1
|
Song WH, Lim YS, Kim JE, Kang HY, Lee C, Rajbongshi L, Hwang SY, Oh SO, Kim BS, Lee D, Song YJ, Yoon S. A Marine Collagen-Based 3D Scaffold for In Vitro Modeling of Human Prostate Cancer Niche and Anti-Cancer Therapeutic Discovery. Mar Drugs 2024; 22:295. [PMID: 39057404 PMCID: PMC11277582 DOI: 10.3390/md22070295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 06/25/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024] Open
Abstract
Recently, the need to develop a robust three-dimensional (3D) cell culture system that serves as a valuable in vitro tumor model has been emphasized. This system should closely mimic the tumor growth behaviors observed in vivo and replicate the key elements and characteristics of human tumors for the effective discovery and development of anti-tumor therapeutics. Therefore, in this study, we developed an effective 3D in vitro model of human prostate cancer (PC) using a marine collagen-based biomimetic 3D scaffold. The model displayed distinctive molecular profiles and cellular properties compared with those of the 2D PC cell culture. This was evidenced by (1) increased cell proliferation, migration, invasion, colony formation, and chemoresistance; (2) upregulated expression of crucial multidrug-resistance- and cancer-stemness-related genes; (3) heightened expression of key molecules associated with malignant progressions, such as epithelial-mesenchymal transition transcription factors, Notch, matrix metalloproteinases, and pluripotency biomarkers; (4) robust enrichment of prostate cancer stem cells (CSCs); and (5) enhanced expression of integrins. These results suggest that our 3D in vitro PC model has the potential to serve as a research platform for studying PC and prostate CSC biology, as well as for screening novel therapies targeting PC and prostate CSCs.
Collapse
Affiliation(s)
- Won Hoon Song
- Department of Urology, Pusan National University Yangsan Hospital and Pusan National University College of Medicine, Yangsan 626-870, Republic of Korea;
| | - Ye Seon Lim
- Department of Anatomy and Convergence Medical Sciences, Pusan National University College of Medicine, Yangsan 626-870, Republic of Korea; (Y.S.L.); (J.-E.K.); (H.Y.K.); (C.L.); (L.R.); (S.Y.H.); (S.-O.O.)
- Immune Reconstitution Research Center of Medical Research Institute, Pusan National University College of Medicine, Yangsan 626-870, Republic of Korea;
| | - Ji-Eun Kim
- Department of Anatomy and Convergence Medical Sciences, Pusan National University College of Medicine, Yangsan 626-870, Republic of Korea; (Y.S.L.); (J.-E.K.); (H.Y.K.); (C.L.); (L.R.); (S.Y.H.); (S.-O.O.)
- Immune Reconstitution Research Center of Medical Research Institute, Pusan National University College of Medicine, Yangsan 626-870, Republic of Korea;
| | - Hae Yeong Kang
- Department of Anatomy and Convergence Medical Sciences, Pusan National University College of Medicine, Yangsan 626-870, Republic of Korea; (Y.S.L.); (J.-E.K.); (H.Y.K.); (C.L.); (L.R.); (S.Y.H.); (S.-O.O.)
| | - Changyong Lee
- Department of Anatomy and Convergence Medical Sciences, Pusan National University College of Medicine, Yangsan 626-870, Republic of Korea; (Y.S.L.); (J.-E.K.); (H.Y.K.); (C.L.); (L.R.); (S.Y.H.); (S.-O.O.)
- Immune Reconstitution Research Center of Medical Research Institute, Pusan National University College of Medicine, Yangsan 626-870, Republic of Korea;
| | - Lata Rajbongshi
- Department of Anatomy and Convergence Medical Sciences, Pusan National University College of Medicine, Yangsan 626-870, Republic of Korea; (Y.S.L.); (J.-E.K.); (H.Y.K.); (C.L.); (L.R.); (S.Y.H.); (S.-O.O.)
- Immune Reconstitution Research Center of Medical Research Institute, Pusan National University College of Medicine, Yangsan 626-870, Republic of Korea;
| | - Seon Yeong Hwang
- Department of Anatomy and Convergence Medical Sciences, Pusan National University College of Medicine, Yangsan 626-870, Republic of Korea; (Y.S.L.); (J.-E.K.); (H.Y.K.); (C.L.); (L.R.); (S.Y.H.); (S.-O.O.)
| | - Sae-Ock Oh
- Department of Anatomy and Convergence Medical Sciences, Pusan National University College of Medicine, Yangsan 626-870, Republic of Korea; (Y.S.L.); (J.-E.K.); (H.Y.K.); (C.L.); (L.R.); (S.Y.H.); (S.-O.O.)
| | - Byoung Soo Kim
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Republic of Korea;
| | - Dongjun Lee
- Department of Convergence Medicine, Pusan National University College of Medicine, Yangsan 50612, Republic of Korea;
| | - Yong Jung Song
- Immune Reconstitution Research Center of Medical Research Institute, Pusan National University College of Medicine, Yangsan 626-870, Republic of Korea;
- Department of Obstetrics and Gynecology, Pusan National University Yangsan Hospital and Pusan National University College of Medicine, Yangsan 626-870, Republic of Korea
| | - Sik Yoon
- Department of Anatomy and Convergence Medical Sciences, Pusan National University College of Medicine, Yangsan 626-870, Republic of Korea; (Y.S.L.); (J.-E.K.); (H.Y.K.); (C.L.); (L.R.); (S.Y.H.); (S.-O.O.)
- Immune Reconstitution Research Center of Medical Research Institute, Pusan National University College of Medicine, Yangsan 626-870, Republic of Korea;
| |
Collapse
|
2
|
Barb AC, Fenesan MP, Pirtea M, Margan MM, Tomescu L, Ceban E, Cimpean AM, Melnic E. Reassessing Breast Cancer-Associated Fibroblasts (CAFs) Interactions with Other Stromal Components and Clinico-Pathologic Parameters by Using Immunohistochemistry and Digital Image Analysis (DIA). Cancers (Basel) 2023; 15:3823. [PMID: 37568639 PMCID: PMC10417678 DOI: 10.3390/cancers15153823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/18/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
BACKGROUND Breast cancer (BC) stroma has CD34- and αSMA-positive cancer-associated fibroblasts (CAFs) differently distributed. During malignant transformation, CD34-positive fibroblasts decrease while αSMA-positive CAFs increase. The prevalence of αSMA-positive CAFs in BC stroma makes microscopic examination difficult without digital image analysis processing (DIA). DIA was used to compare CD34- and αSMA-positive CAFs among breast cancer molecular subgroups. DIA-derived data were linked to age, survival, tumor stroma vessels, tertiary lymphoid structures (TLS), invasion, and recurrence. METHODS Double immunostaining for CD34 and αSMA showed different CAF distribution patterns in normal and BC tissues. Single CD34 immunohistochemistry on supplemental slides quantified tumor stroma CD34_CAFs. Digital image analysis (DIA) data on CAF density, intensity, stromal score, and H-score were correlated with clinico-pathologic factors. RESULTS CD34/αSMA CAF proportion was significantly related to age in Luminal A (LA), Luminal B (LB), and HER2 subtypes. CD34_CAF influence on survival, invasion, and recurrence of LA, LB-HER2, and TNBC subtypes was found to be significant. The CD34/αSMA-expressing CAFs exhibited a heterogeneous impact on stromal vasculature and TLS. CONCLUSION BC stromal CD34_CAFs/αSMA_CAFs have an impact on survival, invasion, and recurrence differently between BC molecular subtypes. The tumor stroma DIA assessment may have predictive potential to prognosis and long-term follow-up of patients with breast cancer.
Collapse
Affiliation(s)
- Alina Cristina Barb
- Department of Microscopic Morphology/Histology, Victor Babes University of Medicine and Pharmacy, 300041 Timisoara, Romania; (A.C.B.); (M.P.F.); (M.P.)
- Doctoral School in Medicine, Victor Babes University of Medicine and Pharmacy, 300041 Timisoara, Romania;
- Department of Clinical Oncology, OncoHelp Hospital, 300239 Timisoara, Romania
| | - Mihaela Pasca Fenesan
- Department of Microscopic Morphology/Histology, Victor Babes University of Medicine and Pharmacy, 300041 Timisoara, Romania; (A.C.B.); (M.P.F.); (M.P.)
- Doctoral School in Medicine, Victor Babes University of Medicine and Pharmacy, 300041 Timisoara, Romania;
- Department of Clinical Oncology, OncoHelp Hospital, 300239 Timisoara, Romania
| | - Marilena Pirtea
- Department of Microscopic Morphology/Histology, Victor Babes University of Medicine and Pharmacy, 300041 Timisoara, Romania; (A.C.B.); (M.P.F.); (M.P.)
| | - Mădălin-Marius Margan
- Department of Functional Sciences/Discipline of Public Health, Victor Babes University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| | - Larisa Tomescu
- Doctoral School in Medicine, Victor Babes University of Medicine and Pharmacy, 300041 Timisoara, Romania;
- Department of Obstetrics and Gynecology, “Victor Babes” University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania
| | - Emil Ceban
- Department of Urology and Surgical Nephrology, Nicolae Testemitanu State University of Medicine and Pharmacy, 2004 Chisinau, Moldova;
- Laboratory of Andrology, Functional Urology and Sexual Medicine, Nicolae Testemitanu State University of Medicine and Pharmacy, 2004 Chisinau, Moldova
| | - Anca Maria Cimpean
- Department of Microscopic Morphology/Histology, Victor Babes University of Medicine and Pharmacy, 300041 Timisoara, Romania; (A.C.B.); (M.P.F.); (M.P.)
- Center of Expertise for Rare Vascular Disease in Children, Emergency Hospital for Children Louis Turcanu, 300011 Timisoara, Romania
| | - Eugen Melnic
- Department of Pathology, Nicolae Testemitanu State University of Medicine and Pharmacy, 2004 Chisinau, Moldova;
| |
Collapse
|
3
|
Zou YW, Ren ZG, Sun Y, Liu ZG, Hu XB, Wang HY, Yu ZJ. The latest research progress on minimally invasive treatments for hepatocellular carcinoma. Hepatobiliary Pancreat Dis Int 2023; 22:54-63. [PMID: 36041973 DOI: 10.1016/j.hbpd.2022.08.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 08/09/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the fourth leading cause of cancer-related death worldwide. Due to the high prevalence of hepatitis B virus (HBV) infection in China, the incidence of HCC in China is high, and liver cirrhosis caused by chronic hepatitis also brings great challenges to treatment. This paper reviewed the latest research progress on minimally invasive treatments for HCC, including percutaneous thermal ablation and new nonthermal ablation techniques, and introduced the principles, advantages, and clinical applications of various therapeutic methods in detail. DATA SOURCES The data of treatments for HCC were systematically collected from the PubMed, ScienceDirect, American Chemical Society and Web of Science databases published in English, using "minimally invasive" and "hepatocellular carcinoma" or "liver cancer" as the keywords. RESULTS Percutaneous thermal ablation is still a first-line strategy for the minimally invasive treatment of HCC. The effect of microwave ablation (MWA) on downgrading treatment before liver transplantation is better than that of radiofrequency ablation (RFA), while RFA is more widely used in the clinical practice. High-intensity focused ultrasound (HIFU) is mainly used for the palliative treatment of advanced liver cancer. Electrochemotherapy (ECT) delivers chemotherapeutic drugs to the target cells while reducing the blood supply around HCC. Irreversible electroporation (IRE) uses a microsecond-pulsed electric field that induces apoptosis and necrosis and triggers a systemic immune response. The nanosecond pulsed electric field (nsPEF) has achieved a good response in the ablation of mice with HCC, but it has not been reported in China for the treatment of human HCC. CONCLUSIONS A variety of minimally invasive treatments provide a sufficient survival advantage for HCC patients. Nonthermal ablation will lead to a new wave with its unique advantage of antitumor recurrence and metastasis.
Collapse
Affiliation(s)
- Ya-Wen Zou
- Department of Infectious Diseases, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Key Laboratory of Pulsed Power Translational Medicine of Zhejiang Province, 2959 Yuhangtang Road, Hangzhou 310000, China; Gene Hospital of Henan Province; Precision Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Zhi-Gang Ren
- Department of Infectious Diseases, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Key Laboratory of Pulsed Power Translational Medicine of Zhejiang Province, 2959 Yuhangtang Road, Hangzhou 310000, China; Gene Hospital of Henan Province; Precision Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Ying Sun
- Department of Infectious Diseases, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Key Laboratory of Pulsed Power Translational Medicine of Zhejiang Province, 2959 Yuhangtang Road, Hangzhou 310000, China; Gene Hospital of Henan Province; Precision Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Zhen-Guo Liu
- Department of Infectious Diseases, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Key Laboratory of Pulsed Power Translational Medicine of Zhejiang Province, 2959 Yuhangtang Road, Hangzhou 310000, China; Gene Hospital of Henan Province; Precision Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Xiao-Bo Hu
- Department of Infectious Diseases, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Gene Hospital of Henan Province; Precision Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Hai-Yu Wang
- Department of Infectious Diseases, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Gene Hospital of Henan Province; Precision Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Zu-Jiang Yu
- Department of Infectious Diseases, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China; Gene Hospital of Henan Province; Precision Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| |
Collapse
|
4
|
Recent advances in atezolizumab-based programmed death-ligand 1 (PD-L1) blockade therapy for breast cancer. Int Immunopharmacol 2022; 113:109334. [DOI: 10.1016/j.intimp.2022.109334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 10/08/2022] [Accepted: 10/08/2022] [Indexed: 11/05/2022]
|
5
|
Moccia C, Haase K. Engineering Breast Cancer On-chip-Moving Toward Subtype Specific Models. Front Bioeng Biotechnol 2021; 9:694218. [PMID: 34249889 PMCID: PMC8261144 DOI: 10.3389/fbioe.2021.694218] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 05/31/2021] [Indexed: 12/12/2022] Open
Abstract
Breast cancer is the second leading cause of death among women worldwide, and while hormone receptor positive subtypes have a clear and effective treatment strategy, other subtypes, such as triple negative breast cancers, do not. Development of new drugs, antibodies, or immune targets requires significant re-consideration of current preclinical models, which frequently fail to mimic the nuances of patient-specific breast cancer subtypes. Each subtype, together with the expression of different markers, genetic and epigenetic profiles, presents a unique tumor microenvironment, which promotes tumor development and progression. For this reason, personalized treatments targeting components of the tumor microenvironment have been proposed to mitigate breast cancer progression, particularly for aggressive triple negative subtypes. To-date, animal models remain the gold standard for examining new therapeutic targets; however, there is room for in vitro tools to bridge the biological gap with humans. Tumor-on-chip technologies allow for precise control and examination of the tumor microenvironment and may add to the toolbox of current preclinical models. These new models include key aspects of the tumor microenvironment (stroma, vasculature and immune cells) which have been employed to understand metastases, multi-organ interactions, and, importantly, to evaluate drug efficacy and toxicity in humanized physiologic systems. This review provides insight into advanced in vitro tumor models specific to breast cancer, and discusses their potential and limitations for use as future preclinical patient-specific tools.
Collapse
Affiliation(s)
| | - Kristina Haase
- European Molecular Biology Laboratory, European Molecular Biology Laboratory Barcelona, Barcelona, Spain
| |
Collapse
|
6
|
Kwon Y. Possible Beneficial Effects of N-Acetylcysteine for Treatment of Triple-Negative Breast Cancer. Antioxidants (Basel) 2021; 10:169. [PMID: 33498875 PMCID: PMC7911701 DOI: 10.3390/antiox10020169] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 01/21/2021] [Accepted: 01/21/2021] [Indexed: 12/24/2022] Open
Abstract
N-acetylcysteine (NAC) is a widely used antioxidant with therapeutic potential. However, the cancer-promoting effect of NAC observed in some preclinical studies has raised concerns regarding its clinical use. Reactive oxygen species (ROS) can mediate signaling that results in both cancer-promoting and cancer-suppressing effects. The beneficial effect of NAC may depend on whether the type of cancer relies on ROS signaling for its survival and metastasis. Triple-negative breast cancer (TNBC) has aggressive phenotypes and is currently treated with standard chemotherapy as the main systemic treatment option. Particularly, basal-like TNBC cells characterized by inactivated BRCA1 and mutated TP53 produce high ROS levels and rely on ROS signaling for their survival and malignant progression. In addition, the high ROS levels in TNBC cells can mediate the interplay between cancer cells and the tissue microenvironment (TME) to trigger the recruitment and conversion of stromal cells and induce hypoxic responses, thus leading to the creation of cancer-supportive TMEs and increased cancer aggressiveness. However, NAC treatment effectively reduces the ROS production and ROS-mediated signaling that contribute to cell survival, metastasis, and drug resistance in TNBC cells. Therefore, the inclusion of NAC in standard chemotherapy could probably provide additional benefits for TNBC patients.
Collapse
Affiliation(s)
- Youngjoo Kwon
- Department of Food Science and Engineering, Ewha Womans University, Seoul 03760, Korea
| |
Collapse
|
7
|
A Marine Collagen-Based Biomimetic Hydrogel Recapitulates Cancer Stem Cell Niche and Enhances Progression and Chemoresistance in Human Ovarian Cancer. Mar Drugs 2020; 18:md18100498. [PMID: 33003514 PMCID: PMC7599646 DOI: 10.3390/md18100498] [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: 08/31/2020] [Revised: 09/25/2020] [Accepted: 09/28/2020] [Indexed: 12/12/2022] Open
Abstract
Recent attention has focused on the development of an effective three-dimensional (3D) cell culture system enabling the rapid enrichment of cancer stem cells (CSCs) that are resistant to therapies and serving as a useful in vitro tumor model that accurately reflects in vivo behaviors of cancer cells. Presently, an effective 3D in vitro model of ovarian cancer (OC) was developed using a marine collagen-based hydrogel. Advantages of the model include simplicity, efficiency, bioactivity, and low cost. Remarkably, OC cells grown in this hydrogel exhibited biochemical and physiological features, including (1) enhanced cell proliferation, migration and invasion, colony formation, and chemoresistance; (2) suppressed apoptosis with altered expression levels of apoptosis-regulating molecules; (3) upregulated expression of crucial multidrug resistance-related genes; (4) accentuated expression of key molecules associated with malignant progression, such as epithelial–mesenchymal transition transcription factors, Notch, and pluripotency biomarkers; and (5) robust enrichment of ovarian CSCs. The findings indicate the potential of our 3D in vitro OC model as an in vitro research platform to study OC and ovarian CSC biology and to screen novel therapies targeting OC and ovarian CSCs.
Collapse
|
8
|
Single-Cell RNA-seq Reveals Obesity-Induced Alterations in the Brca1-Mutated Mammary Gland Microenvironment. Cancers (Basel) 2020; 12:cancers12082235. [PMID: 32785175 PMCID: PMC7464292 DOI: 10.3390/cancers12082235] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 08/06/2020] [Accepted: 08/07/2020] [Indexed: 12/29/2022] Open
Abstract
Clinical and experimental studies have shown that obesity increases the development and progression of breast cancer. The impact of obesity on the tumor microenvironment plays an important role in tumorigenesis, yet the precise mechanisms underlying obesity-mediated effects on cell-to-cell communication within the tumor microenvironment have been difficult to define. In this study, we conducted single-cell RNA sequencing (scRNA-seq) studies to investigate the impact of high-fat diet (HFD)-induced obesity on transcriptomic landscapes of stromal and immune cells in mammary glands of Brca1−/−; p53+/− mice, an animal breast cancer model. Hierarchical clustering and gene pathway enrichment analyses of scRNA-seq data showed that five different subtypes of stromal fibroblasts existed in mouse Brca1-mutated mammary glands. HFD-induced obesity led to upregulated expression of extracellular matrix (ECM) genes (Col3a1, Col6a3, Eln, and Sparc) and downregulated expression of immunoregulatory genes (Iigp1 and Cxcl10) in these stromal subtype cells. These findings, taken together, suggest that obesity alters the ECM composition and immune ecosystem through modulating the functionality of mammary stromal fibroblasts. Moreover, scRNA-seq analysis of mammary immune cells indicated that HFD-induced obesity promoted the generation and/or recruiting of pro-tumorigenic M2 macrophages in mammary glands. Our studies provide new insight into a mechanistic paradigm wherein obesity modulates the functions of stromal and immune cells to create the tumorigenic microenvironment for promoting breast tumorigenesis.
Collapse
|
9
|
Olivares-Urbano MA, Griñán-Lisón C, Marchal JA, Núñez MI. CSC Radioresistance: A Therapeutic Challenge to Improve Radiotherapy Effectiveness in Cancer. Cells 2020; 9:cells9071651. [PMID: 32660072 PMCID: PMC7407195 DOI: 10.3390/cells9071651] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/03/2020] [Accepted: 07/07/2020] [Indexed: 12/12/2022] Open
Abstract
Radiotherapy (RT) is a modality of oncologic treatment that can be used to treat approximately 50% of all cancer patients either alone or in combination with other treatment modalities such as surgery, chemotherapy, immunotherapy, and therapeutic targeting. Despite the technological advances in RT, which allow a more precise delivery of radiation while progressively minimizing the impact on normal tissues, issues like radioresistance and tumor recurrence remain important challenges. Tumor heterogeneity is responsible for the variation in the radiation response of the different tumor subpopulations. A main factor related to radioresistance is the presence of cancer stem cells (CSC) inside tumors, which are responsible for metastases, relapses, RT failure, and a poor prognosis in cancer patients. The plasticity of CSCs, a process highly dependent on the epithelial–mesenchymal transition (EMT) and associated to cell dedifferentiation, complicates the identification and eradication of CSCs and it might be involved in disease relapse and progression after irradiation. The tumor microenvironment and the interactions of CSCs with their niches also play an important role in the response to RT. This review provides a deep insight into the characteristics and radioresistance mechanisms of CSCs and into the role of CSCs and tumor microenvironment in both the primary tumor and metastasis in response to radiation, and the radiobiological principles related to the CSC response to RT. Finally, we summarize the major advances and clinical trials on the development of CSC-based therapies combined with RT to overcome radioresistance. A better understanding of the potential therapeutic targets for CSC radiosensitization will provide safer and more efficient combination strategies, which in turn will improve the live expectancy and curability of cancer patients.
Collapse
Affiliation(s)
| | - Carmen Griñán-Lisón
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, University of Granada, 18100 Granada, Spain;
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, 18016 Granada, Spain
| | - Juan Antonio Marchal
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, University of Granada, 18100 Granada, Spain;
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18016 Granada, Spain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, 18016 Granada, Spain
- Correspondence: (J.A.M.); (M.I.N.); Tel.: +34-958-249321 (J.A.M.); +34-958-242077 (M.I.N.)
| | - María Isabel Núñez
- Department of Radiology and Physical Medicine, University of Granada, 18016 Granada, Spain;
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, University of Granada, 18100 Granada, Spain;
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18012 Granada, Spain
- Correspondence: (J.A.M.); (M.I.N.); Tel.: +34-958-249321 (J.A.M.); +34-958-242077 (M.I.N.)
| |
Collapse
|
10
|
Okuma H, Sudah M, Kettunen T, Niukkanen A, Sutela A, Masarwah A, Kosma VM, Auvinen P, Mannermaa A, Vanninen R. Peritumor to tumor apparent diffusion coefficient ratio is associated with biologically more aggressive breast cancer features and correlates with the prognostication tools. PLoS One 2020; 15:e0235278. [PMID: 32584887 PMCID: PMC7316248 DOI: 10.1371/journal.pone.0235278] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 06/12/2020] [Indexed: 12/11/2022] Open
Abstract
PURPOSE The apparent diffusion coefficient (ADC) is increasingly used to characterize breast cancer. The peritumor/tumor ADC ratio is suggested to be a reliable and generally applicable index. However, its overall prognostication value remains unclear. We aimed to evaluate the associations between the peritumor/tumor ADC ratio and histopathological biomarkers and published prognostic tools in patients with invasive breast cancer. MATERIALS AND METHODS This prospective study included 88 lesions (five bilateral) in 83 patients with primary invasive breast cancer who underwent preoperative 3.0-T magnetic resonance imaging. The lowest intratumoral mean ADC value on the slice with the largest tumor cross-sectional area was designated the tumor ADC, and the highest mean ADC value on the peritumoral breast parenchymal tissue adjacent to the tumor border was designated the peritumor ADC. The peritumor/tumor ADC ratio was then calculated. The tumor and peritumor ADC values and peritumor/tumor ADC ratios were compared with histopathological parameters using an unpaired t test, and their correlations with published prognostic tools were evaluated with Pearson's correlation coefficient. RESULTS The peritumor/tumor ADC ratio was significantly associated with tumor size (p<0.001), histological grade (p = 0.005), Ki-67 index (p = 0.006), axillary-lymph-node metastasis (p = 0.001), and lymphovascular invasion (p = 0.006), but was not associated with estrogen receptor status (p = 0.931), progesterone receptor status (p = 0.160), or human epidermal growth factor receptor 2 status (p = 0.259). The peritumor/tumor ADC ratio showed moderate positive correlations with the Nottingham Prognostic Index (r = 0.498, p<0.001) and mortality predicted using PREDICT (r = 0.436, p<0.001). CONCLUSION The peritumor/tumor ADC ratio was correlated with histopathological biomarkers in patients with invasive breast cancer, showed significant correlations with published prognostic indexes, and may provide an easily applicable imaging index for the preoperative prognostic evaluation of breast cancer.
Collapse
Affiliation(s)
- Hidemi Okuma
- Institute of Clinical Medicine, School of Medicine, Clinical Radiology, University of Eastern Finland, Kuopio, Finland
- Department of Clinical Radiology, Diagnostic Imaging Center, Kuopio University Hospital, Kuopio, Finland
- * E-mail:
| | - Mazen Sudah
- Institute of Clinical Medicine, School of Medicine, Clinical Radiology, University of Eastern Finland, Kuopio, Finland
- Department of Clinical Radiology, Diagnostic Imaging Center, Kuopio University Hospital, Kuopio, Finland
| | - Tiia Kettunen
- Institute of Clinical Medicine, School of Medicine, Oncology, University of Eastern Finland, Kuopio, Finland
- Department of Oncology, Cancer Center, Kuopio University Hospital, Kuopio, Finland
| | - Anton Niukkanen
- Institute of Clinical Medicine, School of Medicine, Clinical Radiology, University of Eastern Finland, Kuopio, Finland
- Department of Clinical Radiology, Diagnostic Imaging Center, Kuopio University Hospital, Kuopio, Finland
| | - Anna Sutela
- Institute of Clinical Medicine, School of Medicine, Clinical Radiology, University of Eastern Finland, Kuopio, Finland
- Department of Clinical Radiology, Diagnostic Imaging Center, Kuopio University Hospital, Kuopio, Finland
| | - Amro Masarwah
- Institute of Clinical Medicine, School of Medicine, Clinical Radiology, University of Eastern Finland, Kuopio, Finland
- Department of Clinical Radiology, Diagnostic Imaging Center, Kuopio University Hospital, Kuopio, Finland
| | - Veli-Matti Kosma
- Institute of Clinical Medicine, School of Medicine, Pathology and Forensic Medicine, and Translational Cancer Research Area, University of Eastern Finland, Kuopio, Finland
- Biobank of Eastern Finland, Kuopio University Hospital, Kuopio, Finland
| | - Päivi Auvinen
- Institute of Clinical Medicine, School of Medicine, Oncology, University of Eastern Finland, Kuopio, Finland
- Department of Oncology, Cancer Center, Kuopio University Hospital, Kuopio, Finland
| | - Arto Mannermaa
- Institute of Clinical Medicine, School of Medicine, Pathology and Forensic Medicine, and Translational Cancer Research Area, University of Eastern Finland, Kuopio, Finland
- Biobank of Eastern Finland, Kuopio University Hospital, Kuopio, Finland
| | - Ritva Vanninen
- Institute of Clinical Medicine, School of Medicine, Clinical Radiology, University of Eastern Finland, Kuopio, Finland
- Department of Clinical Radiology, Diagnostic Imaging Center, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
11
|
Toss MS, Abidi A, Lesche D, Joseph C, Mahale S, Saunders H, Kader T, Miligy IM, Green AR, Gorringe KL, Rakha EA. The prognostic significance of immune microenvironment in breast ductal carcinoma in situ. Br J Cancer 2020; 122:1496-1506. [PMID: 32203210 PMCID: PMC7217899 DOI: 10.1038/s41416-020-0797-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 02/07/2020] [Accepted: 02/26/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The role of different subtypes of tumour infiltrating lymphocytes (TILs) in breast ductal carcinoma in situ (DCIS) is still poorly defined. This study aimed to assess the prognostic significance of B and T lymphocytes and immune checkpoint proteins expression in DCIS. METHODS A well characterised DCIS cohort (n = 700) with long-term follow-up comprising pure DCIS (n = 508) and DCIS mixed with invasive carcinoma (IBC; n = 192) were stained immunohistochemically for CD20, CD3, CD4, CD8, FOXP3, PD1 and PDL1. Copy number variation and TP53 mutation status were assessed in a subset of cases (n = 58). RESULTS CD3+ lymphocytes were the predominant cell subtype in the pure DCIS cohort, while FOXP3 showed the lowest levels. PDL1 expression was mainly seen in the stromal TILs. Higher abundance of TILs subtypes was associated with higher tumour grade, hormone receptor negativity and HER2 positivity. Mutant TP53 variants were associated with higher levels of stromal CD3+, CD4+ and FOXP3+ cells. DCIS coexisting with invasive carcinoma harboured denser stromal infiltrates of all immune cells and checkpoint proteins apart from CD4+ cells. Stromal PD1 was the most differentially expressed protein between DCIS and invasive carcinoma (Z = 5.8, p < 0.0001). Dense TILs, stromal FOXP3 and PDL1 were poor prognostic factors for DCIS recurrence, while dense TILs were independently associated with poor outcome for all recurrences (HR = 7.0; p = 0.024), and invasive recurrence (HR = 2.1; p = 0.029). CONCLUSIONS Immunosuppressive proteins are potential markers for high risk DCIS and disease progression. Different stromal and intratumoural lymphocyte composition between pure DCIS, DCIS associated with IBC and invasive carcinoma play a potential role in their prognostic significance and related to the underlying genomic instability. Assessment of overall TILs provides a promising tool for evaluation of the DCIS immune microenvironment.
Collapse
MESH Headings
- B-Lymphocytes/immunology
- B7-H1 Antigen/genetics
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/immunology
- Breast Neoplasms/genetics
- Breast Neoplasms/pathology
- CD4-Positive T-Lymphocytes/immunology
- Carcinoma, Intraductal, Noninfiltrating/genetics
- Carcinoma, Intraductal, Noninfiltrating/immunology
- Carcinoma, Intraductal, Noninfiltrating/pathology
- Cell Lineage/genetics
- Cell Lineage/immunology
- DNA Copy Number Variations/genetics
- Female
- Forkhead Transcription Factors/genetics
- Humans
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Prognosis
- Receptor, ErbB-2/genetics
- T-Lymphocytes/immunology
- Tumor Microenvironment/immunology
- Tumor Suppressor Protein p53/genetics
Collapse
Affiliation(s)
- Michael S Toss
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham, Nottingham City Hospital, Nottingham, UK.
- Histopathology Department, South Egypt Cancer Institute, Assiut University, Assiut, Egypt.
| | - Asima Abidi
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham, Nottingham City Hospital, Nottingham, UK
| | - Dorothea Lesche
- Cancer Genomics Program, Peter MacCallum Cancer Centre, Melbourne, Australia
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| | - Chitra Joseph
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham, Nottingham City Hospital, Nottingham, UK
| | - Sakshi Mahale
- Cancer Genomics Program, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Hugo Saunders
- Cancer Genomics Program, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Tanjina Kader
- Cancer Genomics Program, Peter MacCallum Cancer Centre, Melbourne, Australia
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| | - Islam M Miligy
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham, Nottingham City Hospital, Nottingham, UK
| | - Andrew R Green
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham, Nottingham City Hospital, Nottingham, UK
| | - Kylie L Gorringe
- Cancer Genomics Program, Peter MacCallum Cancer Centre, Melbourne, Australia
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| | - Emad A Rakha
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, The University of Nottingham, Nottingham City Hospital, Nottingham, UK
| |
Collapse
|
12
|
Obradović A, Matić M, Ognjanović B, Đurđević P, Marinković E, Ušćumlić G, Božić B, Božić Nedeljković B. Antiproliferative and antimigratory effects of 3-(4-substituted benzyl)-5- isopropyl-5-phenylhydantoin derivatives in human breast cancer cells. Saudi Pharm J 2020; 28:246-254. [PMID: 32194325 PMCID: PMC7078530 DOI: 10.1016/j.jsps.2020.01.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 01/19/2020] [Indexed: 02/07/2023] Open
Abstract
In this study, a series of synthesized 3-(4-substituted benzyl)-5-isopropyl-5-phenylhydantoin derivatives as a potential antiproliferative and antimigratory agents were investigated. The possible antitumor mechanisms of investigated hydantoin derivatives were examined on human breast cancer cell line MDA-MB-231. The cells were treated with different concentrations of compounds (from 0.01 µM to 100 µM) during 24 h and 72 h. The proliferation index, nitric oxide production, apoptosis rate, and migration capacity were measured. The cell invasion potential was examined by measuring the level of MMP-9 and COX-2 gene expression. All tested compounds expressed antiproliferative activity and induced dose- and time-dependent increase in the level of nitrites. The investigated molecules significantly decreased cell survival rate, migration capacity and the expression levels of genes included in the process of tumor invasion. Obtained data suggest that the tested hydantoin derivatives express considerable antitumor activity by reducing cell division rate, elevating apoptosis level, and inhibiting the motility and invasiveness of breast cancer cells. The results obtained in this study indicate that investigated compounds express potential as a novel chemotherapeutic agents against breast cancer growth and progression.
Collapse
Affiliation(s)
- Ana Obradović
- Department of Biology and Ecology, Faculty of Science, University of Kragujevac, Kragujevac, Serbia
| | - Miloš Matić
- Department of Biology and Ecology, Faculty of Science, University of Kragujevac, Kragujevac, Serbia
| | - Branka Ognjanović
- Department of Biology and Ecology, Faculty of Science, University of Kragujevac, Kragujevac, Serbia
| | - Predrag Đurđević
- Department of Internal Medicine, Clinic for Hematology Clinical Center Kragujevac, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Emilija Marinković
- Department of Research and Development, Institute of Virology, Vaccines and Sera - TORLAK, Belgrade, Serbia
| | - Gordana Ušćumlić
- Department of Organic Chemistry, Faculty of Technology and Metallurgy, University of Belgrade, Belgrade, Serbia
| | - Bojan Božić
- Institute of Physiology and Biochemistry "Ivan Đaja", Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Biljana Božić Nedeljković
- Institute of Physiology and Biochemistry "Ivan Đaja", Faculty of Biology, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
13
|
Quantitative background parenchymal enhancement to predict recurrence after neoadjuvant chemotherapy for breast cancer. Sci Rep 2019; 9:19185. [PMID: 31844135 PMCID: PMC6914793 DOI: 10.1038/s41598-019-55820-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 11/29/2019] [Indexed: 01/02/2023] Open
Abstract
Breast background parenchymal enhancement (BPE) is an increasingly studied MRI parameter that reflects the microvasculature of normal breast tissue, which has been shown to change during neoadjuvant chemotherapy (NAC) for breast cancer. We aimed at evaluating the BPE in patients undergoing NAC and its prognostic value to predict recurrence. MRI BPE was visually and quantitatively evaluated before and after NAC in a retrospective cohort of 102 women with unilateral biopsy-proven invasive breast cancer. Pre-therapeutic BPE was not predictive of pathological response or recurrence. Quantitative post-therapeutic BPE was significantly decreased compared to pre-therapeutic value. Post-therapeutic quantitative BPE significantly predicted recurrence (HR = 6.38 (0.71, 12.06), p < 0.05).
Collapse
|
14
|
Pu N, Chen Q, Gao S, Liu G, Zhu Y, Yin L, Hu H, Wei L, Wu Y, Maeda S, Lou W, Yu J, Wu W. Genetic landscape of prognostic value in pancreatic ductal adenocarcinoma microenvironment. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:645. [PMID: 31930046 DOI: 10.21037/atm.2019.10.91] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Background The prognosis of pancreatic ductal adenocarcinoma (PDAC) remains dismally poor and is widely considered as an intricate genetic disorder. The mutational landscape of PDAC may directly reflect cancer immunogenicity and dictate the extent and phenotype of immune cell infiltration. In adverse, the microenvironment may also effect the gene expression of cancer cells, which is associated with clinical prognosis. Thus, it is crucial to identify genomic alterations in PDAC microenvironment and its impacts on clinical prognosis. Methods The gene expression profiles, mutation data and clinical information of 179 pancreatic cancer patients with an initial pathologic diagnosis ranging from 2001 to 2013 were retrieved from The Cancer Genome Atlas (TCGA) database. The MAlignant Tumor tissues using Expression data (ESTIMATE) algorithm for calculating immune scores and stromal scores and Tumor IMmune Estimation Resource (TIMER) resource for cell infiltrations were applied in this study. Results The average immune score or stromal score of PDAC subtype was significantly higher than that of other specific subtypes. KRAS mutant cases had significantly lower immune scores (P=0.001) and stromal scores (P=0.007), in concert with lower immune scores in TP53 mutant cases (P=0.030). However, no significant difference was found in SMAD4 and CDKN2A mutations. In the cohort OS/RFS, the infiltration levels of CD8+ T cells, B cells, Macrophages, Neutrophils and DCs in high stromal score group were higher than those in the low score group (all P<0.001), as well as in immune score groups except for Macrophages in the cohort RFS. In the cohort OS/RFS, 317/379 upregulated genes and 9/6 downregulated genes were observed in the high immune score group, while 227/205 upregulated genes and 17/6 downregulated genes in the high stromal score group. With the analysis for prognostic value of DEGs, 82 and 58 DEGs respectively in the high immune and stromal score group were verified to be significantly associated with better OS (P<0.05), while 53 and 17 DEGs respectively with longer RFS (P<0.05). Functional enrichment analysis showed genes of prognostic values were significantly related to immune response. Conclusions A list of genes with prognostic value in PDAC microenvironment were obtained from functional enrichment analysis based on immune and stromal scores, which indicates a series of potential auxiliary prognostic biomarkers for PDAC are available. Further research on these genes may be valuable and helpful to understand the crosstalk between tumor and microenvironment, new immune evasion mechanisms and underlying novel therapeutic targets in an integrated manner.
Collapse
Affiliation(s)
- Ning Pu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China.,Department of Surgery and The Pancreatic Cancer Precision Medicine Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Qiangda Chen
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Shanshan Gao
- Department of Surgery and The Pancreatic Cancer Precision Medicine Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Gao Liu
- Department of Liver Surgery and Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yayun Zhu
- Department of Surgery and The Pancreatic Cancer Precision Medicine Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Liver Surgery and Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Lingdi Yin
- Department of Surgery and The Pancreatic Cancer Precision Medicine Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, and Pancreas Institute of Nanjing Medical University, Nanjing 210029, China
| | - Haijie Hu
- Department of Surgery and The Pancreatic Cancer Precision Medicine Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Biliary Surgery, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Li Wei
- Department of Liver Surgery and Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yong Wu
- Department of Surgery and The Pancreatic Cancer Precision Medicine Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Shimpei Maeda
- Department of Surgery and The Pancreatic Cancer Precision Medicine Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Wenhui Lou
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jun Yu
- Department of Surgery and The Pancreatic Cancer Precision Medicine Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Wenchuan Wu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
15
|
Dos Reis DC, Damasceno KA, de Campos CB, Veloso ES, Pêgas GRA, Kraemer LR, Rodrigues MA, Mattos MS, Gomes DA, Campos PP, Ferreira E, Russo RC, Cassali GD. Versican and Tumor-Associated Macrophages Promotes Tumor Progression and Metastasis in Canine and Murine Models of Breast Carcinoma. Front Oncol 2019; 9:577. [PMID: 31334111 PMCID: PMC6616078 DOI: 10.3389/fonc.2019.00577] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 06/14/2019] [Indexed: 12/31/2022] Open
Abstract
Versican and tumor-associated macrophages (TAMs) are involved in growth and metastases in several cancers. Here, we investigated the potential role of versican, a matrix proteoglycan, and its correlation with TAMs infiltrates in different stages of two different breast cancer models: spontaneous canine mammary gland carcinomas and the murine 4T1 breast cancer model. The stromal versican expression was correlated with TAMs accumulation in tumors with an advanced stage from spontaneous canine mammary carcinoma samples. Versican expression in mice, identified in late stages of tumor progression, was associated to a high number of peri-tumoral infiltrating TAMs. Indeed, TAMs were related to a pro-inflammatory and pro-angiogenic state in the primary tumor. Furthermore, TAMs accumulation was related to versican expression in the lungs and an increased number of pulmonary metastatic nodules with pulmonary mechanical dysfunction, which was due to leukocyte influx in the airways and elevated growth factor levels in the microenvironment. Thus, we suggest that versican and TAMs as attractive targets for breast cancer therapy.
Collapse
Affiliation(s)
- Diego Carlos Dos Reis
- Department of General Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Cecília Bonolo de Campos
- Department of General Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Emerson Soares Veloso
- Department of General Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Gabriela Rafaela Arantes Pêgas
- Department of General Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lucas Rocha Kraemer
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Michele Angela Rodrigues
- Department of General Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Matheus Silvério Mattos
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Dawidson Assis Gomes
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Paula Peixoto Campos
- Department of General Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Enio Ferreira
- Department of General Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Remo Castro Russo
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Geovanni Dantas Cassali
- Department of General Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
16
|
Inhibitory Effect of Alisma canaliculatum Ethanolic Extract on NF-κB-Dependent CXCR3 and CXCL10 Expression in TNFα-Exposed MDA-MB-231 Breast Cancer Cells. Int J Mol Sci 2018; 19:ijms19092607. [PMID: 30177620 PMCID: PMC6165157 DOI: 10.3390/ijms19092607] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 08/29/2018] [Accepted: 08/30/2018] [Indexed: 12/15/2022] Open
Abstract
CXC motif chemokine ligand 10 (CXCL10) and its receptor CXC motif chemokine receptor 3 (CXCR3), play important roles in the motility of breast cancer cells. Alisma canaliculatum is a herb that has been used as a traditional medicine for thousands of years in Korea and China. Whether A. canaliculatum inhibits the motility of metastatic breast cancer cells is not clear yet. In this study, we show that A. canaliculatum ethanolic extract (ACE) prevented tumor necrosis factor-alpha (TNFα)-induced migration of MDA-MB-231 cells. ACE significantly attenuated TNFα-induced upregulation of CXCL10 and CXCR3 expression at the gene promoter level. Mechanistically, ACE inhibits TNFα-induced phosphorylation of inhibitor of κB (IκB) kinase (IKK), IκB and p65/RelA, leading to the suppression of nuclear translocation of p65/RelA nuclear factor kappa-B (NF-κB). Also, ACE inhibited NF-κB-dependent CXCR3 and CXCL10 promoter activities. These results suggest that ACE abrogates TNFα-induced migration of MDA-MB-231 breast cancer cells through down-regulation of IKK-NF-κB-dependent CXCR3 and CXCL10 expression. Our results suggest that ACE has potential as a herbal supplement for the inhibition of breast cancer metastasis.
Collapse
|
17
|
Siersbæk R, Kumar S, Carroll JS. Signaling pathways and steroid receptors modulating estrogen receptor α function in breast cancer. Genes Dev 2018; 32:1141-1154. [PMID: 30181360 PMCID: PMC6120708 DOI: 10.1101/gad.316646.118] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Estrogen receptor α (ER) is the major driver of ∼75% of breast cancers, and multiple ER targeting drugs are routinely used clinically to treat patients with ER+ breast cancer. However, many patients relapse on these targeted therapies and ultimately develop metastatic and incurable disease, and understanding the mechanisms leading to drug resistance is consequently of utmost importance. It is now clear that, in addition to estrogens, ER function is modulated by other steroid receptors and multiple signaling pathways (e.g., growth factor and cytokine signaling), and many of these pathways affect drug resistance and patient outcome. Here, we review the mechanisms through which these pathways impact ER function and drug resistance as well as discuss the clinical implications.
Collapse
Affiliation(s)
- Rasmus Siersbæk
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, United Kingdom
| | - Sanjeev Kumar
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, United Kingdom
- Addenbrookes Hospital, Cambridge CB2 0QQ, United Kingdom
| | - Jason S Carroll
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, United Kingdom
| |
Collapse
|
18
|
Beebe SJ, Lassiter BP, Guo S. Nanopulse Stimulation (NPS) Induces Tumor Ablation and Immunity in Orthotopic 4T1 Mouse Breast Cancer: A Review. Cancers (Basel) 2018; 10:cancers10040097. [PMID: 29601471 PMCID: PMC5923352 DOI: 10.3390/cancers10040097] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 03/13/2018] [Accepted: 03/27/2018] [Indexed: 12/13/2022] Open
Abstract
Nanopulse Stimulation (NPS) eliminates mouse and rat tumor types in several different animal models. NPS induces protective, vaccine-like effects after ablation of orthotopic rat N1-S1 hepatocellular carcinoma. Here we review some general concepts of NPS in the context of studies with mouse metastatic 4T1 mammary cancer showing that the postablation, vaccine-like effect is initiated by dynamic, multilayered immune mechanisms. NPS eliminates primary 4T1 tumors by inducing immunogenic, caspase-independent programmed cell death (PCD). With lower electric fields, like those peripheral to the primary treatment zone, NPS can activate dendritic cells (DCs). The activation of DCs by dead/dying cells leads to increases in memory effector and central memory T-lymphocytes in the blood and spleen. NPS also eliminates immunosuppressive cells in the tumor microenvironment and blood. Finally, NPS treatment of 4T1 breast cancer exhibits an abscopal effect and largely prevents spontaneous metastases to distant organs. NPS with fast rise–fall times and pulse durations near the plasma membrane charging time constant, which exhibits transient, high-frequency components (1/time = Hz), induce responses from mitochondria, endoplasmic reticulum, and nucleus. Such effects may be responsible for release of danger-associated molecular patterns, including ATP, calreticulin, and high mobility group box 1 (HMBG1) from 4T1-Luc cells to induce immunogenic cell death (ICD). This likely leads to immunity and the vaccine-like response. In this way, NPS acts as a unique onco-immunotherapy providing distinct therapeutic advantages showing possible clinical utility for breast cancers as well as for other malignancies.
Collapse
Affiliation(s)
- Stephen J Beebe
- Frank Reidy Research Center for Bioelectrics, 4211 Monarch Ways, Suite 300, Norfolk, VA 23508, USA.
| | - Brittany P Lassiter
- Frank Reidy Research Center for Bioelectrics, 4211 Monarch Ways, Suite 300, Norfolk, VA 23508, USA.
| | - Siqi Guo
- Frank Reidy Research Center for Bioelectrics, 4211 Monarch Ways, Suite 300, Norfolk, VA 23508, USA.
| |
Collapse
|
19
|
Gyamfi J, Eom M, Koo JS, Choi J. Multifaceted Roles of Interleukin-6 in Adipocyte-Breast Cancer Cell Interaction. Transl Oncol 2018; 11:275-285. [PMID: 29413760 PMCID: PMC5884177 DOI: 10.1016/j.tranon.2017.12.009] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 12/18/2017] [Accepted: 12/18/2017] [Indexed: 12/12/2022] Open
Abstract
Breast cancer is the most common malignancy in women worldwide, with a developmental process spanning decades. The malignant cells recruit a variety of cells including fibroblasts, endothelial cells, immune cells, and adipocytes, creating the tumor microenvironment. The tumor microenvironment has emerged as active participants in breast cancer progression and response to treatment through autocrine and paracrine interaction with the malignant cells. Adipose tissue is abundant in the breast cancer microenvironment; interactions with cancer cells create cancer-associated adipocytes which produce a variety of adipokines that influence breast cancer initiation, metastasis, angiogenesis, and cachexia. Interleukin (IL)-6 has emerged as key compound significantly produced by breast cancer cells and adipocytes, with the potential of inducing proliferation, epithelial-mesenchymal phenotype, stem cell phenotype, angiogenesis, cachexia, and therapeutic resistance in breast cancer cells. Our aim is to present a brief knowledge of IL-6’s role in breast cancer. This review summarizes our current understanding of the breast microenvironment, with emphasis on adipocytes as key players in breast cancer tumorigenesis. The effects of key adipocytes such as leptin, adipokines, TGF-b, and IL-6 are discussed. Finally, we discuss the role of IL-6 in various aspects of cancer progression.
Collapse
Affiliation(s)
- Jones Gyamfi
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, Korea
| | - Minseob Eom
- Department of Pathology, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Ja-Seung Koo
- Department of Pathology, Yonsei University College of Medicine.
| | - Junjeong Choi
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, Korea.
| |
Collapse
|
20
|
Study of the tumor microenvironment during breast cancer progression. Cancer Cell Int 2017; 17:123. [PMID: 29299026 PMCID: PMC5741925 DOI: 10.1186/s12935-017-0492-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 12/02/2017] [Indexed: 12/12/2022] Open
Abstract
Background Different cells and mediators in the tumor microenvironment play important roles in the progression of breast cancer. The aim of this study was to determine the composition of the microenvironment during tumor progression in order to discover new related biomarkers and potentials for targeted therapy. Methods In this study, breast cancer biopsies from four different stages, and control breast biopsies were collected. Then, the mRNA expression of several markers related to different CD4+ T cell subsets including regulatory T cells (Treg), T helper (Th) type 1, 2 and 17 were determined. In addition, we investigated the expression of two inflammatory cytokines (TNF-α and IL-6) and inflammatory mediators including FASL, IDO, SOCS1, VEGF, and CCR7. Results The results showed that the expression of Th1 and Th17 genes was decreased in tumor tissues compared to control tissues. In addition, we found that the gene expression related to these two cell subsets decreased during cancer progression. Moreover, the expression level of TNF-α increased with tumor progression. Conclusion We conclude that the expression of genes related to immune response and inflammation is different between tumor tissues and control tissues. In addition, this difference was perpetuated through the different stages of cancer.
Collapse
|
21
|
Spectrum of spontaneous photon emission as a promising biophysical indicator for breast cancer research. Sci Rep 2017; 7:13083. [PMID: 29026159 PMCID: PMC5638945 DOI: 10.1038/s41598-017-13516-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 09/22/2017] [Indexed: 12/14/2022] Open
Abstract
In this study, we investigated the spectral characteristics of Spontaneous Photon Emission (SPE) from the body surface of a human breast cancer-bearing nude mice model during the overall growth process of breast cancers. By comparing and analyzing the data, we found that there was a striking difference between tumor mice and healthy controls in the spectral distribution of SPE from the body surface of lesion site, even when the morphological changes at the lesion site were not obvious. The spectral distribution of SPE from the healthy site of the tumor mice also differed from that of the healthy controls as the breast cancer developed to a certain stage. In addition, the difference in spectrum was related with different growth states of tumors. Interestingly, there was a positive correlation between the spectral ratio (610-630/395-455 nm) and the logarithm of the tumor volume for both the lesion site (R2 = 0.947; p < 0.001) and the normal site (R2 = 0.892; p < 0.001) of the tumor mice. The results suggested that the spectrum of SPE was sensitive to changes in the tumor status.
Collapse
|
22
|
Guo S, Jing Y, Burcus NI, Lassiter BP, Tanaz R, Heller R, Beebe SJ. Nano-pulse stimulation induces potent immune responses, eradicating local breast cancer while reducing distant metastases. Int J Cancer 2017; 142:629-640. [PMID: 28944452 DOI: 10.1002/ijc.31071] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 09/12/2017] [Accepted: 09/18/2017] [Indexed: 12/22/2022]
Abstract
Nano-pulse stimulation (NPS) as a developing technology has been studied for minimally invasive, nonthermal local cancer elimination for more than a decade. Here we show that a single NPS treatment results in complete regression of the poorly immunogenic, metastatic 4T1-Luc mouse mammary carcinoma. Impressively, spontaneous distant organ metastases were largely prevented, even in those animals with incomplete tumor regression. All tumor-free mice were protected from secondary tumor cell challenge, demonstrating a vaccine-like effect. NPS treatment induced antitumor immunity, long-term memory T cells, destruction of tumor microenvironment and reversal of the massive increase of immune suppressor cells in the tumor microenvironment and blood. NPS-treated 4T1 cells exhibited release of damage-associated molecular patterns (DAMPs), including calreticulin, HMGB1 and ATP, and activated dendritic cells. Those findings suggest that NPS is a potent immunogenic cell death inducer that elicits antitumor immunity to prevent distant metastases in addition to local tumor eradication.
Collapse
Affiliation(s)
- Siqi Guo
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, Virginia, 23508
| | - Yu Jing
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, Virginia, 23508
| | - Niculina I Burcus
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, Virginia, 23508
| | - Brittany P Lassiter
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, Virginia, 23508
| | - Royena Tanaz
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, Virginia, 23508
| | - Richard Heller
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, Virginia, 23508
| | - Stephen J Beebe
- Frank Reidy Research Center for Bioelectrics, Old Dominion University, Norfolk, Virginia, 23508
| |
Collapse
|
23
|
Morley ST, Walsh MT, Newport DT. Opportunities for Studying the Hydrodynamic Context for Breast Cancer Cell Spread Through Lymph Flow. Lymphat Res Biol 2017; 15:204-219. [PMID: 28749743 DOI: 10.1089/lrb.2017.0005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The lymphatic system serves as the primary route for the metastatic spread of breast cancer cells (BCCs). A scarcity of information exists with regard to the advection of BCCs in lymph flow and a fundamental understanding of the response of BCCs to the forces in the lymphatics needs to be established. This review summarizes the flow environment metastatic BCCs are exposed to in the lymphatics. Special attention is paid to the behavior of cells/particles in microflows in an attempt to elucidate the behavior of BCCs under lymph flow conditions (Reynolds number <1).
Collapse
Affiliation(s)
- Sinéad T Morley
- 1 Faculty of Science & Engineering, School of Engineering, Bernal Institute, University of Limerick , Limerick, Ireland
| | - Michael T Walsh
- 1 Faculty of Science & Engineering, School of Engineering, Bernal Institute, University of Limerick , Limerick, Ireland .,2 Health Research Institute, University of Limerick , Limerick, Ireland
| | - David T Newport
- 1 Faculty of Science & Engineering, School of Engineering, Bernal Institute, University of Limerick , Limerick, Ireland
| |
Collapse
|
24
|
Morley ST, Walsh MT, Newport DT. The advection of microparticles, MCF-7 and MDA-MB-231 breast cancer cells in response to very low Reynolds numbers. BIOMICROFLUIDICS 2017; 11:034105. [PMID: 28529671 PMCID: PMC5419862 DOI: 10.1063/1.4983149] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 04/26/2017] [Indexed: 05/05/2023]
Abstract
The lymphatic system is an extensive vascular network that serves as the primary route for the metastatic spread of breast cancer cells (BCCs). The dynamics by which BCCs travel in the lymphatics to distant sites, and eventually establish metastatic tumors, remain poorly understood. Particle tracking techniques were employed to analyze the behavior of MCF-7 and MDA-MB-231 BCCs which were exposed to lymphatic flow conditions in a 100 μm square microchannel. The behavior of the BCCs was compared to rigid particles of various diameters (η = dp/H= 0.05-0.32) that have been used to simulate cell flow in lymph. Parabolic velocity profiles were recorded for all particle sizes. All particles were found to lag the fluid velocity, the larger the particle the slower its velocity relative to the local flow (5%-15% velocity lag recorded). A distinct difference between the behavior of BCCs and particles was recorded. The BCCs travelled approximately 40% slower than the undisturbed flow, indicating that morphology and size affects their response to lymphatic flow conditions (Re < 1). BCCs adhered together, forming aggregates whose behavior was irregular. At lymphatic flow rates, MCF-7s were distributed uniformly across the channel in comparison to the MDA-MB-231 cells which travelled in the central region (88% of cells found within 0.35 ≤ W ≤ 0.64), indicating that metastatic MDA-MB-231 cells are subjected to a lower range of shear stresses in vivo. This suggests that both size and deformability need to be considered when modelling BCC behavior in the lymphatics. This finding will inform the development of in vitro lymphatic flow and metastasis models.
Collapse
Affiliation(s)
- Sinéad T Morley
- School of Engineering, Bernal Institute, Faculty of Science and Engineering, University of Limerick, Limerick, Ireland
| | | | - David T Newport
- School of Engineering, Bernal Institute, Faculty of Science and Engineering, University of Limerick, Limerick, Ireland
| |
Collapse
|
25
|
Arrebola JP, Fernández-Rodríguez M, Artacho-Cordón F, Garde C, Perez-Carrascosa F, Linares I, Tovar I, González-Alzaga B, Expósito J, Torne P, Fernández MF, Olea N. Associations of persistent organic pollutants in serum and adipose tissue with breast cancer prognostic markers. THE SCIENCE OF THE TOTAL ENVIRONMENT 2016; 566-567:41-49. [PMID: 27213669 DOI: 10.1016/j.scitotenv.2016.04.188] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 04/15/2016] [Accepted: 04/28/2016] [Indexed: 05/20/2023]
Abstract
This study aimed to evaluate associations between exposure to a group of persistent organic pollutants, measured in both adipose tissue and serum samples from breast cancer patients, and a set of tumor prognostic markers. The study population comprised 103 breast cancer patients recruited in Granada, Southern Spain. Data for tumor prognostic markers were retrieved from hospital clinical records and socio-demographic information was gathered by questionnaire. Persistent organic pollutants were quantified by gas chromatography with electron capture detection. Exposure levels were categorized in quartiles, and associations were evaluated using unconditional logistic regression. Adipose tissue HCB concentrations were associated positively with ER and PR expression (p-trends=0.044 and 0.005, respectively) and negatively with E-Cadherin and p53 expression (p-trends=0.012 and 0.027, respectively). PCB-180 adipose tissue concentrations were positively associated with HER2 expression (p-trend=0.036). Serum PCB-138 concentrations were positively associated with ER and PR expression (p-trends=0.052 and 0.042, respectively). The risk of p53 expression was higher among women in the lowest quartile of serum PCB-138 concentrations, but no significant trend was observed (p-trend=0.161). These findings indicate that human exposure to certain persistent organic pollutants might be related to breast cancer aggressiveness. We also highlight the influence on exposure assessment of the biological matrix selected, given that both serum and adipose tissue might yield relevant information on breast cancer prognosis.
Collapse
Affiliation(s)
- J P Arrebola
- Instituto de Investigación Biosanitaria (ibs. GRANADA), Hospitales Universitarios de Granada, Spain; Virgen de las Nieves University Hospital, Radiation Oncology Department, Oncology Unit, Granada, Spain; CIBER en Epidemiología y Salud Pública (CIBERESP), Spain.
| | - M Fernández-Rodríguez
- Instituto de Investigación Biosanitaria (ibs. GRANADA), Hospitales Universitarios de Granada, Spain; University of Granada, Radiology and Physical Medicine Department, Spain
| | - F Artacho-Cordón
- Instituto de Investigación Biosanitaria (ibs. GRANADA), Hospitales Universitarios de Granada, Spain; University of Granada, Radiology and Physical Medicine Department, Spain
| | - C Garde
- Instituto de Investigación Biosanitaria (ibs. GRANADA), Hospitales Universitarios de Granada, Spain
| | - F Perez-Carrascosa
- Instituto de Investigación Biosanitaria (ibs. GRANADA), Hospitales Universitarios de Granada, Spain; Virgen de las Nieves University Hospital, Radiation Oncology Department, Oncology Unit, Granada, Spain
| | - I Linares
- Instituto de Investigación Biosanitaria (ibs. GRANADA), Hospitales Universitarios de Granada, Spain; Virgen de las Nieves University Hospital, Radiation Oncology Department, Oncology Unit, Granada, Spain
| | - I Tovar
- Instituto de Investigación Biosanitaria (ibs. GRANADA), Hospitales Universitarios de Granada, Spain; Virgen de las Nieves University Hospital, Radiation Oncology Department, Oncology Unit, Granada, Spain
| | - B González-Alzaga
- Instituto de Investigación Biosanitaria (ibs. GRANADA), Hospitales Universitarios de Granada, Spain; Escuela Andaluza de Salud Pública, Granada, Spain
| | - J Expósito
- Instituto de Investigación Biosanitaria (ibs. GRANADA), Hospitales Universitarios de Granada, Spain; Virgen de las Nieves University Hospital, Radiation Oncology Department, Oncology Unit, Granada, Spain
| | - P Torne
- Instituto de Investigación Biosanitaria (ibs. GRANADA), Hospitales Universitarios de Granada, Spain
| | - M F Fernández
- Instituto de Investigación Biosanitaria (ibs. GRANADA), Hospitales Universitarios de Granada, Spain; CIBER en Epidemiología y Salud Pública (CIBERESP), Spain; University of Granada, Radiology and Physical Medicine Department, Spain
| | - N Olea
- Instituto de Investigación Biosanitaria (ibs. GRANADA), Hospitales Universitarios de Granada, Spain; CIBER en Epidemiología y Salud Pública (CIBERESP), Spain; University of Granada, Radiology and Physical Medicine Department, Spain
| |
Collapse
|
26
|
Lian C, Ruan L, Shang D, Wu Y, Lu Y, Lü P, Yang Y, Wei Y, Dong X, Ren D, Chen K, Liu H, Tu Z. Heparin-Binding Epidermal Growth Factor-Like Growth Factor as a Potent Target for Breast Cancer Therapy. Cancer Biother Radiopharm 2016; 31:85-90. [DOI: 10.1089/cbr.2015.1956] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Caixia Lian
- Institute of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Lingling Ruan
- School of Pharmacy, Jiangsu University, Zhenjiang, China
| | - Dongsheng Shang
- Institute of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Yanfang Wu
- Institute of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Yongjin Lu
- School of Pharmacy, Jiangsu University, Zhenjiang, China
| | - Peng Lü
- Institute of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Yuhua Yang
- School of Pharmacy, Jiangsu University, Zhenjiang, China
| | - Yajun Wei
- School of Pharmacy, Jiangsu University, Zhenjiang, China
| | - Xiaojing Dong
- School of Pharmacy, Jiangsu University, Zhenjiang, China
| | - Dewan Ren
- School of Pharmacy, Jiangsu University, Zhenjiang, China
| | - Keping Chen
- Institute of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Hanqing Liu
- School of Pharmacy, Jiangsu University, Zhenjiang, China
| | - Zhigang Tu
- Institute of Life Sciences, Jiangsu University, Zhenjiang, China
| |
Collapse
|
27
|
Goswami S, Sharma-Walia N. Osteoprotegerin secreted by inflammatory and invasive breast cancer cells induces aneuploidy, cell proliferation and angiogenesis. BMC Cancer 2015; 15:935. [PMID: 26608463 PMCID: PMC4660791 DOI: 10.1186/s12885-015-1837-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/19/2015] [Indexed: 12/12/2022] Open
Abstract
Background Osteoprotegerin (OPG) is a glycoprotein that has multifaceted role and is associated with several cancer malignancies like that of bladder carcinoma, gastric carcinoma, prostate cancer, multiple myeloma and breast cancer. Also OPG has been associated with several organ pathologies. The widespread expression of OPG suggests that OPG may have multiple biological activities that are yet to be explored. Methods The anchorage-independent sphere cultures of the adherent cells were instrumental in our study as it provided a deeper insight into the complexity of a 3D tumor. Cytokine profiling was performed for OPG’s detection in the microenvironment. ELISA and western blotting were performed to quantify the OPG secretion and measure the protein levels respectively. OPG expression was detected in human breast cancer tissue samples by IHC. To decipher OPG’s role in tumor aggressiveness both recombinant human OPG as well as OPG rich and depleted breast cancer cell conditioned media were tested. Western blotting and MTT assay were performed to detect changes in signaling pathways and proliferation that were induced in presence of OPG. Onset of aneuploidy, in presence of OPG, was measured by cell cycle analysis and western blotting. Finally, human Breast Cancer qBiomarker Copy Number PCR Array was used to detect how OPG remarkably induced gene copy numbers for oncogenic pathway regulators. Results SUM149PT and SUM1315M02 cells secrete high levels of the cytokine OPG compared to primary human mammary epithelial cells (HMEC). High expression of OPG was also detected in human breast cancer tissue samples compared to the uninvolved tissue from the same patient. OPG induced proliferation of control HMEC spheres and triggered the onset of aneuploidy in HMEC sphere cultures. OPG induced the expression of aneuploidy related kinases Aurora-A Kinase (IAK-1), Bub1 and BubR1 probably through the receptor activator of nuclear factor kappa-B ligand (RANKL) and syndecan-1 receptors via the Erk, AKT and GSK3(3 signaling pathway. Gene copy numbers for oncogenic pathway regulators such AKT1, Aurora-A Kinase (AURKA or IAK-1), epidermal growth factor receptor (EGFR) and MYC with a reduction in the copy numbers of cyclin dependent kinase inhibitor 2A (CDKN2A), PTEN and DNA topoisomerase 2 alpha (TOP2A) were induced in presence of OPG. Conclusions These results highlight the role of OPG in reprogramming normal mammary epithelial cells to a tumorigenic state and suggest promising avenues for treating inflammatory breast cancer as well as highly invasive breast cancer with new therapeutic targets. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1837-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sudeshna Goswami
- Department of Microbiology and Immunology, H. M. Bligh Cancer Research Laboratories, Chicago Medical School, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA.
| | - Neelam Sharma-Walia
- Department of Microbiology and Immunology, H. M. Bligh Cancer Research Laboratories, Chicago Medical School, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL, 60064, USA.
| |
Collapse
|
28
|
Artacho-Cordón F, Ríos-Arrabal S, Olivares-Urbano MA, Storch K, Dickreuter E, Muñoz-Gámez JA, León J, Calvente I, Torné P, Salinas MDM, Cordes N, Núñez MI. Valproic acid modulates radiation-enhanced matrix metalloproteinase activity and invasion of breast cancer cells. Int J Radiat Biol 2015; 91:946-56. [PMID: 26490761 DOI: 10.3109/09553002.2015.1087067] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
PURPOSE To evaluate matrix metalloproteinase (MMP) activity and invasion after ionizing radiation (IR) exposure and to determine whether MMP could be epigenetically modulated by histone deacetylase (HDAC) inhibition. MATERIAL AND METHODS Two human breast cancer cell lines (MDA-MB-231 and MCF-7) were cultured in monolayer (2D) and in laminin-rich extracellular matrix (3D). Invasion capability, collagenolytic and gelatinolytic activity, MMP and TIMP protein and mRNA expression and clonogenic survival were analyzed after IR exposure, with and without a HDAC inhibition treatment [1.5 mM valproic acid (VA) or 1 μM trichostatin-A (TSA)]. RESULTS IR exposure resulted in cell line-dependent stimulation of invasion capacity. In contrast to MCF-7 cells, irradiated MDA-MB-231 showed significantly enhanced mRNA expression of mmp-1, mmp-3 and mmp-13 and of their regulators timp-1 and timp-2 relative to unirradiated controls. This translated into increased collagenolytic and gelatinolytic activity and could be reduced after valproic acid (VA) treatment. Additionally, VA also mitigated IR-enhanced mmp and timp mRNA expression as well as IR-increased invasion capability. Finally, our data confirm the radiosensitizing effect of VA. CONCLUSION These results suggest that IR cell line-dependently induces upregulation of MMP mRNA expression, which appears to be mechanistically linked to a higher invasion capability that is modifiable by HDAC inhibition.
Collapse
Affiliation(s)
- Francisco Artacho-Cordón
- a Department of Radiology and Physical Medicine , University of Granada , Granada , Spain.,b Biosanitary Institute of Granada (ibs.GRANADA), University Hospitals of Granada/University of Granada , Granada , Spain
| | - Sandra Ríos-Arrabal
- a Department of Radiology and Physical Medicine , University of Granada , Granada , Spain.,b Biosanitary Institute of Granada (ibs.GRANADA), University Hospitals of Granada/University of Granada , Granada , Spain.,c Institute of Biopathology and Regenerative Medicine (IBIMER), University of Granada , Armilla, Granada , Spain
| | | | - Katja Storch
- d OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, and Helmholtz-Zentrum Dresden-Rossendorf , Dresden , Germany.,e Department of Radiation Oncology , University Hospital Carl Gustav Carus, Technische Universität Dresden , Dresden , Germany.,f German Cancer Consortium (DKTK), Dresden, Germany.,g German Cancer Research Center (DKFZ) , Heidelberg , Germany.,h Institute of Radiooncology, Helmholtz-Zentrum Dresden-Rossendorf , Dresden , Germany
| | - Ellen Dickreuter
- d OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, and Helmholtz-Zentrum Dresden-Rossendorf , Dresden , Germany.,e Department of Radiation Oncology , University Hospital Carl Gustav Carus, Technische Universität Dresden , Dresden , Germany.,f German Cancer Consortium (DKTK), Dresden, Germany.,g German Cancer Research Center (DKFZ) , Heidelberg , Germany.,h Institute of Radiooncology, Helmholtz-Zentrum Dresden-Rossendorf , Dresden , Germany
| | - José Antonio Muñoz-Gámez
- b Biosanitary Institute of Granada (ibs.GRANADA), University Hospitals of Granada/University of Granada , Granada , Spain.,i CIBER on hepatic and digestive diseases (CIBEREHD) , Spain
| | - Josefa León
- b Biosanitary Institute of Granada (ibs.GRANADA), University Hospitals of Granada/University of Granada , Granada , Spain.,i CIBER on hepatic and digestive diseases (CIBEREHD) , Spain
| | - Irene Calvente
- a Department of Radiology and Physical Medicine , University of Granada , Granada , Spain.,b Biosanitary Institute of Granada (ibs.GRANADA), University Hospitals of Granada/University of Granada , Granada , Spain
| | - Pablo Torné
- j General Surgery Management Unit, San Cecilio University Hospital , Granada , Spain
| | - María del Mar Salinas
- a Department of Radiology and Physical Medicine , University of Granada , Granada , Spain
| | - Nils Cordes
- d OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, and Helmholtz-Zentrum Dresden-Rossendorf , Dresden , Germany.,e Department of Radiation Oncology , University Hospital Carl Gustav Carus, Technische Universität Dresden , Dresden , Germany.,f German Cancer Consortium (DKTK), Dresden, Germany.,g German Cancer Research Center (DKFZ) , Heidelberg , Germany.,h Institute of Radiooncology, Helmholtz-Zentrum Dresden-Rossendorf , Dresden , Germany
| | - María Isabel Núñez
- a Department of Radiology and Physical Medicine , University of Granada , Granada , Spain.,b Biosanitary Institute of Granada (ibs.GRANADA), University Hospitals of Granada/University of Granada , Granada , Spain.,c Institute of Biopathology and Regenerative Medicine (IBIMER), University of Granada , Armilla, Granada , Spain
| |
Collapse
|
29
|
Penfornis P, Vallabhaneni KC, Whitt J, Pochampally R. Extracellular vesicles as carriers of microRNA, proteins and lipids in tumor microenvironment. Int J Cancer 2015; 138:14-21. [PMID: 25559768 DOI: 10.1002/ijc.29417] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Revised: 12/05/2014] [Accepted: 12/23/2014] [Indexed: 12/13/2022]
Abstract
In recent years, the knowledge about the control of tumor microenvironment has increased and emerged as an important player in tumorigenesis. The role of normal stromal cells in the tumor initiation and progression has brought our vision in to the forefront of cell-to-cell communication. In this review, we focus on the mechanism of communication between stromal and tumor cells, which is based on the exchange of extracellular vesicles (EVs). We describe several, evergrowing, pieces of evidence that EVs transfer messages through their miRNA, lipid, protein and nucleic acid contents. A better understanding of this sophisticated method of communication between normal cancer cells may lead to developing novel approaches for personalized diagnostics and therapeutics.
Collapse
Affiliation(s)
- Patrice Penfornis
- Department of Biochemistry and Cancer Institute, University of Mississippi Medical Center, Jackson, MS
| | - Krishna C Vallabhaneni
- Department of Biochemistry and Cancer Institute, University of Mississippi Medical Center, Jackson, MS
| | - Jason Whitt
- Department of Biochemistry and Cancer Institute, University of Mississippi Medical Center, Jackson, MS
| | - Radhika Pochampally
- Department of Biochemistry and Cancer Institute, University of Mississippi Medical Center, Jackson, MS
| |
Collapse
|
30
|
Kang N, Shah VH, Urrutia R. Membrane-to-Nucleus Signals and Epigenetic Mechanisms for Myofibroblastic Activation and Desmoplastic Stroma: Potential Therapeutic Targets for Liver Metastasis? Mol Cancer Res 2014; 13:604-12. [PMID: 25548101 DOI: 10.1158/1541-7786.mcr-14-0542] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 12/05/2014] [Indexed: 12/21/2022]
Abstract
Cancer-associated fibroblasts (CAFs), the most abundant cells in the tumor microenvironment (TME), are a key source of the extracellular matrix (ECM) that constitutes the desmoplastic stroma. Through remodeling of the reactive tumor stroma and paracrine actions, CAFs regulate cancer initiation, progression, and metastasis, as well as tumor resistance to therapies. The CAFs found in stroma-rich primary hepatocellular carcinomas (HCC) and liver metastases of primary cancers of other organs predominantly originate from hepatic stellate cells (HSTC), which are pericytes associated with hepatic sinusoids. During tumor invasion, HSTCs transdifferentiate into myofibroblasts in response to paracrine signals emanating from either tumor cells or a heterogeneous cell population within the hepatic tumor microenvironment. Mechanistically, HSTC-to-myofibroblast transdifferentiation, also known as, HSTC activation, requires cell surface receptor activation, intracellular signal transduction, gene transcription, and epigenetic signals, which combined ultimately modulate distinct gene expression profiles that give rise to and maintain a new phenotype. The current review defines a paradigm that explains how HSTCs are activated into CAFs to promote liver metastasis. Furthermore, a focus on the most relevant intracellular signaling networks and epigenetic mechanisms that control HSTC activation is provided. Finally, we discuss the feasibility of targeting CAF/activated HSTCs, in isolation or in conjunction with targeting cancer cells, which constitutes a promising and viable therapeutic approach for the treatment of primary stroma-rich liver cancers and liver metastasis.
Collapse
Affiliation(s)
- Ningling Kang
- Tumor Microenvironment and Metastasis Section, The Hormel Institute, University of Minnesota, Austin, Minnesota.
| | - Vijay H Shah
- GI Research Unit, Division of Gastroenterology and Hepatology, Epigenomics Translational Program, Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota
| | - Raul Urrutia
- GI Research Unit, Division of Gastroenterology and Hepatology, Epigenomics Translational Program, Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
31
|
Cunha AD, Michelin MA, Murta EFC. Pattern response of dendritic cells in the tumor microenvironment and breast cancer. World J Clin Oncol 2014; 5:495-502. [PMID: 25114862 PMCID: PMC4127618 DOI: 10.5306/wjco.v5.i3.495] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2013] [Revised: 03/07/2014] [Accepted: 05/29/2014] [Indexed: 02/06/2023] Open
Abstract
Breast cancer (BC) is the most common malignant neoplasm and the cause of death by cancer among women worldwide. Its development, including malignancy grade and patient prognosis, is influenced by various mutations that occur in the tumor cell and by the immune system’s status, which has a direct influence on the tumor microenvironment and, consequently, on interactions with non-tumor cells involved in the immunological response. Among the immune response cells, dendritic cells (DCs) play a key role in the induction and maintenance of anti-tumor responses owing to their unique abilities for antigen cross-presentation and promotion of the activation of specific lymphocytes that target neoplasic cells. However, the tumor microenvironment can polarize DCs, transforming them into immunosuppressive regulatory DCs, a tolerogenic phenotype which limits the activity of effector T cells and supports tumor growth and progression. Various factors and signaling pathways have been implicated in the immunosuppressive functioning of DCs in cancer, and researchers are working on resolving processes that can circumvent tumor escape and developing viable therapeutic interventions to prevent or reverse the expression of immunosuppressive DCs in the tumor microenvironment. A better understanding of the pattern of DC response in patients with BC is fundamental to the development of specific therapeutic approaches to enable DCs to function properly. Various studies examining DCs immunotherapy have demonstrated its great potential for inducing immune responses to specific antigens and thereby reversing immunosuppression and related to clinical response in patients with BC. DC-based immunotherapy research has led to immense scientific advances, both in our understanding of the anti-tumor immune response and for the treatment of these patients.
Collapse
|
32
|
Hockemeyer K, Janetopoulos C, Terekhov A, Hofmeister W, Vilgelm A, Costa L, Wikswo JP, Richmond A. Engineered three-dimensional microfluidic device for interrogating cell-cell interactions in the tumor microenvironment. BIOMICROFLUIDICS 2014; 8:044105. [PMID: 25379090 PMCID: PMC4189212 DOI: 10.1063/1.4890330] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Accepted: 07/04/2014] [Indexed: 05/24/2023]
Abstract
Stromal cells in the tumor microenvironment play a key role in the metastatic properties of a tumor. It is recognized that cancer-associated fibroblasts (CAFs) and endothelial cells secrete factors capable of influencing tumor cell migration into the blood or lymphatic vessels. We developed a microfluidic device that can be used to image the interactions between stromal cells and tumor cell spheroids in a three dimensional (3D) microenvironment while enabling external control of interstitial flow at an interface, which supports endothelial cells. The apparatus couples a 200-μm channel with a semicircular well to mimic the interface of a blood vessel with the stroma, and the design allows for visualization of the interactions of interstitial flow, endothelial cells, leukocytes, and fibroblasts with the tumor cells. We observed that normal tissue-associated fibroblasts (NAFs) contribute to the "single file" pattern of migration of tumor cells from the spheroid in the 3D microenvironment. In contrast, CAFs induce a rapid dispersion of tumor cells out of the spheroid with migration into the 3D matrix. Moreover, treatment of tumor spheroid cultures with the chemokine CXCL12 mimics the effect of the CAFs, resulting in similar patterns of dispersal of the tumor cells from the spheroid. Conversely, addition of CXCL12 to co-cultures of NAFs with tumor spheroids did not mimic the effects observed with CAF co-cultures, suggesting that NAFs produce factors that stabilize the tumor spheroids to reduce their migration in response to CXCL12.
Collapse
Affiliation(s)
| | | | - A Terekhov
- Center for Laser Applications, University of Tennessee Space Institute , Tullahoma, Tennessee 37388-9700, USA
| | | | | | | | | | | |
Collapse
|
33
|
Song B, Kim B, Choi SH, Song KY, Chung YG, Lee YS, Park G. Mesenchymal stromal cells promote tumor progression in fibrosarcoma and gastric cancer cells. KOREAN JOURNAL OF PATHOLOGY 2014; 48:217-24. [PMID: 25013420 PMCID: PMC4087135 DOI: 10.4132/koreanjpathol.2014.48.3.217] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Revised: 05/29/2014] [Accepted: 05/30/2014] [Indexed: 01/10/2023]
Abstract
Background Extensive evidence has accumulated regarding the role of mesenchymal stromal cells (MSCs) in tumor progression, but the exact effects and mechanisms underlying this role remain unclear. We investigated the effects of MSC-associated tumor progression in MSC-sarcoma models and a gastric cancer metastatic model. Methods We conducted an in vitro growth kinetics assay and an in vivo tumor progression assay for sarcoma cells and gastric cancer cells in the presence or absence of MSCs. Results MSC-cocultured human fibrosarcoma cells (HT1080) showed accelerated growth compared with HT1080 alone (79- vs 37-fold change, p<.050). For HT1080, human MSC-coinjected tumors showed significantly greater and highly infiltrative growth compared to those of HT1080 alone (p=.035). For mouse fibrosarcoma cells (WEHI164), mouse MSC-coinjected tumors had greater volume than those of WEHI164 alone (p=.141). For rat sarcoma cells (RR1022), rat MSC-coinjected tumors exhibited greater volume and infiltrative growth than those of RR1022 alone (p=.050). For human gastric cancer cells (5FU), tumors of 5FU alone were compact, nodular in shape, and expansile with good demarcation and no definite lung metastatic nodules, whereas tumors grown in the presence of human MSCs showed highly desmoplastic and infiltrative growth and multiple lung metastasis. Conclusions We observed morphological evidence for MSC-associated tumor progression of fibrosarcomas and gastric cancer cells.
Collapse
Affiliation(s)
- Byunghoo Song
- Department of Pathology, Seoul St. Mary's Hospital, The Catholic University of Korea College of Medicine, Seoul, Korea. ; Cancer Research Institute, Seoul St. Mary's Hospital, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Bokyung Kim
- Department of Pathology, Seoul St. Mary's Hospital, The Catholic University of Korea College of Medicine, Seoul, Korea. ; Cancer Research Institute, Seoul St. Mary's Hospital, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Se-Ha Choi
- Cancer Research Institute, Seoul St. Mary's Hospital, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Kyo Young Song
- Department of Surgery, Seoul St. Mary's Hospital, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Yang-Guk Chung
- Department of Orthopedic Surgery, Seoul St. Mary's Hospital, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Youn-Soo Lee
- Department of Pathology, Seoul St. Mary's Hospital, The Catholic University of Korea College of Medicine, Seoul, Korea
| | - Gyeongsin Park
- Department of Pathology, Seoul St. Mary's Hospital, The Catholic University of Korea College of Medicine, Seoul, Korea. ; Cancer Research Institute, Seoul St. Mary's Hospital, The Catholic University of Korea College of Medicine, Seoul, Korea
| |
Collapse
|
34
|
Gu YQ, Gong G, Xu ZL, Wang LY, Fang ML, Zhou H, Xing H, Wang KR, Sun L. miRNA profiling reveals a potential role of milk stasis in breast carcinogenesis. Int J Mol Med 2014; 33:1243-9. [PMID: 24584717 DOI: 10.3892/ijmm.2014.1677] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 02/17/2014] [Indexed: 11/06/2022] Open
Abstract
The tumor microenvironment plays an important role in breast carcinogenesis. Milk acts as an important microenvironment of breast cancer, but its role in breast carcinogenesis is largely unknown. Milk stasis may exist in the breast for a number of years after breastfeeding. In the present study, we reported the first microRNA (miRNA) profiling of milk from patients with milk stasis. We identified 266 known miRNAs and 271 novel miRNAs in 10 milk stasis only samples, 271 known miRNAs and 140 novel miRNAs in 10 milk stasis plus breast neoplasm samples by deep sequencing. miRNA profiles were different between the two groups. Furthermore, nine tumor suppressor miRNAs such as miR-29a, miR-146 and miR-223 were significantly downregulated, while seven oncogenic miRNAs such as miR-451, miR-486, miR-107, miR-92 and miR-10 were significantly upregulated in the milk of milk stasis plus neoplasm patients. Three of the identified miRNAs (miR-140, miR-21 and let-7a) were selected using real-time PCR, confirming that these miRNAs were highly expressed. The results also showed that the three miRNAs detected were more abundant in the milk than in the blood. In summary, the data suggested that miRNAs in milk from milk stasis patients may contribute to breast carcinogenesis and that they are more sensitive biomarkers for breast cancer than miRNAs in the blood.
Collapse
Affiliation(s)
- Yi-Qi Gu
- Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Gu Gong
- Spine Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Zhe-Li Xu
- Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Li-Ying Wang
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Ming-Li Fang
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Hui Zhou
- Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Hua Xing
- Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Ke-Ren Wang
- Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Liang Sun
- Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| |
Collapse
|
35
|
Gromov P, Gromova I, Olsen CJ, Timmermans-Wielenga V, Talman ML, Serizawa RR, Moreira JM. Tumor interstitial fluid — A treasure trove of cancer biomarkers. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2013; 1834:2259-70. [DOI: 10.1016/j.bbapap.2013.01.013] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Revised: 01/09/2013] [Accepted: 01/14/2013] [Indexed: 12/11/2022]
|
36
|
Zhou ZN, Sharma VP, Beaty BT, Roh-Johnson M, Peterson EA, Van Rooijen N, Kenny PA, Wiley HS, Condeelis JS, Segall JE. Autocrine HBEGF expression promotes breast cancer intravasation, metastasis and macrophage-independent invasion in vivo. Oncogene 2013; 33:3784-93. [PMID: 24013225 PMCID: PMC3950352 DOI: 10.1038/onc.2013.363] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Revised: 06/27/2013] [Accepted: 07/16/2013] [Indexed: 01/04/2023]
Abstract
Increased expression of HBEGF in ER negative breast tumors is correlated with enhanced metastasis to distant organ sites and more rapid disease recurrence upon removal of the primary tumor. Our previous work has demonstrated a paracrine loop between breast cancer cells and macrophages in which the tumor cells are capable of stimulating macrophages through the secretion of CSF-1 while the tumor associated macrophages (TAMs) in turn aid in tumor cell invasion by secreting EGF. To determine how the autocrine expression of EGFR ligands by carcinoma cells would affect this paracrine loop mechanism, and in particular whether tumor cell invasion depends on spatial ligand gradients generated by TAMs, we generated cell lines with increased HBEGF expression. We find that autocrine HBEGF expression enhanced in vivo intravasation and metastasis, and resulted in a novel phenomenon in which macrophages were no longer required for in vivo invasion of breast cancer cells. In vitro studies revealed that expression of HBEGF enhanced invadopodium formation, thus providing a mechanism for cell autonomous invasion. The increased invadopodium formation was directly dependent on EGFR signaling, as demonstrated by a rapid decrease in invadopodia upon inhibition of autocrine HBEGF/EGFR signaling as well as inhibition of signaling downstream of EGFR activation. HBEGF expression also resulted in enhanced invadopodium function via upregulation of MMP2 and MMP9 expression. We conclude that high levels of HBEGF expression can short-circuit the tumor cell/macrophage paracrine invasion loop, resulting in enhanced tumor invasion that is independent of macrophage signaling.
Collapse
Affiliation(s)
- Z N Zhou
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - V P Sharma
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - B T Beaty
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - M Roh-Johnson
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - E A Peterson
- Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - N Van Rooijen
- Department of Molecular Cell Biology, Free University Medical Center, Amsterdam, The Netherlands
| | - P A Kenny
- Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - H S Wiley
- 1] Systems Biology Program, Pacific Northwest National Laboratory, Richland, WA, USA [2] Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA, USA
| | - J S Condeelis
- 1] Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY, USA [2] Gruss Lipper Center for Biophotonics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - J E Segall
- 1] Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY, USA [2] Gruss Lipper Center for Biophotonics, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
37
|
Ríos-Arrabal S, Artacho-Cordón F, León J, Román-Marinetto E, del Mar Salinas-Asensio M, Calvente I, Núñez MI. Involvement of free radicals in breast cancer. SPRINGERPLUS 2013; 2:404. [PMID: 24024092 PMCID: PMC3765596 DOI: 10.1186/2193-1801-2-404] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Accepted: 08/22/2013] [Indexed: 12/14/2022]
Abstract
Researchers have recently shown an increased interest in free radicals and their role in the tumor microenvironment. Free radicals are molecules with high instability and reactivity due to the presence of an odd number of electrons in the outermost orbit of their atoms. Free radicals include reactive oxygen and nitrogen species, which are key players in the initiation and progression of tumor cells and enhance their metastatic potential. In fact, they are now considered a hallmark of cancer. However, both reactive species may contribute to improve the outcomes of radiotherapy in cancer patients. Besides, high levels of reactive oxygen species may be indicators of genotoxic damage in non-irradiated normal tissues. The purpose of this article is to review recent research on free radicals and carcinogenesis in order to understand the pathways that contribute to tumor malignancy. This review outlines the involvement of free radicals in relevant cellular events, including their effects on genetic instability through (growth factors and tumor suppressor genes, their enhancement of mitogenic signals, and their participation in cell remodeling, proliferation, senescence, apoptosis, and autophagy processes; the possible relationship between free radicals and inflammation is also explored. This knowledge is crucial for evaluating the relevance of free radicals as therapeutic targets in cancer.
Collapse
Affiliation(s)
- Sandra Ríos-Arrabal
- />Departamento de Radiología y Medicina Física, Universidad de Granada, Av. Madrid s/n, 18012 Granada, Spain
| | - Francisco Artacho-Cordón
- />Departamento de Radiología y Medicina Física, Universidad de Granada, Av. Madrid s/n, 18012 Granada, Spain
- />Instituto de Investigación Biosanitaria de Granada, Granada, Spain
| | - Josefa León
- />Ciber de Enfermedades Hepáticas y Digestivas CIBERehd, Granada, Spain
| | - Elisa Román-Marinetto
- />Departamento de Radiología y Medicina Física, Universidad de Granada, Av. Madrid s/n, 18012 Granada, Spain
| | | | - Irene Calvente
- />Departamento de Radiología y Medicina Física, Universidad de Granada, Av. Madrid s/n, 18012 Granada, Spain
| | - Maria Isabel Núñez
- />Departamento de Radiología y Medicina Física, Universidad de Granada, Av. Madrid s/n, 18012 Granada, Spain
- />Instituto de Investigación Biosanitaria de Granada, Granada, Spain
- />Instituto de Biopatología y Medicina Regenerativa (IBIMER), Universidad de Granada, Av. Conocimiento, s/n, 18100 Armilla Granada, Spain
| |
Collapse
|
38
|
Rong G, Kang H, Wang Y, Hai T, Sun H. Candidate markers that associate with chemotherapy resistance in breast cancer through the study on Taxotere-induced damage to tumor microenvironment and gene expression profiling of carcinoma-associated fibroblasts (CAFs). PLoS One 2013; 8:e70960. [PMID: 23951052 PMCID: PMC3738633 DOI: 10.1371/journal.pone.0070960] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Accepted: 06/24/2013] [Indexed: 11/18/2022] Open
Abstract
Recently, emerging evidence has suggested that carcinoma-associated fibroblasts (CAFs) could contribute to chemotherapy resistances in breast cancer treatment. The aim of this study is to compare the gene expression profiling of CAFs before and after chemotherapy and pick up candidate genes that might associate with chemotherapy resistance and could be used as predictors of treatment response. CAFs were cultured from surgically resected primary breast cancers and identified with immunohistochemistry (IHC) and Flow cytometry (FCM). MDA-MB-231 cells were cultured as the breast cancer cell line. Cell adhesion assay, invasion assay, and proliferation assay (MTT) were performed to compare the function of MDA-MB-231 cells co-cultured with CAFs and MDA-MB-231 cells without co-culture, after chemotherapy. Totally 6 pairs of CAFs were prepared for microarray analysis. Each pair of CAFs were obtained from the same patient and classified into two groups. One group was treated with Taxotere (regarded as after chemotherapy) while the other group was not processed with Taxotere (regarded as before chemotherapy). According to our study, the primary-cultured CAFs exhibited characteristic phenotype. After chemotherapy, MDA-MB-231 cells co-cultured with CAFs displayed increasing adhesion, invasiveness and proliferation abilities, compared with MDA-MB-231 cells without CAFs. Moreover, 35 differentially expressed genes (absolute fold change >2) were identified between CAFs after chemotherapy and before chemotherapy, including 17 up-regulated genes and 18 down-regulated genes. CXCL2, MMP1, IL8, RARRES1, FGF1, and CXCR7 were picked up as the candidate markers, of which the differential expression in CAFs before and after chemotherapy was confirmed. The results indicate the changes of gene expression in CAFs induced by Taxotere treatment and propose the candidate markers that possibly associate with chemotherapy resistance in breast cancer.
Collapse
Affiliation(s)
- Guohua Rong
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, P. R. China
| | - Hua Kang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, P. R. China
- * E-mail:
| | - Yajun Wang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, P. R. China
| | - Tao Hai
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, P. R. China
| | - Haichen Sun
- Surgery Lab, Xuanwu Hospital, Capital Medical University, Beijing, P. R. China
| |
Collapse
|
39
|
Aluminium, carbonyls and cytokines in human nipple aspirate fluids: Possible relationship between inflammation, oxidative stress and breast cancer microenvironment. J Inorg Biochem 2013; 128:250-6. [PMID: 23916117 DOI: 10.1016/j.jinorgbio.2013.07.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 07/02/2013] [Accepted: 07/05/2013] [Indexed: 12/17/2022]
Abstract
The human breast is likely exposed to Al (aluminium) from many sources including diet and personal care products. Underarm applications of aluminium salt-based antiperspirant provide a possible long-term source of exposure, especially after underarm applications to shaved and abraded skin. Al research in breast fluids likely reflects the intraductal microenvironment. We found increased levels of aluminium in noninvasively collected nipple aspirate fluids (NAF) from 19 breast cancer patients compared with 16 healthy control subjects (268 vs 131 μg/l, respectively; p < 0.0001). In the same NAF samples we found significantly increased levels of protein oxidative carbonyls in cancer patients compared to healthy women (2.35 vs 0.41 nmol/mg prot, respectively; p < 0.0001). Aluminium content and carbonyl levels showed a significant positive linear correlation (r(2) 0.6628, p < 0.0001). In cancer NAF samples (containing higher amounts of aluminium salts) we also found a significantly increased levels of pro-inflammatory cytokines (IL-1β, IL-6, IL-12 p70, and TNF-α) and chemoattractant CC and CXC chemokines (IL-8, MIP-1α and MCP-1). In 12 invasive cancer NAF samples we found a significant positive linear correlation among aluminium, carbonyls and pro-inflammatory IL-6 cytokine (Y = 64.79x-39.63, r(2) 0.8192, p < 0.0005), as well as pro-inflammatory monocyte chemoattractant MCP-1 cytokine (Y = 2026x-866, r(2) 0.9495, p < 0.0001). In addition to emerging evidence, our results support the possible involvement of aluminium ions in oxidative and inflammatory status perturbations of breast cancer microenvironment, suggesting aluminium accumulation in breast microenvironment as a possible risk factor for oxidative/inflammatory phenotype of breast cells.
Collapse
|
40
|
Abstract
Combinatorial approaches that integrate conventional pathology with genomic profiling and functional genomics have begun to enhance our understanding of the genetic basis of breast cancer. These methods have identified key genotypic-phenotypic correlations in different breast cancer subtypes that have led to the discovery of genetic dependencies that drive their behavior. Moreover, this knowledge has been applied to define novel tailored therapies for these groups of patients with cancer. With the current emphasis on characterizing the mutational repertoire of breast cancers by next-generation sequencing, the question remains as to what constitutes a driver event. By focusing efforts on homogenous subgroups of breast cancer and integrating orthogonal data-types combined with functional approaches, we can begin to unravel the heterogeneity and identify aberrations that can be therapeutically targeted.
Collapse
Affiliation(s)
- Rachael Natrajan
- Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, United Kingdom.
| | | |
Collapse
|
41
|
Woo SM, Choi YK, Cho SG, Park S, Ko SG. A New Herbal Formula, KSG-002, Suppresses Breast Cancer Growth and Metastasis by Targeting NF- κ B-Dependent TNF α Production in Macrophages. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2013; 2013:728258. [PMID: 23818931 PMCID: PMC3683439 DOI: 10.1155/2013/728258] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Revised: 03/08/2013] [Accepted: 05/09/2013] [Indexed: 12/21/2022]
Abstract
Tumor-associated macrophages (TAMs) in tumor microenvironment regulate cancer progression and metastases. In breast cancer, macrophage infiltration is correlated with a poor prognosis. While metastatic breast cancer is poor prognostic with a severe mortality, therapeutic options are still limited. In this study, we demonstrate that KSG-002, a new herbal composition of radices Astragalus membranaceus and Angelica gigas, suppresses breast cancer via inhibiting TAM recruitment. KSG-002, an extract of radices Astragalus membranaceus and Angelica gigas at 3 : 1 ratio, respectively, inhibited MDA-MB-231 xenograft tumor growth and pulmonary metastasis in nude mice, while KSG-001, another composition (1 : 1 ratio, w/w), enhanced tumor growth, angiogenesis, and pulmonary metastasis, in vivo. KSG-002 further decreased the infiltrated macrophage numbers in xenograft tumor cohorts. In Raw264.7 cells, KSG-002 but not KSG-001 inhibited cell proliferation and migration and reduced TNF-alpha (TNF α ) production by inhibiting NF- κ B pathway. Furthermore, a combinatorial treatment of KSG-002 with TNF α inhibited a proliferation and migration of both MDA-MB-231 and Raw264.7 cells. Taken together, we conclude that KSG-002 suppresses breast cancer growth and metastasis through targeting NF- κ B-mediated TNF α production in macrophages.
Collapse
Affiliation(s)
- Sang-Mi Woo
- Laboratory of Clinical Biology and Pharmacogenomics, Department of Preventive Medicine, Kyung Hee University, 1 Hoegi, Seoul 130-701, Republic of Korea
| | - Youn Kyung Choi
- Laboratory of Clinical Biology and Pharmacogenomics, Department of Preventive Medicine, Kyung Hee University, 1 Hoegi, Seoul 130-701, Republic of Korea
| | - Sung-Gook Cho
- Laboratory of Clinical Biology and Pharmacogenomics, Department of Preventive Medicine, Kyung Hee University, 1 Hoegi, Seoul 130-701, Republic of Korea
| | - Sunju Park
- Center for Clinical Research and Genomics, Department of Preventive Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea
| | - Seong-Gyu Ko
- Laboratory of Clinical Biology and Pharmacogenomics, Department of Preventive Medicine, Kyung Hee University, 1 Hoegi, Seoul 130-701, Republic of Korea
- Center for Clinical Research and Genomics, Department of Preventive Medicine, Kyung Hee University, Seoul 130-701, Republic of Korea
| |
Collapse
|
42
|
Artacho-Cordón F, Ríos-Arrabal S, Lara PC, Artacho-Cordón A, Calvente I, Núñez MI. Matrix metalloproteinases: potential therapy to prevent the development of second malignancies after breast radiotherapy. Surg Oncol 2012; 21:e143-51. [PMID: 22749313 DOI: 10.1016/j.suronc.2012.06.001] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Revised: 05/25/2012] [Accepted: 06/02/2012] [Indexed: 12/24/2022]
Abstract
Radiotherapy is widely used in the treatment of patients with breast cancer, but ionizing radiation-induced carcinogenesis has been described in several studies. Matrix metalloproteinases (MMPs) are a wide family of proteases secreted by tumour and microenvironmental cells that are directly linked with invasion and metastasis through complete extracellular matrix (ECM) breakage. In the past decade, MMPs have been associated with other carcinogenesis steps, including tumour growth and angiogenesis promotion. Moreover, in vitro studies have demonstrated an enhanced migration, invasiveness, and angiogenic ability of cancer cells after radiation exposure through an increase in MMP activity. These findings are consistent with clinical observations of breast cancer metastases raised in bone, lung and brain tissues after radiotherapy. The aim of this review was to analyse the current state of research on MMPs and report new insights into the potential of MMP-targeted therapy in combination with radiotherapy to decrease the risk of radiation-induced second malignancies and to improve the overall survival of breast cancer patients.
Collapse
Affiliation(s)
- F Artacho-Cordón
- Departamento de Radiología y Medicina Física, Universidad de Granada, Av. Madrid s/n, 18012 Granada, Spain
| | | | | | | | | | | |
Collapse
|
43
|
Abstract
The microenvironment of breast cancer comprises predominantly of adipocytes. Adipocytes drive cancer progression through the secretion adipocytokines. Adipocytes induce epithelial mesenchymal transition of breast cancer cells through paracrine IL-6/Stat3 signalling. Treatment approaches that can target adipocytes in the microenvironment and abrogate paracrine signals that drive breast cancer growth and metastasis are urgently needed. Repositioning of old drugs has become an effective approach for discovering new cancer drugs. In this study, niclosamide, an FDA approved anthelminthic drug was evaluated for its anti-breast cancer activity and its ability to inhibit adipocytes induced EMT. Niclosamide potently inhibited proliferation, migration and invasion at low concentration and induced significant apoptosis at high concentrations in human breast cancer cell lines MDA-MB-468 and MCF-7. Additionally, niclosamide reversed adipocyte-induced EMT with a correlated inhibition of IL-6/Stat3 activation and downregulation of EMT-TFs TWIST and SNAIL. Moreover, niclosamide markedly impaired MDA-MB-468 and MCF-7 migration and invasion. We further found that the inhibitory effects of niclosamide on MDA-MB-468 and MCF-7 motility was closely related to destabilization of focal adhesion complex formation. With decreased co-localization of focal adhesion kinase (FAK) and phosphorylated paxillin (pPAX). Collectively, these results demonstrate that niclosamide could be used to inhibit adipocyte-induced breast cancer growth and metastasis.
Collapse
|