1
|
Alkwedhim MAH, Pouresmaeil V, Davoodi-Dehaghani F, Mahavar M, Homayouni Tabrizi M. Synthesis and evaluation of biological effects of modified graphene oxide nanoparticles containing Lawson (Henna extract) on gastric cancer cells. Mol Biol Rep 2023; 50:8971-8983. [PMID: 37715021 DOI: 10.1007/s11033-023-08797-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 09/04/2023] [Indexed: 09/17/2023]
Abstract
PURPOSE Targeted Graphene Oxide (GO) nanoparticles can play an important role in the treatment of cancer by increasing cancer cell targeting. This study was conducted to synthesize GO nanoparticles functionalized with chitosan-folate (CS-FA) to deliver a natural product Lawsone (LA) for cancer treatment. METHODS After characterization of the LA-GO-CS-FA, antioxidant activities of the nanoparticles were investigated by ABTS, DPPH, and FRAP tests. CAM assay was used to study the effect of nanoparticles on angiogenesis. The expression level of inflammatory and angiogenic genes in cells treated with nanoparticles was evaluated by real-time PCR. RESULTS The findings demonstrated the formation of nanoparticles with a size of 113.3 nm, a PDI of 0.31, and a surface charge of + 11.07 mV. The percentages of encapsulation efficiency were reported at 93%. Gastric cancer cells were reported as the most sensitive to treatment compared to the control, and the gastric cancer cells were used to study gene expression changes. The anti-angiogenic effects of nanoparticles were confirmed by reducing the average number and length of blood vessels and reducing the height and weight of embryos in the CAM assay. The reducing the expression of genes involved in angiogenesis in real-time PCR was demonstrated. Nanoparticles displayed high antioxidant properties by inhibiting DPPH and ABTS radicals and reducing iron ions in the FRAP method. The reduction of pro-inflammatory genes in AGS cells which were treated with nanoparticles indicates the anti-inflammatory properties of nanoparticles. CONCLUSION This study showed the efficacy of nanoparticles in inhibiting gastric cancer cells by relying on inhibiting angiogenesis.
Collapse
Affiliation(s)
| | - Vahid Pouresmaeil
- Department of Biochemistry, Faculty of Medicine, Mashhad Medical Sciences, Islamic Azad University, Mashhad, Iran.
| | - Fatemeh Davoodi-Dehaghani
- Department of Biology, Faculty of Basic Sciences, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Mobina Mahavar
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | | |
Collapse
|
2
|
Hamza S, Garanina EE, Alsaadi M, Khaiboullina SF, Tezcan G. Blocking the Hormone Receptors Modulates NLRP3 in LPS-Primed Breast Cancer Cells. Int J Mol Sci 2023; 24:ijms24054846. [PMID: 36902278 PMCID: PMC10002867 DOI: 10.3390/ijms24054846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/22/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
NOD-like receptor protein 3 (NLRP3) may contribute to the growth and propagation of breast cancer (BC). The effect of estrogen receptor-α (ER-α), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) on NLRP3 activation in BC remains unknown. Additionally, our knowledge of the effect of blocking these receptors on NLRP3 expression is limited. We used GEPIA, UALCAN, and the Human Protein Atlas for transcriptomic profiling of NLRP3 in BC. Lipopolysaccharide (LPS) and adenosine 5'-triphosphate (ATP) were used to activate NLRP3 in luminal A MCF-7 and in TNBC MDA-MB-231 and HCC1806 cells. Tamoxifen (Tx), mifepristone (mife), and trastuzumab (Tmab) were used to block ER-α, PR, and HER2, respectively, on inflammasome activation in LPS-primed MCF7 cells. The transcript level of NLRP3 was correlated with ER-ɑ encoding gene ESR1 in luminal A (ER-α+, PR+) and TNBC tumors. NLRP3 protein expression was higher in untreated and LPS/ATP-treated MDA-MB-231 cells than in MCF7 cells. LPS/ATP-mediated NLRP3 activation reduced cell proliferation and recovery of wound healing in both BC cell lines. LPS/ATP treatment prevented spheroid formation in MDA-MB-231 cells but did not affect MCF7. HGF, IL-3, IL-8, M-CSF, MCP-1, and SCGF-b cytokines were secreted in both MDA-MB-231 and MCF7 cells in response to LPS/ATP treatment. Tx (ER-α inhibition) promoted NLRP3 activation and increased migration and sphere formation after LPS treatment of MCF7 cells. Tx-mediated activation of NLRP3 was associated with increased secretion of IL-8 and SCGF-b compared to LPS-only-treated MCF7 cells. In contrast, Tmab (Her2 inhibition) had a limited effect on NLRP3 activation in LPS-treated MCF7 cells. Mife (PR inhibition) opposed NLRP3 activation in LPS-primed MCF7 cells. We have found that Tx increased the expression of NLRP3 in LPS-primed MCF7. These data suggest a link between blocking ER-α and activation of NLRP3, which was associated with increased aggressiveness of the ER-α+ BC cells.
Collapse
Affiliation(s)
- Shaimaa Hamza
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Ekaterina E. Garanina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Mohammad Alsaadi
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
| | - Svetlana F. Khaiboullina
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
- Correspondence: or (S.F.K.); (G.T.); Fax: +1-775682-8258 (S.F.K.); +90-224-294-00-78 (G.T.)
| | - Gulcin Tezcan
- Institute of Fundamental Medicine and Biology, Kazan Federal University, 420008 Kazan, Russia
- Department of Fundamental Sciences, Faculty of Dentistry, Bursa Uludag University, Bursa 16059, Turkey
- Correspondence: or (S.F.K.); (G.T.); Fax: +1-775682-8258 (S.F.K.); +90-224-294-00-78 (G.T.)
| |
Collapse
|
3
|
Singh RD, Dholariya S, Shekher A, Avadhesh, Parchwani D, Gupta SC. Role of IL-1 gene polymorphisms in common solid cancers. MULTIFACETED ROLE OF IL-1 IN CANCER AND INFLAMMATION 2023:1-69. [DOI: 10.1016/b978-0-12-824273-5.00002-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
4
|
Ahmed SA, Mendonca P, Elhag R, Soliman KFA. Anticancer Effects of Fucoxanthin through Cell Cycle Arrest, Apoptosis Induction, Angiogenesis Inhibition, and Autophagy Modulation. Int J Mol Sci 2022; 23:16091. [PMID: 36555740 PMCID: PMC9785196 DOI: 10.3390/ijms232416091] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Cancer accounts for one in seven deaths worldwide and is the second leading cause of death in the United States, after heart disease. One of the standard cancer treatments is chemotherapy which sometimes can lead to chemoresistance and treatment failure. Therefore, there is a great need for novel therapeutic approaches to treat these patients. Novel natural products have exhibited anticancer effects that may be beneficial in treating many kinds of cancer, having fewer side effects, low toxicity, and affordability. Numerous marine natural compounds have been found to inhibit molecular events and signaling pathways associated with various stages of cancer development. Fucoxanthin is a well-known marine carotenoid of the xanthophyll family with bioactive compounds. It is profusely found in brown seaweeds, providing more than 10% of the total creation of natural carotenoids. Fucoxanthin is found in edible brown seaweed macroalgae such as Undaria pinnatifida, Laminaria japonica, and Eisenia bicyclis. Many of fucoxanthin's pharmacological properties include antioxidant, anti-tumor, anti-inflammatory, antiobesity, anticancer, and antihypertensive effects. Fucoxanthin inhibits many cancer cell lines' proliferation, angiogenesis, migration, invasion, and metastasis. In addition, it modulates miRNA and induces cell cycle growth arrest, apoptosis, and autophagy. Moreover, the literature shows fucoxanthin's ability to inhibit cytokines and growth factors such as TNF-α and VEGF, which stimulates the activation of downstream signaling pathways such as PI3K/Akt autophagy, and pathways of apoptosis. This review highlights the different critical mechanisms by which fucoxanthin inhibits diverse cancer types, such as breast, prostate, gastric, lung, and bladder development and progression. Moreover, this article reviews the existing literature and provides critical supportive evidence for fucoxanthin's possible therapeutic use in cancer.
Collapse
Affiliation(s)
- Shade’ A. Ahmed
- Division of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL 32307, USA
| | - Patricia Mendonca
- Department of Biology, College of Science and Technology, Florida A&M University, Tallahassee, FL 32307, USA
| | - Rashid Elhag
- Department of Biology, College of Science and Technology, Florida A&M University, Tallahassee, FL 32307, USA
| | - Karam F. A. Soliman
- Division of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A&M University, Tallahassee, FL 32307, USA
| |
Collapse
|
5
|
Interleukin-1 and Nuclear Factor Kappa B Signaling Promote Breast Cancer Progression and Treatment Resistance. Cells 2022; 11:cells11101673. [PMID: 35626710 PMCID: PMC9139516 DOI: 10.3390/cells11101673] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 05/08/2022] [Accepted: 05/17/2022] [Indexed: 02/01/2023] Open
Abstract
While meant for wound healing and immunity in response to injury and infection, inflammatory signaling is usurped by cancerous tumors to promote disease progression, including treatment resistance. The interleukin-1 (IL-1) inflammatory cytokine family functions in wound healing and innate and adaptive immunity. Two major, closely related IL-1 family members, IL-1α and IL-1β, promote tumorigenic phenotypes and contribute to treatment resistance in cancer. IL-1 signaling converges on transactivation of the Nuclear Factor Kappa B (NF-κB) and Activator protein 1 (AP-1) transcription factors. NF-κB and AP-1 signaling are also activated by the inflammatory cytokine Tumor Necrosis Factor Alpha (TNFα) and microbe-sensing Toll-Like Receptors (TLRs). As reviewed elsewhere, IL-1, TNFα, and TLR can promote cancer progression through NF-κB or AP-1. In this review, we focus on what is known about the role of IL-1α and IL-1β in breast cancer (BCa) progression and therapeutic resistance, and state evidence for the role of NF-κB in mediating IL-1-induced BCa progression and therapeutic resistance. We will present evidence that IL-1 promotes BCa cell proliferation, BCa stem cell expansion, angiogenesis, and metastasis. IL-1 also regulates intracellular signaling and BCa cell hormone receptor expression in a manner that confers a growth advantage to the tumor cells and allows BCa cells to evade therapy. As such, the IL-1 receptor antagonist, anakinra, is in clinical trials to treat BCa and multiple other cancer types. This article presents a review of the literature from the 1990s to the present, outlining the evidence supporting a role for IL-1 and IL-1-NF-κB signaling in BCa progression.
Collapse
|
6
|
Liu Y, Gusev A, Heng YJ, Alexandrov LB, Kraft P. Somatic mutational profiles and germline polygenic risk scores in human cancer. Genome Med 2022; 14:14. [PMID: 35144655 PMCID: PMC8832866 DOI: 10.1186/s13073-022-01016-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 01/24/2022] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND The mutational profile of cancer reflects the activity of the mutagenic processes which have been operative throughout the lineage of the cancer cell. These processes leave characteristic profiles of somatic mutations called mutational signatures. Mutational signatures, including single-base substitution (SBS) signatures, may reflect the effects of exogenous or endogenous exposures. METHODS We used polygenic risk scores (PRS) to summarize common germline variation associated with cancer risk and other cancer-related traits and examined the association between somatic mutational profiles and germline PRS in 12 cancer types from The Cancer Genome Atlas. Somatic mutational profiles were constructed from whole-exome sequencing data of primary tumors. PRS were calculated for the 12 selected cancer types and 9 non-cancer traits, including cancer risk determinants, hormonal factors, and immune-mediated inflammatory diseases, using germline genetic data and published summary statistics from genome-wide association studies. RESULTS We found 17 statistically significant associations between somatic mutational profiles and germline PRS after Bonferroni correction (p < 3.15 × 10-5), including positive associations between germline inflammatory bowel disease PRS and number of somatic mutations attributed to signature SBS1 in prostate cancer and APOBEC-related signatures in breast cancer. Positive associations were also found between age at menarche PRS and mutation counts of SBS1 in overall and estrogen receptor-positive breast cancer. Consistent with prior studies that found an inverse association between the pubertal development PRS and risk of prostate cancer, likely reflecting hormone-related mechanisms, we found an inverse association between age at menarche PRS and mutation counts of SBS1 in prostate cancer. Inverse associations were also found between several cancer PRS and tumor mutation counts. CONCLUSIONS Our analysis suggests that there are robust associations between tumor somatic mutational profiles and germline PRS. These may reflect the mechanisms through hormone regulation and immune responses that contribute to cancer etiology and drive cancer progression.
Collapse
Affiliation(s)
- Yuxi Liu
- grid.38142.3c000000041936754XDepartment of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115 USA ,grid.38142.3c000000041936754XProgram in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Boston, MA 02115 USA
| | - Alexander Gusev
- grid.65499.370000 0001 2106 9910Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02215 USA
| | - Yujing J. Heng
- grid.38142.3c000000041936754XDepartment of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215 USA
| | - Ludmil B. Alexandrov
- grid.266100.30000 0001 2107 4242Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093 USA
| | - Peter Kraft
- grid.38142.3c000000041936754XDepartment of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA 02115 USA ,grid.38142.3c000000041936754XProgram in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, 655 Huntington Avenue, Boston, MA 02115 USA ,grid.38142.3c000000041936754XDepartment of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115 USA
| |
Collapse
|
7
|
Malkova AM, Sharoyko VV, Zhukova NV, Gubal AR, Orlova RV. Laboratory biomarkers of an effective antitumor immune response. Clinical significance. Cancer Treat Res Commun 2021; 29:100489. [PMID: 34837797 DOI: 10.1016/j.ctarc.2021.100489] [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: 05/30/2021] [Revised: 10/18/2021] [Accepted: 11/09/2021] [Indexed: 10/19/2022]
Abstract
The modern checkpoint inhibitors block the programmed death-1 receptor and its ligand, cytotoxic T-lymphocyte-associated antigen 4 on tumor cells and lymphocytes, that induces cytotoxic reactions. Nowadays, there are no approved clinical and laboratory predictor markers of immune therapy efficacy, which would allow a more personalized approach to patient selection and treatment. The aim of this review is to analyze possible biomarkers of efficacy for treatment with checkpoint inhibitors according to the pathogenic mechanisms of drug action. The review revealed possible predictive biomarkers, that could be classified to 3 groups: biomarkers of high mutagenic potential of the tumor, biomarkers of high activity of adaptive immunity, biomarkers of low activity of the tumor microenvironment. The determination of the described markers before the start of therapy can be used to formulate a treatment regimen, in which the use of various immunomodulatory drugs, inhibitors of proinflammatory cytokines, angiogenic molecules, and probiotics can be considered.
Collapse
Affiliation(s)
- A M Malkova
- Saint Petersburg State University, 7/9 Universitetskaya Emb., St Petersburg 199034, Russian Federation.
| | - V V Sharoyko
- Saint Petersburg State University, 7/9 Universitetskaya Emb., St Petersburg 199034, Russian Federation.
| | - N V Zhukova
- Saint Petersburg State University, 7/9 Universitetskaya Emb., St Petersburg 199034, Russian Federation.
| | - A R Gubal
- Saint Petersburg State University, 7/9 Universitetskaya Emb., St Petersburg 199034, Russian Federation.
| | - R V Orlova
- Saint Petersburg State University, 7/9 Universitetskaya Emb., St Petersburg 199034, Russian Federation.
| |
Collapse
|
8
|
You D, Jeong Y, Yoon SY, A Kim S, Kim SW, Nam SJ, Lee JE, Kim S. Celastrol attenuates the inflammatory response by inhibiting IL‑1β expression in triple‑negative breast cancer cells. Oncol Rep 2021; 45:89. [PMID: 33846813 PMCID: PMC8042664 DOI: 10.3892/or.2021.8040] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 03/04/2021] [Indexed: 12/22/2022] Open
Abstract
IL-1 promotes cancer cell proliferation and invasiveness in various malignancies, such as breast and colorectal cancer. In the present study, the functional roles of IL-1β (IL1B) and the inhibitory effect of celastrol on IL1B expression were investigated in triple-negative breast cancer (TNBC) cells. The data revealed that celastrol markedly decreased IL1B expression and suppressed TNBC cell proliferation in a dose-dependent manner. The levels of IL1B and IL8 mRNA were significantly increased in TNBC cells compared with non-TNBC cells. In addition, IL1B augmented the expression levels of IL8 as well as matrix metalloproteinases (MMPs), including MMP-1 and MMP-9, in TNBC cells. Furthermore, IL1B expression was decreased by a specific MEK1/2 inhibitor, MEK162. Celastrol also promoted IL1B downregulation through the suppression of the MEK/ERK-dependent pathway. Furthermore, the results also revealed a decrease in IL1B-induced IL8, MMP-1, and MMP-9 expression in response to celastrol treatment. The induction of cellular invasion by IL1B was also markedly decreased by celastrol. Collectively, the present study results suggested celastrol as an effective drug for the treatment of TNBC, involving a reduction in IL1B expression, activity or signaling pathways.
Collapse
Affiliation(s)
- Daeun You
- Department of Health Sciences and Technology, The Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul 06351, Republic of Korea
| | - Yisun Jeong
- Department of Health Sciences and Technology, The Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul 06351, Republic of Korea
| | - Sun Young Yoon
- Department of Health Sciences and Technology, The Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul 06351, Republic of Korea
| | - Sung A Kim
- Department of Health Sciences and Technology, The Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul 06351, Republic of Korea
| | - Seok Won Kim
- Department of Breast Cancer Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Seok Jin Nam
- Department of Breast Cancer Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Jeong Eon Lee
- Department of Health Sciences and Technology, The Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul 06351, Republic of Korea
| | - Sangmin Kim
- Department of Breast Cancer Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| |
Collapse
|
9
|
Widowati W, Murti H, Widyastuti H, Laksmitawati DR, Rizal R, Widya Kusuma HS, Sumitro SB, Widodo MA, Bachtiar I. Decreased Inhibition of Proliferation and Induction of Apoptosis in Breast Cancer Cell Lines (T47D and MCF7) from Treatment with Conditioned Medium Derived from Hypoxia-Treated Wharton's Jelly MSCs Compared with Normoxia-Treated MSCs. Int J Hematol Oncol Stem Cell Res 2021; 15:77-89. [PMID: 34466206 PMCID: PMC8381107 DOI: 10.18502/ijhoscr.v15i2.6038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 12/15/2020] [Indexed: 11/24/2022] Open
Abstract
Background: Mesenchymal stem cells (MSCs) are an appealing source of adult stem cells for cell therapy due to the high rate of proliferation, self-renewal capability, and applicable therapy. Wharton’s jelly (WJ), the main component of the umbilical cord extracellular matrix, comprises multipotent stem cells with a high proliferation rate and self-renewal capability and has anti-cancer properties. MSCs have been reported to secrete a variety of cytokines that have a cytotoxic effect in various cancers. Oxygen tension affects MSCs proliferation, cytokines level but no in surface markers expression, MSCs’ differentiation. We explored the cytotoxic effect and inducing apoptosis of Wharton’s jelly derived mesenchymal stem cells (WJMSCs) secretions from normoxic WJMSCs (WJMSCs-norCM) (CM: conditioned medium) and hypoxic WJMSCs (WJMSCs-hypoCM) in breast cancer cell lines (T47D and MCF7). Materials and Methods: Cytotoxic activity was determined using the MTS assay. RT-PCR was performed to measure the expression of apoptosis-inducing genes, specifically P53, BAX, and CASP9, and the antiapoptotic gene BCL-2. Results: WJMSCs-norCM and WJMSCs-hypoCM were potent inhibitors of the proliferation in both cell lines. WJMSCs-norCM had more anticancer activity in T47D and MCF7. The IC50 value of WJMSCs-norCM on MCF7 was 42.34%, and on T47D was 42.36%. WJMSCs-norCM significantly induced the gene expression of apoptotic P53, BAX, and CASP9 and insignificantly decreased the antiapoptotic gene BCL-2 in both MCF7 and T47D cells. WJMSCs-CM has anticancer activity by inducing P53, BAX, and CASP9 apoptotic genes. Conclusion: WJMSCs-norCM has more anticancer activity than WJMSCs-hypoCM.
Collapse
Affiliation(s)
- Wahyu Widowati
- Faculty of Medicine, Maranatha Christian University, Jl. Prof. drg.. Suria Sumantri No.65, Bandung 40164, Indonesia
| | - Harry Murti
- Stem Cell and Cancer Institute, Jl. A Yani No 2 Pulo Mas, Jakarta 13210, Indonesia
| | - Halida Widyastuti
- Stem Cell and Cancer Institute, Jl. A Yani No 2 Pulo Mas, Jakarta 13210, Indonesia
| | - Dian Ratih Laksmitawati
- Faculty of Pharmacy, Pancasila University, Jl. Raya Lenteng Agung No.56-80 Jakarta 12640, Indonesia
| | - Rizal Rizal
- Biomolecular and Biomedical Research Center, Aretha Medika Utama,, Jl. Babakan Jeruk II No. 9, Bandung 40163, Indonesia.,Biomedical Engineering, Department of Electrical Engineering, Faculty of Engineering, Universitas Indonesia, Jl. Kampus UI, Depok 16426, West Java, Indonesia
| | - Hanna Sari Widya Kusuma
- Biomolecular and Biomedical Research Center, Aretha Medika Utama,, Jl. Babakan Jeruk II No. 9, Bandung 40163, Indonesia
| | - Sutiman Bambang Sumitro
- Department of Biology, Faculty of Mathematics and Natural Sciences, Brawijaya University, Jl. Veteran, Ketawanggede Malang 65145, Indonesia
| | - M Aris Widodo
- Pharmacology Laboratories, Faculty of Medicine, Brawijaya University Jl. Veteran, Ketawanggede Malang 65145,, Indonesia
| | - Indra Bachtiar
- Stem Cell and Cancer Institute, Jl. A Yani No 2 Pulo Mas, Jakarta 13210, Indonesia
| |
Collapse
|
10
|
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: 10] [Impact Index Per Article: 2.0] [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
|
11
|
Jin H, Kim HJ. NLRC4, ASC and Caspase-1 Are Inflammasome Components that Are Mediated by P2Y 2R Activation in Breast Cancer Cells. Int J Mol Sci 2020; 21:ijms21093337. [PMID: 32397236 PMCID: PMC7246622 DOI: 10.3390/ijms21093337] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 04/24/2020] [Accepted: 05/06/2020] [Indexed: 12/21/2022] Open
Abstract
The inflammasomes are reported to be associated with tumor progression. In our previous study, we determined that extracellular ATP enhances invasion and tumor growth by inducing inflammasome activation in a P2Y purinergic receptor 2 (P2Y2R)-dependent manner. However, it is not clear which inflammasome among the diverse complexes is associated with P2Y2R activation in breast cancer. Thus, in this study, we determined which inflammasome components are regulated by P2Y2R activation and are involved in tumor progression in breast cancer cells and radiotherapy-resistant (RT-R)-breast cancer cells. First, we found that NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3); NLR family caspase activation and recruitment domain (CARD) containing 4 (NLRC4); apoptosis-associated speck-like protein containing a CARD complex (ASC); and caspase-1 mRNA levels were upregulated in RT-R-MDA-MB-231 cells compared to MDA-MB-231 cells, whereas tumor necrosis factor-α (TNF-α) or ATP treatment induced NLRC4, ASC, and caspase-1 but not NLRP3 protein levels. Moreover, TNF-α or ATP increased protein levels of NLRC4, ASC, and caspase-1 in a P2Y2R-dependent manner in MDA-MB-231 and RT-R-MDA-MB-231 cells. In addition, P2Y2R activation by ATP induced the secretion of IL-1β and VEGF-A, as well as invasion, in MDA-MB-231 and RT-R-MDA-MB-231 cells, which was inhibited by NLRC4, ASC, and caspase-1 small interfering RNA (siRNA). Taken together, this report suggests that P2Y2R activation by ATP induces tumor invasion and angiogenesis through inflammasome activation, specifically by regulating the inflammasome components NLRC4, ASC, and caspase-1.
Collapse
|
12
|
Nawas AF, Kanchwala M, Thomas-Jardin SE, Dahl H, Daescu K, Bautista M, Anunobi V, Wong A, Meade R, Mistry R, Ghatwai N, Bayerl F, Xing C, Delk NA. IL-1-conferred gene expression pattern in ERα + BCa and AR + PCa cells is intrinsic to ERα - BCa and AR - PCa cells and promotes cell survival. BMC Cancer 2020; 20:46. [PMID: 31959131 PMCID: PMC6971947 DOI: 10.1186/s12885-020-6529-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 01/10/2020] [Indexed: 02/07/2023] Open
Abstract
Background Breast (BCa) and prostate (PCa) cancers are hormone receptor (HR)-driven cancers. Thus, BCa and PCa patients are given therapies that reduce hormone levels or directly block HR activity; but most patients eventually develop treatment resistance. We have previously reported that interleukin-1 (IL-1) inflammatory cytokine downregulates ERα and AR mRNA in HR-positive (HR+) BCa and PCa cell lines, yet the cells can remain viable. Additionally, we identified pro-survival proteins and processes upregulated by IL-1 in HR+ BCa and PCa cells, that are basally high in HR− BCa and PCa cells. Therefore, we hypothesize that IL-1 confers a conserved gene expression pattern in HR+ BCa and PCa cells that mimics conserved basal gene expression patterns in HR− BCa and PCa cells to promote HR-independent survival and tumorigenicity. Methods We performed RNA sequencing (RNA-seq) for HR+ BCa and PCa cell lines exposed to IL-1 and for untreated HR− BCa and PCa cell lines. We confirmed expression patterns of select genes by RT-qPCR and used siRNA and/or drug inhibition to silence select genes in the BCa and PCa cell lines. Finally, we performed Ingenuity Pathway Analysis (IPA) and used the gene ontology web-based tool, GOrilla, to identify signaling pathways encoded by our RNA-seq data set. Results We identified 350 genes in common between BCa and PCa cells that are induced or repressed by IL-1 in HR+ cells that are, respectively, basally high or low in HR− cells. Among these genes, we identified Sequestome-1 (SQSTM1/p62) and SRY (Sex-Determining Region Y)-Box 9 (SOX9) to be essential for survival of HR− BCa and PCa cell lines. Analysis of publicly available data indicates that p62 and SOX9 expression are elevated in HR-independent BCa and PCa sublines generated in vitro, suggesting that p62 and SOX9 have a role in acquired hormone receptor independence and treatment resistance. We also assessed HR− cell line viability in response to the p62-targeting drug, verteporfin, and found that verteporfin is cytotoxic for HR− cell lines. Conclusions Our 350 gene set can be used to identify novel therapeutic targets and/or biomarkers conserved among acquired (e.g. due to inflammation) or intrinsic HR-independent BCa and PCa.
Collapse
Affiliation(s)
- Afshan F Nawas
- Biological Sciences Department, The University of Texas at Dallas, 800 West Campbell Road, FO-1, Richardson, TX, 75080, USA
| | - Mohammed Kanchwala
- McDermott Center of Human Growth and Development, The University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Shayna E Thomas-Jardin
- Biological Sciences Department, The University of Texas at Dallas, 800 West Campbell Road, FO-1, Richardson, TX, 75080, USA
| | - Haley Dahl
- Biological Sciences Department, The University of Texas at Dallas, 800 West Campbell Road, FO-1, Richardson, TX, 75080, USA
| | - Kelly Daescu
- Biological Sciences Department, The University of Texas at Dallas, 800 West Campbell Road, FO-1, Richardson, TX, 75080, USA
| | - Monica Bautista
- Biological Sciences Department, The University of Texas at Dallas, 800 West Campbell Road, FO-1, Richardson, TX, 75080, USA
| | - Vanessa Anunobi
- Biological Sciences Department, The University of Texas at Dallas, 800 West Campbell Road, FO-1, Richardson, TX, 75080, USA
| | - Ally Wong
- Biological Sciences Department, The University of Texas at Dallas, 800 West Campbell Road, FO-1, Richardson, TX, 75080, USA
| | - Rachel Meade
- Biological Sciences Department, The University of Texas at Dallas, 800 West Campbell Road, FO-1, Richardson, TX, 75080, USA
| | - Ragini Mistry
- Biological Sciences Department, The University of Texas at Dallas, 800 West Campbell Road, FO-1, Richardson, TX, 75080, USA
| | - Nisha Ghatwai
- Biological Sciences Department, The University of Texas at Dallas, 800 West Campbell Road, FO-1, Richardson, TX, 75080, USA
| | - Felix Bayerl
- Biological Sciences Department, The University of Texas at Dallas, 800 West Campbell Road, FO-1, Richardson, TX, 75080, USA
| | - Chao Xing
- McDermott Center of Human Growth and Development, The University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.,Department of Bioinformatics, The University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.,Department of Clinical Sciences, The University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Nikki A Delk
- Biological Sciences Department, The University of Texas at Dallas, 800 West Campbell Road, FO-1, Richardson, TX, 75080, USA.
| |
Collapse
|
13
|
Does Direct and Indirect Exposure to Ionising Radiation Influence the Metastatic Potential of Breast Cancer Cells. Cancers (Basel) 2020; 12:cancers12010236. [PMID: 31963587 PMCID: PMC7016586 DOI: 10.3390/cancers12010236] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 01/14/2020] [Accepted: 01/15/2020] [Indexed: 12/13/2022] Open
Abstract
Ionising radiation (IR) is commonly used for cancer therapy; however, its potential influence on the metastatic ability of surviving cancer cells exposed directly or indirectly to IR remains controversial. Metastasis is a multistep process by which the cancer cells dissociate from the initial site, invade, travel through the blood stream or lymphatic system, and colonise distant sites. This complex process has been reported to require cancer cells to undergo epithelial-mesenchymal transition (EMT) by which the cancer cells convert from an adhesive, epithelial to motile, mesenchymal form and is also associated with changes in glycosylation of cell surface proteins, which may be functionally involved in metastasis. In this paper, we give an overview of metastatic mechanisms and of the fundamentals of cancer-associated glycosylation changes. While not attempting a comprehensive review of this wide and fast moving field, we highlight some of the accumulating evidence from in vitro and in vivo models for increased metastatic potential in cancer cells that survive IR, focusing on angiogenesis, cancer cell motility, invasion, and EMT and glycosylation. We also explore the indirect effects in cells exposed to exosomes released from irradiated cells. The results of such studies need to be interpreted with caution and there remains limited evidence that radiotherapy enhances the metastatic capacity of cancers in a clinical setting and undoubtedly has a very positive clinical benefit. However, there is potential that this therapeutic benefit may ultimately be enhanced through a better understanding of the direct and indirect effects of IR on cancer cell behaviour.
Collapse
|
14
|
Eras N, Daloglu FT, Çolak T, Guler M, Akbas E. The Correlation between IL-1β-C31T Gene Polymorphism and Susceptibility to Breast Cancer. J Breast Cancer 2019; 22:210-218. [PMID: 31281724 PMCID: PMC6597413 DOI: 10.4048/jbc.2019.22.e27] [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: 11/23/2018] [Accepted: 05/09/2019] [Indexed: 12/18/2022] Open
Abstract
Purpose Interleukin-1 beta (IL-1β), a pro-inflammatory cytokine, has been shown to influence breast cancer susceptibility. The relationship between its risk of breast cancer and IL-1β-C31T polymorphism has been demonstrated, but the results remain controversial. Therefore, our study aimed to investigate the correlation between the IL-1β-C31T gene polymorphism and susceptibility to breast cancer. Methods The genotype frequencies of IL-1β-C31T polymorphism were compared between 204 breast cancer cases and 210 controls using polymerase chain reaction and restriction fragment length polymorphism techinques. Further multivariate binary logistic regression analyses were used to assess the association between IL-1β-C31T polymorphism and breast cancer risk. Results The frequency of the T allele of IL-1β-C31T polymorphism in breast cancer cases was significantly higher than that in the controls (56.1% vs. 47.9%). The frequencies of genotypes CC, CT, and TT in the cases were 22.1%, 43.6%, and 34.3%, respectively, while in the control group they were 24.3%, 55.7%, and 20.0%, respectively. There was a significant difference between the prevalence of TT genotype in the 2 groups (adjusted odds ratio [OR], 2.06; 95% confidence interval [CI], 1.16–3.66; p = 0.014). Breast cancer risk increased in women with TT genotype, body mass index (BMI) ≥ 25 kg/m2 (OR, 2.19; 95% CI, 1.09–4.36), late age at first birth (OR, 2.43; 95% CI, 1.29–4.56), postmenopausal status (OR, 3.15; 95% CI, 1.39–7.16), and negative smoking history (OR, 2.52; 95% CI, 1.32–4.82). Furthermore, increase in breast cancer risk among women diagnosed with invasive ductal carcinoma was associated with CT/TT genotypes (OR, 2.82; 95% CI, 1.38–5.76). Conclusion The IL-1β-C31T polymorphism affects breast cancer susceptibility, especially in women with late age at first birth, high BMI, postmenopausal status, negative smoking history, and invasive ductal carcinoma. Our study adds to the evidence about the importance of IL-1β-C31T polymorphism in breast cancer susceptibility.
Collapse
Affiliation(s)
- Nazan Eras
- Department of Medical Genetics, Faculty of Medicine, Mersin University, Mersin, Turkey
| | | | - Tahsin Çolak
- Department of General Surgery, Faculty of Medicine, Mersin University, Mersin, Turkey
| | - Mehmet Guler
- Department of General Surgery, Medical Park Hospital, Antalya, Turkey
| | - Etem Akbas
- Department of Medical Biology, Faculty of Medicine, Mersin University, Mersin, Turkey
| |
Collapse
|
15
|
Tawara K, Scott H, Emathinger J, Wolf C, LaJoie D, Hedeen D, Bond L, Montgomery P, Jorcyk C. HIGH expression of OSM and IL-6 are associated with decreased breast cancer survival: synergistic induction of IL-6 secretion by OSM and IL-1β. Oncotarget 2019; 10:2068-2085. [PMID: 31007849 PMCID: PMC6459341 DOI: 10.18632/oncotarget.26699] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 01/31/2019] [Indexed: 02/07/2023] Open
Abstract
Chronic inflammation has been recognized as a risk factor for the development and maintenance of malignant disease. Cytokines such as interleukin-6 (IL-6), oncostatin M (OSM), and interleukin-1 beta (IL-1β) promote the development of both acute and chronic inflammation while promoting in vitro metrics of breast cancer metastasis. However, anti-IL-6 and anti-IL-1β therapeutics have not yielded significant results against solid tumors in clinical trials. Here we show that these three cytokines are interrelated in expression. Using the Curtis TCGA™ dataset, we have determined that there is a correlation between expression levels of OSM, IL-6, and IL-1β and reduced breast cancer patient survival (r = 0.6, p = 2.2 x 10−23). Importantly, we confirm that OSM induces at least a 4-fold increase in IL-6 production from estrogen receptor-negative (ER−) breast cancer cells in a manner that is dependent on STAT3 signaling. Furthermore, OSM induces STAT3 phosphorylation and IL-1β promotes p65 phosphorylation to synergistically induce IL-6 secretion in ER− MDA-MB-231 and to a lesser extent in ER+ MCF7 human breast cancer cells. Induction may be reduced in the ER+ MCF7 cells due to a previously known suppressive interaction between ER and STAT3. Interestingly, we show in MCF7 cells that ER’s interaction with STAT3 is reduced by 50% through both OSM and IL-1β treatment, suggesting a role for ER in mitigating STAT3-mediated inflammatory cascades. Here, we provide a rationale for a breast cancer treatment regime that simultaneously suppresses multiple targets, as these cytokines possess many overlapping functions that increase metastasis and worsen patient survival.
Collapse
Affiliation(s)
- Ken Tawara
- Boise State University, Biomolecular Sciences Program, Boise, ID, USA
| | - Hannah Scott
- Boise State University, Department of Biological Sciences, Boise, ID, USA
| | | | - Cody Wolf
- Boise State University, Biomolecular Sciences Program, Boise, ID, USA.,Boise State University, Department of Biological Sciences, Boise, ID, USA
| | - Dollie LaJoie
- Boise State University, Department of Biological Sciences, Boise, ID, USA.,University of Utah, Department of Oncological Sciences, Salt Lake City, UT, USA
| | - Danielle Hedeen
- Boise State University, Department of Biological Sciences, Boise, ID, USA.,University of Utah, Department of Oncological Sciences, Salt Lake City, UT, USA
| | - Laura Bond
- Boise State University, Biomolecular Research Center, Boise, ID, USA
| | | | - Cheryl Jorcyk
- Boise State University, Biomolecular Sciences Program, Boise, ID, USA.,Boise State University, Department of Biological Sciences, Boise, ID, USA
| |
Collapse
|
16
|
Nawas A, Narayanan S, Mistry R, Thomas-Jardin S, Ramachandran J, Ravichandran J, Neduvelil E, Luangpanh K, Delk NA. IL-1 induces p62/SQSTM1 and autophagy in ERα + /PR + BCa cell lines concomitant with ERα and PR repression, conferring an ERα - /PR - BCa-like phenotype. J Cell Biochem 2019; 120:1477-1491. [PMID: 30324661 PMCID: PMC6465183 DOI: 10.1002/jcb.27340] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 06/26/2018] [Indexed: 02/06/2023]
Abstract
Estrogen receptor α (ERα)low/- tumors are associated with breast cancer (BCa) endocrine resistance, where ERα low tumors show a poor prognosis and a molecular profile similar to triple negative BCa tumors. Interleukin-1 (IL-1) downregulates ERα accumulation in BCa cell lines, yet the cells can remain viable. In kind, IL-1 and ERα show inverse accumulation in BCa patient tumors and IL-1 is implicated in BCa progression. IL-1 represses the androgen receptor hormone receptor in prostate cancer cells concomitant with the upregulation of the prosurvival, autophagy-related protein, Sequestome-1 (p62/SQSTM1; hereinafter, p62); and given their similar etiology, we hypothesized that IL-1 also upregulates p62 in BCa cells concomitant with hormone receptor repression. To test our hypothesis, BCa cell lines were exposed to conditioned medium from IL-1-secreting bone marrow stromal cells (BMSCs), IL-1, or IL-1 receptor antagonist. Cells were analyzed for the accumulation of ERα, progesterone receptor (PR), p62, or the autophagosome membrane protein, microtubule-associated protein 1 light chain 3 (LC3), and for p62-LC3 interaction. We found that IL-1 is sufficient to mediate BMSC-induced ERα and PR repression, p62 and autophagy upregulation, and p62-LC3 interaction in ERα+ /PR+ BCa cell lines. However, IL-1 does not significantly elevate the high basal p62 accumulation or high basal autophagy in the ERα- /PR- BCa cell lines. Thus, our observations imply that IL-1 confers a prosurvival ERα- /PR- molecular phenotype in ERα+ /PR+ BCa cells that may be dependent on p62 function and autophagy and may underlie endocrine resistance.
Collapse
Affiliation(s)
- A.F. Nawas
- Biological Sciences Department, University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080
| | - S. Narayanan
- Biological Sciences Department, University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080
| | - R. Mistry
- Biological Sciences Department, University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080
| | - S.E. Thomas-Jardin
- Biological Sciences Department, University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080
| | - J. Ramachandran
- Biological Sciences Department, University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080
| | - J. Ravichandran
- Biological Sciences Department, University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080
| | - E. Neduvelil
- Biological Sciences Department, University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080
| | - K. Luangpanh
- Biological Sciences Department, University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080
| | - N. A. Delk
- Biological Sciences Department, University of Texas at Dallas, 800 West Campbell Road, Richardson, TX 75080
| |
Collapse
|
17
|
Todorović-Raković N, Radulovic M, Vujasinović T, Milovanović J, Nikolić-Vukosavljević D. The time-dependent prognostic value of intratumoral cytokine expression profiles in a natural course of primary breast cancer with a long-term follow-up. Cytokine 2018; 102:12-17. [PMID: 29245048 DOI: 10.1016/j.cyto.2017.11.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 11/27/2017] [Accepted: 11/29/2017] [Indexed: 01/05/2023]
Abstract
Despite the increasing evidence for the importance of immunity in breast cancer, the contradictory role of inflammation has not been thoroughly researched. In this study, we investigate the prognostic value of intratumoral inflammation as evaluated by cytokine mRNA levels. Intratumoral mRNA was measured for IL1β, IL6, IL8, IL10 and IL17A, using Taqman quantitative PCR. By the AUC criteria, none of the cytokines associated with metastasis outcome over the entire follow-up period. However, separation of the follow-up period has revealed a time-dependent and robust prognostic association of IL β. It discriminated between patients with and without metastasis relapse by AUCs of 0.21 and 0.82 during the early and late follow-up of 0-7 and 7-14 years, respectively. Interestingly, the prognostic effect by IL1β shifted during follow-up from good prognosis in the first seven years to bad prognosis thereafter. By the less stringent criteria of Cox regression analysis, other cytokines also significantly associated positively or negatively with metastasis outcome. IL17A associated with good prognosis in the first 7 years of follow up while IL6 associated with poor and IL10 with good prognosis from 7 to 14 years. The revealed time-dependent prognostic effects of cytokine mRNA levels are intriguing and may reflect valuable biological information which should be considered in breast cancer immunotherapy research.
Collapse
Affiliation(s)
- Nataša Todorović-Raković
- Department of Experimental Oncology, Institute of Oncology and Radiology of Serbia, Belgrade, Serbia.
| | - Marko Radulovic
- Department of Experimental Oncology, Institute of Oncology and Radiology of Serbia, Belgrade, Serbia
| | - Tijana Vujasinović
- Department of Experimental Oncology, Institute of Oncology and Radiology of Serbia, Belgrade, Serbia
| | - Jelena Milovanović
- Department of Experimental Oncology, Institute of Oncology and Radiology of Serbia, Belgrade, Serbia
| | | |
Collapse
|
18
|
Setrerrahmane S, Xu H. Tumor-related interleukins: old validated targets for new anti-cancer drug development. Mol Cancer 2017; 16:153. [PMID: 28927416 PMCID: PMC5606116 DOI: 10.1186/s12943-017-0721-9] [Citation(s) in RCA: 149] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 09/05/2017] [Indexed: 02/07/2023] Open
Abstract
In-depth knowledge of cancer molecular and cellular mechanisms have revealed a strong regulation of cancer development and progression by the inflammation which orchestrates the tumor microenvironment. Immune cells, residents or recruited, in the inflammation milieu can have rather contrasting effects during cancer development. Accumulated clinical and experimental data support the notion that acute inflammation could exert an immunoprotective effect leading to tumor eradication. However, chronic immune response promotes tumor growth and invasion. These reactions are mediated by soluble mediators or cytokines produced by either host immune cells or tumor cells themselves. Herein, we provide an overview of the current understanding of the role of the best-validated cytokines involved in tumor progression, IL-1, IL-4 and IL-6; in addition to IL-2 cytokines family, which is known to promote tumor eradication by immune cells. Furthermore, we summarize the clinical attempts to block or bolster the effect of these tumor-related interleukins in anti-cancer therapy development.
Collapse
Affiliation(s)
- Sarra Setrerrahmane
- The Engineering Research Center of Peptide Drug Discovery and Development, China Pharmaceutical University, Nanjing, Jiangsu, 210009, People's Republic of China
| | - Hanmei Xu
- The Engineering Research Center of Peptide Drug Discovery and Development, China Pharmaceutical University, Nanjing, Jiangsu, 210009, People's Republic of China. .,State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing, Jiangsu, 210009, People's Republic of China.
| |
Collapse
|
19
|
Espinoza-Sánchez NA, Chimal-Ramírez GK, Mantilla A, Fuentes-Pananá EM. IL-1β, IL-8, and Matrix Metalloproteinases-1, -2, and -10 Are Enriched upon Monocyte-Breast Cancer Cell Cocultivation in a Matrigel-Based Three-Dimensional System. Front Immunol 2017; 8:205. [PMID: 28337194 PMCID: PMC5340783 DOI: 10.3389/fimmu.2017.00205] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 02/14/2017] [Indexed: 12/21/2022] Open
Abstract
Breast cancer remains the first cancer-related cause of death in women worldwide, particularly in developing countries in which most cases are diagnosed in late stages. Although most cancer studies are based in the genetic or epigenetic changes of the tumor cells, immune cells within the tumor stroma often cooperate with cancer progression. Particularly, monocytes are attracted to the tumor primary site in which they are differentiated into tumor-associated macrophages that facilitate tumor cell invasion and metastasis. In this study, we used three-dimensional cultures to form acini-like structures to analyze the inflammatory secretion profile of tumor cells individually or in co-culture with monocytes. Breast cancer cell lines and primary isolates from eight Mexican patients with breast cancer were used. We found high levels of RANTES/CCL5, MCP-1/CCL2, and G-CSF in the breast cancer individual cultures, supporting an important recruitment capacity of monocytes, but also of neutrophils. The co-cultures of the tumor cells and monocytes were significantly enriched with the potent pro-inflammatory cytokines interleukin (IL)-1β and IL-8, known to support malignant progression. We also found that the interaction of tumor cells with monocytes promoted high levels of matrix metalloproteinases (MMP)-1, MMP-2, and MMP-10. Our study supports that a key event for malignant progression is the recruitment of different immune cell populations, which help to sustain and enhance a chronic inflammatory microenvironment that highly favors tumor malignancy.
Collapse
Affiliation(s)
- Nancy Adriana Espinoza-Sánchez
- Unidad de Investigación en Virología y Cáncer, Hospital Infantil de México Federico Gómez, Ciudad de México, México; Programa de Doctorado en Ciencias Biomédicas, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Gloria Karina Chimal-Ramírez
- Unidad de Investigación en Virología y Cáncer, Hospital Infantil de México Federico Gómez , Ciudad de México , México
| | - Alejandra Mantilla
- Departamento de Patología, Hospital de Oncología, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social , Ciudad de México , México
| | | |
Collapse
|
20
|
Chung STM, Geerts D, Roseman K, Renaud A, Connelly L. Osteoprotegerin mediates tumor-promoting effects of Interleukin-1beta in breast cancer cells. Mol Cancer 2017; 16:27. [PMID: 28143606 PMCID: PMC5286681 DOI: 10.1186/s12943-017-0606-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 01/25/2017] [Indexed: 12/26/2022] Open
Abstract
Background It is widely recognized that inflammation promotes breast cancer invasion and metastasis. Given the complex nature of the breast tumor inflammatory microenvironment, much remains to be understood of the molecular mechanisms that govern these effects. We have previously shown that osteoprotegerin knockdown in breast cancer cells resulted in reduced invasion and metastasis. Here we present novel insight into the role of osteoprotegerin in inflammation-driven tumor progression in breast cancer by investigating the link between osteoprotegerin, macrophages and the potent pro-inflammatory cytokine Interleukin-1beta. Methods We used human breast cancer cell lines to investigate the effects of Interleukin-1beta treatment on osteoprotegerin secretion as measured by ELISA. We analyzed public datasets containing human breast cancer genome-wide mRNA expression data to reveal a significant and positive correlation between osteoprotegerin mRNA expression and the mRNA expression of Interleukin-1beta and of monocyte chemoattractant protein CC-chemokine ligand 2. Osteoprotegerin, Interleukin-1beta and CC-chemokine ligand 2 mRNA levels were also examined by qPCR on cDNA from normal and cancerous human breast tissue. We determined the effect of Interleukin-1beta–producing macrophages on osteoprotegerin expression by co-culturing breast cancer cells and differentiated THP-1 macrophages. Immunohistochemistry was performed on human breast tumor tissue microarrays to assess macrophage infiltration and osteoprotegerin expression. To demonstrate that osteoprotegerin mediated functional effects of Interleukin-1beta we performed cell invasion studies with control and OPG siRNA knockdown on Interleukin-1beta-treated breast cancer cells. Results We report that Interleukin-1beta induces osteoprotegerin secretion, independent of breast cancer subtype and basal osteoprotegerin levels. Co-culture of breast cancer cells with Interleukin-1beta-secreting macrophages resulted in a similar increase in osteoprotegerin secretion in breast cancer cells as Interleukin-1beta treatment. Macrophage infiltration correlates with osteoprotegerin secretion in human breast tumor tissue samples. We show that osteoprotegerin secretion is regulated by Interleukin-1beta in a p38- and p42/44-dependent manner. We also demonstrate that osteoprotegerin knockdown represses Interleukin-1beta expression, Interleukin-1beta-mediated breast cancer cell invasion and MMP3 expression. Conclusions These data indicate a novel role for osteoprotegerin as a mediator of inflammation- promoted breast cancer progression. Electronic supplementary material The online version of this article (doi:10.1186/s12943-017-0606-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Stephanie Tsang Mui Chung
- Department of Pharmaceutical Sciences, Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, Hilo, Hawaii, USA
| | - Dirk Geerts
- Department of Pediatric Oncology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Kim Roseman
- Department of Pharmaceutical Sciences, Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, Hilo, Hawaii, USA
| | - Ashleigh Renaud
- Department of Pharmaceutical Sciences, Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, Hilo, Hawaii, USA
| | - Linda Connelly
- Department of Pharmaceutical Sciences, Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, Hilo, Hawaii, USA.
| |
Collapse
|
21
|
Hickish T, Andre T, Wyrwicz L, Saunders M, Sarosiek T, Kocsis J, Nemecek R, Rogowski W, Lesniewski-Kmak K, Petruzelka L, Apte RN, Mohanty P, Stecher M, Simard J, de Gramont A. MABp1 as a novel antibody treatment for advanced colorectal cancer: a randomised, double-blind, placebo-controlled, phase 3 study. Lancet Oncol 2017; 18:192-201. [PMID: 28094194 DOI: 10.1016/s1470-2045(17)30006-2] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 10/07/2016] [Accepted: 10/19/2016] [Indexed: 12/29/2022]
Abstract
BACKGROUND MABp1, an antibody that targets interleukin 1α, has been associated with antitumour activity and relief of debilitating symptoms in patients with advanced colorectal cancer. We sought to establish the effect of MABp1 with a new primary endpoint in patients with advanced colorectal cancer. METHODS Eligible patients for the double-blind phase of this ongoing, placebo-controlled, randomised, phase 3 trial, had metastatic or unresectable disease, Eastern Cooperative Oncology Group performance status score 1 or 2, systemic inflammation, weight loss, and other disease-related morbidities associated with poor prognosis, and were refractory to oxaliplatin and irinotecan. Patients were randomly assigned 2:1 to receive either MABp1 or placebo. Randomisation codes were obtained from a centrally held list via an interactive web response system. Patients received an intravenous infusion of 7·5 mg/kg MABp1 or placebo given every 2 weeks for 8 weeks. The primary endpoint was assessed in patients who received at least one dose of MABp1 or placebo (modified intention-to-treat population), and was a composite of stable or increased lean body mass and stability or improvement in two of three symptoms (pain, fatigue, or anorexia) at week 8 compared with baseline measurements. This study is registered with ClinicalTrials.gov, number NCT02138422. FINDINGS Patients were enrolled between May 20, 2014, and Sept 2, 2015. The double-blind phase of the study was completed on Nov 3, 2015. Of 333 patients randomly assigned treatment, 207 received at least one dose of MABp1 and 102 at least one dose of placebo. 68 (33%) and 19 (19%) patients, respectively, achieved the primary endpoint (relative risk 1·76, 95% CI 1·12-2·77, p=0·0045). The most common grade 3-4 adverse events in the MABp1 group compared with in the placebo group were anaemia (eight [4%] of 207 vs five [5%] of 102 patients), increased concentration of alkaline phosphatase (nine [4%] vs two [2%]), fatigue (six [3%] vs seven [7%]), and increased concentration of aspartate aminotransferase (six [3%] vs two [2%]). After 8 weeks, 17 (8%) patients in the MABp1 group and 11 (11%) in the placebo group had died, but no death was judged to be related to treatment. The incidence of serious adverse events was not significantly different in the MABp1 group and placebo groups (47 [23%] vs 33 [32%], p=0·07). INTERPRETATION The primary endpoint was a useful means of measuring clinical performance in patients. MABp1 might represent a new standard in the management of advanced colorectal cancer. FUNDING XBiotech.
Collapse
Affiliation(s)
- Tamas Hickish
- Poole Hospital NHS Foundation Trust, Poole, Dorset, UK; Oncology Department, Royal Bournemouth Hospital NHS Foundation Trust Bournemouth, UK; Department of Oncology, Bournemouth University, Bournemouth, UK.
| | - Thierry Andre
- Oncology Department, Saint Antoine Hospital, and Pierre and Marie Curie University (Paris 6), Paris, France
| | - Lucjan Wyrwicz
- Maria Sklodowska-Curie Memorial Cancer Centre and Institute of Oncology, Warsaw, Poland
| | | | | | | | - Radim Nemecek
- Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - Wojciech Rogowski
- Clinical Department of Chemotherapy, Hospital Ministry of the Interior and Administration and Warmia and Mazury Oncology Centre, Olsztyn, Poland
| | | | | | - Ron N Apte
- Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | | | | | | | - Aimery de Gramont
- Oncology Department, Institut Hospitalier Franco-Britannique, Levallois-Perret, France
| |
Collapse
|
22
|
Simsa P, Mihalyi A, Kyama CM, Mwenda JM, Fülöp V, D'Hooghe TM. Selective Estrogen-Receptor Modulators and Aromatase Inhibitors: Promising New Medical Therapies for Endometriosis? WOMENS HEALTH 2016; 3:617-28. [DOI: 10.2217/17455057.3.5.617] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Endometriosis is an estrogen-dependent disease and estrogen-related pathways are imbalanced in women with endometriosis. One of the key enzymes in estrogen synthesis is aromatase. Inhibiting this pathway at several points is a promising idea for the treatment of endometriosis. The third generation of aromatase inhibitors is becoming more potent in efficacy, with fewer side effects than previous generations, but cotreatment with other hormones is needed to inhibit ovarian stimulation. Other components that promote estrogen synthesis such as COX-2 can also be potentially targeted. Selective estrogen-receptor modulators could also be interesting in view of their tissue-specific effect. However, all these new drugs are still in an early phase of development. At present, it is too early to conclude that aromatase inhibitors, COX-2 inhibitors or selective estrogen-receptor modulators really present any added value compared with the existing drugs that can be used to achieve hormonal suppression in the medical treatment of endometriosis.
Collapse
Affiliation(s)
- Peter Simsa
- Leuven University Fertility Centre, Department of Obstetrics & Gynaecology, University Hospital Gasthuisberg, Leuven, Belgium, Tel: +32 1634 3624; Fax: +32 1634 3607
- National Institute of Health, Budapest, Hungary
| | - Attila Mihalyi
- Leuven University Fertility Centre, Department of Obstetrics & Gynaecology, University Hospital Gasthuisberg, Leuven, Belgium, Tel: +32 1634 3624; Fax: +32 1634 3607
| | - Cleophas M Kyama
- Leuven University Fertility Centre, Department of Obstetrics & Gynaecology, University Hospital Gasthuisberg, Leuven, Belgium, Tel: +32 1634 3624; Fax: +32 1634 3607
- Institute of Primate Research, Division of Reproduction, PO Box 24481, Karen, Nairobi, Kenya
| | | | | | - Thomas M D'Hooghe
- Leuven University Fertility Centre, Department of Obstetrics & Gynaecology, University Hospital Gasthuisberg, Leuven, Belgium, Tel: +32 1634 3624; Fax: +32 1634 3607
- Institute of Primate Research, Division of Reproduction, PO Box 24481, Karen, Nairobi, Kenya
| |
Collapse
|
23
|
Kohnz RA, Roberts LS, DeTomaso D, Bideyan L, Yan P, Bandyopadhyay S, Goga A, Yosef N, Nomura DK. Protein Sialylation Regulates a Gene Expression Signature that Promotes Breast Cancer Cell Pathogenicity. ACS Chem Biol 2016; 11:2131-9. [PMID: 27380425 PMCID: PMC4994060 DOI: 10.1021/acschembio.6b00433] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
![]()
Many mechanisms have been proposed
for how heightened aerobic glycolytic
metabolism fuels cancer pathogenicity, but there are still many unexplored
pathways. Here, we have performed metabolomic profiling to map glucose
incorporation into metabolic pathways upon transformation of mammary
epithelial cells by 11 commonly mutated human oncogenes. We show that
transformation of mammary epithelial cells by oncogenic stimuli commonly
shunts glucose-derived carbons into synthesis of sialic acid, a hexosamine
pathway metabolite that is converted to CMP-sialic acid by cytidine
monophosphate N-acetylneuraminic acid synthase (CMAS)
as a precursor to glycoprotein and glycolipid sialylation. We show
that CMAS knockdown leads to elevations in intracellular sialic acid
levels, a depletion of cellular sialylation, and alterations in the
expression of many cancer-relevant genes to impair breast cancer pathogenicity.
Our study reveals the heretofore unrecognized role of sialic acid
metabolism and protein sialylation in regulating the expression of
genes that maintain breast cancer pathogenicity.
Collapse
Affiliation(s)
- Rebecca A. Kohnz
- Departments
of Chemistry, Molecular and Cell Biology, and Nutritional Sciences
and Toxicology, University of California, Berkeley, Berkeley, California 94720, United States
| | - Lindsay S. Roberts
- Departments
of Chemistry, Molecular and Cell Biology, and Nutritional Sciences
and Toxicology, University of California, Berkeley, Berkeley, California 94720, United States
| | - David DeTomaso
- Department
of Electrical Engineering and Computer Science, University of California, Berkeley, Berkeley, California 94720, United States
| | - Lara Bideyan
- Departments
of Chemistry, Molecular and Cell Biology, and Nutritional Sciences
and Toxicology, University of California, Berkeley, Berkeley, California 94720, United States
| | - Peter Yan
- Departments
of Chemistry, Molecular and Cell Biology, and Nutritional Sciences
and Toxicology, University of California, Berkeley, Berkeley, California 94720, United States
| | - Sourav Bandyopadhyay
- Division
of Hematology/Oncology, Department of Cell and Tissue Biology, University of California, San Francisco, 513 Parnassus Avenue HSW616, San Francisco, California 94143, United States
- University of California, San Francisco Helen Diller
Family Comprehensive Cancer Center, Box
0128, San Francisco, California 94143, United States
| | - Andrei Goga
- Division
of Hematology/Oncology, Department of Cell and Tissue Biology, University of California, San Francisco, 513 Parnassus Avenue HSW616, San Francisco, California 94143, United States
- University of California, San Francisco Helen Diller
Family Comprehensive Cancer Center, Box
0128, San Francisco, California 94143, United States
| | - Nir Yosef
- Department
of Electrical Engineering and Computer Science, University of California, Berkeley, Berkeley, California 94720, United States
| | - Daniel K. Nomura
- Departments
of Chemistry, Molecular and Cell Biology, and Nutritional Sciences
and Toxicology, University of California, Berkeley, Berkeley, California 94720, United States
| |
Collapse
|
24
|
Abstract
The human body combats infection and promotes wound healing through the remarkable process of inflammation. Inflammation is characterized by the recruitment of stromal cell activity including recruitment of immune cells and induction of angiogenesis. These cellular processes are regulated by a class of soluble molecules called cytokines. Based on function, cell target, and structure, cytokines are subdivided into several classes including: interleukins, chemokines, and lymphokines. While cytokines regulate normal physiological processes, chronic deregulation of cytokine expression and activity contributes to cancer in many ways. Gene polymorphisms of all types of cytokines are associated with risk of disease development. Deregulation RNA and protein expression of interleukins, chemokines, and lymphokines have been detected in many solid tumors and hematopoetic malignancies, correlating with poor patient prognosis. The current body of literature suggests that in some tumor types, interleukins and chemokines work against the human body by signaling to cancer cells and remodeling the local microenvironment to support the growth, survival, and invasion of primary tumors and enhance metastatic colonization. Some lymphokines are downregulated to suppress tumor progression by enhancing cytotoxic T cell activity and inhibiting tumor cell survival. In this review, we will describe the structure/function of several cytokine families and review our current understanding on the roles and mechanisms of cytokines in tumor progression. In addition, we will also discuss strategies for exploiting the expression and activity of cytokines in therapeutic intervention.
Collapse
Affiliation(s)
- M Yao
- University of Kansas Medical Center, Kansas City, KS, United States
| | - G Brummer
- University of Kansas Medical Center, Kansas City, KS, United States
| | - D Acevedo
- University of Kansas Medical Center, Kansas City, KS, United States
| | - N Cheng
- University of Kansas Medical Center, Kansas City, KS, United States.
| |
Collapse
|
25
|
Celik B, Yalcin AD, Genc GE, Bulut T, Kuloglu Genc S, Gumuslu S. CXCL8, IL-1β and sCD200 are pro-inflammatory cytokines and their levels increase in the circulation of breast carcinoma patients. Biomed Rep 2016; 5:259-263. [PMID: 27446554 PMCID: PMC4950671 DOI: 10.3892/br.2016.709] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 06/03/2016] [Indexed: 02/05/2023] Open
Abstract
The influence of biomarkers on carcinogenesis has been investigated extensively. Whether they promote carcinogenesis or work against cancer development remains to be elucidated. To the best of our knowledge, the novel molecule cluster of differentiation 200 (CD200) has not been studied on human breast cancer subjects. The present study aimed to evaluate interleukin-1β (IL-1β), C-X-C motif chemokine ligand 8 (CXCL8), cancer antigen 15.3 (CA 15.3) and the soluble CD200 (sCD200) levels in the serum samples of breast carcinoma patients in order to predict their role in breast carcinoma. The subjects included individuals with early and advanced stage breast cancers, as well as healthy controls. Commercially available ELISA kits were used to measure the serum concentrations of sCD200, IL-1β, CXCL8, CA 15.3, C-reactive protein (CRP) and leukocyte count. A total of 130 subjects were recruited; 50 early stage cancer, 50 advanced stage and 30 control subjects. Serum sCD200, CXCL8, IL-1β and CRP levels were significantly higher in the early as well as the advanced stage breast cancer patients compared to the control group. The level of CA 15.3 was statistically different between early and advanced stage. There were significant positive correlations between IL-1β and CXCL8, and IL-1β and serum sCD200 levels in the control group. These correlations did not persist in the early or the advanced stage cancer groups except CRP and CA 15.3, but new correlations appeared between serum sCD200 level and leukocyte count for advanced stage breast cancer group. Multivariate regression correlation analysis revealed positive correlation between IL-1β and sCD200; and IL-1β and CXCL8. In conclusion, sCD200, CXCL8, CA 15.3 and IL-1β are proinflammatory molecules and their levels are influenced in breast cancer patients.
Collapse
Affiliation(s)
- Betul Celik
- Department of Pathology, Antalya Training and Research Hospital, 07100 Antalya, Turkey
| | - Arzu Didem Yalcin
- Department of Internal Medicine, Allergy and Clinical Immunology Unit, Antalya Training and Research Hospital, 07100 Antalya, Turkey
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan, R.O.C
| | - Gizem Esra Genc
- Department of Medical Biochemistry, Faculty of Medicine, Akdeniz University, 07070 Antalya, Turkey
| | - Tangul Bulut
- Department of Pathology, Antalya Training and Research Hospital, 07100 Antalya, Turkey
| | - Sibel Kuloglu Genc
- Department of Medical Biochemistry, Faculty of Medicine, Akdeniz University, 07070 Antalya, Turkey
| | - Saadet Gumuslu
- Department of Medical Biochemistry, Faculty of Medicine, Akdeniz University, 07070 Antalya, Turkey
| |
Collapse
|
26
|
Esquivel-Velázquez M, Ostoa-Saloma P, Palacios-Arreola MI, Nava-Castro KE, Castro JI, Morales-Montor J. The role of cytokines in breast cancer development and progression. J Interferon Cytokine Res 2015; 35:1-16. [PMID: 25068787 PMCID: PMC4291218 DOI: 10.1089/jir.2014.0026] [Citation(s) in RCA: 337] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 05/23/2014] [Indexed: 12/12/2022] Open
Abstract
Cytokines are highly inducible, secretory proteins that mediate intercellular communication in the immune system. They are grouped into several protein families that are referred to as tumor necrosis factors, interleukins, interferons, and colony-stimulating factors. In recent years, it has become clear that some of these proteins as well as their receptors are produced in the organisms under physiological and pathological conditions. The exact initiation process of breast cancer is unknown, although several hypotheses have emerged. Inflammation has been proposed as an important player in tumor initiation, promotion, angiogenesis, and metastasis, all phenomena in which cytokines are prominent players. The data here suggest that cytokines play an important role in the regulation of both induction and protection in breast cancer. This knowledge could be fundamental for the proposal of new therapeutic approaches to particularly breast cancer and other cancer-related disorders.
Collapse
Affiliation(s)
- Marcela Esquivel-Velázquez
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México City, México
| | - Pedro Ostoa-Saloma
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México City, México
| | | | - Karen E. Nava-Castro
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, SSA, Cuernavaca, Morelos, México
| | - Julieta Ivonne Castro
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, SSA, Cuernavaca, Morelos, México
| | - Jorge Morales-Montor
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México City, México
| |
Collapse
|
27
|
El-Ghonaimy EA, El-Shinawi M, Ibrahim SA, El-Ghazaly H, Abd-El-Tawab R, Nouh MA, El-Mamlouk T, Mohamed MM. Positive lymph-node breast cancer patients - activation of NF-κB in tumor-associated leukocytes stimulates cytokine secretion that promotes metastasis via C-C chemokine receptor CCR7. FEBS J 2014; 282:271-82. [DOI: 10.1111/febs.13124] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2014] [Revised: 08/23/2014] [Accepted: 10/17/2014] [Indexed: 12/20/2022]
Affiliation(s)
| | | | | | | | | | - Mohamed A. Nouh
- Department of Pathology; National Cancer institute; Cairo University; Giza Egypt
| | | | | |
Collapse
|
28
|
Slattery ML, Herrick JS, Torres-Mejia G, John EM, Giuliano AR, Hines LM, Stern MC, Baumgartner KB, Presson AP, Wolff RK. Genetic variants in interleukin genes are associated with breast cancer risk and survival in a genetically admixed population: the Breast Cancer Health Disparities Study. Carcinogenesis 2014; 35:1750-9. [PMID: 24670917 DOI: 10.1093/carcin/bgu078] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Interleukins (ILs) are key regulators of immune response. Genetic variation in IL genes may influence breast cancer risk and mortality given their role in cell growth, angiogenesis and regulation of inflammatory process. We examined 16 IL genes with breast cancer risk and mortality in an admixed population of Hispanic/Native American (NA) (2111 cases and 2597 controls) and non-Hispanic white (NHW) (1481 cases and 1585 controls) women. Adaptive Rank Truncated Product (ARTP) analysis was conducted to determine gene significance and lasso (least absolute shrinkage and selection operator) was used to identify potential gene by gene and gene by lifestyle interactions. The pathway was statistically significant for breast cancer risk overall (P ARTP = 0.0006), for women with low NA ancestry (P(ARTP) = 0.01), for premenopausal women (P(ARTP) = 0.02), for estrogen receptor (ER)+/progesterone receptor (PR)+ tumors (P(ARTP) = 0.03) and ER-/PR- tumors (P(ARTP) = 0.02). Eight of the 16 genes evaluated were associated with breast cancer risk (IL1A, IL1B, IL1RN, IL2, IL2RA, IL4, IL6 and IL10); four genes were associated with breast cancer risk among women with low NA ancestry (IL1B, IL6, IL6R and IL10), two were associated with breast cancer risk among women with high NA ancestry (IL2 and IL2RA) and four genes were associated with premenopausal breast cancer risk (IL1A, IL1B, IL2 and IL3). IL4, IL6R, IL8 and IL17A were associated with breast cancer-specific mortality. We confirmed associations with several functional polymorphisms previously associated with breast cancer risk and provide support that their combined effect influences the carcinogenic process.
Collapse
Affiliation(s)
- Martha L Slattery
- Department of Medicine, University of Utah, 383 Colorow, Salt Lake City, UT 84108, USA, Instituto Nacional de Salud Pública, Centro de Investigación en Salud Poblacional, Av. Universidad No. 655, Col. Sta. Ma. Ahuacatitlán, Cuernavaca Morelos CP 62100, México, Cancer Prevention Institute of California, Fremont, CA 84108, USA, Division of Epidemiology, Department of Health Research and Policy and Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 62508, USA, Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA, Department of Biology, University of Colorado at Colorado Springs, 1420 Austin Bluffs Parkway, Colorado Springs, CO 80918, USA, Department of Preventive Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA 90089, USA and Department of Epidemiology and Population Health, School of Public Health & Information Sciences, James Graham Brown Cancer Center, University of Louisville, Louisville, KY 90089-9031, USA
| | - Jennifer S Herrick
- Department of Medicine, University of Utah, 383 Colorow, Salt Lake City, UT 84108, USA, Instituto Nacional de Salud Pública, Centro de Investigación en Salud Poblacional, Av. Universidad No. 655, Col. Sta. Ma. Ahuacatitlán, Cuernavaca Morelos CP 62100, México, Cancer Prevention Institute of California, Fremont, CA 84108, USA, Division of Epidemiology, Department of Health Research and Policy and Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 62508, USA, Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA, Department of Biology, University of Colorado at Colorado Springs, 1420 Austin Bluffs Parkway, Colorado Springs, CO 80918, USA, Department of Preventive Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA 90089, USA and Department of Epidemiology and Population Health, School of Public Health & Information Sciences, James Graham Brown Cancer Center, University of Louisville, Louisville, KY 90089-9031, USA
| | - Gabriella Torres-Mejia
- Instituto Nacional de Salud Pública, Centro de Investigación en Salud Poblacional, Av. Universidad No. 655, Col. Sta. Ma. Ahuacatitlán, Cuernavaca Morelos CP 62100, México
| | - Esther M John
- Cancer Prevention Institute of California, Fremont, CA 84108, USA, Division of Epidemiology, Department of Health Research and Policy and Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 62508, USA
| | - Anna R Giuliano
- Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Lisa M Hines
- Department of Biology, University of Colorado at Colorado Springs, 1420 Austin Bluffs Parkway, Colorado Springs, CO 80918, USA
| | - Mariana C Stern
- Department of Preventive Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA 90089, USA and
| | - Kathy B Baumgartner
- Department of Epidemiology and Population Health, School of Public Health & Information Sciences, James Graham Brown Cancer Center, University of Louisville, Louisville, KY 90089-9031, USA
| | - Angela P Presson
- Department of Medicine, University of Utah, 383 Colorow, Salt Lake City, UT 84108, USA, Instituto Nacional de Salud Pública, Centro de Investigación en Salud Poblacional, Av. Universidad No. 655, Col. Sta. Ma. Ahuacatitlán, Cuernavaca Morelos CP 62100, México, Cancer Prevention Institute of California, Fremont, CA 84108, USA, Division of Epidemiology, Department of Health Research and Policy and Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 62508, USA, Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA, Department of Biology, University of Colorado at Colorado Springs, 1420 Austin Bluffs Parkway, Colorado Springs, CO 80918, USA, Department of Preventive Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA 90089, USA and Department of Epidemiology and Population Health, School of Public Health & Information Sciences, James Graham Brown Cancer Center, University of Louisville, Louisville, KY 90089-9031, USA
| | - Roger K Wolff
- Department of Medicine, University of Utah, 383 Colorow, Salt Lake City, UT 84108, USA, Instituto Nacional de Salud Pública, Centro de Investigación en Salud Poblacional, Av. Universidad No. 655, Col. Sta. Ma. Ahuacatitlán, Cuernavaca Morelos CP 62100, México, Cancer Prevention Institute of California, Fremont, CA 84108, USA, Division of Epidemiology, Department of Health Research and Policy and Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 62508, USA, Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA, Department of Biology, University of Colorado at Colorado Springs, 1420 Austin Bluffs Parkway, Colorado Springs, CO 80918, USA, Department of Preventive Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA 90089, USA and Department of Epidemiology and Population Health, School of Public Health & Information Sciences, James Graham Brown Cancer Center, University of Louisville, Louisville, KY 90089-9031, USA
| |
Collapse
|
29
|
Newman G, Gonzalez-Perez RR. Leptin-cytokine crosstalk in breast cancer. Mol Cell Endocrinol 2014; 382:570-582. [PMID: 23562747 PMCID: PMC3844060 DOI: 10.1016/j.mce.2013.03.025] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2013] [Accepted: 03/26/2013] [Indexed: 02/07/2023]
Abstract
Despite accumulating evidence suggesting a positive correlation between leptin levels, obesity, post-menopause and breast cancer incidence, our current knowledge on the mechanisms involved in these relationships is still incomplete. Since the cloning of leptin in 1994 and its receptor (OB-R) 1 year later by Friedman's laboratory (Zhang et al., 1994) and Tartaglia et al. (Tartaglia et al., 1995), respectively, more than 22,000 papers related to leptin functions in several biological systems have been published (Pubmed, 2012). The ob gene product, leptin, is an important circulating signal for the regulation of body weight. Additionally, leptin plays critical roles in the regulation of glucose homeostasis, reproduction, growth and the immune response. Supporting evidence for leptin roles in cancer has been shown in more than 1000 published papers, with almost 300 papers related to breast cancer (Pubmed, 2012). Specific leptin-induced signaling pathways are involved in the increased levels of inflammatory, mitogenic and pro-angiogenic factors in breast cancer. In obesity, a mild inflammatory condition, deregulated secretion of proinflammatory cytokines and adipokines such as IL-1, IL-6, TNF-α and leptin from adipose tissue, inflammatory and cancer cells could contribute to the onset and progression of cancer. We used an in silico software program, Pathway Studio 9, and found 4587 references citing these various interactions. Functional crosstalk between leptin, IL-1 and Notch signaling (NILCO) found in breast cancer cells could represent the integration of developmental, proinflammatory and pro-angiogenic signals critical for leptin-induced breast cancer cell proliferation/migration, tumor angiogenesis and breast cancer stem cells (BCSCs). Remarkably, the inhibition of leptin signaling via leptin peptide receptor antagonists (LPrAs) significantly reduced the establishment and growth of syngeneic, xenograft and carcinogen-induced breast cancer and, simultaneously decreased the levels of VEGF/VEGFR2, IL-1 and Notch. Inhibition of leptin-cytokine crosstalk might serve as a preventative or adjuvant measure to target breast cancer, particularly in obese women. This review is intended to present an update analysis of leptin actions in breast cancer, highlighting its crosstalk to inflammatory cytokines and growth factors essential for tumor development, angiogenesis and potential role in BCSC.
Collapse
Affiliation(s)
- Gale Newman
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, United States.
| | - Ruben Rene Gonzalez-Perez
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, United States.
| |
Collapse
|
30
|
Bhat IA, Naykoo NA, Qasim I, Ganie FA, Yousuf Q, Bhat BA, Rasool R, Aziz SA, Shah ZA. Association of interleukin 1 beta (IL-1β) polymorphism with mRNA expression and risk of non small cell lung cancer. Meta Gene 2014; 2:123-33. [PMID: 25606396 PMCID: PMC4287803 DOI: 10.1016/j.mgene.2013.12.002] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Revised: 12/09/2013] [Accepted: 12/09/2013] [Indexed: 11/29/2022] Open
Abstract
Introduction Interleukin 1 beta (IL- 1β), a key proinflammatory cytokine encoded by the interleukin 1 beta gene, has been associated with chronic inflammation and plays an important role in lung inflammatory diseases including lung cancer. Elevated levels of Interleukin 1proteins, in particular interleukin 1 beta greatly enhance the intensity of the inflammatory response. Aim To study the role of interleukin 1 beta-31C > T and -511 T > C polymorphism in the pathogenesis of non small cell lung cancer (NSCLC). Materials and methods One hundred and ninety non small cell lung cancer patients and 200 healthy age, sex, smoking and dwelling matched controls were used for polymorphic analysis by polymerase chain reaction—restriction fragment length polymorphism (PCR-RFLP) followed by sequencing. Normal tissues of 48 histopathologically confirmed non small cell lung cancer patients were taken for mRNA expression analysis. Quantitation of interleukin 1 beta was carried out by quantitative real time PCR. Result The T/T genotype of interleukin 1 beta-31 gene was significantly associated with increased risk of NSCLC [(P = 0.001, OR – 2.8 (95%CI 1.52–5.26)]. The interleukin 1 beta − 511 T > C does not show any difference between the NSCLC and control group (P = 0.3, OR – 0.72 (95%CI 0.41–1.28). Quantitative analysis of mRNA showed significant association with interleukin 1 beta T allele as compared to the interleukin 1 beta-31C allele (P = 0.006). Conclusion We conclude that lung cancer risk genotype interleukin 1 beta-31TT results in increased expression of interleukin 1 beta mRNA in lung cancer patients. Our data suggest that this genotype (IL1β -31TT) in the interleukin 1 beta regulatory region provide a microenvironment with elevated inflammatory stimuli and thus increasing the risk for lung cancer.
Collapse
Affiliation(s)
- Imtiyaz A Bhat
- Department of Immunology & Molecular Medicine, Sher-i-Kashmir Institute of Medical Sciences, Srinagar, India
| | - Niyaz A Naykoo
- Department of Animal Biotechnology, Sher-i-Kashmir University of Agricultural Sciences, Srinagar, India
| | - Iqbal Qasim
- Department of Immunology & Molecular Medicine, Sher-i-Kashmir Institute of Medical Sciences, Srinagar, India
| | - Farooq A Ganie
- Department of Cardio Vascular and Thoracic Surgery, Sher-i-Kashmir Institute of Medical Sciences, Srinagar India
| | - Qaiser Yousuf
- Advanced Center for Human Genetics, Sher-i-Kashmir Institute of Medical Sciences Srinagar, India
| | - Bashir A Bhat
- Department of Plastic Surgery, Sher-i-Kashmir Institute of Medical Sciences Srinagar India
| | - Roohi Rasool
- Department of Immunology & Molecular Medicine, Sher-i-Kashmir Institute of Medical Sciences, Srinagar, India
| | - S A Aziz
- Department of Medical Oncology, Sher-i-Kashmir Institute of Medical Sciences Srinagar India
| | - Zafar Amin Shah
- Department of Immunology & Molecular Medicine, Sher-i-Kashmir Institute of Medical Sciences, Srinagar, India
| |
Collapse
|
31
|
Mohamed MM, Al-Raawi D, Sabet SF, El-Shinawi M. Inflammatory breast cancer: New factors contribute to disease etiology: A review. J Adv Res 2013; 5:525-36. [PMID: 25685520 PMCID: PMC4294279 DOI: 10.1016/j.jare.2013.06.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2013] [Revised: 05/16/2013] [Accepted: 06/07/2013] [Indexed: 12/11/2022] Open
Abstract
Inflammatory breast cancer (IBC) is a highly metastatic and fatal form of breast cancer. In fact, IBC is characterized by specific morphological, phenotypic, and biological properties that distinguish it from non-IBC. The aggressive behavior of IBC being more common among young women and the low survival rate alarmed researchers to explore the disease biology. Despite the basic and translational studies needed to understand IBC disease biology and identify specific biomarkers, studies are limited by few available IBC cell lines, experimental models, and paucity of patient samples. Above all, in the last decade, researchers were able to identify new factors that may play a crucial role in IBC progression. Among identified factors are cytokines, chemokines, growth factors, and proteases. In addition, viral infection was also suggested to participate in the etiology of IBC disease. In this review, we present novel factors suggested by different studies to contribute to the etiology of IBC and the proposed new therapeutic insights.
Collapse
Affiliation(s)
- Mona M Mohamed
- Department of Zoology, Faculty of Science, Cairo University, Giza 12613, Egypt
| | - Diaa Al-Raawi
- Department of Zoology, Faculty of Science, Sana'a University, Yemen
| | - Salwa F Sabet
- Department of Zoology, Faculty of Science, Cairo University, Giza 12613, Egypt
| | - Mohamed El-Shinawi
- Department of General Surgery, Faculty of Medicine, Ain Shams University, Cairo 11566, Egypt
| |
Collapse
|
32
|
Epidermal growth factor protects squamous cell carcinoma against cisplatin-induced cytotoxicity through increased interleukin-1β expression. PLoS One 2013; 8:e55795. [PMID: 23383347 PMCID: PMC3562190 DOI: 10.1371/journal.pone.0055795] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2012] [Accepted: 01/02/2013] [Indexed: 11/19/2022] Open
Abstract
The expression of cytokines, such as IL-1β, and the activation of the epidermal growth factor receptor (EGFR) are crucial regulators in the process of carcinogenesis. The correlation between growth factor and activated cytokine signals in the control of tumor development is a critical issue to be clarified. In our study, we found that the IL-1β gene and protein expression were induced by EGF in squamous cell carcinoma. To clarify the mechanism involved in EGF-regulated IL-1β expression, we examined the transcriptional activity and mRNA stability of IL-1β in EGF-treated cells. We found that EGF induced the expression of IL-1β and was mediated through transcriptional activation, but not through mRNA stability. The involvement of Akt and NF-κB signaling pathways in the EGF-induced IL-1β gene expression was confirmed by knockdown of RelA and Akt in cells or treating cells with Akt and NF-κB inhibitors, LY294002 and parthenolide, respectively. The expression of dominant negative IκB also repressed the activation of NF-κB and inhibited EGF-induced IL-1β expression. Using immunofluorescence staining assay, the EGF-stimulated nuclear translocation of NF-κB (p65) was inhibited by pre-treating cells with LY294002 and parthenolide. Furthermore, EGF increased the binding of NF-κB to the NF-κB binding site of the IL-1β promoter through the activation of the Akt/NF-κB pathway, which resulted in activating IL-1β promoter activity. The expression and secretion of IL-1β induced by EGF considerably reduced chemotherapeutic drug cisplatin-induced cell death. These results showed that EGF enhanced the expression of IL-1β, which was mediated by the Akt/NF-κB pathway. The activation of EGF signaling and increase of IL-1β contributed to chemotherapeutic resistance of cancer cells, suggesting that the expression of IL-1β may be used as a biomarker to evaluate successful cancer treatment.
Collapse
|
33
|
A systematic review of the association between immunogenomic markers and cancer-related fatigue. Brain Behav Immun 2012; 26:830-48. [PMID: 22595751 PMCID: PMC3398196 DOI: 10.1016/j.bbi.2012.05.004] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Revised: 04/24/2012] [Accepted: 05/08/2012] [Indexed: 01/04/2023] Open
Abstract
Fatigue, which is one of the most commonly reported symptoms in cancer, can negatively impact the functional status and the health-related quality of life of individuals. This paper systematically reviews 34 studies to determine patterns of associations between immunogenomic markers and levels of cancer-related fatigue (CRF). Findings from the longitudinal studies revealed that elevated fatigue symptoms especially of women with early stages of breast cancer were associated with high levels of neutrophil/monocyte, IL-1ra, and IL-6 during radiation therapy; high levels of CD4+, IL-1β, and IL-6 with stressing stimuli; high levels of IL-1β during chemotherapy; low NK cell levels after chemotherapy; and presence of homozygous IL-6 and TNF alleles. In the cross-sectional studies, associations between levels of fatigue and immune/inflammatory markers were not consistently found, especially when covariates such as BMI, ethnicity, menopausal status, and educational level were controlled in the statistical analyses. However, a number of genomic markers were observed to be elevated mostly in fatigued breast cancer survivors in the cross-sectional studies. Gaps in knowledge and recommendations for future research are discussed.
Collapse
|
34
|
Li HJ, Reinhardt F, Herschman HR, Weinberg RA. Cancer-stimulated mesenchymal stem cells create a carcinoma stem cell niche via prostaglandin E2 signaling. Cancer Discov 2012; 2:840-55. [PMID: 22763855 DOI: 10.1158/2159-8290.cd-12-0101] [Citation(s) in RCA: 289] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
UNLABELLED Mesenchymal cells of the tumor-associated stroma are critical determinants of carcinoma cell behavior. We focus here on interactions of carcinoma cells with mesenchymal stem cells (MSC), which are recruited to the tumor stroma and, once present, are able to influence the phenotype of the carcinoma cells. We find that carcinoma cell-derived interleukin-1 (IL-1) induces prostaglandin E(2) (PGE(2)) secretion by MSCs. The resulting PGE(2) operates in an autocrine manner, cooperating with ongoing paracrine IL-1 signaling, to induce expression of a group of cytokines by the MSCs. The PGE(2) and cytokines then proceed to act in a paracrine fashion on the carcinoma cells to induce activation of β-catenin signaling and formation of cancer stem cells. These observations indicate that MSCs and derived cell types create a cancer stem cell niche to enable tumor progression via release of PGE(2) and cytokines. SIGNIFICANCE Although PGE2 has been implicated time and again in fostering tumorigenesis, its effects on carcinoma cells that contribute specifically to tumor formation are poorly understood. Here we show that tumor cells are able to elicit a strong induction of the COX-2/microsomal prostaglandin-E synthase-1 (mPGES-1)/PGE(2) axis in MSCs recruited to the tumor-associated stroma by releasing IL-1, which in turn elicits a mesenchymal/stem cell–like phenotype in the carcinoma cells.
Collapse
Affiliation(s)
- Hua-Jung Li
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts 02142, USA
| | | | | | | |
Collapse
|
35
|
Liu L, Mills PJ, Rissling M, Fiorentino L, Natarajan L, Dimsdale JE, Sadler GR, Parker BA, Ancoli-Israel S. Fatigue and sleep quality are associated with changes in inflammatory markers in breast cancer patients undergoing chemotherapy. Brain Behav Immun 2012; 26:706-13. [PMID: 22406004 PMCID: PMC3372667 DOI: 10.1016/j.bbi.2012.02.001] [Citation(s) in RCA: 154] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2011] [Revised: 02/07/2012] [Accepted: 02/07/2012] [Indexed: 10/28/2022] Open
Abstract
Fatigue and sleep disturbances are two of the most common and distressing symptoms reported by cancer patients. Fatigue and sleep are also correlated with each other. While fatigue has been reported to be associated with some inflammatory markers, data about the relationship between cancer-related sleep disturbances and inflammatory markers are limited. This study examined the relationship between fatigue and sleep, measured both subjectively and objectively, and inflammatory markers in a sample of breast cancer patients before and during chemotherapy. Fifty-three women with newly diagnosed stage I-III breast cancer scheduled to receive at least four 3-week cycles of chemotherapy participated in this longitudinal study. Fatigue was assessed with the Multidimensional Fatigue Symptom Inventory-Short Form (MFSI-SF), sleep quality was assessed with the Pittsburgh Sleep Quality Index (PSQI) and objective sleep was measured with actigraphy. Three inflammatory markers were examined: Interleukin-6 (IL-6), Interleukin-1 receptor antagonist (IL-1RA) and C-reactive protein (CRP). Data were collected before (baseline) and during cycle 1 and cycle 4 of chemotherapy. Compared to baseline, more fatigue was reported, levels of IL-6 increased and IL-1RA decreased during chemotherapy. Reports of sleep quality remained poor. Mixed model analyses examining changes from baseline to each treatment time point revealed overall positive relationships between changes in total MFSI-SF scores and IL-6, between changes in total PSQI scores and IL-6 and IL-1RA, and between total wake time at night and CRP (all p's<0.05). These relationships suggest that cancer-related fatigue and sleep disturbances may share common underlying biochemical mechanisms.
Collapse
Affiliation(s)
- Lianqi Liu
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Paul J. Mills
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA,SDSU/UCSD Joint Doctoral Program in Clinical Psychology, San Diego, CA, USA,Moores UCSD Cancer Center, La Jolla, CA, USA
| | - Michelle Rissling
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA,SDSU/UCSD Joint Doctoral Program in Clinical Psychology, San Diego, CA, USA
| | - Lavinia Fiorentino
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA,Moores UCSD Cancer Center, La Jolla, CA, USA
| | - Loki Natarajan
- Department of Family and Preventive Medicine, University of California, San Diego, La Jolla, CA, USA,Moores UCSD Cancer Center, La Jolla, CA, USA
| | - Joel E. Dimsdale
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA,SDSU/UCSD Joint Doctoral Program in Clinical Psychology, San Diego, CA, USA,Moores UCSD Cancer Center, La Jolla, CA, USA
| | - Georgia Robins Sadler
- Moores UCSD Cancer Center, La Jolla, CA, USA,Department of Surgery, University of California, San Diego, La Jolla, CA, USA
| | - Barbara A. Parker
- Moores UCSD Cancer Center, La Jolla, CA, USA,Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Sonia Ancoli-Israel
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA,SDSU/UCSD Joint Doctoral Program in Clinical Psychology, San Diego, CA, USA,Moores UCSD Cancer Center, La Jolla, CA, USA,Department of Medicine, University of California, San Diego, La Jolla, CA, USA,Corresponding author: Sonia Ancoli-Israel, PhD, Professor of Psychiatry, Department of Psychiatry. University of California, San Diego; 9500 Gilman Drive, # 0733, La Jolla, California 92093-0733, Phone: 858 822-7710, Fax: 858 822-7712,
| |
Collapse
|
36
|
Inflammatory mediators in breast cancer: coordinated expression of TNFα & IL-1β with CCL2 & CCL5 and effects on epithelial-to-mesenchymal transition. BMC Cancer 2011; 11:130. [PMID: 21486440 PMCID: PMC3095565 DOI: 10.1186/1471-2407-11-130] [Citation(s) in RCA: 217] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2010] [Accepted: 04/12/2011] [Indexed: 01/08/2023] Open
Abstract
Background The inflammatory chemokines CCL2 (MCP-1) & CCL5 (RANTES) and the inflammatory cytokines TNFα & IL-1β were shown to contribute to breast cancer development and metastasis. In this study, we wished to determine whether there are associations between these factors along stages of breast cancer progression, and to identify the possible implications of these factors to disease course. Methods The expression of CCL2, CCL5, TNFα and IL-1β was determined by immunohistochemistry in patients diagnosed with: (1) Benign breast disorders (=healthy individuals); (2) Ductal Carcinoma In Situ (DCIS); (3) Invasive Ducal Carcinoma without relapse (IDC-no-relapse); (4) IDC-with-relapse. Based on the results obtained, breast tumor cells were stimulated by the inflammatory cytokines, and epithelial-to-mesenchymal transition (EMT) was determined by flow cytometry, confocal analyses and adhesion, migration and invasion experiments. Results CCL2, CCL5, TNFα and IL-1β were expressed at very low incidence in normal breast epithelial cells, but their incidence was significantly elevated in tumor cells of the three groups of cancer patients. Significant associations were found between CCL2 & CCL5 and TNFα & IL-1β in the tumor cells in DCIS and IDC-no-relapse patients. In the IDC-with-relapse group, the expression of CCL2 & CCL5 was accompanied by further elevated incidence of TNFα & IL-1β expression. These results suggest progression-related roles for TNFα and IL-1β in breast cancer, as indeed indicated by the following: (1) Tumors of the IDC-with-relapse group had significantly higher persistence of TNFα and IL-1β compared to tumors of DCIS or IDC-no-relapse; (2) Continuous stimulation of the tumor cells by TNFα (and to some extent IL-1β) has led to EMT in the tumor cells; (3) Combined analyses with relevant clinical parameters suggested that IL-1β acts jointly with other pro-malignancy factors to promote disease relapse. Conclusions Our findings suggest that the coordinated expression of CCL2 & CCL5 and TNFα & IL-1β may be important for disease course, and that TNFα & IL-1β may promote disease relapse. Further in vitro and in vivo studies are needed for determination of the joint powers of the four factors in breast cancer, as well as analyses of their combined targeting in breast cancer.
Collapse
|
37
|
Zhou W, Guo S, Gonzalez-Perez RR. Leptin pro-angiogenic signature in breast cancer is linked to IL-1 signalling. Br J Cancer 2011; 104:128-37. [PMID: 21139583 PMCID: PMC3039812 DOI: 10.1038/sj.bjc.6606013] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2010] [Revised: 10/15/2010] [Accepted: 10/26/2010] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Leptin and interleukin-1 (IL-1) upregulate vascular endothelial growth factor (VEGF), promote angiogenesis and are related to worse prognosis of breast cancer. However, it is unknown whether leptin regulates IL-1, and whether these effects are related to leptin-induction of VEGF/VEGFR2 in breast cancer. METHODS Several genetic and pharmacological approaches were used to determine the mechanisms involved in leptin regulation of IL-1 system (IL-1α, IL-1β, IL-1Ra and IL-1R tI) and the impact of IL-1 signalling on leptin-induced VEGF/VEGFR2 expression in mouse mammary cancer 4T1 cells (a model that resembles invasive and highly metastatic human breast cancer). RESULTS Leptin increased protein and mRNA levels of all components of the IL-1 system. IL-1 upregulation involved leptin activation of JAK2/STAT3, MAPK/ERK 1/2, PI-3K/AKT1, PKC, p38 and JNK. Leptin-induced phosphorylation of mTOR/4E-BP1 increased IL-1β and IL-1Ra expression, but downregulated IL-1α. Leptin upregulation of IL-1α promoter was linked to SP1 and NF-κB transcription factors. In addition, leptin receptor (Ob-Rb) was upregulated by leptin. Interestingly, leptin upregulation of VEGF/VEGFR2 was partially mediated by IL-1/IL-1R tI signalling. CONCLUSIONS We show for the first time that leptin induces several signalling pathways to upregulate the translational and transcriptional expression of IL-1 system in breast cancer cells. Moreover, leptin upregulation of VEGF/VEGFR2 was impaired by IL-1 signalling blockade. These data suggest that leptin pro-angiogenic signature in breast cancer is linked to, or regulated, in part by IL-1 signalling.
Collapse
Affiliation(s)
- W Zhou
- Department of Microbiology, Biochemistry & Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA
- Clinic Medicine & Pharmacy College of China Medical University, Shenyang City, Liaoning Province 110002, People's Republic of China
| | - S Guo
- Department of Microbiology, Biochemistry & Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - R R Gonzalez-Perez
- Department of Microbiology, Biochemistry & Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| |
Collapse
|
38
|
Nicolini A, Carpi A. Immune manipulation of advanced breast cancer: an interpretative model of the relationship between immune system and tumor cell biology. Med Res Rev 2009; 29:436-71. [PMID: 19105214 DOI: 10.1002/med.20143] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
This review summarizes some recent clinical immunological approaches with cytokines and/or antibodies for therapy of advanced breast cancer. It considers the recent advances in genetics and molecular tumor biology related to impaired immunosurveillance involving cytokines and growth factors to explain clinical results. Evasion of the host immune attack might be induced by the following groups of mechanisms: (a) tumor dependent (genomic instability, HLA class I antigen abnormalities, upregulation of fetal type nonclassical HLA class I molecules, epitope immunodominance, apoptosis inhibition by defective death receptor signaling, apoptosis of activated T cells, tumor cannibalism and constitutive activation of signal transducer, and activator of transcription-3 (Stat 3) and nuclear factor-kappaB (NF-kappaB) signaling); (b) host dependent (CD4+CD25+ regulatory T cells (T reg), CD4+ T cells anergy, Th2 antitumor immunity diversion and myeloid suppressor cells); (c) tumor and host dependent (lack of co-stimulation molecules, immunosuppressive cytokines (vascular endothelial growth factor (VEGF), interleukin (IL)-10, prostaglandin (PG)E2, transforming growth factor (TGF)-beta)). Cytokines and growth factors are involved in virtually all three types of mechanisms. These mechanisms are integrated with the current knowledge of tumor growth and inhibited apoptosis primarily mediated by cytokines and growth factors to propose an interpretation of the relationships among tumor cells, tumor stroma, and tumor-infiltrating lymphocytes. Tumor growth, defective immunorecognition and immunosuppression are the three principal effects considered responsible for immune evasion.
Collapse
Affiliation(s)
- Andrea Nicolini
- Department of Internal Medicine, University of Pisa, Pisa, Italy.
| | | |
Collapse
|
39
|
Péant B, Diallo JS, Dufour F, Le Page C, Delvoye N, Saad F, Mes-Masson AM. Over-expression of IkappaB-kinase-epsilon (IKKepsilon/IKKi) induces secretion of inflammatory cytokines in prostate cancer cell lines. Prostate 2009; 69:706-18. [PMID: 19170126 DOI: 10.1002/pros.20912] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
BACKGROUND Elevated inflammatory cytokine levels in serum have been associated with advanced stage metastasis-related morbidity in prostate cancer. Several studies have shown that IL-6 and IL-8 can accelerate the growth of human prostate cancer cell lines. Previous studies, in murine embryonic fibroblasts, have shown that Ikappa-B kinase-epsilon (IKKepsilon/IKKi)-deficiency results in the reduction of lipopolysaccharide-mediated expression of IL-6. RESULTS In this study, we report that over-expression of IKKepsilon in hormone-sensitive 22Rv1 and LNCaP prostate cancer cells induces the secretion of several inflammatory cytokines including IL-6 and IL-8. Both of these cytokines are secreted by hormone-refractory PC-3 prostate cancer cells and IKKepsilon knock-down in these cells correlates with a strong decrease in IL-6 secretion. Furthermore, we demonstrate that IKKepsilon over-expression does not induce the activation of the IKKepsilon classical targets NF-kappaB and IRF-3, two transcription factors involved in the regulation of several cytokines. Finally, we observe that high IKKepsilon expression results in its nuclear translocation, a phenomena that is TBK1-independent. CONCLUSIONS This study identifies IKKepsilon as a potential prostate cancer gene that may favor chronic inflammation and create a tumor-supporting microenvironment that promotes prostate cancer progression, particularly by the induction of IL-6 secretion that may act as a positive growth factor in prostate cancer.
Collapse
Affiliation(s)
- Benjamin Péant
- Centre de recherche du Centre hospitalier de l'Université de Montréal/Institut du cancer de Montréal, Québec, Canada
| | | | | | | | | | | | | |
Collapse
|
40
|
Grimm C, Kantelhardt E, Heinze G, Polterauer S, Zeillinger R, Kölbl H, Reinthaller A, Hefler L. The prognostic value of four interleukin-1 gene polymorphisms in Caucasian women with breast cancer: a multicenter study. BMC Cancer 2009; 9:78. [PMID: 19267917 PMCID: PMC2656545 DOI: 10.1186/1471-2407-9-78] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2008] [Accepted: 03/06/2009] [Indexed: 11/23/2022] Open
Abstract
Background The proinflammatory cytokine interleukin-1 (IL-1) is known to play an important role in the carcinogenesis of breast cancer. Although IL-1 gene polymorphisms were reported to be associated with increased risk of breast cancer, their influence on survival of Caucasian breast cancer patients remains to be shown. Methods We studied the influence of four common gene polymorphisms (IL1A -889C/T, IL1B -511C/T, IL1B +3953E1/E2, and IL1RN long/2) of the IL-1 family on survival in 262 Caucasian patients with breast cancer by univariate and multivariate survival analysis. The combined effect of the four gene polymorphisms on overall survival was studied by haplotype analysis. Results In the present study 38 cases of cancer related death and a median time of follow-up (range) of 55.3 (0.4–175.8) months was observed. IL1RN 2/2 (homozygous mutant) gene polymorphism was associated with shortened disease free and overall survival in a univariate (p = 0.001 and p = 0.01, respectively) and multivariate analysis (p = 0.002, Odds Ratio [95% Confidence Interval] = 3.6 [1.6–8.0] and p = 0.05, Odds Ratio = 3.0 [1.1–9.3], respectively). Presence of the homozygous mutant genotype of the IL1A -889 and IL1B +3953 gene polymorphism was associated with overall survival in the univariate (p = 0.004 and p = 0.002, respectively), but not in the multivariate analysis. No association was observed between all possible haplotype combinations and overall survival. Conclusion Carriage of the mutant alleles of IL1RN was independently associated with shortened disease free and overall survival rates in Caucasian patients with breast cancer.
Collapse
Affiliation(s)
- Christoph Grimm
- Department of Obstetrics and Gynecology, Medical University of Vienna, Vienna, Austria.
| | | | | | | | | | | | | | | |
Collapse
|
41
|
IL-1 family in breast cancer: potential interplay with leptin and other adipocytokines. FEBS Lett 2008; 583:259-65. [PMID: 19111549 DOI: 10.1016/j.febslet.2008.12.030] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2008] [Revised: 11/17/2008] [Accepted: 12/11/2008] [Indexed: 12/15/2022]
Abstract
Obesity is associated with an increased risk of breast cancer. interleukin-1 (IL-1), a pro-inflammatory cytokine secreted by adipose tissue, is involved in breast cancer development. There is also convincing evidence that other adipocytokines including leptin not only have a role in haematopoiesis, reproduction and immunity but are also growth factors in cancer. Therefore, IL-1 family and leptin family are adipocytokines which could represent a major link between obesity and breast cancer progression. This minireview provides insight into recent findings on the prognostic significance of IL-1 and leptin in mammary tumours, and discusses the potential interplay between IL-1 family members and adipocyte-derived hormones in breast cancer.
Collapse
|
42
|
Lavigne JA, Takahashi Y, Chandramouli GVR, Liu H, Perkins SN, Hursting SD, Wang TTY. Concentration-dependent effects of genistein on global gene expression in MCF-7 breast cancer cells: an oligo microarray study. Breast Cancer Res Treat 2007; 110:85-98. [PMID: 17687646 DOI: 10.1007/s10549-007-9705-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2007] [Accepted: 07/17/2007] [Indexed: 01/11/2023]
Abstract
Breast cancer is the most commonly diagnosed cancer among US women; there is therefore great interest in developing preventive and treatment strategies for this disease. Because breast cancer incidence is much lower in countries where women consume high levels of soy, bioactive compounds in this food source have been studied for their effects on breast cancer. Genistein, found at high levels in soybeans and soy foods, is a controversial candidate breast cancer preventive phytochemical whose effects on breast cells are complex. To understand more clearly the molecular mechanisms underlying the effects of genistein on breast cancer cells, we used a DNA oligo microarray approach to examine the global gene expression patterns in MCF-7 breast cancer cells at both physiologic (1 or 5 microM) and pharmacologic (25 microM) genistein concentrations. Microarray analyses were performed on MCF-7 cells after 48 h of either vehicle or 1, 5, or 25 microM genistein treatment. We found that genistein altered the expression of genes belonging to a wide range of pathways, including estrogen- and p53-mediated pathways. At 1 and 5 microM, genistein elicited an expression pattern suggestive of increased mitogenic activity, confirming the proliferative response to genistein observed in cultured MCF-7 cells, while at 25 microM genistein effected a pattern that likely contributes to increased apoptosis, decreased proliferation and decreased total cell number, also consistent with cell culture results. These findings provide evidence for a molecular signature of genistein's effects in MCF-7 cells and lay the foundation for elucidating the mechanisms of genistein's biological impact in breast cancer cells.
Collapse
Affiliation(s)
- Jackie A Lavigne
- Division of Cancer Prevention, National Cancer Institute, NIH, Bethesda, MD, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Roy D, Cai Q, Felty Q, Narayan S. Estrogen-induced generation of reactive oxygen and nitrogen species, gene damage, and estrogen-dependent cancers. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2007; 10:235-57. [PMID: 17620201 DOI: 10.1080/15287390600974924] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
In addition to the direct effect of estrogen on mitochondria and the redox cycling of catechol estrogen, estrogen-induced proinflammatory cytokines, such as interleukin-1 beta (IL-1beta) and tumor necrosis factor alpha (TNF-alpha), also generate reactive oxygen and nitrogen species (RO/NS). Different cellular signaling pathways may operate in response to varying levels of estrogen-induced RO/NS, leading to genotoxic damage, cell apoptosis, or cell growth. At high levels of RO/NS, cells receiving genotoxic insults, if not repaired, may engage the apoptotic pathways. There is increasing evidence supporting that estrogen-induced alterations in the genome of cells is produced by oxidative attack. Furthermore, ROS generated by estrogen exposure and/or active metabolites of estrogen in combination with receptor-mediated proliferation of genetically damaged cells may be involved in tumor development. This view is supported by the findings of DNA modifications produced in vitro or in vivo by natural and synthetic estrogens in the target organs of cancer both in experimental models and in humans. Interaction of estrogen-induced oxidants and estrogen metabolites with DNA was shown to generate mutations in genes. Cotreatment with an inhibitor of IL-1beta and TNF-alpha synthesis, pentoxifylline, decreased stilbene estrogen-induced levels of myeloperoxidase (MPO), 8-hydroxydeoxyguanosine formation, and gene mutations, and prevented stilbene estrogen-induced lesions. Stable MCF-7 clones overexpressing IL-1beta resulted in a high level of IL-1beta peptide secretion undergoing cell apoptosis, and an elevated level of p53 protein in response to high oxidative stress when compared to nontransfected cells, whereas MCF-7 clones overexpressing IL-1beta that resulted in a moderate level of IL-1beta secretion stimulated the clonal expansion of MCF-7 and TM3 cells. Estrogen-induced MCF-7 cell growth and cyclin D1 expression were suppressed by antioxidants and mitochondrial blockers. These studies support that in addition to ovarian estrogen-mediated ER signaling, mitogenic signals may also come from estrogen-induced RO/NS. Further validation of this concept that the concentration of the RO/NS within the cellular microenvironment determines its stimulatory or inhibitory growth signals as well as its genotoxic effects regulating the growth of estrogen-dependent tumors may result in novel preventive strategies.
Collapse
Affiliation(s)
- Deodutta Roy
- Department of Environmental and Occupational Health, Florida International University, Miami, Florida 33199, USA.
| | | | | | | |
Collapse
|
44
|
Chavey C, Bibeau F, Gourgou-Bourgade S, Burlinchon S, Boissière F, Laune D, Roques S, Lazennec G. Oestrogen receptor negative breast cancers exhibit high cytokine content. Breast Cancer Res 2007; 9:R15. [PMID: 17261184 PMCID: PMC1851386 DOI: 10.1186/bcr1648] [Citation(s) in RCA: 262] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2006] [Revised: 12/29/2006] [Accepted: 01/29/2007] [Indexed: 11/11/2022] Open
Abstract
Introduction An emerging hypothesis suggests that cytokines could play an important role in cancer as potential modulators of angiogenesis and leucocyte infiltration. Methods A novel multiplexed flow cytometry technology was used to measure the expression of 17 cytokines (IL-1β, IL-2, IL-4, IL-5, IL-6, IL-7, IL-8, IL-10, IL-12 [p70], IL-13, IL-17, granulocyte colony-stimulating factor [CSF], granulocyte-macrophage CSF, IFN-γ, monocyte chemoattractant protein [MCP]-1, macrophage inflammatory protein [MIP]-1β, tumour necrosis factor [TNF]-α) at the protein level in 105 breast carcinoma. B lymphocyte, T lymphocyte and macrophage levels were determined by immunohistochemistry. Results Fourteen of the 17 cytokines were expressed in breast carcinoma, whereas only nine cytokines could be detected in normal breast. Most cytokines were more abundant in breast carcinoma than in normal breast, with IL-6, IL-8, granulocyte CSF, IFN-γ, MCP-1 and MIP-1β being very abundant. IL-2, IL-6, IL-8, IL-10, IFN-γ, MCP-1, MIP-1β and TNF-α, and to a lesser extent IL-1β and IL-13 exhibited levels of expression that were inversely correlated to oestrogen receptor and progesterone receptor status. Most cytokines were not correlated with age at cancer diagnosis, tumour size, histological type, or lymph node status. However, IL-1β, IL-6, IL-8, IL-10, IL-12, MCP-1 and MIP-1β were more abundant in high-grade tumours than in low-grade tumours. In addition, IL-8 and MIP-1β were expressed to a greater degree in HER2-positive than in HER2-negative patients. The expression of most of the studied cytokines was correlated to levels of activator protein-1, which is known to regulate numerous cytokines. Overexpression of MCP-1 and MIP-1β were linked to B lymphocyte, T lymphocyte and macrophage infiltration, whereas high levels of IL-8 were correlated with high macrophage content in tumour. Moreover, IL-8 positive tumours exhibited increased vascularization. Conclusion We found that multiple cytokines were overexpressed in oestrogen receptor negative breast carcinoma, and that the three major cytokines – MCP-1, MIP-1β and IL-8 – were correlated with inflammatory cell component, which could account for the aggressiveness of these tumours.
Collapse
Affiliation(s)
- Carine Chavey
- INSERM, U844, Site Saint Eloi, Bâtiment INM, University of Montpellier I, rue Augustin Fliche, Montpellier, F-34091, France; Montpellier, F-34090, France
| | - Frédéric Bibeau
- CRLC Val d'Aurelle, Pathology Department, rue des Apothicaires, Montpellier, F-34298, France
| | | | - Sandrine Burlinchon
- INSERM, U844, Site Saint Eloi, Bâtiment INM, University of Montpellier I, rue Augustin Fliche, Montpellier, F-34091, France; Montpellier, F-34090, France
| | - Florence Boissière
- CRLC Val d'Aurelle, Pathology Department, rue des Apothicaires, Montpellier, F-34298, France
| | - Daniel Laune
- CNRS, UMR 5160, Centre de Pharmacologie et Biotechnologie pour la Sante, Faculte de Pharmacie, avenue Charles Flahault, Montpellier, F-34093, France
| | - Sylvie Roques
- CRLC Val d'Aurelle, Pathology Department, rue des Apothicaires, Montpellier, F-34298, France
| | - Gwendal Lazennec
- INSERM, U844, Site Saint Eloi, Bâtiment INM, University of Montpellier I, rue Augustin Fliche, Montpellier, F-34091, France; Montpellier, F-34090, France
| |
Collapse
|
45
|
Apte RN, Dotan S, Elkabets M, White MR, Reich E, Carmi Y, Song X, Dvozkin T, Krelin Y, Voronov E. The involvement of IL-1 in tumorigenesis, tumor invasiveness, metastasis and tumor-host interactions. Cancer Metastasis Rev 2007; 25:387-408. [PMID: 17043764 DOI: 10.1007/s10555-006-9004-4] [Citation(s) in RCA: 447] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Interleukin-1 (IL-1) includes a family of closely related genes; the two major agonistic proteins, IL-1alpha and IL-1beta, are pleiotropic and affect mainly inflammation, immunity and hemopoiesis. The IL-1Ra antagonist is a physiological inhibitor of pre-formed IL-1. Recombinant IL-1alpha and IL-1beta bind to the same receptors and induce the same biological functions. As such, the IL-1 molecules have been considered identical in normal homeostasis and in disease. However, the IL-1 molecules differ in their compartmentalization within the producing cell or the microenvironment. Thus, IL-1beta is solely active in its secreted form, whereas IL-1alpha is mainly active in cell-associated forms (intracellular precursor and membrane-bound IL-1alpha) and only rarely as a secreted cytokine, as it is secreted only in a limited manner. IL-1 is abundant at tumor sites, where it may affect the process of carcinogenesis, tumor growth and invasiveness and also the patterns of tumor-host interactions. Here, we review the effects of microenvironment- and tumor cell-derived IL-1 on malignant processes in experimental tumor models and in cancer patients. We propose that membrane-associated IL-1alpha expressed on malignant cells stimulates anti-tumor immunity, while secretable IL-1beta, derived from the microenvironment or the malignant cells, activates inflammation that promotes invasiveness and also induces tumor-mediated suppression. Inhibition of the function of IL-1 by the IL-1Ra, reduces tumor invasiveness and alleviates tumor-mediated suppression, pointing to its feasibility in cancer therapy. Differential manipulation of IL-1alpha and IL-1beta in malignant cells or in the tumor's microenvironment can open new avenues for using IL-1 in cancer therapy.
Collapse
Affiliation(s)
- Ron N Apte
- Department of Microbiology and Immunology, Faculty of Health Sciences and The Cancer Research Center, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
In recent decades many advances have occurred in the understanding of the role of cytokines in breast cancer. New signalling pathways of interleukin (IL)-1 family, IL-6, IL-11, IL-18, interferons (IFNs) and interferon regulatory factors 1 (IRF-1) and 2 (IRF-2) have been found within tumour microenvironments and in metastatic sites. Some cytokines (IL-1, IL-6, IL-11, TGFbeta) stimulate while others (IL-12, IL-18, IFNs) inhibit breast cancer proliferation and/or invasion. Similarly, high circulating levels of some cytokines seem to be favourable (soluble IL-2R) while others are unfavourable (IL-1beta, IL-6, IL-8, IL-10, IL-18, gp130) prognostic indicators. So far IL-2, IFNalpha, IFNbeta and occasionally IFNgamma, IL-6, IL-12 have been the cytokines used for anti tumour treatment of advanced breast cancer either to induce or increase hormone sensitivity and/or to stimulate cellular immunity. Disappointing results occurred in most trials; however, two long-term pilot studies suggest that IL-2 and IFNbeta, when used appropriately can have a positive effect on clinical benefit and overall survival of patients with minimal residual disease after chemotherapy or with disseminated disease controlled by conventional endocrine therapy.
Collapse
Affiliation(s)
- A Nicolini
- Department of Internal Medicine, University of Pisa, Via Roma 67, 56126 Pisa, Italy.
| | | | | |
Collapse
|
47
|
Balasubramanian SP, Azmy IAF, Higham SE, Wilson AG, Cross SS, Cox A, Brown NJ, Reed MW. Interleukin gene polymorphisms and breast cancer: a case control study and systematic literature review. BMC Cancer 2006; 6:188. [PMID: 16842617 PMCID: PMC1553474 DOI: 10.1186/1471-2407-6-188] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2006] [Accepted: 07/14/2006] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Interleukins and cytokines play an important role in the pathogenesis of many solid cancers. Several single nucleotide polymorphisms (SNPs) identified in cytokine genes are thought to influence the expression or function of these proteins and many have been evaluated for their role in inflammatory disease and cancer predisposition. The aim of this study was to evaluate any role of specific SNPs in the interleukin genes IL1A, IL1B, IL1RN, IL4R, IL6 and IL10 in predisposition to breast cancer susceptibility and severity. METHODS Candidate single nucleotide polymorphisms (SNPs) in key cytokine genes were genotyped in breast cancer patients and in appropriate healthy volunteers who were similar in age, race and sex. Genotyping was performed using a high throughput allelic discrimination method. Data on clinico-pathological details and survival were collected. A systematic review of Medline English literature was done to retrieve previous studies of these polymorphisms in breast cancer. RESULTS None of the polymorphisms studied showed any overall predisposition to breast cancer susceptibility, severity or to time to death or occurrence of distant metastases. The results of the systematic review are summarised. CONCLUSION Polymorphisms within key interleukin genes (IL1A, IL1B, IL1RN, IL4R, IL6 and IL10 do not appear to play a significant overall role in breast cancer susceptibility or severity.
Collapse
Affiliation(s)
| | - IAF Azmy
- Academic Surgical Oncology Unit, University of Sheffield, Sheffield, UK
| | - SE Higham
- Academic Surgical Oncology Unit, University of Sheffield, Sheffield, UK
| | - AG Wilson
- Academic Rheumatology Unit, University of Sheffield, Sheffield, UK
| | - SS Cross
- Academic Unit of Pathology, University of Sheffield, Sheffield, UK
| | - A Cox
- Institute of Cancer Studies, University of Sheffield, Sheffield, UK
| | - NJ Brown
- Academic Surgical Oncology Unit, University of Sheffield, Sheffield, UK
| | - MW Reed
- Academic Surgical Oncology Unit, University of Sheffield, Sheffield, UK
| |
Collapse
|
48
|
Hefler LA, Grimm C, Lantzsch T, Lampe D, Leodolter S, Koelbl H, Heinze G, Reinthaller A, Tong-Cacsire D, Tempfer C, Zeillinger R. Interleukin-1 and interleukin-6 gene polymorphisms and the risk of breast cancer in caucasian women. Clin Cancer Res 2005; 11:5718-21. [PMID: 16115908 DOI: 10.1158/1078-0432.ccr-05-0001] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Genetic polymorphisms of cytokine-encoding genes are known to predispose to malignant disease. Interleukin (IL)-1 and IL-6 are crucially involved in breast carcinogenesis. Whether polymorphisms of the genes encoding IL-1 (IL1) and IL-6 (IL6) also influence breast cancer risk is unknown. EXPERIMENTAL DESIGN In the present case-control study, we ascertained three polymorphisms of the IL1 gene cluster [-889 C/T polymorphism of the IL1alpha gene (IL1A), -511 C/T polymorphism of the IL1beta promoter (IL1B promoter), a polymorphism of IL1beta exon 5 (IL1B exon 5)], an 86-bp repeat in intron 2 of the IL1 receptor antagonist gene (IL1RN), and the -174 G/C polymorphism of the IL6 gene (IL6) in 269 patients with breast cancer and 227 healthy controls using PCR and pyrosequencing. RESULTS Polymorphisms within the IL1 gene cluster and the respective haplotypes were not associated with the presence and the phenotype of breast cancer. The IL6 polymorphism was significantly associated with breast cancer. Odds ratios for women with one or two high-risk alleles versus women homozygous for the low-risk allele were 1.5 (95% confidence interval, 1.04-2.3; P = 0.04) and 2.0 (95% confidence interval, 1.1-3.6; P = 0.02), respectively. No association was ascertained between presence of the IL6 polymorphism and various clinicopathologic variables. CONCLUSIONS Although polymorphisms within the IL1 gene cluster do not seem to influence breast cancer risk or phenotype, presence of the -174C IL6 allele increases the risk of breast cancer in Caucasian women in a dose-dependent fashion.
Collapse
Affiliation(s)
- Lukas A Hefler
- Department of Obstetrics and Gynecology, Medical University of Vienna, Austria.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Lee KM, Park SK, Hamajima N, Tajima K, Choi JY, Noh DY, Ahn SH, Yoo KY, Hirvonen A, Kang D. Genetic polymorphisms of interleukin-1 beta (IL-1B) and IL-1 receptor antagonist (IL-1RN) and breast cancer risk in Korean women. Breast Cancer Res Treat 2005; 96:197-202. [PMID: 16319982 DOI: 10.1007/s10549-005-9079-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2005] [Accepted: 09/12/2005] [Indexed: 11/30/2022]
Abstract
OBJECTIVE To evaluate the potential role of genetic polymorphisms of interleukin-1 beta (IL-1B) and IL-1 receptor antagonist (IL-1RN) on breast cancer development, a hospital-based case-control study was conducted in Korea. METHODS Histologically confirmed breast cancer cases (n = 560) and controls (n = 509) without cancer history were recruited from three teaching hospitals in Seoul between September 1998 and January 2002. Information on risk factors of breast cancer were collected by interviewed questionnaire. Genotypes of IL-1B (-31C/T) and IL-1RN (86 bp variable number tandom repeats in intron 2) were determined by PCR-CTPP (confronting two-pair primers) and PCR, respectively. Adjusted odds ratios (ORs) and 95% confidence intervals (CIs) were estimated by unconditional logistic regression model. RESULTS The IL-1RN *2-allele was associated with decreased breast cancer risk with marginal significance (OR = 0.7, 95% CI = 0.48-1.05). The IL-1B CC or TC genotype was not associated with decreased risk of breast cancer (OR = 0.9, 95% CI = 0.65-1.16). However, combination of IL-1B C-allele (CT or CC) and IL-1RN *2-allele containing genotypes significantly decreased the risk of breast cancer (OR = 0.6, 95% CI = 0.39-0.99). A moderately decreasing trend of risk was observed as the number of 'putative low risk' allele increased (p for trend = 0.07). Suggestive combined effect on breast cancer risk was also observed between body mass index (BMI) and IL-1RN non-*2 allele: women with higher BMI and IL-1RN non-*2 allele had 1.7-fold higher risk than women with lower BMI and IL-1RN*2 genotypes. CONCLUSION Our results suggest that genetic polymorphisms of interleukin-1 may play a role in the individual susceptibility for breast cancer development in Korean women.
Collapse
Affiliation(s)
- Kyoung-Mu Lee
- Cancer Research Institute, Institute of Environmental Medicine, SNUMRC, and Department of Preventive Medicine, Seoul National University College of Medicine, 28 Yongon-Dong, Chongno-Gu, Seoul, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Bulun SE, Lin Z, Imir G, Amin S, Demura M, Yilmaz B, Martin R, Utsunomiya H, Thung S, Gurates B, Tamura M, Langoi D, Deb S. Regulation of aromatase expression in estrogen-responsive breast and uterine disease: from bench to treatment. Pharmacol Rev 2005; 57:359-83. [PMID: 16109840 DOI: 10.1124/pr.57.3.6] [Citation(s) in RCA: 403] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
A single gene encodes the key enzyme for estrogen biosynthesis termed aromatase, inhibition of which effectively eliminates estrogen production. Aromatase inhibitors successfully treat breast cancer and endometriosis, whereas their roles in endometrial cancer, uterine fibroids, and aromatase excess syndrome are less clear. Ovary, testis, adipose tissue, skin, hypothalamus, and placenta express aromatase normally, whereas breast and endometrial cancers, endometriosis, and uterine fibroids overexpress aromatase and produce local estrogen that exerts paracrine and intracrine effects. Tissue-specific promoters distributed over a 93-kilobase regulatory region upstream of a common coding region alternatively control aromatase expression. A distinct set of transcription factors regulates each promoter in a signaling pathway- and tissue-specific manner. Three mechanisms are responsible for aromatase overexpression in a pathologic tissue versus its normal counterpart. First, cellular composition is altered to increase aromatase-expressing cell types that use distinct promoters (breast cancer). Second, molecular alterations in stromal cells favor binding of transcriptional enhancers versus inhibitors to a normally quiescent aromatase promoter and initiate transcription (breast/endometrial cancer, endometriosis, and uterine fibroids). Third, heterozygous mutations, which cause the aromatase coding region to lie adjacent to constitutively active cryptic promoters that normally transcribe other genes, result in excessive estrogen formation owing to the overexpression of aromatase in many tissues.
Collapse
Affiliation(s)
- Serdar E Bulun
- Division of Reproductive Biology Research, Department of Obstetric and Gynecology, Northwestern University, Chicago, IL 60611, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|