1
|
Yin P, Jiang H, Ji X, Xia L, Su Z, Tian Y. Soluble TREM2 drives triple-negative breast cancer progression via activation of the AKT pathway. Int Immunopharmacol 2024; 145:113750. [PMID: 39672020 DOI: 10.1016/j.intimp.2024.113750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 12/15/2024]
Abstract
Triggering receptor expressed on myeloid cells 2 (TREM2) plays a key role in immune regulation, particularly within tumor-associated macrophages (TAMs). In triple-negative breast cancer (TNBC), TREM2+ TAMs have been shown to modulate the tumor microenvironment, but the role of its soluble form: soluble triggering receptor expressed on myeloid cells 2 (sTREM2), produced through proteolytic cleavage, remains unclear. In this study, we investigated the effects of sTREM2 on TNBC progression. In vitro, treatment of TNBC cells with recombinant sTREM2 or sTREM2-containing culture supernatant significantly enhanced cell proliferation, invasion, and migration. These effects were further confirmed by the use of TREM2-neutralizing antibodies, which abrogated sTREM2's tumor-promoting activities. In vivo, peri-tumoral injections of recombinant sTREM2 led to a notable acceleration of tumor growth in mouse models. Mechanistically, we found that the effects of sTREM2 were mediated through its binding to the TG2 protein in 4T1 cells, thereby activating the AKT signaling pathway. Collectively, our findings suggest that sTREM2 drives TNBC progression by enhancing critical tumor cell functions, positioning it as a potential therapeutic target for TNBC treatment.
Collapse
Affiliation(s)
- Peng Yin
- Institute of Hematological Disease, Jiangsu University, Zhenjiang 212001, China; School of Life Sciences, Jiangsu University, Zhenjiang 212013, China
| | - Haiqiang Jiang
- Department of Laboratory Medicine, Jiangyin Hospital of Traditional Chinese Medicine, Wuxi 214499, China
| | - Xiaoyun Ji
- Institute of Hematological Disease, Jiangsu University, Zhenjiang 212001, China; School of Life Sciences, Jiangsu University, Zhenjiang 212013, China
| | - Lin Xia
- Institute of Hematological Disease, Jiangsu University, Zhenjiang 212001, China; School of Life Sciences, Jiangsu University, Zhenjiang 212013, China
| | - Zhaoliang Su
- Institute of Hematological Disease, Jiangsu University, Zhenjiang 212001, China; School of Life Sciences, Jiangsu University, Zhenjiang 212013, China
| | - Yu Tian
- Institute of Hematological Disease, Jiangsu University, Zhenjiang 212001, China; School of Life Sciences, Jiangsu University, Zhenjiang 212013, China.
| |
Collapse
|
2
|
Dong Y, Zhang J, Xie A, Yue X, Li M, Zhou Q. Garlic peel extract as an antioxidant inhibits triple-negative breast tumor growth and angiogenesis by inhibiting cyclooxygenase-2 expression. Food Sci Nutr 2024; 12:6886-6895. [PMID: 39554336 PMCID: PMC11561839 DOI: 10.1002/fsn3.4320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 06/23/2024] [Accepted: 06/26/2024] [Indexed: 11/19/2024] Open
Abstract
Garlic peels are frequently disposed of as agro-waste; their bioactivity and physiological activity for health benefits and disease protection are neglected. This study aims to examine the potential inhibitory effects of garlic peel extract as an antioxidant on 4 T1 triple-negative breast cancer (TNBC) tumors in mice. The bioactive constituents of garlic peel were identified through HPLC-MS/MS analysis, while the antioxidant properties of garlic peel extract were assessed using peroxyl radical scavenging capacity (PSC) and cellular antioxidant activity (CAA) assays. Subsequently, the inhibitory effects of garlic peel extract on 4T1 tumor growth were evaluated using a 4T1 model. The results showed that 433 polyphenol compounds were found in garlic peel extract; among them, flavonoids and phenolic acid are the primary polyphenols with natural antioxidant activity, and both high and low concentrations of the extract exhibited tumor-suppressive effects. Immunohistochemistry was employed to assess the expression levels of COX-2, CD31, VEGFA, MMP2, and MMP9 in tumor tissues in order to investigate the antioxidant properties of garlic peel extract, specifically its ability to suppress COX-2 expression. The findings of this study offer a foundation for the advancement of garlic peel-based functional products and contribute to the identification of potential anti-cancer agents and therapeutic targets.
Collapse
Affiliation(s)
- Yushi Dong
- College of Food ScienceShenyang Agricultural UniversityShenyangChina
| | - Jiyue Zhang
- Spice and Beverage Research Institute, Chinese Academy of Tropical Agricultural SciencesWanningChina
| | - Aijun Xie
- Department of Chemical and Biomolecular EngineeringNational University of SingaporeSingaporeSingapore
| | - Xiqing Yue
- College of Food ScienceShenyang Agricultural UniversityShenyangChina
| | - Mohan Li
- College of Food ScienceShenyang Agricultural UniversityShenyangChina
| | - Qian Zhou
- College of Food ScienceShenyang Agricultural UniversityShenyangChina
| |
Collapse
|
3
|
Shen GY, Yang PJ, Zhang WS, Chen JB, Tian QY, Zhang Y, Han B. Identification of a Prognostic Gene Signature Based on Lipid Metabolism-Related Genes in Esophageal Squamous Cell Carcinoma. Pharmgenomics Pers Med 2023; 16:959-972. [PMID: 38023824 PMCID: PMC10631388 DOI: 10.2147/pgpm.s430786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Background Dysregulation of lipid metabolism is common in cancer. However, the molecular mechanism underlying lipid metabolism in esophageal squamous cell carcinoma (ESCC) and its effect on patient prognosis are not well understood. The objective of our study was to construct a lipid metabolism-related prognostic model to improve prognosis prediction in ESCC. Methods We downloaded the mRNA expression profiles and corresponding survival data of patients with ESCC from the Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) databases. We performed differential expression analysis to identify differentially expressed lipid metabolism-related genes (DELMGs). We used Univariate Cox regression and least absolute shrinkage and selection operator (LASSO) analyses to establish a risk model in the GEO cohort and used data of patients with ESCC from the TCGA cohort for validation. We also explored the relationship between the risk model and the immune microenvironment via infiltrated immune cells and immune checkpoints. Results The result showed that 132 unique DELMGs distinguished patients with ESCC from the controls. We identified four genes (ACAA1, ACOT11, B4GALNT1, and DDHD1) as prognostic gene expression signatures to construct a risk model. Patients were classified into high- and low-risk groups as per the signature-based risk score. We used the receiver operating characteristic (ROC) curve and the Kaplan-Meier (KM) survival analysis to validate the predictive performance of the 4-gene signature in both the training and validation sets. Infiltrated immune cells and immune checkpoints indicated a difference in the immune status between the two risk groups. Conclusion The results of our study indicated that a prognostic model based on the 4-gene signature related to lipid metabolism was useful for the prediction of prognosis in patients with ESCC.
Collapse
Affiliation(s)
- Guo-Yi Shen
- Department of Cardiothoracic Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, 363000, People’s Republic of China
| | - Peng-Jie Yang
- Department of Thoracic Surgery, Inner Mongolia Cancer Hospital & Affiliated People’s Hospital of Inner Mongolia Medical University, Huhhot, Inner Mongolia Autonomous Region, 010020, People’s Republic of China
| | - Wen-Shan Zhang
- Department of Cardiothoracic Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, 363000, People’s Republic of China
| | - Jun-Biao Chen
- Department of Cardiothoracic Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, 363000, People’s Republic of China
| | - Qin-Yong Tian
- Department of Cardiothoracic Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, 363000, People’s Republic of China
| | - Yi Zhang
- Department of Cardiothoracic Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, 363000, People’s Republic of China
| | - Bater Han
- Department of Thoracic Surgery, Inner Mongolia Cancer Hospital & Affiliated People’s Hospital of Inner Mongolia Medical University, Huhhot, Inner Mongolia Autonomous Region, 010020, People’s Republic of China
| |
Collapse
|
4
|
Steinhäuser S, Silva P, Lenk L, Beder T, Hartmann A, Hänzelmann S, Fransecky L, Neumann M, Bastian L, Lipinski S, Richter K, Bultmann M, Hübner E, Xia S, Röllig C, Vogiatzi F, Schewe DM, Yumiceba V, Schultz K, Spielmann M, Baldus CD. Isocitrate dehydrogenase 1 mutation drives leukemogenesis by PDGFRA activation due to insulator disruption in acute myeloid leukemia (AML). Leukemia 2023; 37:134-142. [PMID: 36411356 PMCID: PMC9883162 DOI: 10.1038/s41375-022-01751-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 10/24/2022] [Accepted: 10/28/2022] [Indexed: 11/22/2022]
Abstract
Acute myeloid leukemia (AML) is characterized by complex molecular alterations and driver mutations. Elderly patients show increased frequencies of IDH mutations with high chemoresistance and relapse rates despite recent therapeutic advances. Besides being associated with global promoter hypermethylation, IDH1 mutation facilitated changes in 3D DNA-conformation by CTCF-anchor methylation and upregulated oncogene expression in glioma, correlating with poor prognosis. Here, we investigated the role of IDH1 p.R132H mutation in altering 3D DNA-architecture and subsequent oncogene activation in AML. Using public RNA-Seq data, we identified upregulation of tyrosine kinase PDGFRA in IDH1-mutant patients, correlating with poor prognosis. DNA methylation analysis identified CpG hypermethylation within a CTCF-anchor upstream of PDGFRA in IDH1-mutant patients. Increased PDGFRA expression, PDGFRA-CTCF methylation and decreased CTCF binding were confirmed in AML CRISPR cells with heterozygous IDH1 p.R132H mutation and upon exogenous 2-HG treatment. IDH1-mutant cells showed higher sensitivity to tyrosine kinase inhibitor dasatinib, which was supported by reduced blast count in a patient with refractory IDH1-mutant AML after dasatinib treatment. Our data illustrate that IDH1 p.R132H mutation leads to CTCF hypermethylation, disrupting DNA-looping and insulation of PDGFRA, resulting in PDGFRA upregulation in IDH1-mutant AML. Treatment with dasatinib may offer a novel treatment strategy for IDH1-mutant AML.
Collapse
Affiliation(s)
- Sophie Steinhäuser
- Department of Inner Medicine II (Hematology/Oncology), University Hospital Schleswig-Holstein, Kiel, Germany
| | - Patricia Silva
- Department of Hematology and Oncology, Charité University Hospital, Berlin, Germany
| | - Lennart Lenk
- Department of Pediatrics I, ALL-BFM Study Group, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Thomas Beder
- Department of Inner Medicine II (Hematology/Oncology), University Hospital Schleswig-Holstein, Kiel, Germany
| | - Alina Hartmann
- Department of Inner Medicine II (Hematology/Oncology), University Hospital Schleswig-Holstein, Kiel, Germany
| | - Sonja Hänzelmann
- Department of Inner Medicine II (Hematology/Oncology), University Hospital Schleswig-Holstein, Kiel, Germany
| | - Lars Fransecky
- Department of Inner Medicine II (Hematology/Oncology), University Hospital Schleswig-Holstein, Kiel, Germany
| | - Martin Neumann
- Department of Inner Medicine II (Hematology/Oncology), University Hospital Schleswig-Holstein, Kiel, Germany
| | - Lorenz Bastian
- Department of Inner Medicine II (Hematology/Oncology), University Hospital Schleswig-Holstein, Kiel, Germany
| | - Simone Lipinski
- Department of Inner Medicine II (Hematology/Oncology), University Hospital Schleswig-Holstein, Kiel, Germany
- University Cancer Center Schleswig-Holstein (UCCSH), University Hospital Schleswig-Holstein, Kiel, Germany
| | - Kathrin Richter
- Department of Inner Medicine II (Hematology/Oncology), University Hospital Schleswig-Holstein, Kiel, Germany
| | - Miriam Bultmann
- Department of Inner Medicine II (Hematology/Oncology), University Hospital Schleswig-Holstein, Kiel, Germany
| | - Emely Hübner
- Department of Inner Medicine II (Hematology/Oncology), University Hospital Schleswig-Holstein, Kiel, Germany
| | - Shuli Xia
- Kennedy Krieger Institute, Baltimore, MD, USA
- School of Medicine, Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Christoph Röllig
- Department of Internal Medicine I, University Hospital Carl-Gustav-Carus, Dresden, Germany
| | - Fotini Vogiatzi
- Department of Pediatrics I, ALL-BFM Study Group, University Hospital Schleswig-Holstein, Kiel, Germany
| | | | - Veronica Yumiceba
- Institute for Human Genetics, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Kristin Schultz
- Institute for Human Genetics, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Malte Spielmann
- Institute for Human Genetics, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Claudia Dorothea Baldus
- Department of Inner Medicine II (Hematology/Oncology), University Hospital Schleswig-Holstein, Kiel, Germany.
- University Cancer Center Schleswig-Holstein (UCCSH), University Hospital Schleswig-Holstein, Kiel, Germany.
| |
Collapse
|
5
|
Bhuin S, Sharma P, Chakraborty P, Kulkarni OP, Chakravarty M. Solid-state emitting twisted π-conjugate as AIE-active DSE-gen: in vitro anticancer properties against FaDu and 4T1 with biocompatibility and bioimaging. J Mater Chem B 2022; 11:188-203. [PMID: 36477106 DOI: 10.1039/d2tb02078e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Dual-state emissive fluorogens (DSE-gens) are currently defining their importance as a transpiring tool in biological and biomedical applications. This work focuses on designing and synthesizing indole-anthracene-based solid-state emitting twisted π-conjugates using a metal-free protocol to achieve AIE-active DSE-gens, expanding their scope in biological applications. Special effort has been made to introduce proficient and photo/thermostable DSE-gens that inhibit cancer but not normal cells. Here, the lead DSE-gen initially detects cancer and normal cells by bioimaging; however, it could also confirm and distinguish cancer cells from normal cells by its abated fluorescence signal after killing cancer cells. In contrast, the fluorescence signals for a normal cell remain unscathed. Surprisingly, these molecules displayed decent anticancer properties against FaDu and 4T1 but not MCF-7 cell lines. From a series of newly designed indole-based molecules, we report one single 2,3,4-trimethoxybenzene-linked DSE-gen (the lead), exhibiting high ROS generation, less haemolysis, and less cytotoxicity than doxorubicin (DOX) for normal cells, crucial parameters for a biocompatible in vitro anticancer probe. Thus, we present a potentially applicable anticancer drug, offering a bioactive material with bioimaging efficacy and a way to detect dead cancer cells selectively. The primary mechanism behind the identified outcomes is deciphered with the support of experimental (steady-state and time-resolved fluorescence, biological assays, cellular uptake) and molecular docking studies.
Collapse
Affiliation(s)
- Shouvik Bhuin
- Department of Chemistry, Birla Institute of Technology and Science-Pilani, Hyderabad Campus Jawahar Nagar, Shamirpet, Hyderabad, Telangana, 500078, India.
| | - Pravesh Sharma
- Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad Campus Jawahar Nagar, Shamirpet, Hyderabad, Telangana, 500078, India
| | - Purbali Chakraborty
- Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad Campus Jawahar Nagar, Shamirpet, Hyderabad, Telangana, 500078, India
| | - Onkar Prakash Kulkarni
- Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad Campus Jawahar Nagar, Shamirpet, Hyderabad, Telangana, 500078, India
| | - Manab Chakravarty
- Department of Chemistry, Birla Institute of Technology and Science-Pilani, Hyderabad Campus Jawahar Nagar, Shamirpet, Hyderabad, Telangana, 500078, India.
| |
Collapse
|
6
|
Reimche I, Yu H, Ariantari NP, Liu Z, Merkens K, Rotfuß S, Peter K, Jungwirth U, Bauer N, Kiefer F, Neudörfl JM, Schmalz HG, Proksch P, Teusch N. Phenanthroindolizidine Alkaloids Isolated from Tylophora ovata as Potent Inhibitors of Inflammation, Spheroid Growth, and Invasion of Triple-Negative Breast Cancer. Int J Mol Sci 2022; 23:ijms231810319. [PMID: 36142230 PMCID: PMC9499467 DOI: 10.3390/ijms231810319] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 09/01/2022] [Accepted: 09/02/2022] [Indexed: 11/16/2022] Open
Abstract
Triple-negative breast cancer (TNBC), representing the most aggressive form of breast cancer with currently no targeted therapy available, is characterized by an inflammatory and hypoxic tumor microenvironment. To date, a broad spectrum of anti-tumor activities has been reported for phenanthroindolizidine alkaloids (PAs), however, their mode of action in TNBC remains elusive. Thus, we investigated six naturally occurring PAs extracted from the plant Tylophora ovata: O-methyltylophorinidine (1) and its five derivatives tylophorinidine (2), tylophoridicine E (3), 2-demethoxytylophorine (4), tylophoridicine D (5), and anhydrodehydrotylophorinidine (6). In comparison to natural (1) and for more-in depth studies, we also utilized a sample of synthetic O-methyltylophorinidine (1s). Our results indicate a remarkably effective blockade of nuclear factor kappa B (NFκB) within 2 h for compounds (1) and (1s) (IC50 = 17.1 ± 2.0 nM and 3.3 ± 0.2 nM) that is different from its effect on cell viability within 24 h (IC50 = 13.6 ± 0.4 nM and 4.2 ± 1 nM). Furthermore, NFκB inhibition data for the additional five analogues indicate a structure–activity relationship (SAR). Mechanistically, NFκB is significantly blocked through the stabilization of its inhibitor protein kappa B alpha (IκBα) under normoxic as well as hypoxic conditions. To better mimic the TNBC microenvironment in vitro, we established a 3D co-culture by combining the human TNBC cell line MDA-MB-231 with primary murine cancer-associated fibroblasts (CAF) and type I collagen. Compound (1) demonstrates superiority against the therapeutic gold standard paclitaxel by diminishing spheroid growth by 40% at 100 nM. The anti-proliferative effect of (1s) is distinct from paclitaxel in that it arrests the cell cycle at the G0/G1 state, thereby mediating a time-dependent delay in cell cycle progression. Furthermore, (1s) inhibited invasion of TNBC monoculture spheroids into a matrigel®-based environment at 10 nM. In conclusion, PAs serve as promising agents with presumably multiple target sites to combat inflammatory and hypoxia-driven cancer, such as TNBC, with a different mode of action than the currently applied chemotherapeutic drugs.
Collapse
Affiliation(s)
- Irene Reimche
- Department of Biomedical Sciences, Institute of Health Research and Education, University of Osnabrück, 49090 Osnabrück, Germany
- Institute of Pharmaceutical Biology and Biotechnology, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Haiqian Yu
- Institute of Pharmaceutical Biology and Biotechnology, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Ni Putu Ariantari
- Institute of Pharmaceutical Biology and Biotechnology, Heinrich Heine University, 40225 Düsseldorf, Germany
- Department of Pharmacy, Faculty of Mathematics and Natural Sciences, Udayana University, Bali 80361, Indonesia
| | - Zhen Liu
- Institute of Pharmaceutical Biology and Biotechnology, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Kay Merkens
- Department of Chemistry, University of Cologne, 50923 Cologne, Germany
| | - Stella Rotfuß
- Department of Biomedical Sciences, Institute of Health Research and Education, University of Osnabrück, 49090 Osnabrück, Germany
| | - Karin Peter
- Department of Biomedical Sciences, Institute of Health Research and Education, University of Osnabrück, 49090 Osnabrück, Germany
| | - Ute Jungwirth
- Department of Life Sciences, Centre for Therapeutic Innovation, University of Bath, Bath BA2 7AY, UK
| | - Nadine Bauer
- European Institute of Molecular Imaging, University of Münster, 48149 Münster, Germany
| | - Friedemann Kiefer
- European Institute of Molecular Imaging, University of Münster, 48149 Münster, Germany
- Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany
| | | | | | - Peter Proksch
- Institute of Pharmaceutical Biology and Biotechnology, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Nicole Teusch
- Department of Biomedical Sciences, Institute of Health Research and Education, University of Osnabrück, 49090 Osnabrück, Germany
- Institute of Pharmaceutical Biology and Biotechnology, Heinrich Heine University, 40225 Düsseldorf, Germany
- Correspondence: ; Tel.: +49-211-81-14163
| |
Collapse
|
7
|
Collery P, Lagadec P, Krossa I, Cohen C, Antomarchi J, Varlet D, Lucio M, Guigonis JM, Scimeca JC, Schmid-Antomarchi H, Schmid-Alliana A. Relationship between the oxidative status and the tumor growth in transplanted triple-negative 4T1 breast tumor mice after oral administration of rhenium(I)-diselenoether. J Trace Elem Med Biol 2022; 71:126931. [PMID: 35063816 DOI: 10.1016/j.jtemb.2022.126931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 01/12/2022] [Accepted: 01/14/2022] [Indexed: 10/19/2022]
Abstract
BACKGROUND Selective inhibitory effects of rhenium(I)-diselenoether (Re-diSe) were observed in cultured breast malignant cells. They were attributed to a decrease in Reactive Oxygen Species (ROS) production. A concomitant decrease in the production of Transforming Growth Factor-beta (TGFβ1), Insulin Growth Factor 1 (IGF1), and Vascular Endothelial Growth Factor A (VEGFA) by the malignant cells was also observed. AIM The study aimed to investigate the anti-tumor effects of Re-diSe on mice bearing 4T1 breast tumors, an experimental model of triple-negative breast cancer, and correlate them with several biomarkers. MATERIAL AND METHODS 4T1 mammary breast cancer cells were orthotopically inoculated into syngenic BALB/c Jack mice. Different doses of Re-diSe (1, 10, and 60 mg/kg) were administered orally for 23 consecutive days to assess the efficacy and toxicity. The oxidative status was evaluated by assaying Advanced Oxidative Protein Products (AOPP), and by the dinitrophenylhydrazone (DNPH) test in plasma of healthy mice, non-treated tumor-bearing mice (controls), treated tumor-bearing mice, and tumors in all tumor-bearing mice. Tumor necrosis factor (TNFα), VEGFA, VEGFB, TGFβ1, Interferon, and selenoprotein P (selenoP) were selected as biomarkers. RESULTS Doses of 1 and 10 mg/kg did not affect the tumor weights. There was a significant increase in the tumor weights in mice treated with the maximum dose of 60 mg/kg, concomitantly with a significant decrease in AOPP, TNFα, and TGFβ1 in the tumors. SelenoP concentrations increased in the plasma but not in the tumors. CONCLUSION We did not confirm the anti-tumor activity of the Re-diSe compound in this experiment. However, the transplantation of the tumor cells did not induce an expected pro-oxidative status without any increase of the oxidative biomarkers in the plasma of controls compared to healthy mice. This condition could be essential to evaluate the effect of an antioxidant drug. The choice of the experimental model will be primordial to assess the effects of the Re-diSe compound in further studies.
Collapse
Affiliation(s)
- Philippe Collery
- Société de Coordination de Recherches Thérapeutiques, 20220, Algajola, France.
| | - Patricia Lagadec
- Université Nice Sophia Antipolis, CNRS, Inserm, iBV, UFR de médecine Pasteur, 06107, Nice cedex 2, France
| | - Imène Krossa
- Université Nice Sophia Antipolis, CNRS, Inserm, iBV, UFR de médecine Pasteur, 06107, Nice cedex 2, France
| | - Charlotte Cohen
- Université Nice Sophia Antipolis, CNRS, Inserm, iBV, UFR de médecine Pasteur, 06107, Nice cedex 2, France
| | - Julie Antomarchi
- Université Nice Sophia Antipolis, CNRS, Inserm, iBV, UFR de médecine Pasteur, 06107, Nice cedex 2, France
| | | | - Marianna Lucio
- Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt, 85764, Neuherberg, Germany
| | - Jean-Marie Guigonis
- Université Nice Sophia Antipolis, Plateforme "Bernard Rossi", UFR de médecine Pasteur, UMR 4320, CEA TIRO, 06107, Nice cedex 2, France
| | - Jean-Claude Scimeca
- Université Nice Sophia Antipolis, CNRS, Inserm, iBV, UFR de médecine Pasteur, 06107, Nice cedex 2, France
| | - Heidy Schmid-Antomarchi
- Université Nice Sophia Antipolis, CNRS, Inserm, iBV, UFR de médecine Pasteur, 06107, Nice cedex 2, France
| | - Annie Schmid-Alliana
- Université Nice Sophia Antipolis, CNRS, Inserm, iBV, UFR de médecine Pasteur, 06107, Nice cedex 2, France
| |
Collapse
|
8
|
Romayor I, García-Vaquero ML, Márquez J, Arteta B, Barceló R, Benedicto A. Discoidin Domain Receptor 2 Expression as Worse Prognostic Marker in Invasive Breast Cancer. Breast J 2022; 2022:5169405. [PMID: 35711892 PMCID: PMC9187291 DOI: 10.1155/2022/5169405] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 02/10/2022] [Indexed: 01/01/2023]
Abstract
Discoidin domain receptor 2 (DDR2) is arising as a promising therapeutic target in breast carcinoma (BC). The ability of DDR2 to bind to collagen promotes protumoral responses in cancer cells that influence the tumor microenvironment (TME). Nonetheless, the interrelation between DDR2 expression and TME modulation during BC progression remains poorly known. For this reason, we aim to evaluate the correlation between intratumoral expression of DDR2 and the infiltration of the main TME cell populations, cancer-associated fibroblasts (CAFs), and tumor-associated macrophages (TAMs). First, collagen and DDR2 expression levels were analyzed in human invasive BC samples. Then, DDR2 status correlation with tumor aggressiveness and patient survival were retrieved from different databases. Subsequently, the main pathways, cell types, and tissues correlated with DDR2 expression in BC were obtained through bioinformatics approach. Finally, we studied the association of DDR2 expression with the recruitment of CAFs and TAMs. Our findings showed that, together with the expected overexpression of TME markers, DDR2 was upregulated in tumor samples. Besides, we uncovered that altered TME markers were linked to DDR2 expression in invasive BC patients. Consequently, DDR2 modulates the stromal reaction through CAFs and TAMs infiltration and could be used as a potential worse prognostic factor in the treatment response of invasive BC.
Collapse
Affiliation(s)
- Irene Romayor
- Department of Cell Biology and Histology, School of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
| | - Marina Luque García-Vaquero
- Department of Medicine and Cytometry General Service-Nucleus, CIBERONC, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca9, Spain
| | - Joana Márquez
- Department of Cell Biology and Histology, School of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
| | - Beatriz Arteta
- Department of Cell Biology and Histology, School of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
| | - Ramón Barceló
- Department of Cell Biology and Histology, School of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
- Oncology Service, Basurto University Hospital, 48002 Bilbao, Spain
| | - Aitor Benedicto
- Department of Cell Biology and Histology, School of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
| |
Collapse
|
9
|
Mollah F, Varamini P. Overcoming Therapy Resistance and Relapse in TNBC: Emerging Technologies to Target Breast Cancer-Associated Fibroblasts. Biomedicines 2021; 9:1921. [PMID: 34944738 PMCID: PMC8698629 DOI: 10.3390/biomedicines9121921] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 12/10/2021] [Accepted: 12/11/2021] [Indexed: 12/12/2022] Open
Abstract
Breast cancer is the most diagnosed cancer and is the leading cause of cancer mortality in women. Triple-negative breast cancer (TNBC) is an aggressive form of breast cancer. Often, TNBC is not effectively treated due to the lack of specificity of conventional therapies and results in relapse and metastasis. Breast cancer-associated fibroblasts (BCAFs) are the predominant cells that reside in the tumor microenvironment (TME) and regulate tumorigenesis, progression and metastasis, and therapy resistance. BCAFs secrete a wide range of factors, including growth factors, chemokines, and cytokines, some of which have been proved to lead to a poor prognosis and clinical outcomes. This TME component has been emerging as a promising target due to its crucial role in cancer progression and chemotherapy resistance. A number of therapeutic candidates are designed to effectively target BCAFs with a focus on their tumor-promoting properties and tumor immune response. This review explores various agents targeting BCAFs in TNBC, including small molecules, nucleic acid-based agents, antibodies, proteins, and finally, nanoparticles.
Collapse
Affiliation(s)
- Farhana Mollah
- Faculty of Medicine and Health, School of Pharmacy, University of Sydney, Sydney, NSW 2006, Australia;
| | - Pegah Varamini
- Faculty of Medicine and Health, School of Pharmacy, University of Sydney, Sydney, NSW 2006, Australia;
- Sydney Nano Institute, University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
10
|
Li M, Zheng K, Ma S, Hu P, Yuan B, Yue X, Li Q. Pilose antler polypeptides promote chemosensitization and T-cell infiltration of triple-negative breast cancer. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104664] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
|
11
|
Fernández-Nogueira P, Fuster G, Gutierrez-Uzquiza Á, Gascón P, Carbó N, Bragado P. Cancer-Associated Fibroblasts in Breast Cancer Treatment Response and Metastasis. Cancers (Basel) 2021; 13:3146. [PMID: 34201840 PMCID: PMC8268405 DOI: 10.3390/cancers13133146] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/15/2021] [Accepted: 06/16/2021] [Indexed: 12/21/2022] Open
Abstract
Breast cancer (BrCa) is the leading cause of death among women worldwide, with about one million new cases diagnosed each year. In spite of the improvements in diagnosis, early detection and treatment, there is still a high incidence of mortality and failure to respond to current therapies. With the use of several well-established biomarkers, such as hormone receptors and human epidermal growth factor receptor-2 (HER2), as well as genetic analysis, BrCa patients can be categorized into multiple subgroups: Luminal A, Luminal B, HER2-enriched, and Basal-like, with specific treatment strategies. Although chemotherapy and targeted therapies have greatly improved the survival of patients with BrCa, there is still a large number of patients who relapse or who fail to respond. The role of the tumor microenvironment in BrCa progression is becoming increasingly understood. Cancer-associated fibroblasts (CAFs) are the principal population of stromal cells in breast tumors. In this review, we discuss the current understanding of CAFs' role in altering the tumor response to therapeutic agents as well as in fostering metastasis in BrCa. In addition, we also review the available CAFs-directed molecular therapies and their potential implications for BrCa management.
Collapse
Affiliation(s)
- Patricia Fernández-Nogueira
- Department of Biochemistry and Molecular Biomedicine, Institute of Biomedicine, University of Barcelona (IBUB), 08028 Barcelona, Spain; (G.F.); (P.G.); (N.C.)
- Department of Biomedicine, School of Medicine, University of Barcelona, 08028 Barcelona, Spain
| | - Gemma Fuster
- Department of Biochemistry and Molecular Biomedicine, Institute of Biomedicine, University of Barcelona (IBUB), 08028 Barcelona, Spain; (G.F.); (P.G.); (N.C.)
- Department of Biochemistry & Physiology, School of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain
- Department of Biosciences, Faculty of Sciences and Technology, University of Vic, 08500 Vic, Spain
| | - Álvaro Gutierrez-Uzquiza
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain;
- Health Research Institute of the Hospital Clínico San Carlos, 28040 Madrid, Spain
| | - Pere Gascón
- Department of Biochemistry and Molecular Biomedicine, Institute of Biomedicine, University of Barcelona (IBUB), 08028 Barcelona, Spain; (G.F.); (P.G.); (N.C.)
| | - Neus Carbó
- Department of Biochemistry and Molecular Biomedicine, Institute of Biomedicine, University of Barcelona (IBUB), 08028 Barcelona, Spain; (G.F.); (P.G.); (N.C.)
| | - Paloma Bragado
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain;
- Health Research Institute of the Hospital Clínico San Carlos, 28040 Madrid, Spain
| |
Collapse
|