School of Medicine, Osaka University, Yamadaoka 1-7, Suita, Osaka 565-0871, Japan
| | - Yoshihiro Morimoto
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Yamadaoka 2-2, Suita, Osaka 565-0871, Japan
| | - Tomohiro Adachi
- Department of Surgery, Hiroshima City North Medical Center Asa Citizens Hospital, 1-2-1, Kameyama-minami, Asakita-ku, Horoshima 731-0293, Japan
| | - Haruka Hirose
- Department of Molecular Pathology, Division of Health Sciences, Graduate School of Medicine, Osaka University, Yamadaoka 1-7, Suita, Osaka 565-0871, Japan
| | - Yuhki Yokoyama
- Department of Molecular Pathology, Division of Health Sciences, Graduate School of Medicine, Osaka University, Yamadaoka 1-7, Suita, Osaka 565-0871, Japan
| | - Takayuki Ogino
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Yamadaoka 2-2, Suita, Osaka 565-0871, Japan
| | - Norikatsu Miyoshi
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Yamadaoka 2-2, Suita, Osaka 565-0871, Japan
| | - Hidekazu Takahashi
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Yamadaoka 2-2, Suita, Osaka 565-0871, Japan
| | - Mamoru Uemura
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Yamadaoka 2-2, Suita, Osaka 565-0871, Japan
| | - Tsunekazu Mizushima
- Department of Surgery, Osaka Police Hospital, 10-31, Kitayama-town, Tennoji-ku, Osaka city, Osaka 543-0035, Japan
| | - Takao Hinoi
- Department of Clinical and Molecular Genetics, Hiroshima University Hospital, 1-2-3, Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Masaki Mori
- Department of Surgery, Graduate School of Medical Sciences, Tokai University, 143, Shimokasuya, Isehara, Kanagawa 259-1193, Japan
| | - Yuichiro Doki
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Yamadaoka 2-2, Suita, Osaka 565-0871, Japan
| | - Hidetoshi Eguchi
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Yamadaoka 2-2, Suita, Osaka 565-0871, Japan
| | - Hirofumi Yamamoto
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Yamadaoka 2-2, Suita, Osaka 565-0871, Japan
- Department of Molecular Pathology, Division of Health Sciences, Graduate School of Medicine, Osaka University, Yamadaoka 1-7, Suita, Osaka 565-0871, Japan
- Corresponding author
| |
Collapse
|
2
|
Pei H, Wu S, Zheng L, Wang H, Zhang X. Identification of the active compounds and their mechanisms of medicinal and edible Shanzha based on network pharmacology and molecular docking. J Food Biochem 2021; 46:e14020. [PMID: 34825377 DOI: 10.1111/jfbc.14020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 10/30/2021] [Accepted: 11/02/2021] [Indexed: 12/18/2022]
Abstract
Shanzha (Crataegus pinnatifida Bunge), an edible traditional Chinese medicine (TCM), has an effect on dyspepsia. However, the investigations of the pharmacological effects have not been carried out. This study aimed to identify the potential targets and pharmacological mechanisms of Shanzha in the treatment of dyspepsia by network pharmacology and molecular docking. Five active compounds and 13 key targets were obtained by a set of bioinformatics assays. Vitexin 7-glucoside, suchilactone, and 20-hexadecanoylingenol were the main compounds acting on dyspepsia. The key targets were prostaglandin-endoperoxide synthase 2 (PTGS2), serine/threonine-protein kinase mTOR (MTOR), heat shock protein HSP 90-alpha (HSP90AA1), mitogen-activated protein kinase 1 (MAPK1), MAPK3, E3 ubiquitin-protein ligase Mdm2 (MDM2), receptor tyrosine-protein kinase erbB-2 (ERBB2), caspase-3 (CASP3), matrix metalloproteinase-9 (MMP9), estrogen receptor (ESR1), tumor necrosis factor (TNF), phosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit alpha isoform (PIK3CA), and peroxisome proliferator-activated receptor gamma (PPARG), which played the vital roles in TNF, prostate cancer, thyroid hormone, hepatitis B and estrogen signaling pathway. The molecular mechanisms of Shanzha regulating dyspepsia were mainly related to reduction of inflammatory response, controlling cell proliferation and survival, increasing intestinal moisture, and promoting intestinal motility. PRACTICAL APPLICATIONS: Shanzha has been used as an edible TCM to improve digestion for a long time. However, the ingredients and mechanisms of Shanzha in the treatment of dyspepsia are not clear. In this research, network pharmacological analysis integrated with molecular docking was conducted to investigate the molecular mechanism. The results suggested that the core targets alleviated dyspepsia by reducing the intestinal inflammatory response, increasing intestinal movement, controlling cell physiological activities, and reducing constipation. In summary, this study demonstrated the multiple compounds, targets, and pathways characteristics of Shanzha in the treatment of dyspepsia, which may provide guidance and foundations for further application of edible medicine.
Collapse
Affiliation(s)
- Huimin Pei
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, China
| | - Shaokang Wu
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, China
| | - Lijun Zheng
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, China
| | - Hanxun Wang
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, China
| | - Xiangrong Zhang
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
3
|
Okada F, Izutsu R, Goto K, Osaki M. Inflammation-Related Carcinogenesis: Lessons from Animal Models to Clinical Aspects. Cancers (Basel) 2021; 13:cancers13040921. [PMID: 33671768 PMCID: PMC7926701 DOI: 10.3390/cancers13040921] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/11/2021] [Accepted: 02/12/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary In multicellular organisms, inflammation is the body’s most primitive and essential protective response against any external agent. Inflammation, however, not only causes various modern diseases such as cardiovascular disorders, neurological disorders, autoimmune diseases, metabolic syndrome, infectious diseases, and cancer but also shortens the healthy life expectancy. This review focuses on the onset of carcinogenesis due to chronic inflammation caused by pathogen infections and inhalation/ingestion of foreign substances. This study summarizes animal models associated with inflammation-related carcinogenesis by organ. By determining factors common to inflammatory carcinogenesis models, we examined strategies for the prevention and treatment of inflammatory carcinogenesis in humans. Abstract Inflammation-related carcinogenesis has long been known as one of the carcinogenesis patterns in humans. Common carcinogenic factors are inflammation caused by infection with pathogens or the uptake of foreign substances from the environment into the body. Inflammation-related carcinogenesis as a cause for cancer-related death worldwide accounts for approximately 20%, and the incidence varies widely by continent, country, and even region of the country and can be affected by economic status or development. Many novel approaches are currently available concerning the development of animal models to elucidate inflammation-related carcinogenesis. By learning from the oldest to the latest animal models for each organ, we sought to uncover the essential common causes of inflammation-related carcinogenesis. This review confirmed that a common etiology of organ-specific animal models that mimic human inflammation-related carcinogenesis is prolonged exudation of inflammatory cells. Genotoxicity or epigenetic modifications by inflammatory cells resulted in gene mutations or altered gene expression, respectively. Inflammatory cytokines/growth factors released from inflammatory cells promote cell proliferation and repair tissue injury, and inflammation serves as a “carcinogenic niche”, because these fundamental biological events are common to all types of carcinogenesis, not just inflammation-related carcinogenesis. Since clinical strategies are needed to prevent carcinogenesis, we propose the therapeutic apheresis of inflammatory cells as a means of eliminating fundamental cause of inflammation-related carcinogenesis.
Collapse
Affiliation(s)
- Futoshi Okada
- Division of Experimental Pathology, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan; (R.I.); (K.G.); (M.O.)
- Chromosome Engineering Research Center, Tottori University, Yonago 683-8503, Japan
- Correspondence: ; Tel.: +81-859-38-6241
| | - Runa Izutsu
- Division of Experimental Pathology, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan; (R.I.); (K.G.); (M.O.)
| | - Keisuke Goto
- Division of Experimental Pathology, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan; (R.I.); (K.G.); (M.O.)
- Division of Gastrointestinal and Pediatric Surgery, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan
| | - Mitsuhiko Osaki
- Division of Experimental Pathology, Faculty of Medicine, Tottori University, Yonago 683-8503, Japan; (R.I.); (K.G.); (M.O.)
- Chromosome Engineering Research Center, Tottori University, Yonago 683-8503, Japan
| |
Collapse
|
4
|
Guey B, Bodnar-Wachtel M, Drouillard A, Eberhardt A, Pratviel M, Goutagny N, Bendriss-Vermare N, Puisieux I, Caux C, Walzer T, Petrilli V. Inflammasome Deletion Promotes Anti-tumor NK Cell Function in an IL-1/IL-18 Independent Way in Murine Invasive Breast Cancer. Front Oncol 2020; 10:1683. [PMID: 33042810 PMCID: PMC7526436 DOI: 10.3389/fonc.2020.01683] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 07/29/2020] [Indexed: 12/22/2022] Open
Abstract
Inflammasomes are molecular complexes that trigger an inflammatory response upon detection of pathogens or danger signals. Recent studies suggest that they are also involved in cancer progression. However, their roles during tumorigenesis remain poorly understood and controversial. Here, we investigated whether inflammasome activation supports mammary tumor growth. Using mouse models of invasive breast cancer, our results demonstrate that the absence of a functional inflammasome impairs tumor growth. Importantly, tumors implanted into inflammasome-deficient mice recruited significantly less neutrophils and more natural killer (NK) cells, and these latter cells displayed a more active phenotype. Interestingly, NK cell depletion abolished the anti-tumoral effect observed in inflammasome-deficient mice, although inflammasome-regulated cytokine neutralization had no effect. Thus, our work identifies a novel role for the inflammasome in supporting mammary tumor growth by attenuating NK cell recruitment and activity. These results suggest that inflammasome inhibition could be a putative target for treating invasive breast cancers.
Collapse
Affiliation(s)
- Baptiste Guey
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR5286, Université de Lyon, Université Lyon 1, Centre Léon Bérard, Lyon, France
| | - Mélanie Bodnar-Wachtel
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR5286, Université de Lyon, Université Lyon 1, Centre Léon Bérard, Lyon, France
| | - Annabelle Drouillard
- Centre International de Recherche en Infectiologie, INSERM U1111 - CNRS UMR5308, Université de Lyon, ENS de Lyon, Université Lyon 1, Lyon, France
| | - Anaïs Eberhardt
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR5286, Université de Lyon, Université Lyon 1, Centre Léon Bérard, Lyon, France
| | - Manon Pratviel
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR5286, Université de Lyon, Université Lyon 1, Centre Léon Bérard, Lyon, France
| | - Nadège Goutagny
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR5286, Université de Lyon, Université Lyon 1, Centre Léon Bérard, Lyon, France
| | - Nathalie Bendriss-Vermare
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR5286, Université de Lyon, Université Lyon 1, Centre Léon Bérard, Lyon, France
| | - Isabelle Puisieux
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR5286, Université de Lyon, Université Lyon 1, Centre Léon Bérard, Lyon, France
| | - Christophe Caux
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR5286, Université de Lyon, Université Lyon 1, Centre Léon Bérard, Lyon, France
| | - Thierry Walzer
- Centre International de Recherche en Infectiologie, INSERM U1111 - CNRS UMR5308, Université de Lyon, ENS de Lyon, Université Lyon 1, Lyon, France
| | - Virginie Petrilli
- Centre de Recherche en Cancérologie de Lyon, INSERM U1052, CNRS UMR5286, Université de Lyon, Université Lyon 1, Centre Léon Bérard, Lyon, France
| |
Collapse
|
5
|
Improvement in the Anti-Tumor Efficacy of Doxorubicin Nanosponges in In Vitro and in Mice Bearing Breast Tumor Models. Cancers (Basel) 2020; 12:cancers12010162. [PMID: 31936526 PMCID: PMC7016577 DOI: 10.3390/cancers12010162] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 01/07/2020] [Accepted: 01/08/2020] [Indexed: 12/14/2022] Open
Abstract
Doxorubicin (DOX) is an anthracycline widely used in cancer therapy and in particular in breast cancer treatment. The treatment with DOX appears successful, but it is limited by a severe cardiotoxicity. This work evaluated the in vitro and in vivo anticancer effect of a new formulation of β-cyclodextrin nanosponges containing DOX (BNS-DOX). The BNS-DOX effectiveness was evaluated in human and mouse breast cancer cell lines in vitro in terms of effect on cell growth, cell cycle distribution, and apoptosis induction; and in vivo in BALB-neuT mice developing spontaneous breast cancer in terms of biodistribution, cancer growth inhibition, and heart toxicity. BNS-DOX significantly inhibited cancer cell proliferation, through the induction of apoptosis, with higher efficiency than free DOX. The breast cancer growth in BALB-neuT mice was inhibited by 60% by a BNS-DOX dose five times lower than the DOX therapeutic dose, with substantial reduction of tumor neoangiogenesis and lymphangiogenesis. Biodistribution after BNS-DOX treatment revealed a high accumulation of DOX in the tumor site and a low accumulation in the hearts of mice. Results indicated that use of BNS may be an efficient strategy to deliver DOX in the treatment of breast cancer, since it improves the anti-cancer effectiveness and reduces cardiotoxicity.
Collapse
|
6
|
Liu G, Khanna V, Kirtane A, Grill A, Panyam J. Chemopreventive efficacy of oral curcumin: a prodrug hypothesis. FASEB J 2019; 33:9453-9465. [DOI: 10.1096/fj.201900166r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Garvey Liu
- Department of Pharmaceutics College of Pharmacy University of Minnesota Minneapolis Minnesota USA
| | - Vidhi Khanna
- Department of Pharmaceutics College of Pharmacy University of Minnesota Minneapolis Minnesota USA
| | - Ameya Kirtane
- Department of Pharmaceutics College of Pharmacy University of Minnesota Minneapolis Minnesota USA
| | - Alex Grill
- Department of Pharmaceutics College of Pharmacy University of Minnesota Minneapolis Minnesota USA
| | - Jayanth Panyam
- Department of Pharmaceutics College of Pharmacy University of Minnesota Minneapolis Minnesota USA
| |
Collapse
|
7
|
Liu S, Lee JS, Jie C, Park MH, Iwakura Y, Patel Y, Soni M, Reisman D, Chen H. HER2 Overexpression Triggers an IL1α Proinflammatory Circuit to Drive Tumorigenesis and Promote Chemotherapy Resistance. Cancer Res 2018; 78:2040-2051. [PMID: 29382706 DOI: 10.1158/0008-5472.can-17-2761] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 11/14/2017] [Accepted: 01/26/2018] [Indexed: 01/16/2023]
Abstract
Systemic inflammation in breast cancer correlates with poor prognosis, but the molecular underpinnings of this connection are not well understood. In this study, we explored the relationship between HER2 overexpression, inflammation, and expansion of the mammary stem/progenitor and cancer stem-like cell (CSC) population in breast cancer. HER2-positive epithelial cells initiated and sustained an inflammatory milieu needed to promote tumorigenesis. HER2 induced a feedforward activation loop of IL1α and IL6 that stimulated NFκB and STAT3 pathways for generation and maintenance of breast CSC. In mice, Il1a genetic deficiency delayed MMTV-Her2-induced tumorigenesis and reduced inflammatory cytokine expression as well as CSC in primary tumors. In clinical specimens of human breast tumor tissues, tissue microarray analysis revealed a strong positive correlation between IL1α/IL6 expression and CSC-positive phenotype. Pharmacologic blockade of IL1α signaling reduced the CSC population and improved chemotherapeutic efficacy. Our findings suggest new therapeutic or prevention strategies for HER2-positive breast cancers.Significance: IL1α signaling driven by HER2 promotes chronic inflammation needed to support cancer stem-like cell maintenance in HER2-positive breast cancers. Cancer Res; 78(8); 2040-51. ©2018 AACR.
Collapse
Affiliation(s)
- Shou Liu
- Department of Biological Science, University of South Carolina, Columbia, South Carolina.,Center for Colon Cancer Research, University of South Carolina, Columbia, South Carolina
| | - Ji Shin Lee
- Department of Pathology, Chonnam National University Hwasun Hospital, Jeonnam, Republic of Korea
| | - Chunfa Jie
- Master of Science in Biomedical Sciences Program, Des Moines University, Des Moines, Iowa
| | - Min Ho Park
- Department of Surgery, Chonnam National University Hwasun Hospital, Jeonnam, Republic of Korea
| | - Yoichiro Iwakura
- Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Yogin Patel
- Department of Biological Science, University of South Carolina, Columbia, South Carolina.,Center for Colon Cancer Research, University of South Carolina, Columbia, South Carolina
| | - Mithil Soni
- Department of Biological Science, University of South Carolina, Columbia, South Carolina.,Center for Colon Cancer Research, University of South Carolina, Columbia, South Carolina
| | - David Reisman
- Department of Biological Science, University of South Carolina, Columbia, South Carolina.,Center for Colon Cancer Research, University of South Carolina, Columbia, South Carolina
| | - Hexin Chen
- Department of Biological Science, University of South Carolina, Columbia, South Carolina. .,Center for Colon Cancer Research, University of South Carolina, Columbia, South Carolina
| |
Collapse
|
8
|
Reiter RJ, Rosales-Corral SA, Tan DX, Acuna-Castroviejo D, Qin L, Yang SF, Xu K. Melatonin, a Full Service Anti-Cancer Agent: Inhibition of Initiation, Progression and Metastasis. Int J Mol Sci 2017; 18:E843. [PMID: 28420185 PMCID: PMC5412427 DOI: 10.3390/ijms18040843] [Citation(s) in RCA: 313] [Impact Index Per Article: 44.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 04/05/2017] [Accepted: 04/06/2017] [Indexed: 12/21/2022] Open
Abstract
There is highly credible evidence that melatonin mitigates cancer at the initiation, progression and metastasis phases. In many cases, the molecular mechanisms underpinning these inhibitory actions have been proposed. What is rather perplexing, however, is the large number of processes by which melatonin reportedly restrains cancer development and growth. These diverse actions suggest that what is being observed are merely epiphenomena of an underlying more fundamental action of melatonin that remains to be disclosed. Some of the arresting actions of melatonin on cancer are clearly membrane receptor-mediated while others are membrane receptor-independent and involve direct intracellular actions of this ubiquitously-distributed molecule. While the emphasis of melatonin/cancer research has been on the role of the indoleamine in restraining breast cancer, this is changing quickly with many cancer types having been shown to be susceptible to inhibition by melatonin. There are several facets of this research which could have immediate applications at the clinical level. Many studies have shown that melatonin's co-administration improves the sensitivity of cancers to inhibition by conventional drugs. Even more important are the findings that melatonin renders cancers previously totally resistant to treatment sensitive to these same therapies. Melatonin also inhibits molecular processes associated with metastasis by limiting the entrance of cancer cells into the vascular system and preventing them from establishing secondary growths at distant sites. This is of particular importance since cancer metastasis often significantly contributes to death of the patient. Another area that deserves additional consideration is related to the capacity of melatonin in reducing the toxic consequences of anti-cancer drugs while increasing their efficacy. Although this information has been available for more than a decade, it has not been adequately exploited at the clinical level. Even if the only beneficial actions of melatonin in cancer patients are its ability to attenuate acute and long-term drug toxicity, melatonin should be used to improve the physical wellbeing of the patients. The experimental findings, however, suggest that the advantages of using melatonin as a co-treatment with conventional cancer therapies would far exceed improvements in the wellbeing of the patients.
Collapse
Affiliation(s)
- Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health, San Antonio, TX 78229, USA.
| | - Sergio A Rosales-Corral
- Centro de Investigacion Biomedica de Occidente, Del Instituto Mexicano del Seguro Social, Guadalajara 44340, Mexico.
| | - Dun-Xian Tan
- Department of Cell Systems and Anatomy, UT Health, San Antonio, TX 78229, USA.
| | | | - Lilan Qin
- Department of Cell Systems and Anatomy, UT Health, San Antonio, TX 78229, USA.
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan, Medical University, Taichung 40201, Taiwan.
| | - Kexin Xu
- Department of Molecular Medicine, UT Health, San Antonio, TX 78229, USA.
| |
Collapse
|
9
|
Abstract
This chapter is focused on the role of the plasma form of platelet-activating factor-acetylhydrolase (PAF-AH), heretofore referred to as PAF-AH, in tumorigenic responses. Biochemical and other properties of this enzyme were discussed in detail in chapter "Plasma PAF-AH (PLA2G7): Biochemical Properties, Association with LDLs and HDLs, and Regulation of Expression" by Stafforini and in other chapters. Although phospholipases tend not to be drivers of tumorigenesis themselves, these enzymes and the lipid mediators whose levels they regulate interact with a variety of oncogenes and tumor suppressors [1]. Like other phospholipases, the functions of PAF-AH in cancer likely are related to its ability to regulate the levels of lipid mediators that participate in cellular processes related to initial tumorigenic events (e.g., proliferation, growth, inflammation) and/or spreading of the disease (e.g., matrix metalloproteinase secretion, actin cytoskeleton reorganization, migration, and angiogenesis) [1]. The importance of substrates and products of PAF-AH on key cellular functions has been evaluated in cell-based analyses which revealed that these metabolites can have pro- and antitumorigenic functions. Studies in genetically engineered mice lacking PAF-AH expression and genetic manipulation of PAF-AH levels in cancer cells demonstrated diverse functions of the protein in models of melanoma, prostate cancer, colon cancer, and others. The following sections highlight lessons learned from studies in cell lines and in mouse models regarding the diversity of functions of PAF-AH in cancer, and the potential of PAFAH transcripts, protein, and/or activity levels to become cancer biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Diana M Stafforini
- Huntsman Cancer Institute and Division of Cardiovascular Medicine, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA.
| |
Collapse
|
10
|
Cornelissen B, Able S, Kartsonaki C, Kersemans V, Allen PD, Cavallo F, Cazier JB, Iezzi M, Knight J, Muschel R, Smart S, Vallis KA. Imaging DNA damage allows detection of preneoplasia in the BALB-neuT model of breast cancer. J Nucl Med 2014; 55:2026-31. [PMID: 25453049 DOI: 10.2967/jnumed.114.142083] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
UNLABELLED A prominent feature of many human cancers is oncogene-driven activation of the DNA damage response (DDR) during early tumorigenesis. It has been shown previously that noninvasive imaging of the phosphorylated histone H2A variant H2AX, γH2AX, a DNA damage signaling protein, is possible using (111)In-labeled anti-γH2AX antibody conjugated to the cell-penetrating peptide transactivator of transcription (TAT). The purpose of this study was to investigate whether (111)In-anti-γH2AX-TAT detects the DDR during mammary oncogenesis in BALB-neuT mice. METHODS Mammary fat pads from BALB-neuT and wild-type mice (age, 40-106 d) were immunostained for γH2AX. (111)In-anti-γH2AX-TAT or a control probe was administered intravenously to BALB-neuT mice. SPECT was performed weekly and compared with tumor detection using palpation and dynamic contrast-enhanced MR imaging. RESULTS γH2AX expression was elevated in hyperplastic lesions in the mammary fat pads of BALB-neuT mice aged 76-106 d, compared with normal fat pads from younger mice and carcinomas from older mice (13.5 ± 1.2 γH2AX foci/cell vs. 5.2 ± 1.5 [P < 0.05] and 3.4 ± 1.1 [P < 0.001], respectively). Serial SPECT imaging revealed a 2.5-fold increase in (111)In-anti-γH2AX-TAT accumulation in the mammary fat pads of mice aged 76-106 d, compared with control probe (P = 0.01). The median time to detection of neoplastic lesions by (111)In-anti-γH2AX-TAT (defined as >5% injected dose per gram of tissue) was 96 d, compared with 120 and 131 d for dynamic contrast-enhanced MR imaging and palpation, respectively (P < 0.001). CONCLUSION DDR imaging using (111)In-anti-γH2AX-TAT identified mammary tumors significantly earlier than MR imaging. Imaging the DDR holds promise for the detection of preneoplasia and as a technique for screening cancer-prone individuals.
Collapse
Affiliation(s)
- Bart Cornelissen
- Department of Oncology, CRUK/MRC Oxford Institute for Radiation Oncology, Oxford University, Oxford, United Kingdom
| | - Sarah Able
- Department of Oncology, CRUK/MRC Oxford Institute for Radiation Oncology, Oxford University, Oxford, United Kingdom
| | - Christiana Kartsonaki
- Department of Oncology, CRUK/MRC Oxford Institute for Radiation Oncology, Oxford University, Oxford, United Kingdom
| | - Veerle Kersemans
- Department of Oncology, CRUK/MRC Oxford Institute for Radiation Oncology, Oxford University, Oxford, United Kingdom
| | - P Danny Allen
- Department of Oncology, CRUK/MRC Oxford Institute for Radiation Oncology, Oxford University, Oxford, United Kingdom
| | - Federica Cavallo
- Molecular Biotechnology Center, University of Turin, Turin, Italy; and
| | - Jean-Baptiste Cazier
- Department of Oncology, CRUK/MRC Oxford Institute for Radiation Oncology, Oxford University, Oxford, United Kingdom
| | - Manuela Iezzi
- CeSI Foundation, University G. d' Annunzio, Chieti, Italy
| | - James Knight
- Department of Oncology, CRUK/MRC Oxford Institute for Radiation Oncology, Oxford University, Oxford, United Kingdom
| | - Ruth Muschel
- Department of Oncology, CRUK/MRC Oxford Institute for Radiation Oncology, Oxford University, Oxford, United Kingdom
| | - Sean Smart
- Department of Oncology, CRUK/MRC Oxford Institute for Radiation Oncology, Oxford University, Oxford, United Kingdom
| | - Katherine A Vallis
- Department of Oncology, CRUK/MRC Oxford Institute for Radiation Oncology, Oxford University, Oxford, United Kingdom
| |
Collapse
|
11
|
Gorczynski RM, Chen Z, Erin N, Khatri I, Podnos A. Comparison of immunity in mice cured of primary/metastatic growth of EMT6 or 4THM breast cancer by chemotherapy or immunotherapy. PLoS One 2014; 9:e113597. [PMID: 25409195 PMCID: PMC4237434 DOI: 10.1371/journal.pone.0113597] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 10/29/2014] [Indexed: 12/19/2022] Open
Abstract
PURPOSE We have compared cure from local/metastatic tumor growth in BALB/c mice receiving EMT6 or the poorly immunogenic, highly metastatic 4THM, breast cancer cells following manipulation of immunosuppressive CD200:CD200R interactions or conventional chemotherapy. METHODS We reported previously that EMT6 tumors are cured in CD200R1KO mice following surgical resection and immunization with irradiated EMT6 cells and CpG oligodeoxynucleotide (CpG), while wild-type (WT) animals developed pulmonary and liver metastases within 30 days of surgery. We report growth and metastasis of both EMT6 and a highly metastatic 4THM tumor in WT mice receiving iv infusions of Fab anti-CD200R1 along with CpG/tumor cell immunization. Metastasis was followed both macroscopically (lung/liver nodules) and microscopically by cloning tumor cells at limiting dilution in vitro from draining lymph nodes (DLN) harvested at surgery. We compared these results with local/metastatic tumor growth in mice receiving 4 courses of combination treatment with anti-VEGF and paclitaxel. RESULTS In WT mice receiving Fab anti-CD200R, no tumor cells are detectable following immunotherapy, and CD4+ cells produced increased TNFα/IL-2/IFNγ on stimulation with EMT6 in vitro. No long-term cure was seen following surgery/immunotherapy of 4THM, with both microscopic (tumors in DLN at limiting dilution) and macroscopic metastases present within 14 d of surgery. Chemotherapy attenuated growth/metastases in 4THM tumor-bearers and produced a decline in lung/liver metastases, with no detectable DLN metastases in EMT6 tumor-bearing mice-these latter mice nevertheless showed no significantly increased cytokine production after restimulation with EMT6 in vitro. EMT6 mice receiving immunotherapy were resistant to subsequent re-challenge with EMT6 tumor cells, but not those receiving curative chemotherapy. Anti-CD4 treatment caused tumor recurrence after immunotherapy, but produced no apparent effect in either EMT6 or 4THM tumor bearers after chemotherapy treatment. CONCLUSION Immunotherapy, but not chemotherapy, enhances CD4+ immunity and affords long-term control of breast cancer growth and resistance to new tumor foci.
Collapse
MESH Headings
- Animals
- Antibodies/therapeutic use
- Antigens, CD/immunology
- Antineoplastic Agents, Phytogenic/therapeutic use
- Breast Neoplasms/pathology
- Breast Neoplasms/surgery
- Breast Neoplasms/therapy
- CD4 Antigens/immunology
- CD4-Positive T-Lymphocytes/cytology
- CD4-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/cytology
- CD8-Positive T-Lymphocytes/immunology
- Cell Line, Tumor
- Female
- Humans
- Immunoglobulin Fab Fragments/therapeutic use
- Immunotherapy
- Interferon-gamma/metabolism
- Interleukin-2/metabolism
- Liver Neoplasms/pathology
- Liver Neoplasms/prevention & control
- Liver Neoplasms/secondary
- Lung Neoplasms/pathology
- Lung Neoplasms/prevention & control
- Lung Neoplasms/secondary
- Lymph Nodes/pathology
- Mammary Neoplasms, Experimental/pathology
- Mammary Neoplasms, Experimental/surgery
- Mammary Neoplasms, Experimental/therapy
- Mice
- Mice, Inbred BALB C
- Mice, Knockout
- Neoplasm Recurrence, Local/prevention & control
- Oligodeoxyribonucleotides/immunology
- Orexin Receptors/deficiency
- Orexin Receptors/genetics
- Orexin Receptors/metabolism
- Paclitaxel/therapeutic use
- Spleen/cytology
- Spleen/transplantation
- Transplantation, Homologous
- Tumor Necrosis Factor-alpha/metabolism
Collapse
Affiliation(s)
- Reginald M. Gorczynski
- University Health Network, Toronto General Hospital, Toronto, Canada
- Department of Immunology, Faculty of Medicine, University of Toronto, and Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- * E-mail:
| | - Zhiqi Chen
- University Health Network, Toronto General Hospital, Toronto, Canada
| | - Nuray Erin
- Department of Medical Pharmacology, Akdeniz University, School of Medicine, Antalya, Turkey
| | - Ismat Khatri
- University Health Network, Toronto General Hospital, Toronto, Canada
| | - Anna Podnos
- University Health Network, Toronto General Hospital, Toronto, Canada
| |
Collapse
|
12
|
Zhao Y, Tan YS, Aupperlee MD, Langohr IM, Kirk EL, Troester MA, Schwartz RC, Haslam SZ. Pubertal high fat diet: effects on mammary cancer development. Breast Cancer Res 2014; 15:R100. [PMID: 24156623 PMCID: PMC3978633 DOI: 10.1186/bcr3561] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 10/11/2013] [Indexed: 02/08/2023] Open
Abstract
INTRODUCTION Epidemiological studies linking dietary fat intake and obesity to breast cancer risk have produced inconsistent results. This may be due to the difficulty of dissociating fat intake from obesity, and/or the lack of defined periods of exposure in these studies. The pubertal mammary gland is highly sensitive to cancer-causing agents. We assessed how high fat diet (HFD) affects inflammation, proliferative, and developmental events in the pubertal gland, since dysregulation of these can promote mammary tumorigenesis. To test the effect of HFD initiated during puberty on tumorigenesis, we utilized BALB/c mice, for which HFD neither induces obesity nor metabolic syndrome, allowing dissociation of HFD effects from other conditions associated with HFD. METHODS Pubertal BALB/c mice were fed a low fat diet (12% kcal fat) or a HFD (60% kcal fat), and subjected to carcinogen 7,12-dimethylbenz[a]anthracene (DMBA)-induced tumorigenesis. RESULTS HFD elevated mammary gland expression of inflammatory and growth factor genes at 3 and 4 weeks of diet. Receptor activator of nuclear factor kappa-B ligand (RANKL), robustly induced at 4 weeks, has direct mitogenic activity in mammary epithelial cells and, as a potent inducer of NF-κB activity, may induce inflammatory genes. Three weeks of HFD induced a transient influx of eosinophils into the mammary gland, consistent with elevated inflammatory factors. At 10 weeks, prior to the appearance of palpable tumors, there were increased numbers of abnormal mammary epithelial lesions, enhanced cellular proliferation, increased growth factors, chemokines associated with immune-suppressive regulatory T cells, increased vascularization, and elevated M2 macrophages. HFD dramatically reduced tumor latency. Early developing tumors were more proliferative and were associated with increased levels of tumor-related growth factors, including increased plasma levels of HGF in tumor-bearing animals. Early HFD tumors also had increased vascularization, and more intra-tumor and stromal M2 macrophages. CONCLUSIONS Taken together in this non-obesogenic context, HFD promotion of inflammatory processes, as well as local and systemically increased growth factor expression, are likely responsible for the enhanced tumorigenesis. It is noteworthy that although DMBA mutagenesis is virtually random in its targeting of genes in tumorigenesis, the short latency tumors arising in animals on HFD showed a unique gene expression profile, highlighting the potent overarching influence of HFD.
Collapse
|
13
|
Microenvironment, oncoantigens, and antitumor vaccination: lessons learned from BALB-neuT mice. BIOMED RESEARCH INTERNATIONAL 2014; 2014:534969. [PMID: 25136593 PMCID: PMC4065702 DOI: 10.1155/2014/534969] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Accepted: 05/12/2014] [Indexed: 12/20/2022]
Abstract
The tyrosine kinase human epidermal growth factor receptor 2 (HER2) gene is amplified in approximately 20% of human breast cancers and is associated with an aggressive clinical course and the early development of metastasis. Its crucial role in tumor growth and progression makes HER2 a prototypic oncoantigen, the targeting of which may be critical for the development of effective anticancer therapies. The setup of anti-HER2 targeting strategies has revolutionized the clinical outcome of HER2+ breast cancer. However, their initial success has been overshadowed by the onset of pharmacological resistance that renders them ineffective. Since the tumor microenvironment (TME) plays a crucial role in drug resistance, the design of more effective anticancer therapies should depend on the targeting of both cancer cells and their TME as a whole. In this review, starting from the successful know-how obtained with a HER2+ mouse model of mammary carcinogenesis, the BALB-neuT mice, we discuss the role of TME in mammary tumor development. Indeed, a deeper knowledge of antigens critical for cancer outbreak and progression and of the mechanisms that regulate the interplay between cancer and stromal cell populations could advise promising ways for the development of the best anticancer strategy.
Collapse
|
14
|
Bandini S, Curcio C, Macagno M, Quaglino E, Arigoni M, Lanzardo S, Hysi A, Barutello G, Consolino L, Longo DL, Musiani P, Forni G, Iezzi M, Cavallo F. Early onset and enhanced growth of autochthonous mammary carcinomas in C3-deficient Her2/neu transgenic mice. Oncoimmunology 2013; 2:e26137. [PMID: 24228231 PMCID: PMC3820812 DOI: 10.4161/onci.26137] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2013] [Revised: 08/12/2013] [Accepted: 08/12/2013] [Indexed: 12/26/2022] Open
Abstract
Aside from its classical role in fighting infections, complement is an important, although poorly understood, component of the tumor microenvironment. In particular, the tumor growth-regulatory activities of complement remain under debate. To assess the role of the complement system in the progression of autochthonous mammary carcinomas, we have crossed complement component 3 (C3)-deficient (C3−/−) BALB/c male mice with BALB/c females expressing the activated rat Her2/neu oncogene (neuT). Although neuT transgenic mice develop spontaneous mammary cancers with 100% penetrance, a significantly shorter tumor latency (i.e., earlier onset of the first palpable tumor), a higher frequency of multiple tumors (multiplicity), and a dramatic increase in the tumor growth rate were found in neuT-C3−/− animals. The accelerated tumor onset observed in neuT-C3−/− mice was paralleled by an earlier onset of spontaneous lung metastases and by an increase in Her2 expression levels, primarily on the surface of tumor cells. The percentage of immune cells infiltrating neuT carcinomas was similar in C3-deficient and C3-proficient mice, with the exception of a significant increase in the frequency of regulatory T cells in neuT-C3−/− tumors. Of particular interest, the enhanced immunosuppression imparted by C3 deficiency clearly influenced the immunogenic phenotype of autochthonous mammary tumors as neuT-C3−/− malignant cells transplanted into syngeneic immunocompetent hosts gave rise to lesions with a significantly delayed kinetics and reduced incidence as compared with cells obtained from neuT C3-proficient tumors. Finally, increased blood vessel permeability was evident in neuT-C3−/− tumors, although a similar number of tumor vessels was found in neuT and neuT-C3−/− lesions. Altogether, these data suggest that complement plays a crucial role in the immunosurveillance and, possibly, the immunoediting of Her2-driven autochthonous mammary tumors.
Collapse
Affiliation(s)
- Silvio Bandini
- Department of Molecular Biotechnology and Health Sciences; Molecular Biotechnology Center; University of Torino; Torino, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Cancer Associated Fibroblasts express pro-inflammatory factors in human breast and ovarian tumors. Biochem Biophys Res Commun 2013; 437:397-402. [DOI: 10.1016/j.bbrc.2013.06.089] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Accepted: 06/24/2013] [Indexed: 11/17/2022]
|
16
|
Lanara Z, Giannopoulou E, Fullen M, Kostantinopoulos E, Nebel JC, Kalofonos HP, Patrinos GP, Pavlidis C. Comparative study and meta-analysis of meta-analysis studies for the correlation of genomic markers with early cancer detection. Hum Genomics 2013; 7:14. [PMID: 23738773 PMCID: PMC3686617 DOI: 10.1186/1479-7364-7-14] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Accepted: 05/01/2013] [Indexed: 12/12/2022] Open
Abstract
A large number of common disorders, including cancer, have complex genetic traits, with multiple genetic and environmental components contributing to susceptibility. A literature search revealed that even among several meta-analyses, there were ambiguous results and conclusions. In the current study, we conducted a thorough meta-analysis gathering the published meta-analysis studies previously reported to correlate any random effect or predictive value of genome variations in certain genes for various types of cancer. The overall analysis was initially aimed to result in associations (1) among genes which when mutated lead to different types of cancer (e.g. common metabolic pathways) and (2) between groups of genes and types of cancer. We have meta-analysed 150 meta-analysis articles which included 4,474 studies, 2,452,510 cases and 3,091,626 controls (5,544,136 individuals in total) including various racial groups and other population groups (native Americans, Latinos, Aborigines, etc.). Our results were not only consistent with previously published literature but also depicted novel correlations of genes with new cancer types. Our analysis revealed a total of 17 gene-disease pairs that are affected and generated gene/disease clusters, many of which proved to be independent of the criteria used, which suggests that these clusters are biologically meaningful.
Collapse
Affiliation(s)
- Zoi Lanara
- Faculty of Mathematical, Physical and Natural Sciences, Department of Biological Sciences, University of Trieste, Trieste, 34128, Italy
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Koman IE, Commane M, Paszkiewicz G, Hoonjan B, Pal S, Safina A, Toshkov I, Purmal AA, Wang D, Liu S, Morrison C, Gudkov AV, Gurova KV. Targeting FACT complex suppresses mammary tumorigenesis in Her2/neu transgenic mice. Cancer Prev Res (Phila) 2012; 5:1025-35. [PMID: 22689915 DOI: 10.1158/1940-6207.capr-11-0529] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Development of safe and effective tumor-preventive treatments for high-risk patient populations and therapies for early-stage cancer remains a critical need in oncology. We have recently discovered compound with anticancer activity, Curaxin-137, which modulates several important signaling pathways involved in even the very early stages of cancer. In tumor cells, Curaxin-137 inhibits NF-κB- and HSF1-dependent transcription (prosurvival pathways) and activates p53 (a proapoptotic pathway) without inducing DNA damage. These effects result from chromatin trapping and inhibition of activity of the FACT (facilitates chromatin transcription) complex by Curaxin-137. FACT has not been previously implicated in cancer, but we found that its subunits are overexpressed in breast cancer. On the basis of this background, we tested whether Curaxin-137 could suppress tumorigenesis in MMTV-neu transgenic mice, which spontaneously develop mammary carcinoma due to steroid receptor-regulated expression of the Her2 proto-oncogene. We found that chronic administration of Curaxin-137 in a preventive regimen to MMTV-neu mice did not cause any detectable changes in normal organs and tissues, yet inhibited tumor onset, delayed tumor progression, and prolonged survival of mice in a dose-dependent manner. Curaxin-137 induced changes in FACT, altered NF-κB localization, and activated p53 in tumor cells as expected from its defined mechanism of action. These results support further investigation of Curaxin-137 as a potential preventive and/or early-stage therapeutic agent for breast cancer.
Collapse
Affiliation(s)
- Igor E Koman
- Department of Cell Stress Biology, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY, 14263, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Cheriyath V, Kuhns MA, Jacobs BS, Evangelista P, Elson P, Downs-Kelly E, Tubbs R, Borden EC. G1P3, an interferon- and estrogen-induced survival protein contributes to hyperplasia, tamoxifen resistance and poor outcomes in breast cancer. Oncogene 2012; 31:2222-36. [PMID: 21996729 DOI: 10.1038/onc.2011.393] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2010] [Revised: 06/26/2011] [Accepted: 08/04/2011] [Indexed: 02/06/2023]
Abstract
Hormonally regulated survival factors can have an important role in breast cancer. Here we elucidate G1P3, a survival protein induced by interferons (IFNs), as a target of estrogen signaling and a contributor to poor outcomes in estrogen receptor-positive (ER(+)) breast cancer. Compared with normal breast tissue, G1P3 was upregulated in the malignant epithelium (50 × higher) and was induced by estrogen ex vivo. In accord with its overexpression in early stages of breast cancer (hyperplasia and ductal carcinoma in situ), in morphogenesis assays G1P3 enhanced the survival of MCF10A acinar luminal cells causing hyperplasia by suppressing detachment-induced loss of mitochondrial potential and apoptosis (anoikis). In cells undergoing anoikis, G1P3 attenuated the induction of Bim protein, a proapoptotic member of the Bcl-2 family and reversed the downmodulation of Bcl-2 protein. Downregulation of G1P3 induced spontaneous apoptosis in BT-549 breast cancer cells and significantly reduced the growth of ER(+) breast cancer cell MCF7 (P≤0.01), further suggesting its prosurvival activity. In agreement with its induction by estrogen, G1P3 antagonized tamoxifen, an inhibitor of ER in MCF7 cells. More importantly, elevated expression of G1P3 was significantly associated with decreased relapse-free and overall survival in ER(+) breast cancer patients (P≤0.01). Our studies suggest that elevated expression of G1P3 may perturb canonical tumor-suppressing activity of IFNs partly by affecting the balance of pro- and antiapoptotic members of Bcl-2 family proteins, leading to breast cancer development and resistance to therapies.
Collapse
Affiliation(s)
- V Cheriyath
- Translational Hematology and Oncology Research, Taussig Cancer Institute, The Cleveland Clinic, Cleveland, OH 44195, USA.
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Bourguignon LYW, Wong G, Earle CA, Xia W. Interaction of low molecular weight hyaluronan with CD44 and toll-like receptors promotes the actin filament-associated protein 110-actin binding and MyD88-NFκB signaling leading to proinflammatory cytokine/chemokine production and breast tumor invasion. Cytoskeleton (Hoboken) 2011; 68:671-93. [PMID: 22031535 DOI: 10.1002/cm.20544] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Revised: 09/30/2011] [Accepted: 10/12/2011] [Indexed: 12/23/2022]
Abstract
Both high and low molecular weight hyaluronan (HMW-HA vs. LMW-HA) exist in various tissues and cells. In this study, we investigated LMW-HA-mediated CD44 interaction with Toll-like receptors (TLRs), the actin filament-associated protein (AFAP-110), and a myeloid differentiation factor (MyD88) in breast tumor cells (MDA-MB-231 cells). Our data indicate that LMW-HA (but not HMW-HA) preferentially stimulates a physical association between CD44 and TLRs followed by a concomitant recruitment of AFAP-110 and MyD88 into receptor-containing complexes in breast tumor cells. LMW-HA-activated AFAP-110 then binds to filamentous actin (F-actin) resulting in MyD88/nuclear factor-κB (NF-κB) nuclear translocation, NF-κB-specific transcription, and target gene [interleukine 1β and interleukine-8 (IL-1β and IL-8)] expression. These signaling events lead to proinflammatory cytokine/chemokine production in the breast tumor cells. AFAP-110-F-actin (activated by LMW-HA) also promotes tumor cell invasion. Downregulation of AFAP-110 or MyD88 by transfecting breast tumor cells with AFAP-110 siRNA or MyD88 siRNA, respectively not only blocks the ability of LMW-HA to stimulate AFAP-110-actin function, but also impairs MyD88-NF-κB nuclear translocation and NF-κB transcriptional activation. Consequently, both IL-1β/IL-8 production and tumor cell invasion are impaired. Taken together, these findings suggest that LMW-HA plays an important role in CD44-TLR-associated AFAP-110-actin interaction and MyD88-NF-κB signaling required for tumor cell behaviors, which may contribute to the progression of breast cancer.
Collapse
|
20
|
Serão NVL, Delfino KR, Southey BR, Beever JE, Rodriguez-Zas SL. Cell cycle and aging, morphogenesis, and response to stimuli genes are individualized biomarkers of glioblastoma progression and survival. BMC Med Genomics 2011; 4:49. [PMID: 21649900 PMCID: PMC3127972 DOI: 10.1186/1755-8794-4-49] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2010] [Accepted: 06/07/2011] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Glioblastoma is a complex multifactorial disorder that has swift and devastating consequences. Few genes have been consistently identified as prognostic biomarkers of glioblastoma survival. The goal of this study was to identify general and clinical-dependent biomarker genes and biological processes of three complementary events: lifetime, overall and progression-free glioblastoma survival. METHODS A novel analytical strategy was developed to identify general associations between the biomarkers and glioblastoma, and associations that depend on cohort groups, such as race, gender, and therapy. Gene network inference, cross-validation and functional analyses further supported the identified biomarkers. RESULTS A total of 61, 47 and 60 gene expression profiles were significantly associated with lifetime, overall, and progression-free survival, respectively. The vast majority of these genes have been previously reported to be associated with glioblastoma (35, 24, and 35 genes, respectively) or with other cancers (10, 19, and 15 genes, respectively) and the rest (16, 4, and 10 genes, respectively) are novel associations. Pik3r1, E2f3, Akr1c3, Csf1, Jag2, Plcg1, Rpl37a, Sod2, Topors, Hras, Mdm2, Camk2g, Fstl1, Il13ra1, Mtap and Tp53 were associated with multiple survival events.Most genes (from 90 to 96%) were associated with survival in a general or cohort-independent manner and thus the same trend is observed across all clinical levels studied. The most extreme associations between profiles and survival were observed for Syne1, Pdcd4, Ighg1, Tgfa, Pla2g7, and Paics. Several genes were found to have a cohort-dependent association with survival and these associations are the basis for individualized prognostic and gene-based therapies. C2, Egfr, Prkcb, Igf2bp3, and Gdf10 had gender-dependent associations; Sox10, Rps20, Rab31, and Vav3 had race-dependent associations; Chi3l1, Prkcb, Polr2d, and Apool had therapy-dependent associations. Biological processes associated glioblastoma survival included morphogenesis, cell cycle, aging, response to stimuli, and programmed cell death. CONCLUSIONS Known biomarkers of glioblastoma survival were confirmed, and new general and clinical-dependent gene profiles were uncovered. The comparison of biomarkers across glioblastoma phases and functional analyses offered insights into the role of genes. These findings support the development of more accurate and personalized prognostic tools and gene-based therapies that improve the survival and quality of life of individuals afflicted by glioblastoma multiforme.
Collapse
Affiliation(s)
- Nicola VL Serão
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Kristin R Delfino
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Bruce R Southey
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Jonathan E Beever
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Sandra L Rodriguez-Zas
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Department of Statistics, University of Illinois at Urbana-Champaign, Champaign, Illinois, USA
- Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| |
Collapse
|
21
|
Cavallo F, De Giovanni C, Nanni P, Forni G, Lollini PL. 2011: the immune hallmarks of cancer. Cancer Immunol Immunother 2011; 60:319-26. [PMID: 21267721 PMCID: PMC3042096 DOI: 10.1007/s00262-010-0968-0] [Citation(s) in RCA: 237] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2010] [Accepted: 12/27/2010] [Indexed: 12/26/2022]
Abstract
Ten years after the publication of the position paper “The hallmarks of cancer” (Hanahan and Weinberg Cell 100:57–70, 2000), it has become increasingly clear that mutated cells on their way to giving rise to a tumor have also to learn how to thrive in a chronically inflamed microenvironment, evade immune recognition, and suppress immune reactivity. Genetic and molecular definition of these three immune hallmarks of cancer offers the opportunity to learn how to deploy specific countermeasures to reverse the situation in favor of the immune system and, eventually, the patient. This new information could be channeled to address what seem to be the three major hallmarks for the immune control of cancer progression: effective procedures to activate immune reactivity; characterization of not-disposable oncoantigens; and counteraction of immune suppression.
Collapse
Affiliation(s)
- Federica Cavallo
- Department of Clinical and Biological Sciences, University of Turin, Molecular Biotechnology Center, Via Nizza 52, 10126 Turin, Italy.
| | | | | | | | | |
Collapse
|
22
|
Croci S, Recktenwald CV, Lichtenfels R, Nicoletti G, Dressler SP, De Giovanni C, Astolfi A, Palladini A, Shin-ya K, Landuzzi L, Nanni P, Lollini PL, Seliger B. Proteomic and PROTEOMEX profiling of mammary cancer progression in a HER-2/neu oncogene-driven animal model system. Proteomics 2010; 10:3835-53. [DOI: 10.1002/pmic.200900643] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
23
|
Xie X, Feng S, Vuong H, Liu Y, Goodison S, Lubman DM. A comparative phosphoproteomic analysis of a human tumor metastasis model using a label-free quantitative approach. Electrophoresis 2010; 31:1842-52. [PMID: 20446291 DOI: 10.1002/elps.200900752] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Alterations in cellular phosphorylation patterns have been implicated in a number of diseases, including cancer, through multiple mechanisms. Herein we present a survey of the phosphorylation profiles of an isogenic pair of human cancer cell lines with opposite metastatic phenotype. Phosphopeptides were enriched from tumor cell lysates with titanium dioxide and zirconium dioxide, and identified with nano-LC-MS/MS using an automatic cross-validation of MS/MS and MS/MS/MS (MS2+MS3) data-dependent neutral loss method. A spectral counting quantitative strategy was applied to the two cell line samples on the MS2-only scan, which was implemented successively after each MS2+MS3 scan in the same sample. For all regulated phosphopeptides reported by spectral counting analysis, sequence and phosphorylation site assignments were validated by a MS2+MS3 data-dependent neutral loss method. With this approach, we identified over 70 phosphorylated sites on 27 phosphoproteins as being differentially expressed with respect to tumor cell phenotype. The altered expression levels of proteins identified by LC-MS/MS were validated using Western blotting. Using network pathway analysis, we observed that the majority of the differentially expressed proteins were highly interconnected and belong to two major intracellular signaling pathways. Our findings suggest that the phosphorylation of isoform A of lamin A/C and GTPase activating protein binding protein 1 is associated with metastatic propensity. The study demonstrates a quantitative and comparative proteomics strategy to identify differential phosphorylation patterns in complex biological samples.
Collapse
Affiliation(s)
- Xiaolei Xie
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, MI 48109-0656, USA
| | | | | | | | | | | |
Collapse
|
24
|
Inflammation, aging, and cancer vaccines. Biogerontology 2010; 11:615-26. [PMID: 20455022 DOI: 10.1007/s10522-010-9280-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2010] [Accepted: 04/27/2010] [Indexed: 10/19/2022]
Abstract
Immunosenescence is characterized by a series of changes of immune pathways, including a chronic state of low-grade inflammation. Mounting evidence from experimental and clinical studies suggests that persistent inflammation increases the risk of cancer and the progression of the disease. Cancer vaccination, which came into view in the last years as the most intriguing means of activating an immune response capable of effectively hampering the progression of the preclinical stages of a tumour, has been shown to be less effective in older age than in young adults. Available evidence on the use of inhibitors of inflammation has indicated their potential enhancement of cancer vaccines, suggesting the possibility to improve the low effectiveness of cancer vaccines in old age employing pharmacological or natural compounds-based anti-inflammatory intervention. This review addresses the effects of age and inflammation on cancer development and progression, and speculates as to whether the modulation of inflammation may influence the response to cancer immunization.
Collapse
|
25
|
Gritzapis AD, Voutsas IF, Lekka E, Papamichail M, Baxevanis CN. Peptide Vaccination Breaks Tolerance to HER-2/neu by Generating Vaccine-Specific FasL+ CD4+ T Cells: First Evidence for Intratumor Apoptotic Regulatory T Cells. Cancer Res 2010; 70:2686-96. [DOI: 10.1158/0008-5472.can-09-2517] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
26
|
Abe F, Dafferner AJ, Donkor M, Westphal SN, Scholar EM, Solheim JC, Singh RK, Hoke TA, Talmadge JE. Myeloid-derived suppressor cells in mammary tumor progression in FVB Neu transgenic mice. Cancer Immunol Immunother 2010; 59:47-62. [PMID: 19449184 PMCID: PMC11030983 DOI: 10.1007/s00262-009-0719-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2008] [Accepted: 04/22/2009] [Indexed: 01/13/2023]
Abstract
Female mice transgenic for the rat proto-oncogene c-erb-B2, under control of the mouse mammary tumor virus (MMTV) promoter (neuN), spontaneously develop metastatic mammary carcinomas. The development of these mammary tumors is associated with increased number of GR-1(+)CD11b(+) myeloid derived suppressor cells (MDSCs) in the peripheral blood (PB), spleen and tumor. We report a complex relationship between tumor growth, MDSCs and immune regulatory molecules in non-mutated neu transgenic mice on a FVB background (FVB-neuN). The first and second tumors in FVB-neuN mice develop at a median of 265 (147-579) and 329 (161-523) days, respectively, resulting in a median survival time (MST) of 432 (201 to >500) days. During tumor growth, significantly increased number of MDSCs is observed in the PB and spleen, as well as, in infiltrating the mammary tumors. Our results demonstrate a direct correlation between tumor size and the number of MDSCs infiltrating the tumor and an inverse relationship between the frequency of CD4(+) T-cells and MDSCs in the spleen. Quantitative reverse transcription-polymerase chain reaction (qRT-PCR) assessment of enzyme and cytokine transcript levels in the spleen, tumor, tumor-infiltrating non-parenchymal cells (NPCs) and mammary glands revealed a significant increase in transcript levels from grossly normal mammary glands and tumor-infiltrating NPCs during tumor progression. Tumor NPCs, as compared to spleen cells from wild-type (w/t) mice, expressed significantly higher levels of arginase-1 (ARG-1), nitric oxide synthase (NOS-2), vascular endothelial growth factor (VEGF-A) and significantly lower levels of interferon (IFN)-gamma, interleukin (IL)-2 and fms-like tyrosine kinase-3 ligand (Flt3L) transcript levels. Transcript levels in the spleens of tumor-bearing (TB) mice also differed from normal mice, although to a lesser extent than transcript levels from tumor-infiltrating NPCs. Furthermore, both spleen cells and NPCs from TB mice, but not control mice, suppressed alloantigen responses by syngeneic control spleen cells. Correlative studies revealed that the number of MDSCs in the spleen was directly associated with granulocyte colony stimulating factor (G-CSF) transcript levels in the spleen; while the number of MDSCs in the tumors was directly correlated with splenic granulocyte macrophage stimulating factor (GM-CSF) transcript levels, tumor volume and tumor cell number. Together our results support a role for MDSCs in tumor initiation and progressive, T-cell depression and loss of function provide evidence which support multiple mechanisms of MDSC expansion in a site-dependent manner.
Collapse
Affiliation(s)
- Fuminori Abe
- Laboratory of Transplantation Immunology, Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198-7660 USA
| | - Alicia J. Dafferner
- Laboratory of Transplantation Immunology, Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198-7660 USA
| | - Moses Donkor
- Laboratory of Transplantation Immunology, Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198-7660 USA
| | - Sherry N. Westphal
- Laboratory of Transplantation Immunology, Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198-7660 USA
| | - Eric M. Scholar
- Laboratory of Transplantation Immunology, Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198-7660 USA
| | - Joyce C. Solheim
- Laboratory of Transplantation Immunology, Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198-7660 USA
| | - Rakesh K. Singh
- Laboratory of Transplantation Immunology, Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198-7660 USA
| | - Traci A. Hoke
- Laboratory of Transplantation Immunology, Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198-7660 USA
| | - James E. Talmadge
- Laboratory of Transplantation Immunology, Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198-7660 USA
| |
Collapse
|
27
|
Differences in the tumor microenvironment between African-American and European-American breast cancer patients. PLoS One 2009; 4:e4531. [PMID: 19225562 PMCID: PMC2638012 DOI: 10.1371/journal.pone.0004531] [Citation(s) in RCA: 161] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2008] [Accepted: 01/06/2009] [Indexed: 12/20/2022] Open
Abstract
Background African-American breast cancer patients experience higher mortality rates than European-American patients despite having a lower incidence of the disease. We tested the hypothesis that intrinsic differences in the tumor biology may contribute to this cancer health disparity. Methods and Results Using laser capture microdissection, we examined genome-wide mRNA expression specific to tumor epithelium and tumor stroma in 18 African-American and 17 European-American patients. Numerous genes were differentially expressed between these two patient groups and a two-gene signature in the tumor epithelium distinguished between them. To identify the biological processes in tumors that are different by race/ethnicity, Gene Ontology and disease association analyses were performed. Several biological processes were identified which may contribute to enhanced disease aggressiveness in African-American patients, including angiogenesis and chemotaxis. African-American tumors also contained a prominent interferon signature. The role of angiogenesis in the tumor biology of African-Americans was further investigated by examining the extent of vascularization and macrophage infiltration in an expanded set of 248 breast tumors. Immunohistochemistry revealed that microvessel density and macrophage infiltration is higher in tumors of African-Americans than in tumors of European-Americans. Lastly, using an in silico approach, we explored the potential of tailored treatment options for African-American patients based on their gene expression profile. This exploratory approach generated lists of therapeutics that may have specific antagonistic activity against tumors of African-American patients, e.g., sirolimus, resveratrol, and chlorpromazine in estrogen receptor-negative tumors. Conclusions The gene expression profiles of breast tumors indicate that differences in tumor biology may exist between African-American and European-American patients beyond the knowledge of current markers. Notably, pathways related to tumor angiogenesis and chemotaxis could be functionally different in these two patient groups.
Collapse
|
28
|
Calogero RA, Quaglino E, Saviozzi S, Forni G, Cavallo F. Oncoantigens as anti-tumor vaccination targets: the chance of a lucky strike? Cancer Immunol Immunother 2008; 57:1685-94. [PMID: 18286283 PMCID: PMC11030840 DOI: 10.1007/s00262-008-0481-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2007] [Accepted: 02/05/2008] [Indexed: 11/25/2022]
Abstract
Neoplastic transformation is a multistage process and distinct gene products of specific cell regulatory pathways are involved at each stage. Identification of genes overexpressed at a specific stage provides an unprecedented opportunity to address the immune system against antigens with a driving role in tumor progression (oncoantigens). The ERBB2 oncogene is a prototype of deregulated oncogenic protein kinase membrane receptors. Mice transgenic for rat ERBB2 (BALB-neuT mice) were used in this study to identify an additional set of oncoantigens expressed at defined stages by most breast carcinomas to be used as alternatives to ERBB2-driven vaccination. To address this question, we integrated the transcription data generated by comparing preneoplastic lesions and neoplasia in BALB-neuT mice with a meta-analysis on transcription profiles generated from normal and breast tumor human specimens. Forty-six putative oncoantigens identified and prioritized according to their expression on the cell membrane or in the extra cellular space, cytoplasm and nucleus were chosen for preclinical investigation as vaccination targets.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- Cancer Vaccines/genetics
- Cancer Vaccines/metabolism
- Carcinoma, Ductal, Breast/genetics
- Carcinoma, Ductal, Breast/immunology
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Lobular/genetics
- Carcinoma, Lobular/immunology
- Carcinoma, Lobular/pathology
- Female
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic
- Mammary Neoplasms, Experimental/genetics
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/therapy
- Mice
- Mice, Transgenic
- Oligonucleotide Array Sequence Analysis
- Precancerous Conditions/genetics
- Precancerous Conditions/immunology
- Precancerous Conditions/pathology
- Rats
- Receptor, ErbB-2/physiology
- Signal Transduction
- Transcription, Genetic
Collapse
Affiliation(s)
- Raffaele Adolfo Calogero
- Molecular Biotechnology Center, Department of Clinical and Biological Sciences, University of Turin, 10126 Turin, Italy
| | - Elena Quaglino
- Molecular Biotechnology Center, Department of Clinical and Biological Sciences, University of Turin, 10126 Turin, Italy
| | - Silvia Saviozzi
- Molecular Biotechnology Center, Department of Clinical and Biological Sciences, University of Turin, 10126 Turin, Italy
| | - Guido Forni
- Molecular Biotechnology Center, Department of Clinical and Biological Sciences, University of Turin, 10126 Turin, Italy
| | - Federica Cavallo
- Molecular Biotechnology Center, Department of Clinical and Biological Sciences, University of Turin, 10126 Turin, Italy
| |
Collapse
|
29
|
Quaglino E, Mastini C, Forni G, Cavallo F. ErbB2 transgenic mice: a tool for investigation of the immune prevention and treatment of mammary carcinomas. CURRENT PROTOCOLS IN IMMUNOLOGY 2008; Chapter 20:Unit 20.9.1-20.9-10. [PMID: 18729063 DOI: 10.1002/0471142735.im2009s82] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
The epidermal growth factor receptor belongs to a superfamily of receptor tyrosine kinases (RTK) that includes ErbB2. ErbB2 is involved in normal physiological processes, such as embryogenesis, cell proliferation, differentiation, adhesion motility, and apoptosis, while its malfunction or overexpression is responsible for development defects, diabetes, and cancer. The human ortholog of ErbB2 is referred as Her-2 (human ErbB2) while the rat ortholog is referred as neu (rat ErbB2). As ErbB2 is directly involved in carcinogenesis, mice transgenic for the rat neu oncogene allow straightforward assessment of the ability of drugs and vaccines to inhibit the progression of neu-driven cancer. Information from this model may provide indications on the efficacy of similar treatments in patients. This commentary provides key information regarding the use of these transgenic mouse models for evaluation of the efficacy of anti-tumor strategies.
Collapse
Affiliation(s)
- Elena Quaglino
- Molecular Biotechnology Center, Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | | | | | | |
Collapse
|
30
|
Dong B, Matsumura F. Roles of Cytosolic Phospholipase A2 and Src Kinase in the Early Action of 2,3,7,8-Tetrachlorodibenzo-p-dioxin through a Nongenomic Pathway in MCF10A Cells. Mol Pharmacol 2008; 74:255-63. [DOI: 10.1124/mol.107.044669] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
|
31
|
Standish LJ, Sweet ES, Novack J, Wenner CA, Bridge C, Nelson A, Martzen M, Torkelson C. Breast cancer and the immune system. JOURNAL OF THE SOCIETY FOR INTEGRATIVE ONCOLOGY 2008; 6:158-168. [PMID: 19134448 PMCID: PMC2845458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
This article reviews the evidence that the functioning of both the innate and the adaptive immune system plays a role in preventing relapse in women with breast cancer. Lymphocytes, including T cells, T regulatory cells, and natural killer cells, and their cytokine release patterns are implicated in both primary prevention and recurrence of breast cancer. Cancer prognosis may be related to immune system functional status. The hypothesis that the immune system has a causal role in breast cancer etiology is supported by epidemiologic, preclinical, and clinical research. Empirical support for the concept that immune status and immunomodulatory therapy have important roles in comprehensive breast cancer treatment is provided.
Collapse
|