1
|
Quoniou R, Moreau E, Cachin F, Blavignac C, Bortoli E, Chautard E, Peyrode C. Chondrosarcoma Co-Culture 3D Model─An Insight to Evaluate Drugs Acting on TAMs. ACS Biomater Sci Eng 2024; 10:5832-5843. [PMID: 39121344 DOI: 10.1021/acsbiomaterials.4c00625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/11/2024]
Abstract
Chondrosarcoma (CHS), also known as malignant cartilage tumors, is the second most common bone cancer after osteosarcoma. This tumor is particularly chemo- and radioresistant, and the only therapeutic alternative is surgery with wide margins. The tumor immune microenvironment reveals an infiltration of tumor-associated macrophages (TAMs) sometimes approaching 50% of the tumor mass, mainly differentiated into M2-like phenotype and correlated with poor prognosis and metastasis. Thus, macrophage-targeting therapies could have an interest in the management of CHS. To evaluate these strategies, we propose here the development of a three-dimensional (3D) tumoroid co-culture model between two human CHS cell lines (JJ012 and CH2879) and a human leukemia monocytic cell line (THP-1) in a methylcellulose matrix. These two models were compared to the in vivo xenograft models in terms of macrophage phenotypes, proteoglycans, MMP-9, and COX-2 expression. Finally, mifamurtide, an immunomodulator acting on TAMs, was evaluated on the most in vitro relevant model: 3D co-culture CH2879 model. Our results showed that it is now possible to develop 3D models that very accurately mimic what is found in vivo with the possibility of evaluating treatments specific to a tumor cell component.
Collapse
Affiliation(s)
- Rohan Quoniou
- Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, Université Clermont Auvergne, INSERM, 63000 Clermont-Ferrand, France
| | - Emmanuel Moreau
- Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, Université Clermont Auvergne, INSERM, 63000 Clermont-Ferrand, France
| | - Florent Cachin
- Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, Université Clermont Auvergne, INSERM, 63000 Clermont-Ferrand, France
- Service de Médecine Nucléaire, Centre Jean PERRIN, 63000 Clermont-Ferrand, France
| | | | - Elisa Bortoli
- Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, Université Clermont Auvergne, INSERM, 63000 Clermont-Ferrand, France
| | - Emmanuel Chautard
- Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, Université Clermont Auvergne, INSERM, 63000 Clermont-Ferrand, France
- Service de Pathologie, Centre Jean PERRIN, 63000 Clermont-Ferrand, France
| | - Caroline Peyrode
- Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, Université Clermont Auvergne, INSERM, 63000 Clermont-Ferrand, France
| |
Collapse
|
2
|
Bao R, Qu H, Li B, Cheng K, Miao Y, Wang J. The role of metabolic reprogramming in immune escape of triple-negative breast cancer. Front Immunol 2024; 15:1424237. [PMID: 39192979 PMCID: PMC11347331 DOI: 10.3389/fimmu.2024.1424237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 07/29/2024] [Indexed: 08/29/2024] Open
Abstract
Triple-negative breast cancer (TNBC) has become a thorny problem in the treatment of breast cancer because of its high invasiveness, metastasis and recurrence. Although immunotherapy has made important progress in TNBC, immune escape caused by many factors, especially metabolic reprogramming, is still the bottleneck of TNBC immunotherapy. Regrettably, the mechanisms responsible for immune escape remain poorly understood. Exploring the mechanism of TNBC immune escape at the metabolic level provides a target and direction for follow-up targeting or immunotherapy. In this review, we focus on the mechanism that TNBC affects immune cells and interstitial cells through hypoxia, glucose metabolism, lipid metabolism and amino acid metabolism, and changes tumor metabolism and tumor microenvironment. This will help to find new targets and strategies for TNBC immunotherapy.
Collapse
Affiliation(s)
- Ruochen Bao
- Thyroid and Breast Surgery, Yantai Affiliated Hospital of Binzhou Medical University, The 2Medical College of Binzhou Medical University, Yantai, China
| | - Hongtao Qu
- Emergency Department of Yantai Mountain Hospital, Yantai, China
| | - Baifeng Li
- Thyroid and Breast Surgery, Yantai Affiliated Hospital of Binzhou Medical University, The 2Medical College of Binzhou Medical University, Yantai, China
| | - Kai Cheng
- Thyroid and Breast Surgery, Yantai Affiliated Hospital of Binzhou Medical University, The 2Medical College of Binzhou Medical University, Yantai, China
| | - Yandong Miao
- Cancer Center, Yantai Affiliated Hospital of Binzhou Medical University, The 2 Medical College of Binzhou Medical University, Yantai, China
| | - Jiangtao Wang
- Thyroid and Breast Surgery, Yantai Affiliated Hospital of Binzhou Medical University, The 2Medical College of Binzhou Medical University, Yantai, China
| |
Collapse
|
3
|
Sui C, Wu H, Li X, Wang Y, Wei J, Yu J, Wu X. Cancer immunotherapy and its facilitation by nanomedicine. Biomark Res 2024; 12:77. [PMID: 39097732 PMCID: PMC11297660 DOI: 10.1186/s40364-024-00625-6] [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: 04/21/2024] [Accepted: 07/22/2024] [Indexed: 08/05/2024] Open
Abstract
Cancer immunotherapy has sparked a wave of cancer research, driven by recent successful proof-of-concept clinical trials. However, barriers are emerging during its rapid development, including broad adverse effects, a lack of reliable biomarkers, tumor relapses, and drug resistance. Integration of nanomedicine may ameliorate current cancer immunotherapy. Ultra-large surface-to-volume ratio, extremely small size, and easy modification surface of nanoparticles enable them to selectively detect cells and kill cancer cells in vivo. Exciting synergistic applications of the two approaches have emerged in treating various cancers at the intersection of cancer immunotherapy and cancer nanomedicine, indicating the potential that the combination of these two therapeutic modalities can lead to new paradigms in the treatment of cancer. This review discusses the status of current immunotherapy and explores the possible opportunities that the nanomedicine platform can make cancer immunotherapy more powerful and precise by synergizing the two approaches.
Collapse
Affiliation(s)
- Chao Sui
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, 1500 East Duarte, Los Angeles, CA, 91010, USA
| | - Heqing Wu
- The First Affiliated Hospital of Soochow University, Suzhou, China
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Xinxin Li
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an Shaanxi, 710072, China
| | - Yuhang Wang
- The First Affiliated Hospital of Soochow University, Suzhou, China
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Jiaqi Wei
- The First Affiliated Hospital of Soochow University, Suzhou, China
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Jianhua Yu
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, 1500 East Duarte, Los Angeles, CA, 91010, USA.
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA, 91010, USA.
| | - Xiaojin Wu
- The First Affiliated Hospital of Soochow University, Suzhou, China.
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Suzhou, China.
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.
| |
Collapse
|
4
|
Ammarah U, Pereira‐Nunes A, Delfini M, Mazzone M. From monocyte-derived macrophages to resident macrophages-how metabolism leads their way in cancer. Mol Oncol 2024; 18:1739-1758. [PMID: 38411356 PMCID: PMC11223613 DOI: 10.1002/1878-0261.13618] [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: 10/23/2023] [Revised: 01/24/2024] [Accepted: 02/16/2024] [Indexed: 02/28/2024] Open
Abstract
Macrophages are innate immune cells that play key roles during both homeostasis and disease. Depending on the microenvironmental cues sensed in different tissues, macrophages are known to acquire specific phenotypes and exhibit unique features that, ultimately, orchestrate tissue homeostasis, defense, and repair. Within the tumor microenvironment, macrophages are referred to as tumor-associated macrophages (TAMs) and constitute a heterogeneous population. Like their tissue resident counterpart, TAMs are plastic and can switch function and phenotype according to the niche-derived stimuli sensed. While changes in TAM phenotype are known to be accompanied by adaptive alterations in their cell metabolism, it is reported that metabolic reprogramming of macrophages can dictate their activation state and function. In line with these observations, recent research efforts have been focused on defining the metabolic traits of TAM subsets in different tumor malignancies and understanding their role in cancer progression and metastasis formation. This knowledge will pave the way to novel therapeutic strategies tailored to cancer subtype-specific metabolic landscapes. This review outlines the metabolic characteristics of distinct TAM subsets and their implications in tumorigenesis across multiple cancer types.
Collapse
Affiliation(s)
- Ummi Ammarah
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer BiologyVIBLeuvenBelgium
- Laboratory of Tumor Inflammation and Angiogenesis, Department of Oncology, Center for Cancer BiologyKU LeuvenBelgium
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology CentreUniversity of TorinoItaly
| | - Andreia Pereira‐Nunes
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer BiologyVIBLeuvenBelgium
- Laboratory of Tumor Inflammation and Angiogenesis, Department of Oncology, Center for Cancer BiologyKU LeuvenBelgium
- Life and Health Sciences Research Institute (ICVS), School of MedicineUniversity of MinhoBragaPortugal
- ICVS/3B's‐PT Government Associate LaboratoryBraga/GuimarãesPortugal
| | - Marcello Delfini
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer BiologyVIBLeuvenBelgium
- Laboratory of Tumor Inflammation and Angiogenesis, Department of Oncology, Center for Cancer BiologyKU LeuvenBelgium
| | - Massimiliano Mazzone
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer BiologyVIBLeuvenBelgium
- Laboratory of Tumor Inflammation and Angiogenesis, Department of Oncology, Center for Cancer BiologyKU LeuvenBelgium
| |
Collapse
|
5
|
Wang F, Fu K, Wang Y, Pan C, Wang X, Liu Z, Yang C, Zheng Y, Li X, Lu Y, To KKW, Xia C, Zhang J, Shi Z, Hu Z, Huang M, Fu L. Small-molecule agents for cancer immunotherapy. Acta Pharm Sin B 2024; 14:905-952. [PMID: 38486980 PMCID: PMC10935485 DOI: 10.1016/j.apsb.2023.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/28/2023] [Accepted: 12/06/2023] [Indexed: 03/17/2024] Open
Abstract
Cancer immunotherapy, exemplified by the remarkable clinical benefits of the immune checkpoint blockade and chimeric antigen receptor T-cell therapy, is revolutionizing cancer therapy. They induce long-term tumor regression and overall survival benefit in many types of cancer. With the advances in our knowledge about the tumor immune microenvironment, remarkable progress has been made in the development of small-molecule drugs for immunotherapy. Small molecules targeting PRR-associated pathways, immune checkpoints, oncogenic signaling, metabolic pathways, cytokine/chemokine signaling, and immune-related kinases have been extensively investigated. Monotherapy of small-molecule immunotherapeutic drugs and their combinations with other antitumor modalities are under active clinical investigations to overcome immune tolerance and circumvent immune checkpoint inhibitor resistance. Here, we review the latest development of small-molecule agents for cancer immunotherapy by targeting defined pathways and highlighting their progress in recent clinical investigations.
Collapse
Affiliation(s)
- Fang Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Kai Fu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Yujue Wang
- School of Pharmaceutical Sciences, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, Tsinghua University, Beijing 100084, China
| | - Can Pan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Xueping Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Zeyu Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Chuan Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Ying Zheng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xiaopeng Li
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Yu Lu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Kenneth Kin Wah To
- School of Pharmacy, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Chenglai Xia
- Affiliated Foshan Maternity and Child Healthcare Hospital, Southern Medical University, Foshan 528000, China
| | - Jianye Zhang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Zhi Shi
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Zeping Hu
- School of Pharmaceutical Sciences, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, Tsinghua University, Beijing 100084, China
| | - Min Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Liwu Fu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| |
Collapse
|
6
|
Stachowicz-Suhs M, Łabędź N, Anisiewicz A, Banach J, Kłopotowska D, Milczarek M, Piotrowska A, Dzięgiel P, Maciejczyk A, Matkowski R, Wietrzyk J. Calcitriol promotes M2 polarization of tumor-associated macrophages in 4T1 mouse mammary gland cancer via the induction of proinflammatory cytokines. Sci Rep 2024; 14:3778. [PMID: 38355711 PMCID: PMC10866890 DOI: 10.1038/s41598-024-54433-x] [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: 10/31/2023] [Accepted: 02/13/2024] [Indexed: 02/16/2024] Open
Abstract
Our research found that vitamin D3 (VD3) treatment increased lung metastasis in mice with 4T1 murine breast cancer (BC). This study aims to investigate the impact of VD3 on the activation of tumor-associated macrophages (TAMs) in BC. Mice bearing 4T1, E0771, 67NR BC cells, and healthy mice, were fed diets with varying VD3 contents (100-deficient, 1000-normal, and 5000 IU/kg-elevated). Some mice in the 1000 and 100 IU/kg groups received calcitriol. We studied bone metastasis and characterized TAMs and bone marrow-derived macrophages (BMDMs). 4T1 cells had higher bone metastasis potential in the 5000 IU/kg and calcitriol groups. In the same mice, an elevated tumor osteopontin level and M2 polarization of TAMs (MHCIIlow CD44high phenotype) were observed. Gene expression analysis confirmed M2 polarization of 4T1 (but not 67NR) TAMs and BMDMs, particularly in the 100 IU + cal group (increased Mrc1, Il23, and Il6). This polarization was likely due to COX-2/PGE2 induction in 4T1 calcitriol-treated cells, leading to increased proinflammatory cytokines like IL-6 and IL-23. Future studies will explore COX-2/PGE2 as a primary mediator of calcitriol-stimulated inflammation in the BC microenvironment, especially relevant for BC patients with VD3 deficiency and supplementation.
Collapse
Affiliation(s)
- Martyna Stachowicz-Suhs
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Weigla 12, 53-114, Wroclaw, Poland
| | - Natalia Łabędź
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Weigla 12, 53-114, Wroclaw, Poland
| | - Artur Anisiewicz
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Weigla 12, 53-114, Wroclaw, Poland
| | - Joanna Banach
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Weigla 12, 53-114, Wroclaw, Poland
| | - Dagmara Kłopotowska
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Weigla 12, 53-114, Wroclaw, Poland
| | - Magdalena Milczarek
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Weigla 12, 53-114, Wroclaw, Poland
| | - Aleksandra Piotrowska
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Chałubińskiego 6a, 50-368, Wroclaw, Poland
| | - Piotr Dzięgiel
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Chałubińskiego 6a, 50-368, Wroclaw, Poland
| | - Adam Maciejczyk
- Department of Oncology, Wroclaw Medical University, Pl. Ludwika Hirszfelda 12, 53-413, Wrocław, Poland
- Lower Silesian Oncology, Pulmonology and Hematology Center, Pl. Ludwika Hirszfelda 12, 53-413, Wrocław, Poland
| | - Rafał Matkowski
- Department of Oncology, Wroclaw Medical University, Pl. Ludwika Hirszfelda 12, 53-413, Wrocław, Poland
- Lower Silesian Oncology, Pulmonology and Hematology Center, Pl. Ludwika Hirszfelda 12, 53-413, Wrocław, Poland
| | - Joanna Wietrzyk
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Weigla 12, 53-114, Wroclaw, Poland.
| |
Collapse
|
7
|
Hao M, Huang B, Wu R, Peng Z, Luo KQ. The Interaction between Macrophages and Triple-negative Breast Cancer Cells Induces ROS-Mediated Interleukin 1α Expression to Enhance Tumorigenesis and Metastasis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302857. [PMID: 37551997 PMCID: PMC10582438 DOI: 10.1002/advs.202302857] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/24/2023] [Indexed: 08/09/2023]
Abstract
Triple-negative breast cancer (TNBC) has higher mortality than non-TNBC because of its stronger metastatic capacity. Increasing studies reported that TNBC tumors had more macrophage infiltration than non-TNBC tumors, which promoted the metastasis of TNBC cells. However, how TNBC cells become more malignant after interacting with macrophages is less reported. In this study, it is observed that when TNBC cells are co-cultured with macrophages, they display higher viability and stronger metastatic ability than non-TNBC cells. Mechanistic studies reveal that TNBC cells acquired these abilities via interactions with macrophages in three phases. First, within 12 h of co-culture with macrophages, some TNBC cells have significantly elevated levels of reactive oxygen species (ROS), which upregulate interleukin 1α (IL1α) expression in ERK1/2-c-Jun- and NF-κB-dependent manners at 24-48 h. Second, the secreted IL1α bound to IL1R1 activates the ERK1/2-ZEB1-VIM pathway which increases metastasis. Third, IL1α/IL1R1 facilitates its own synthesis and induces the expression of IL1β and IL8 at 72-96 h through the MKK4-JNK-c-Jun and NF-κB signaling pathways. Moreover, a higher level of IL1α is positively correlated with more macrophage infiltration and shorter overall survival in breast cancer patients. Thus, reducing ROS elevation or downregulating IL1α expression can serve as new strategies to decrease metastasis of TNBC.
Collapse
Affiliation(s)
- Meng Hao
- Department of Biomedical SciencesFaculty of Health SciencesUniversity of MacauTaipaMacao SAR99078China
| | - Bin Huang
- Department of Biomedical SciencesFaculty of Health SciencesUniversity of MacauTaipaMacao SAR99078China
| | - Renfei Wu
- Department of Biomedical SciencesFaculty of Health SciencesUniversity of MacauTaipaMacao SAR99078China
| | - Zheng Peng
- Department of Biomedical SciencesFaculty of Health SciencesUniversity of MacauTaipaMacao SAR99078China
| | - Kathy Qian Luo
- Department of Biomedical SciencesFaculty of Health SciencesUniversity of MacauTaipaMacao SAR99078China
- Ministry of Education Frontiers Science Center for Precision OncologyUniversity of MacauTaipaMacao SAR99078China
| |
Collapse
|
8
|
Liang Y, He J, Chen X, Yin L, Yuan Q, Zeng Q, Zu X, Shen Y. The emerging roles of metabolism in the crosstalk between breast cancer cells and tumor-associated macrophages. Int J Biol Sci 2023; 19:4915-4930. [PMID: 37781517 PMCID: PMC10539698 DOI: 10.7150/ijbs.86039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 09/11/2023] [Indexed: 10/03/2023] Open
Abstract
Breast cancer is the most common cancer affecting women worldwide. Investigating metabolism in breast cancer may accelerate the exploitation of new therapeutic options for immunotherapies. Metabolic reprogramming can confer breast cancer cells (BCCs) with a survival advantage in the tumor microenvironment (TME) and metabolic alterations in breast cancer, and the corresponding metabolic byproducts can affect the function of tumor-associated macrophages (TAMs). Additionally, TAMs undergo metabolic reprogramming in response to signals present in the TME, which can affect their function and breast cancer progression. Here, we review the metabolic crosstalk between BCCs and TAMs in terms of glucose, lipids, amino acids, iron, and adenosine metabolism. Summaries of inhibitors that target metabolism-related processes in BCCs or TAMs within breast cancer have also served as valuable inspiration for novel therapeutic approaches in the fight against this disease. This review provides new perspectives on targeted anticancer therapies for breast cancer that combine immunity with metabolism.
Collapse
Affiliation(s)
- Yuxin Liang
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Jun He
- Department of Spine Surgery, The Nanhua Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Xiguang Chen
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Liyang Yin
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Qiong Yuan
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Qiting Zeng
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Xuyu Zu
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Yingying Shen
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| |
Collapse
|
9
|
Liu S, Shen YY, Yin LY, Liu J, Zu X. Lipid Metabolic Regulatory Crosstalk Between Cancer Cells and Tumor-Associated Macrophages. DNA Cell Biol 2023; 42:445-455. [PMID: 37535386 DOI: 10.1089/dna.2023.0071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2023] Open
Abstract
In the tumor microenvironment, tumor-associated macrophages (TAMs) are one of the most abundant cell populations, playing key roles in tumorigenesis, chemoresistance, immune evasion, and metastasis. There is an important interaction between TAMs and cancer cells: on the one hand, tumors control the function of infiltrating macrophages, contributing to reprogramming of TAMs, and on the other hand, TAMs affect the growth of cancer cells. This review focuses on lipid metabolism changes in the complex relationship between cancer cells and TAMs. We discuss how lipid metabolism in cancer cells affects macrophage phenotypic and metabolic changes and, subsequently, how altered lipid metabolism of TAMs influences tumor progression. Identifying the metabolic changes that influence the complex interaction between tumor cells and TAMs is also an important step in exploring new therapeutic approaches that target metabolic reprogramming of immune cells to enhance their tumoricidal potential and bypass therapy resistance. Our work may provide new targets for antitumor therapies.
Collapse
Affiliation(s)
- Shu Liu
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Ying Ying Shen
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Li Yang Yin
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jianghua Liu
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xuyu Zu
- Cancer Research Institute, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
10
|
Yamaguchi-Tanaka M, Takagi K, Miki Y, Sato A, Iwabuchi E, Miyashita M, Suzuki T. The Pro-Tumorigenic Role of Chemotherapy-Induced Extracellular HSP70 from Breast Cancer Cells via Intratumoral Macrophages. Cancers (Basel) 2023; 15:cancers15061903. [PMID: 36980788 PMCID: PMC10047178 DOI: 10.3390/cancers15061903] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/17/2023] [Accepted: 03/18/2023] [Indexed: 03/30/2023] Open
Abstract
Tumor-associated macrophages (TAMs) contribute to tumor progression and chemoresistance; it is therefore important to clarify the altered functions of macrophages following chemotherapy. While extracellular heat shock protein (HSP) 70 is associated with therapeutic resistance, the effects of HSP70 on TAMs remain largely unknown. Here, we conducted in vitro experiments and immunohistochemistry in 116 breast carcinoma specimens to determine whether the secretion of HSP70 from breast cancer cells following chemotherapy affects macrophage function. It was revealed that the interaction of epirubicin (EPI)-exposed breast cancer cells with macrophages enhanced tumor progression, and EPI promoted the secretion of extracellular HSP70 from breast cancer cells. The expression of pro-tumorigenic macrophage marker CD163 was decreased in macrophages treated with a conditioned medium (CM) from HSP70-silenced breast cancer cells. Breast cancer cells treated with CM from HSP70-silenced breast cancer cells showed decreased expression of transforming growth factor (TGF)-β, and the pro-tumorigenic effects of macrophages were impaired when TGF-β signaling was inhibited. Immunohistochemistry demonstrated that HSP70 served as a poor prognostic factor in conjunction with macrophage infiltration. It was therefore concluded that extracellular HSP70 levels increased following chemotherapy and enhanced the pro-tumorigenic effects of TAMs, either directly or indirectly, by regulating TGF-β expression in breast cancer cells.
Collapse
Affiliation(s)
- Mio Yamaguchi-Tanaka
- Department of Pathology and Histotechnology, Graduate School of Medicine, Tohoku University, Sendai 980-8575, Japan
| | - Kiyoshi Takagi
- Department of Pathology and Histotechnology, Graduate School of Medicine, Tohoku University, Sendai 980-8575, Japan
| | - Yasuhiro Miki
- Department of Nursing, Faculty of Medical Science & Welfare, Tohoku Bunka Gakuen University, Sendai 981-8551, Japan
| | - Ai Sato
- Department of Pathology and Histotechnology, Graduate School of Medicine, Tohoku University, Sendai 980-8575, Japan
| | - Erina Iwabuchi
- Department of Pathology and Histotechnology, Graduate School of Medicine, Tohoku University, Sendai 980-8575, Japan
| | - Minoru Miyashita
- Department of Breast and Endocrine Surgical Oncology, Graduate School of Medicine, Tohoku University, Sendai 980-8575, Japan
| | - Takashi Suzuki
- Department of Pathology and Histotechnology, Graduate School of Medicine, Tohoku University, Sendai 980-8575, Japan
- Department of Anatomic Pathology, Graduate School of Medicine, Tohoku University, Sendai 980-8575, Japan
- Department of Pathology, Tohoku University Hospital, Sendai 980-8574, Japan
| |
Collapse
|
11
|
Ruiz TFR, Colleta SJ, Dos Santos DD, Castro NFC, Cabral ÁS, Calmon MF, Rahal P, Gil CD, Girol AP, Vilamaior PSL, Leonel ECR, Taboga SR. Bisphenol A disruption promotes mammary tumor microenvironment via phenotypic cell polarization and inflammatory response. Cell Biol Int 2023; 47:1136-1146. [PMID: 36906806 DOI: 10.1002/cbin.12007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 01/04/2023] [Accepted: 02/20/2023] [Indexed: 03/13/2023]
Abstract
Inflammation in the established tumor microenvironment (TME) is often associated with a poor prognosis of breast cancer. Bisphenol A (BPA) is an endocrine-disrupting chemical that acts as inflammatory promoter and tumoral facilitator in mammary tissue. Previous studies demonstrated the onset of mammary carcinogenesis at aging when BPA exposure occurred in windows of development/susceptibility. We aim to investigate the inflammatory repercussions of BPA in TME in mammary gland (MG) during neoplastic development in aging. Female Mongolian gerbils were exposed to low (50 µg/kg) or high BPA (5000 µg/kg) doses during pregnancy and lactation. They were euthanized at 18 months of age (aging) and the MG were collected for inflammatory markers and histopathological analysis. Contrarily to control MG, BPA induced carcinogenic development mediated by COX-2 and p-STAT3 expression. BPA was also able to promote macrophage and mast cell (MC) polarization in tumoral phenotype, evidenced by pathways for recruitment and activation of these inflammatory cells and tissue invasiveness triggered by tumor necrosis factor-alpha and transforming growth factor-beta 1 (TGF-β1). Increase of tumor-associated macrophages, M1 (CD68 + iNOS+) and M2 (CD163+) expressing pro-tumoral mediators and metalloproteases was observed; this aspect greatly contributed to stromal remodeling and invasion of neoplastic cells. In addition, the MC population drastically increased in BPA-exposed MG. Tryptase-positive MCs increased in disrupted MG and expressed TGF-β1, contributing to EMT process during carcinogenesis mediated by BPA. BPA exposure interfered in inflammatory response by releasing and enhancing the expression of mediators that contribute to tumor growth and recruitment of inflammatory cells that promote a malignant profile.
Collapse
Affiliation(s)
- Thalles F R Ruiz
- Department of Biological Sciences, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil
| | - Simone J Colleta
- Department of Biological Sciences, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil
| | - Diego D Dos Santos
- Department of Biological Sciences, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil.,Department of Morphology and Genetics, Paulista School of Medicine, Federal University of São Paulo (UNIFESP), São Paulo, São Paulo, Brazil
| | - Nayara F C Castro
- Department of Biological Sciences, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil
| | - Ágata S Cabral
- Laboratory of Genomic Studies, São Paulo State University, São José do Rio Preto, São Paulo, Brazil
| | - Marilia F Calmon
- Laboratory of Genomic Studies, São Paulo State University, São José do Rio Preto, São Paulo, Brazil
| | - Paula Rahal
- Laboratory of Genomic Studies, São Paulo State University, São José do Rio Preto, São Paulo, Brazil
| | - Cristiane D Gil
- Department of Biological Sciences, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil.,Department of Morphology and Genetics, Paulista School of Medicine, Federal University of São Paulo (UNIFESP), São Paulo, São Paulo, Brazil
| | - Ana Paula Girol
- Department of Basics Sciences, University Center Padre Albino (UNIFIPA), Catanduva, São Paulo, Brazil
| | - Patricia S L Vilamaior
- Department of Biological Sciences, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil
| | - Ellen C R Leonel
- Department of Histology, Embryology and Cell Biology, Institute of Biological Sciences (ICB III), Federal University of Goiás (UFG), Goiânia, Goiás, Brazil
| | - Sebastião R Taboga
- Department of Biological Sciences, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil
| |
Collapse
|
12
|
Selective COX-2 Inhibitor Etoricoxib's Liposomal Formulation Attenuates M2 Polarization of TAMs and Enhances its Anti-metastatic Potential. Pharm Res 2023; 40:551-566. [PMID: 36670330 DOI: 10.1007/s11095-022-03444-2] [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: 04/15/2022] [Accepted: 11/20/2022] [Indexed: 01/21/2023]
Abstract
INTRODUCTION COX-2 inhibition in pro-tumoral M2 polarization of Tumor-Associated Macrophages (TAMs) underscore the improved prognosis and response to cancer therapy. Thus, etoricoxib, a COX-2 inhibiting NSAID drug is highly effective against tumorigenesis, but its compromised solubility and associated hepatotoxicity, and cardiotoxicity limit its clinical translation. OBJECTIVE In view of the consequences, the proposed study entails the development of a liposomal formulation for etoricoxib and evaluates its anticancer potential. METHODS AND RESULT Etoricoxib loaded liposome was prepared by thin layer hydration method and characterized as a nearly monodisperse system with particle size (91.64 nm), zeta potential (-44.5 mV), drug loading (17.22%), and entrapment efficiency (94.76%). The developed formulation was administered subcutaneously into the orthotopic 4T1/Balb/c mice model. Its treatment significantly reduced tumor size and skewed M2 polarization of TAMs to a greater extent against free etoricoxib. Furthermore, Tumor tissues analyzed through immunoblotting study confirmed the reduction in Akt phosphorylation at Thr308 residue and pro-tumoral VEGF, MMP-9, and MMP-2 proteins; Moreover, histology studies and microCT analysis of bones revealed the enhanced anti-metastatic potential of etoricoxib delivered through developed formulation against free etoricoxib. CONCLUSION As an epilogue, the developed formulation efficiently delivers poorly soluble etoricoxib, enhances its therapeutic potential as an anti-tumor and anti-metastatic agent, and directs explorative research for clinical translation.
Collapse
|
13
|
Dubey S, Ghosh S, Goswami D, Ghatak D, De R. Immunometabolic attributes and mitochondria-associated signaling of Tumor-Associated Macrophages in tumor microenvironment modulate cancer progression. Biochem Pharmacol 2023; 208:115369. [PMID: 36481347 DOI: 10.1016/j.bcp.2022.115369] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/24/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022]
Abstract
Macrophages are specialized immune cells, which have the capacity to phagocytize and destroy the target cells, including tumor cells. Some macrophages, however on their way to devour the cancer cells undergo a change due to a complex set of signaling pathways. They are induced to change into a polarized state known as M2. The M2 macrophages help in metastasis, tumor suppression, and angiogenesis. The macrophage which gets associated with this TME, are referred to as tumor-associated macrophages (TAMs). TAMS undergo a metabolic reprogramming toward oxidative metabolism for bioenergetic purposes (OXPHOS), fatty acid oxidation (FAO), decreased glycolysis, decreased metabolism via the PPP, and upregulation of arginase 1 (ARG1) which triggers immunosuppressive pro-tumor signaling in the tumor microenvironment (TME) in which mitochondria plays an instrumental role. Reports have suggested that a complex series of interactions and exchange of materials, such as cytokines, metabolic intermediates and sometimes even transfer of mitochondria take place between TAMS and other TME components most importantly cancer cells that reprogram their metabolism to encourage cell growth, division, epithelial to mesenchymal transition, that ultimately play an important role in tumor progression. This review will try to focus on the crosstalk between the TAMs with several other components of TME, what instrumental role mitochondria play in that and also try to explore some of the therapeutic options available in cancer patients.
Collapse
Affiliation(s)
- Srijan Dubey
- Amity Institute of Biotechnology, Amity University, Kolkata, Plot No: 36, 37 & 38, Major Arterial Road, Action Area II, Kadampukur Village, Newtown, Kolkata 700135, West Bengal, India
| | - Sayak Ghosh
- Amity Institute of Biotechnology, Amity University, Kolkata, Plot No: 36, 37 & 38, Major Arterial Road, Action Area II, Kadampukur Village, Newtown, Kolkata 700135, West Bengal, India
| | - Debosmita Goswami
- Amity Institute of Biotechnology, Amity University, Kolkata, Plot No: 36, 37 & 38, Major Arterial Road, Action Area II, Kadampukur Village, Newtown, Kolkata 700135, West Bengal, India
| | - Debapriya Ghatak
- Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| | - Rudranil De
- Amity Institute of Biotechnology, Amity University, Kolkata, Plot No: 36, 37 & 38, Major Arterial Road, Action Area II, Kadampukur Village, Newtown, Kolkata 700135, West Bengal, India.
| |
Collapse
|
14
|
Piperine Reduces Neoplastic Progression in Cervical Cancer Cells by Downregulating the Cyclooxygenase 2 Pathway. Pharmaceuticals (Basel) 2023; 16:ph16010103. [PMID: 36678600 PMCID: PMC9866887 DOI: 10.3390/ph16010103] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 12/22/2022] [Accepted: 12/27/2022] [Indexed: 01/13/2023] Open
Abstract
Cervical cancer is the fourth-most common type of cancer in the world that causes death in women. It is mainly caused by persistent infection by human papillomavirus (HPV) that triggers a chronic inflammatory process. Therefore, the use of anti-inflammatory drugs is a potential treatment option. The effects of piperine, an amino alkaloid derived from Piper nigrum, are poorly understood in cervical cancer inflammation, making it a target of research. This work aimed to investigate the antitumor effect of piperine on cervical cancer and to determine whether this effect is modulated by the cyclooxygenase 2 (PTGS2) pathway using in vitro model of cervical cancer (HeLa, SiHa, CaSki), and non-tumoral (HaCaT) cell lines. The results showed that piperine reduces in vitro parameters associated with neoplastic evolution such as proliferation, viability and migration by cell cycle arrest in the G1/G0 and G2/M phases, with subsequent induction of apoptosis. This action was modulated by downregulation of cyclooxygenase 2 (PTGS2) pathway, which in turn regulates the secretion of cytokines and the expression of mitogen-activated protein kinases (MAPKs), metalloproteinases (MMPs), and their antagonists (TIMPs). These findings indicate the phytotherapeutic potential of piperine as complementary treatment in cervical cancer.
Collapse
|
15
|
Fang P, Han Y, Qu Y, Wang X, Zhang Y, Zhang W, Zhang N, Li G, Ma W. EIF3B stabilizes PTGS2 expression by counteracting MDM2-mediated ubiquitination to promote the development and progression of malignant melanoma. Cancer Sci 2022; 113:4181-4192. [PMID: 36050601 DOI: 10.1111/cas.15543] [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: 08/02/2021] [Revised: 08/05/2022] [Accepted: 08/10/2022] [Indexed: 12/15/2022] Open
Abstract
Malignant melanoma (MM) is a neoplasm that develops from human melanocytes. It was reported that eukaryotic translation initiation factor 3 subunit B (EIF3B) is associated with multiple types of cancers, but its role in MM has not been reported. In the present study, we found that EIF3B was abundantly expressed in MM and was strongly related to lymphatic metastasis and pathological stage of MM patients. In addition, EIF3B depletion could block the progression of MM in vitro and in vivo. In contrast, EIF3B overexpression increased cell proliferation and migration in melanoma cells. More importantly, we identified that EIF3B's driver role in MM was mediated by PTGS2. In detail, we found that EIF3B stabilized PTGS2 expression by inhibiting PTGS2 ubiquitination, which is mediated by the E3 ligase MDM2. Moreover, like EIF3B, silencing PTGS2 could suppress MM development, and more interestingly, it could reverse the situation caused by overexpression of EIF3B in vitro and in vivo. Furthermore, the proliferation and migration inhibited by silencing of EIF3B were also partially recovered by overexpression of PTGS2. Overall, our findings revealed the potential of EIF3B as a therapeutic target for MM. Identification of EIF3B's function in MM may pave the way for future development of more specific and more effective targeted therapy strategies against MM.
Collapse
Affiliation(s)
- Pengli Fang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yikai Han
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanhong Qu
- Oncology Department of Laiyang People's Hospital, Laiyang, China
| | - Xin Wang
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yong Zhang
- Department of Cancer Biotherapy Center, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Wei Zhang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Na Zhang
- Department of Dermatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guangshuai Li
- Department of Plastic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wang Ma
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
16
|
Song Y, Wang J, Wang X, Zhang H, Niu X, Yang Y, Yang X, Yin L, Wang Y, Zhang C, Shui R, Zhang Q, Ji H. Analyzing the multi-target pharmacological mechanism of folium Artemisia argyi acting on breast cancer: a network pharmacology approach. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:1368. [PMID: 36660662 PMCID: PMC9843367 DOI: 10.21037/atm-22-5769] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 12/15/2022] [Indexed: 12/30/2022]
Abstract
Background Folium Artemisia argyi (FAA) is a traditional Chinese herbal medicine that is widely used in the clinic. However, the underlying mechanisms of its anticancer effects have not been fully elucidated. Methods In this study, we applied a network pharmacology approach to identify the potential mechanisms of FAA against breast cancer. To be specific, we screened the active ingredients and potential targets of the FAA through the Traditional Chinese Medicine Systems Pharmacology (TCMSP) database. Meanwhile, we employed the oral bioavailability (OB) and drug-likeness (DL) to search for potential bioactive compounds of FAA. Breast cancer-related target genes data were gathered from the GeneCards and Online Mendelian Inheritance in Man (OMIM) databases, and the protein-protein interaction (PPI) data were acquired from the Search Tool for the Retrieval of Interacting Genes (STRING) database. In addition, we constructed the network and performed Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) Pathway Enrichment Analysis. Results We obtained a total of nine active ingredients and 236 potential targets from FAA to construct a network, which showed that quercetin served as the major ingredient in FAA. AKT1 (RAC-alpha serine/threonine-protein kinase), MYC (Myc proto-oncogene protein), CASP3 (Caspase-3), EGFR (Epidermal growth factor receptor), JUN (Transcription factor AP-1), CCND1 (G1/S-specific cyclin-D1), VEGFA (Vascular endothelial growth factor A), ESR1 (Estrogen receptor), MAPK1 (Mitogen-activated protein kinase 1), and EGF (pro-epidermal growth factor) were identified as key targets of FAA in the treatment of breast cancer. The PPI cluster demonstrated that AKT1 was the seed in this cluster, indicating that AKT1 played a crucial role in connecting other nodes in the PPI network. This enrichment demonstrated that FAA was highly related to signal transduction, endocrine system, replication and repair, as well as cell growth and death. The enrichment results also verified that the underlying mechanisms of FAA against breast cancer might be attributed to the coordinated regulation of several cancer-related pathways, such as the MAPK and mammalian target of rapamycin (mTOR) signaling pathways, among others. Conclusions This study identified the potential targets and pathways of FAA in the treatment of breast cancer using a network pharmacology approach, and systematically elucidated the mechanisms of FAA in the treatment of breast cancer.
Collapse
Affiliation(s)
- Ying Song
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, China
| | - Jinlu Wang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, China
| | - Xiuli Wang
- Department of Clinical Laboratory, The Seventh Hospital in Qiqihar, Qiqihar, China
| | - Han Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, China
| | - Xingjian Niu
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, China
| | - Yue Yang
- Institute of Cancer Prevention and Treatment, Harbin Medical University, Harbin, China
- Heilongjiang Academy of Medical Sciences, Harbin, China
| | - Xudong Yang
- Institute of Cancer Prevention and Treatment, Harbin Medical University, Harbin, China
- Heilongjiang Academy of Medical Sciences, Harbin, China
| | - Lei Yin
- Institute of Cancer Prevention and Treatment, Harbin Medical University, Harbin, China
- Heilongjiang Academy of Medical Sciences, Harbin, China
| | - Yiran Wang
- Institute of Cancer Prevention and Treatment, Harbin Medical University, Harbin, China
- Heilongjiang Academy of Medical Sciences, Harbin, China
| | - Cuiying Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, China
| | - Ruixue Shui
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, China
| | - Qingyuan Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, China
- Institute of Cancer Prevention and Treatment, Harbin Medical University, Harbin, China
- Heilongjiang Academy of Medical Sciences, Harbin, China
| | - Hongfei Ji
- Institute of Cancer Prevention and Treatment, Harbin Medical University, Harbin, China
- Heilongjiang Academy of Medical Sciences, Harbin, China
| |
Collapse
|
17
|
Amer HT, Eissa RA, El Tayebi HM. A cutting-edge immunomodulatory interlinkage between HOTAIR and MALAT1 in tumor-associated macrophages in breast cancer: A personalized immunotherapeutic approach. Front Mol Biosci 2022; 9:1032517. [PMID: 36387279 PMCID: PMC9649622 DOI: 10.3389/fmolb.2022.1032517] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 10/10/2022] [Indexed: 07/30/2023] Open
Abstract
Breast cancer (BC) is one of the most common cancers, accounting for 2.3 million cases worldwide. BC can be molecularly subclassified into luminal A, luminal B HER2-, luminal B HER2+, HER2+, and triple-negative breast cancer (TNBC). These molecular subtypes differ in their prognosis and treatment strategies; thus, understanding the tumor microenvironment (TME) of BC could lead to new potential treatment strategies. The TME hosts a population of cells that act as antitumorigenic such as tumor-associated eosinophils or pro-tumorigenic such as cancer-associated fibroblasts (CAFs), tumor-associated neutrophils (TANs), monocytic-derived populations such as MDSCs, or most importantly "tumor-associated macrophages (TAMs)," which are derived from CD14+ monocytes. TAMs are reported to have the pro-inflammatory phenotype M1, which is found only in the very early stages of tumor and is not correlated with progression; however, the M2 phenotype is anti-inflammatory that is correlated with tumor progression and metastasis. The current study focused on controlling the anti-inflammatory activity in TAMs of hormonal, HER2+, and TNBC by epigenetic fine-tuning of two immunomodulatory proteins, namely, CD80 and mesothelin (MSLN), which are known to be overexpressed in BC with pro-tumorigenic activity. Long non-coding RNAs are crucial key players in tumor progression whether acting as oncogenic or tumor suppressors. We focused on the regulatory role of MALAT1 and HOTAIR lncRNAs and their role in controlling the tumorigenic activity of TAMs. This study observed the impact of manipulation of MALAT1 and HOTAIR on the expression of both CD80 and MSLN in TAMs of BC. Moreover, we analyzed the interlinkage between HOTAIR and MALAT1 as regulators to one another in TAMs of BC. The current study reported an upstream regulatory effect of HOTAIR on MALAT1. Moreover, our results showed a promising use of MALAT1 and HOTAIR in regulating oncogenic immune-modulatory proteins MSLN and CD80 in TAMs of HER2+ and TNBC. The downregulation of MALAT1 and HOTAIR resulted in the upregulation of CD80 and MSLN, which indicates that they might have a cell-specific activity in TAMs. These data shed light on novel key players affecting the anti-inflammatory activity of TAMs as a possible therapeutic target in HER2+ and TNBC.
Collapse
Affiliation(s)
- Hoda T. Amer
- Department of Pharmacology and Toxicology, The Molecular Pharmacology Research Group, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| | - Reda A. Eissa
- Department of Surgery, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Hend M. El Tayebi
- Department of Pharmacology and Toxicology, The Molecular Pharmacology Research Group, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| |
Collapse
|
18
|
The origins of resident macrophages in mammary gland influence the tumorigenesis of breast cancer. Int Immunopharmacol 2022; 110:109047. [DOI: 10.1016/j.intimp.2022.109047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 07/06/2022] [Accepted: 07/08/2022] [Indexed: 11/05/2022]
|
19
|
A review on valorization of different byproducts of mango (Mangifera indica L.) for functional food and human health. FOOD BIOSCI 2022. [DOI: 10.1016/j.fbio.2022.101783] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
20
|
Li J, DeNicola GM, Ruffell B. Metabolism in tumor-associated macrophages. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 367:65-100. [PMID: 35461660 PMCID: PMC9094395 DOI: 10.1016/bs.ircmb.2022.01.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Macrophages functionally adapt to a diverse set of signals, a process that is critical for their role in maintaining or restoring tissue homeostasis. This process extends to cancer, where macrophages respond to a series of inflammatory and metabolic cues that direct a maladaptive healing response. Tumor-associated macrophages (TAMs) have altered glucose, amino acid, and lipid metabolic profiles, and interfering with this metabolic shift can blunt the ability of macrophages to promote tumor growth, metastasis, and the creation of an immunosuppressive microenvironment. Here we will review changes in metabolites and metabolic pathways in TAMs and link these with the phenotypic and functional properties of the cells. We will also discuss current strategies targeting TAM metabolism as a therapeutic intervention in cancer.
Collapse
Affiliation(s)
- Jie Li
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States; Cancer Biology PhD Program, University of South Florida, Tampa, FL, United States
| | - Gina M DeNicola
- Department of Cancer Physiology, Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | - Brian Ruffell
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States; Department of Breast Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States.
| |
Collapse
|
21
|
Liu Y, Wang X, Zhu Y, Cao Y, Wang L, Li F, Zhang Y, Li Y, Zhang Z, Luo J, Deng X, Peng C, Wei G, Chen H, Shen B. The CTCF/LncRNA-PACERR complex recruits E1A binding protein p300 to induce pro-tumour macrophages in pancreatic ductal adenocarcinoma via directly regulating PTGS2 expression. Clin Transl Med 2022; 12:e654. [PMID: 35184402 PMCID: PMC8858628 DOI: 10.1002/ctm2.654] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 10/31/2021] [Accepted: 11/06/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Tumour-associated macrophages (TAMs) play an important role in promoting the progression of pancreatic ductal adenocarcinoma (PDAC). Here, we aimed to study the epigenetic mechanisms in regulating pro-tumour M2-polarised TAMs in the PDAC tumour microenvironment. METHODS This study was conducted based on ex vivo TAMs isolated from PDAC tissues and in vitro THP1-derived TAM model. RNA-sequencing (RNA-seq), assay for transposase-accessible chromatin with sequencing and chromatin immunoprecipitation sequencing were performed to investigate gene expression, chromatin accessibility, transcription factor binding sites and histone modifications. Gene knockdown in THP1-derived TAMs was performed with lentivirus, and the impact of THP1-derived TAMs on invasion and metastasis ability of PDAC cells were investigated with in vitro and in vivo functional assays. RNA-chromatin interaction was analysed by chromatin isolation through RNA purification with sequencing. RNA-protein interaction was studied by RNA immunoprecipitation and RNA pull-down. RESULTS Our data showed that the transcription factor CTCF (CCCTC-binding factor) was highly expressed in TAMs and predicted to be significantly enriched in hyper-accessible chromatin regions when compared to monocytes. High infiltration of CTCF+ TAMs was significantly associated with poor prognosis in PDAC patients. Knockdown of CTCF in THP1-derived TAMs led to the down-regulation of specific markers for M2-polarised TAMs, including CD206 and CD163. When THP1-derived TAMs with CTCF knockdown, they showed a decreased ability of invasion and metastasis. Further integrative analysis of multi-omics data revealed that prostaglandin-endoperoxide synthase 2 (PTGS2) and PTGS2 antisense NF-κB1 complex-mediated expression regulator RNA (PACERR) were critical downstream targets of CTCF and positively correlated with each other, which are closely situated on a chromosome. Knockdown of PACERR exhibited a similar phenotype as observed in CTCF knockdown THP1-derived TAMs. Moreover, PACERR could directly bind to CTCF and recruit histone acetyltransferase E1A binding protein p300 to the promoter regions of PACERR and PTGS2, thereby enhancing histone acetylation and gene transcription, promoting the M2 polarization of TAMs in PDAC. CONCLUSIONS Our study demonstrated a novel epigenetic regulation mechanism of promoting pro-tumour M2-polarised TAMs in the PDAC tumour microenvironment.
Collapse
Affiliation(s)
- Yihao Liu
- Department of General SurgeryPancreatic Disease CenterRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- CAS Key Laboratory of Computational BiologyShanghai Institute of Nutrition and Health, University of Chinese Academy of SciencesChinese Academy of SciencesShanghaiChina
- Research Institute of Pancreatic DiseasesShanghai Jiao Tong University School of MedicineShanghaiChina
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational MedicineShanghai Jiao Tong UniversityShanghaiChina
- Institute of Translational MedicineShanghai Jiaotong UniversityShanghaiChina
| | - Xuelong Wang
- Department of General SurgeryPancreatic Disease CenterRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- CAS Key Laboratory of Computational BiologyShanghai Institute of Nutrition and Health, University of Chinese Academy of SciencesChinese Academy of SciencesShanghaiChina
- Research Institute of Pancreatic DiseasesShanghai Jiao Tong University School of MedicineShanghaiChina
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational MedicineShanghai Jiao Tong UniversityShanghaiChina
- Institute of Translational MedicineShanghai Jiaotong UniversityShanghaiChina
| | - Youwei Zhu
- Department of General SurgeryPancreatic Disease CenterRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Research Institute of Pancreatic DiseasesShanghai Jiao Tong University School of MedicineShanghaiChina
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational MedicineShanghai Jiao Tong UniversityShanghaiChina
- Institute of Translational MedicineShanghai Jiaotong UniversityShanghaiChina
| | - Yizhi Cao
- Department of General SurgeryPancreatic Disease CenterRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Research Institute of Pancreatic DiseasesShanghai Jiao Tong University School of MedicineShanghaiChina
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational MedicineShanghai Jiao Tong UniversityShanghaiChina
- Institute of Translational MedicineShanghai Jiaotong UniversityShanghaiChina
| | - Liwen Wang
- Department of General SurgeryPancreatic Disease CenterRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Research Institute of Pancreatic DiseasesShanghai Jiao Tong University School of MedicineShanghaiChina
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational MedicineShanghai Jiao Tong UniversityShanghaiChina
- Institute of Translational MedicineShanghai Jiaotong UniversityShanghaiChina
| | - Fanlu Li
- Department of General SurgeryPancreatic Disease CenterRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Research Institute of Pancreatic DiseasesShanghai Jiao Tong University School of MedicineShanghaiChina
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational MedicineShanghai Jiao Tong UniversityShanghaiChina
- Institute of Translational MedicineShanghai Jiaotong UniversityShanghaiChina
| | - Yu Zhang
- CAS Key Laboratory of Computational BiologyShanghai Institute of Nutrition and Health, University of Chinese Academy of SciencesChinese Academy of SciencesShanghaiChina
| | - Ying Li
- CAS Key Laboratory of Computational BiologyShanghai Institute of Nutrition and Health, University of Chinese Academy of SciencesChinese Academy of SciencesShanghaiChina
| | - Zhiqiang Zhang
- CAS Key Laboratory of Computational BiologyShanghai Institute of Nutrition and Health, University of Chinese Academy of SciencesChinese Academy of SciencesShanghaiChina
| | - Jiaxin Luo
- Department of General SurgeryPancreatic Disease CenterRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Research Institute of Pancreatic DiseasesShanghai Jiao Tong University School of MedicineShanghaiChina
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational MedicineShanghai Jiao Tong UniversityShanghaiChina
- Institute of Translational MedicineShanghai Jiaotong UniversityShanghaiChina
| | - Xiaxing Deng
- Department of General SurgeryPancreatic Disease CenterRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Research Institute of Pancreatic DiseasesShanghai Jiao Tong University School of MedicineShanghaiChina
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational MedicineShanghai Jiao Tong UniversityShanghaiChina
- Institute of Translational MedicineShanghai Jiaotong UniversityShanghaiChina
| | - Chenghong Peng
- Department of General SurgeryPancreatic Disease CenterRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Research Institute of Pancreatic DiseasesShanghai Jiao Tong University School of MedicineShanghaiChina
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational MedicineShanghai Jiao Tong UniversityShanghaiChina
- Institute of Translational MedicineShanghai Jiaotong UniversityShanghaiChina
| | - Gang Wei
- CAS Key Laboratory of Computational BiologyShanghai Institute of Nutrition and Health, University of Chinese Academy of SciencesChinese Academy of SciencesShanghaiChina
| | - Hao Chen
- Department of General SurgeryPancreatic Disease CenterRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Research Institute of Pancreatic DiseasesShanghai Jiao Tong University School of MedicineShanghaiChina
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational MedicineShanghai Jiao Tong UniversityShanghaiChina
- Institute of Translational MedicineShanghai Jiaotong UniversityShanghaiChina
| | - Baiyong Shen
- Department of General SurgeryPancreatic Disease CenterRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Research Institute of Pancreatic DiseasesShanghai Jiao Tong University School of MedicineShanghaiChina
- State Key Laboratory of Oncogenes and Related Genes, Institute of Translational MedicineShanghai Jiao Tong UniversityShanghaiChina
- Institute of Translational MedicineShanghai Jiaotong UniversityShanghaiChina
| |
Collapse
|
22
|
Ruiz TFR, Leonel ECR, Colleta SJ, Bedolo CM, Pegorin de Campos SG, Taboga SR. Gestational and lactational xenoestrogen exposure disrupts morphology and inflammatory aspects in mammary gland of gerbil mothers during involution. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2022; 89:103785. [PMID: 34896274 DOI: 10.1016/j.etap.2021.103785] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/30/2021] [Accepted: 12/06/2021] [Indexed: 06/14/2023]
Abstract
In the mammary gland (MG), the developmental window for gestational/lactational differentiation and growth is highly vulnerable to hormonal disruption. Here we describe that the MG involution process in female gerbil mothers is delayed by bisphenol A (BPA) exposure during gestation and lactation. The process is directly influenced by changes in expression of extracellular matrix proteases MMP-2, MMP-9, and FAP, and the incidence of collagen and elastin is reduced after 7 and 14 days of weaning. A pro-inflammatory environment in the late involution process was confirmed by higher expression of TNF-α, COX-2 and phospho-STAT3 n the MG stroma, allied to increases in the incidence of macrophages and mast cells. These aspects impacted the proliferative pattern of epithelial cells, which decreased on the 14th post-weaning day. These data confirm that the milk production window of susceptibility is vulnerable to the impact of BPA, which promotes a suggestive pro-tumoral microenvironment during mammary involution.
Collapse
Affiliation(s)
- Thalles Fernando Rocha Ruiz
- Department of Biology, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (Unesp), Rua Cristóvão Colombo, 2265, Jardim Nazareth, 15054-000 São José do Rio Preto, São Paulo, Brazil.
| | - Ellen Cristina Rivas Leonel
- Department of Biology, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (Unesp), Rua Cristóvão Colombo, 2265, Jardim Nazareth, 15054-000 São José do Rio Preto, São Paulo, Brazil; Department of Histology, Embryology and Cell Biology, Institute of Biological Sciences, Federal University of Goiás (UFG), Avenida Esperança, s/n, Câmpus Samambaia, 74690-900 Goiânia, Goiás, Brazil.
| | - Simone Jacovaci Colleta
- Department of Biology, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (Unesp), Rua Cristóvão Colombo, 2265, Jardim Nazareth, 15054-000 São José do Rio Preto, São Paulo, Brazil
| | - Carolina Marques Bedolo
- Department of Biology, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (Unesp), Rua Cristóvão Colombo, 2265, Jardim Nazareth, 15054-000 São José do Rio Preto, São Paulo, Brazil
| | - Silvana Gisele Pegorin de Campos
- Department of Biology, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (Unesp), Rua Cristóvão Colombo, 2265, Jardim Nazareth, 15054-000 São José do Rio Preto, São Paulo, Brazil
| | - Sebastião Roberto Taboga
- Department of Biology, Institute of Biosciences, Humanities and Exact Sciences, São Paulo State University (Unesp), Rua Cristóvão Colombo, 2265, Jardim Nazareth, 15054-000 São José do Rio Preto, São Paulo, Brazil.
| |
Collapse
|
23
|
Wu H, Xu J, Gong G, Zhang Y, Wu S. CircARL8B Contributes to the Development of Breast Cancer Via Regulating miR-653-5p/HMGA2 Axis. Biochem Genet 2021; 59:1648-1665. [PMID: 34050452 DOI: 10.1007/s10528-021-10082-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 05/15/2021] [Indexed: 02/05/2023]
Abstract
Circular RNAs (circRNAs) act as essential regulators in breast cancer (BC) progression. In this paper, we aimed to investigate the functions of circARL8B in BC. The levels of circARL8B, ADP Ribosylation Factor Like GTPase 8B (ARL8B), miR-653-5p and high-mobility group AT-hook 2 (HMGA2) mRNA were examined by qRT-PCR. The stability of circARL8B was determined by RNase R assay and Actinomycin D assay. Cell viability and metastasis were evaluated by Cell Counting Kit-8 (CCK-8) assay and transwell assay, respectively. The levels of cellular phospholipids and triglycerides were measured using relevant kits. Protein levels were measured by western blot analysis. The association between miR-653-5p and circARL8B or HMGA2 was verified by dual-luciferase reporter assay. A murine xenograft model was established to explore the function of circARL8B in vivo. CircARL8B was increased in BC tissues and cells. CircARL8B silencing inhibited cell viability, migration, invasion and fatty acid metabolism in BC cells in vitro and blocked tumor growth in vivo. MiR-653-5p was identified as the target of circARL8B and miR-653-5p was negatively modulated by circARL8B. The suppressive role of circARL8B silencing in BC cell progression was abolished by miR-653-5p downregulation. Moreover, HMGA2 was the target gene of miR-653-5p. HMGA2 overexpression abrogated the effect of miR-653-5p on BC cell development. In addition, circARL8B knockdown might block PGE2/PI3K/AKT/GSK-3β/Wnt/β-catenin pathway. Silencing of circARL8B inhibited cell viability, migration, invasion and fatty acid metabolism via miR-653-5p/HMGA2 axis in BC.
Collapse
Affiliation(s)
- Hansheng Wu
- Department of Thoracic Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Jingyun Xu
- Department of Pathology, The First Affiliated Hospital of Shantou University Medical College, No. 57, Changping Road, Shantou, 515041, Guangdong, China
| | - Guoliang Gong
- Department of Pathology, The First Affiliated Hospital of Shantou University Medical College, No. 57, Changping Road, Shantou, 515041, Guangdong, China.
- Department of Pathology, Chaonan Minsheng Hospital of Shantou, Shantou, Guangdong, China.
| | - Yuanxin Zhang
- Department of Pathology, Chaonan Minsheng Hospital of Shantou, Shantou, Guangdong, China
| | - Shenggui Wu
- Department of Pathology, Chaonan Minsheng Hospital of Shantou, Shantou, Guangdong, China
| |
Collapse
|
24
|
Chiloiro G, Cusumano D, de Franco P, Lenkowicz J, Boldrini L, Carano D, Barbaro B, Corvari B, Dinapoli N, Giraffa M, Meldolesi E, Manfredi R, Valentini V, Gambacorta MA. Does restaging MRI radiomics analysis improve pathological complete response prediction in rectal cancer patients? A prognostic model development. Radiol Med 2021; 127:11-20. [PMID: 34725772 DOI: 10.1007/s11547-021-01421-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 10/14/2021] [Indexed: 12/23/2022]
Abstract
PURPOSE Our study investigated the contribution that the application of radiomics analysis on post-treatment magnetic resonance imaging can add to the assessments performed by an experienced disease-specific multidisciplinary tumor board (MTB) for the prediction of pathological complete response (pCR) after neoadjuvant chemoradiotherapy (nCRT) in locally advanced rectal cancer (LARC). MATERIALS AND METHODS This analysis included consecutively retrospective LARC patients who obtained a complete or near-complete response after nCRT and/or a pCR after surgery between January 2010 and September 2019. A three-step radiomics features selection was performed and three models were generated: a radiomics model (rRM), a multidisciplinary tumor board model (yMTB) and a combined model (CM). The predictive performance of models was quantified using the receiver operating characteristic (ROC) curve, evaluating the area under curve (AUC). RESULTS The analysis involved 144 LARC patients; a total of 232 radiomics features were extracted from the MR images acquired post-nCRT. The yMTB, rRM and CM predicted pCR with an AUC of 0.82, 0.73 and 0.84, respectively. ROC comparison was not significant (p = 0.6) between yMTB and CM. CONCLUSION Radiomics analysis showed good performance in identifying complete responders, which increased when combined with standard clinical evaluation; this increase was not statistically significant but did improve the prediction of clinical response.
Collapse
Affiliation(s)
- Giuditta Chiloiro
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Roma, Italy
| | - Davide Cusumano
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Roma, Italy
| | - Paola de Franco
- Università Cattolica del Sacro Cuore, Largo Francesco Vito, 1, 00168, Roma, Italy.
| | - Jacopo Lenkowicz
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Roma, Italy
| | - Luca Boldrini
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Roma, Italy
| | - Davide Carano
- Università Cattolica del Sacro Cuore, Largo Francesco Vito, 1, 00168, Roma, Italy
| | - Brunella Barbaro
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Roma, Italy
- Università Cattolica del Sacro Cuore, Largo Francesco Vito, 1, 00168, Roma, Italy
| | - Barbara Corvari
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Roma, Italy
| | - Nicola Dinapoli
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Roma, Italy
| | - Martina Giraffa
- Università Cattolica del Sacro Cuore, Largo Francesco Vito, 1, 00168, Roma, Italy
| | - Elisa Meldolesi
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Roma, Italy
| | - Riccardo Manfredi
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Roma, Italy
- Università Cattolica del Sacro Cuore, Largo Francesco Vito, 1, 00168, Roma, Italy
| | - Vincenzo Valentini
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Roma, Italy
- Università Cattolica del Sacro Cuore, Largo Francesco Vito, 1, 00168, Roma, Italy
| | - Maria Antonietta Gambacorta
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168, Roma, Italy
- Università Cattolica del Sacro Cuore, Largo Francesco Vito, 1, 00168, Roma, Italy
| |
Collapse
|
25
|
Mehraj U, Ganai RA, Macha MA, Hamid A, Zargar MA, Bhat AA, Nasser MW, Haris M, Batra SK, Alshehri B, Al-Baradie RS, Mir MA, Wani NA. The tumor microenvironment as driver of stemness and therapeutic resistance in breast cancer: New challenges and therapeutic opportunities. Cell Oncol (Dordr) 2021; 44:1209-1229. [PMID: 34528143 DOI: 10.1007/s13402-021-00634-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/30/2021] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Breast cancer (BC), the second most common cause of cancer-related deaths, remains a significant threat to the health and wellness of women worldwide. The tumor microenvironment (TME), comprising cellular components, such as cancer-associated fibroblasts (CAFs), immune cells, endothelial cells and adipocytes, and noncellular components such as extracellular matrix (ECM), has been recognized as a critical contributor to the development and progression of BC. The interplay between TME components and cancer cells promotes phenotypic heterogeneity, cell plasticity and cancer cell stemness that impart tumor dormancy, enhanced invasion and metastasis, and the development of therapeutic resistance. While most previous studies have focused on targeting cancer cells with a dismal prognosis, novel therapies targeting stromal components are currently being evaluated in preclinical and clinical studies, and are already showing improved efficacies. As such, they may offer better means to eliminate the disease effectively. CONCLUSIONS In this review, we focus on the evolving concept of the TME as a key player regulating tumor growth, metastasis, stemness, and the development of therapeutic resistance. Despite significant advances over the last decade, several clinical trials focusing on the TME have failed to demonstrate promising effectiveness in cancer patients. To expedite clinical efficacy of TME-directed therapies, a deeper understanding of the TME is of utmost importance. Secondly, the efficacy of TME-directed therapies when used alone or in combination with chemo- or radiotherapy, and the tumor stage needs to be studied. Likewise, identifying molecular signatures and biomarkers indicating the type of TME will help in determining precise TME-directed therapies.
Collapse
Affiliation(s)
- Umar Mehraj
- Department of Bioresources, University of Kashmir, Srinagar, Jammu & Kashmir, India
| | - Rais A Ganai
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science & Technology , Awantipora, Jammu & Kashmir, India
| | - Muzafar A Macha
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science & Technology , Awantipora, Jammu & Kashmir, India
| | - Abid Hamid
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal, J&K, India
| | - Mohammed A Zargar
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal, J&K, India
| | - Ajaz A Bhat
- Translational Medicine, Research Branch, Sidra Medicine, Doha, Qatar
| | - Mohd Wasim Nasser
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Mohammad Haris
- Translational Medicine, Research Branch, Sidra Medicine, Doha, Qatar.,Laboratory of Animal Research, Qatar University, Doha, Qatar
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.,Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska, Lincoln, NE, USA.,Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Bader Alshehri
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Almajmaah, Kingdom of Saudi Arabia
| | - Raid Saleem Al-Baradie
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Almajmaah, Kingdom of Saudi Arabia
| | - Manzoor A Mir
- Department of Bioresources, University of Kashmir, Srinagar, Jammu & Kashmir, India.
| | - Nissar Ahmad Wani
- Department of Biotechnology, School of Life Sciences, Central University of Kashmir, Ganderbal, J&K, India.
| |
Collapse
|
26
|
Androgens enhance the ability of intratumoral macrophages to promote breast cancer progression. Oncol Rep 2021; 46:188. [PMID: 34278480 DOI: 10.3892/or.2021.8139] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 05/28/2021] [Indexed: 11/05/2022] Open
Abstract
Androgens are produced locally in breast carcinoma tissues by androgen‑producing enzymes such as 5α‑reductase type 1 (5αRed1) and affect not only breast cancer cells but the tumor microenvironment as well. Tumor‑associated macrophages (TAMs) are primary components of the tumor microenvironment and contribute to tumor progression. Although previous studies suggest that androgen/androgen receptor (AR) signaling in macrophages has important roles in human diseases, androgen action on TAMs has remained largely unknown. We immunolocalized macrophage marker CD163 as well as AR and 5αRed1 in 116 breast carcinomas and correlated them with clinicopathological parameters and clinical outcomes. Moreover, we examined the roles of androgens on macrophages in breast cancer progression using cell lines 4T1 (mouse breast cancer) and RAW264.7 (macrophage) in a tumor‑bearing female BALB/c mouse model. Double immunohistochemistry revealed that AR was sporadically expressed in the macrophages in breast carcinoma tissues. Macrophage infiltration was significantly correlated with an aggressive phenotype of breast carcinomas and worse prognosis, especially in the 5αRed1‑positive group. In a sphere‑forming assay using 4T1 and RAW‑AR cells, which stably express AR, the sphere size was significantly increased due to androgens when 4T1 cells were cocultured with RAW‑AR cells. Furthermore, in vivo experiments revealed that tumor growth and Ki67, a cell proliferation marker, were increased when androgens were stably produced in breast cancer cells and AR was expressed in macrophages. In conclusion, AR is expressed in intratumoral macrophages and is associated with an aggressive phenotype of breast carcinomas, especially when breast cancer cells actively produce androgens. Thus, androgens may enhance the ability of macrophages to promote breast cancer progression.
Collapse
|
27
|
Gómez-Valenzuela F, Escobar E, Pérez-Tomás R, Montecinos VP. The Inflammatory Profile of the Tumor Microenvironment, Orchestrated by Cyclooxygenase-2, Promotes Epithelial-Mesenchymal Transition. Front Oncol 2021; 11:686792. [PMID: 34178680 PMCID: PMC8222670 DOI: 10.3389/fonc.2021.686792] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 05/13/2021] [Indexed: 12/11/2022] Open
Abstract
The tumor microenvironment (TME) corresponds to a complex and dynamic interconnection between the extracellular matrix and malignant cells and their surrounding stroma composed of immune and mesenchymal cells. The TME has constant cellular communication through cytokines that sustain an inflammatory profile, which favors tumor progression, angiogenesis, cell invasion, and metastasis. Although the epithelial-mesenchymal transition (EMT) represents a relevant metastasis-initiating event that promotes an invasive phenotype in malignant epithelial cells, its relationship with the inflammatory profile of the TME is poorly understood. Previous evidence strongly suggests that cyclooxygenase-2 (COX-2) overexpression, a pro-inflammatory enzyme related to chronic unresolved inflammation, is associated with common EMT-signaling pathways. This review article summarizes how COX-2 overexpression, within the context of the TME, orchestrates the EMT process and promotes initial metastatic-related events.
Collapse
Affiliation(s)
- Fernán Gómez-Valenzuela
- Department of Hematology-Oncology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Enrico Escobar
- Department of Oral Pathology and Medicine, Faculty of Dentistry, University of Chile, Santiago, Chile
| | - Ricardo Pérez-Tomás
- Department of Pathology and Experimental Therapy - Bellvitge, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Viviana P Montecinos
- Department of Hematology-Oncology, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
28
|
Mei S, Ma H, Chen X. Anticancer and anti-inflammatory properties of mangiferin: A review of its molecular mechanisms. Food Chem Toxicol 2021; 149:111997. [DOI: 10.1016/j.fct.2021.111997] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 01/04/2021] [Accepted: 01/11/2021] [Indexed: 02/07/2023]
|
29
|
Stromal CCL5 Promotes Breast Cancer Progression by Interacting with CCR3 in Tumor Cells. Int J Mol Sci 2021; 22:ijms22041918. [PMID: 33671956 PMCID: PMC7919043 DOI: 10.3390/ijms22041918] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/12/2021] [Accepted: 02/12/2021] [Indexed: 02/06/2023] Open
Abstract
Chemokines secreted from stromal cells have important roles for interactions with carcinoma cells and regulating tumor progression. C-C motif chemokine ligand (CCL) 5 is expressed in various types of stromal cells and associated with tumor progression, interacting with C-C chemokine receptor (CCR) 1, 3 and 5 expressed in tumor cells. However, the expression on CCL5 and its receptors have so far not been well-examined in human breast carcinoma tissues. We therefore immunolocalized CCL5, as well as CCR1, 3 and 5, in 111 human breast carcinoma tissues and correlated them with clinicopathological characteristics. Stromal CCL5 immunoreactivity was significantly correlated with the aggressive phenotype of breast carcinomas. Importantly, this tendency was observed especially in the CCR3-positive group. Furthermore, the risk of recurrence was significantly higher in the patients with breast carcinomas positive for CCL5 and CCR3 but negative for CCR1 and CCR5, as compared with other patients. In summary, the CCL5-CCR3 axis might contribute to a worse prognosis in breast cancer patients, and these findings will contribute to a better understanding of the significance of the CCL5/CCRs axis in breast carcinoma microenvironment.
Collapse
|
30
|
Makabe T, Koga K, Nagabukuro H, Asada M, Satake E, Taguchi A, Takeuchi A, Miyashita M, Harada M, Hirata T, Hirota Y, Wada-Hiraike O, Fujii T, Osuga Y. Use of selective PGE2 receptor antagonists on human endometriotic stromal cells and peritoneal macrophages. Mol Hum Reprod 2021; 27:gaaa077. [PMID: 33543288 PMCID: PMC7846198 DOI: 10.1093/molehr/gaaa077] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 11/10/2020] [Indexed: 12/16/2022] Open
Abstract
Non-hormonal therapeutic strategies for endometriosis are needed. The aim of this study was to characterize the effects of prostaglandin (PG)E2 receptor inhibitors to explore their potential as novel therapeutic strategies for endometriosis. The expression of PGE2 receptors (EP2 and EP4) in donated tissues from human ovarian endometriosis, adenomyosis and peritoneal endometriosis was examined using immunohistochemistry. Human endometriotic stromal cells (ESC) isolated from ovarian endometriotic tissue and peritoneal macrophages were treated with EP2 and EP4 antagonists. cAMP accumulation and the effect of EP antagonists were measured using cAMP assays. DNA synthesis in ESC was detected using bromodeoxyuridine incorporation analysis. Interleukin (IL)-6 and IL-8 protein levels in ESC supernatants were measured using ELISAs. mRNA expression level for aromatase by ESC, and selected cytokines by peritoneal macrophages was measured using RT-PCR. EP2 and EP4 receptors were expressed in cells derived from control and diseased tissue, ovarian endometriotic, adenomyotic and peritoneal lesions. A selective EP2 antagonist reduced DNA synthesis, cAMP accumulation and IL-1β-induced proinflammatory cytokine secretion and aromatase expression. A selective EP4 antagonist negated IL-1β-induced IL-6 secretion and aromatase expression. In peritoneal macrophages, EP expression was elevated in endometriosis samples but the EP4 antagonist reduced cAMP levels and expression of vascular endothelial growth factor, chemokine ligand 2 and chemokine ligand 3 mRNA. EP2 and EP4 are functioning in endometriosis lesions and peritoneal macrophages, and their selective antagonists can reduce EP-mediated actions, therefore, the EP antagonists are potential therapeutic agents for controlling endometriosis.
Collapse
Affiliation(s)
- Tomoko Makabe
- Department of Obstetrics and Gynecology, The University of Tokyo, Tokyo 113-8655, Japan
| | - Kaori Koga
- Department of Obstetrics and Gynecology, The University of Tokyo, Tokyo 113-8655, Japan
| | - Hiroshi Nagabukuro
- Extra Value Generation Drug Discovery Unit, Takeda Pharmaceutical Company, Fujisawa, Kanagawa 251-8555, Japan
| | - Mari Asada
- Extra Value Generation Drug Discovery Unit, Takeda Pharmaceutical Company, Fujisawa, Kanagawa 251-8555, Japan
| | - Erina Satake
- Department of Obstetrics and Gynecology, The University of Tokyo, Tokyo 113-8655, Japan
| | - Ayumi Taguchi
- Department of Obstetrics and Gynecology, The University of Tokyo, Tokyo 113-8655, Japan
| | - Arisa Takeuchi
- Department of Obstetrics and Gynecology, The University of Tokyo, Tokyo 113-8655, Japan
| | - Mariko Miyashita
- Department of Obstetrics and Gynecology, The University of Tokyo, Tokyo 113-8655, Japan
| | - Miyuki Harada
- Department of Obstetrics and Gynecology, The University of Tokyo, Tokyo 113-8655, Japan
| | - Tetsuya Hirata
- Department of Obstetrics and Gynecology, The University of Tokyo, Tokyo 113-8655, Japan
| | - Yasushi Hirota
- Department of Obstetrics and Gynecology, The University of Tokyo, Tokyo 113-8655, Japan
| | - Osamu Wada-Hiraike
- Department of Obstetrics and Gynecology, The University of Tokyo, Tokyo 113-8655, Japan
| | - Tomoyuki Fujii
- Department of Obstetrics and Gynecology, The University of Tokyo, Tokyo 113-8655, Japan
| | - Yutaka Osuga
- Department of Obstetrics and Gynecology, The University of Tokyo, Tokyo 113-8655, Japan
| |
Collapse
|
31
|
Li D, Zhong S, Zhu Z, Jiang X, Zhang J, Gu J, Chen F. LncRNA MAFG-AS1 Promotes the Progression of Bladder Cancer by Targeting the miR-143-3p/COX-2 Axis. Pathobiology 2020; 87:345-355. [DOI: 10.1159/000509957] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 07/05/2020] [Indexed: 11/19/2022] Open
Abstract
<b><i>Background:</i></b> Long noncoding RNAs (lncRNAs) are potential biomarkers that are very important for the development of cancer. Studies show that lncRNAs are significantly correlated with the carcinogenesis and progression of bladder cancer (BLCA). In this research, we aimed at probing into the role of lncRNA MAFG-AS1 in the tumorigenesis of BLCA. <b><i>Methods:</i></b> RT-qPCR was employed to detect MAFG-AS1 expression in BLCA tissues and cells. MAFG-AS1 siRNA and overexpression plasmid were transfected into 5637 and T24 BLCA cell lines to inhibit or upregulate MAFG-AS1 expression, respectively, and then the regulatory functions of MAFG-AS1 on BLCA cell proliferation, migration, and invasion were assessed using cell counting kit-8 (CCK-8) assay, EdU method, and Transwell experiments, respectively. Dual-luciferase reporter assay and RNA immunoprecipitation were conducted to validate the targeting relationships between MAFG-AS1 and miR-143-3p, and miR-143-3p and COX-2. In addition, miR-143-3p was repressed in MAFG-AS1-silenced 5637 and T24 cell lines, and the function of MAFG-AS1/miR-143-3p axis in BLCA cells was further evaluated. The regulatory effects of MAFG-AS1 and miR-143-3p on the expression of COX-2 protein were detected by Western blot. <b><i>Results:</i></b> MAFG-AS1 was remarkably upregulated in BLCA patient tissues and cell lines, and its high expression was closely related to histological grade, tumor size, and lymph node metastasis. Silencing of MAFG-AS1 inhibited BLCA cell proliferation, metastasis, and invasion, while overexpression of MAFG-AS1 in BLCA cells had opposite biological effects. MAFG-AS1 was proved to target miR-143-3p to repress its expression. Moreover, it was confirmed that MAFG-AS1 and miR-143-3p could modulate COX-2 expression. <b><i>Conclusion:</i></b> The MAFG-AS1/miR-143-3p/COX-2 axis contributes to BLCA progression.
Collapse
|
32
|
Ge Z, Ding S. The Crosstalk Between Tumor-Associated Macrophages (TAMs) and Tumor Cells and the Corresponding Targeted Therapy. Front Oncol 2020; 10:590941. [PMID: 33224886 PMCID: PMC7670061 DOI: 10.3389/fonc.2020.590941] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/12/2020] [Indexed: 12/14/2022] Open
Abstract
Tumor microenvironment (TME) is composed of tumor cells and surrounding non-tumor stromal cells, mainly including tumor associated macrophages (TAMs), endothelial cells, and carcinoma-associated fibroblasts (CAFs). The TAMs are the major components of non-tumor stromal cells, and play an important role in promoting the occurrence and development of tumors. Macrophages originate from bone marrow hematopoietic stem cells and embryonic yolk sacs. There is close crosstalk between TAMs and tumor cells. With the occurrence of tumors, tumor cells secrete various chemokines to recruit monocytes to infiltrate tumor tissues and further promote their M2-type polarization. Importantly, M2-like TAMs can in turn accelerate tumor growth, promote tumor cell invasion and metastasis, and inhibit immune killing to promote tumor progression. Therefore, targeting TAMs in tumor tissues has become one of the principal strategies in current tumor immunotherapy. Current treatment strategies focus on reducing macrophage infiltration in tumor tissues and reprogramming TAMs to M1-like to kill tumors. Although these treatments have had some success, their effects are still limited. This paper mainly summarized the recruitment and polarization of macrophages by tumors, the support of TAMs for the growth of tumors, and the research progress of TAMs targeting tumors, to provide new treatment strategies for tumor immunotherapy.
Collapse
Affiliation(s)
- Zhe Ge
- School of Physical Education & Health Care, East China Normal University, Shanghai, China.,Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, China
| | - Shuzhe Ding
- School of Physical Education & Health Care, East China Normal University, Shanghai, China.,Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, China
| |
Collapse
|
33
|
Liu M, Zhong YB, Shao J, Zhang C, Shi C. Tumor-associated macrophages promote human hepatoma Huh-7 cell migration and invasion through the Gli2/IGF-II/ERK1/2 axis by secreting TGF-β1. Cancer Biol Ther 2020; 21:1041-1050. [PMID: 33081566 DOI: 10.1080/15384047.2020.1824478] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
AIM In this study, we explored the ability of TAMs to affect the malignant phenotype of human hepatoma Huh-7 cells through the Gli2/IGF-II/ERK1/2 pathway. METHODS The TAMs were characterized by flow cytometry and ELISA assays. Huh-7 cells were treated with conditioned medium of TAMs (TAMs-CM), and the proliferation, migration and invasion abilities were measured by CCK-8, Transwell and scratch assays. The levels of TGF-β1, Gli2, IGF-II and related proteins in the ERK1/2 pathway and the epithelial-mesenchymal transition (EMT) process were examined by RT-qPCR and western blot. Huh-7 cells were injected subcutaneously into nude mice with TAMs to explore the role of TAMs in tumor growth. RESULTS The expression levels of TGF-β1, Gli2 and IGF-II and the cell proliferation, migration and invasion abilities were elevated in Huh-7 cells treated with TAMs-CM. TGF-β1 was upregulated in the conditioned medium and was found to be involved in the promotion of migration, invasion and the EMT of Huh-7 cells. The activation of TGF-β1 signaling increased the expression of Gli2. Knockdown of Gli2 decreased the expression of IGF-II and also reversed the promotional effect of the conditioned medium on migration, invasion and the EMT of Huh-7 cells. TGF-β1/Gli2/IGF-II signaling was shown to promote the malignant phenotype of Huh-7 cells by activating the ERK1/2 signaling pathway. Further, TGF-β1 knockdown attenuated the influence of TAMs on tumor growth in mouse model. CONCLUSION The TGF-β1 secreted by TAMs promotes the migration, invasion and EMT of human hepatoma Huh-7 cells through the Gli2/IGF-II/ERK1/2 pathway.
Collapse
Affiliation(s)
- Mei Liu
- Department of Oncology, The First Affiliated Hospital of Nanchang University , Nanchang, Jiangxi Province, P. R China
| | - Yuan-Bin Zhong
- Department of Infectious Diseases & Key Laboratory of Liver Regenerative Medicine of Jiangxi Province, The First Affiliated Hospital of Nanchang University , Nanchang, Jiangxi Province, P. R China
| | - Jia Shao
- Department of Assisted Reproductive, The First Affiliated Hospital of Nanchang University , Nanchang, Jiangxi Province, P. R China
| | - Cheng Zhang
- School of Basic Medical Sciences, Nanchang University , Nanchang, Jiangxi Province, P. R China
| | - Chao Shi
- Department of Oncology, The First Affiliated Hospital of Nanchang University , Nanchang, Jiangxi Province, P. R China
| |
Collapse
|
34
|
Wang J, Zhang Y, Liu L, Cui Z, Shi R, Hou J, Liu Z, Yang L, Wang L, Li Y. NFAT2 overexpression suppresses the malignancy of hepatocellular carcinoma through inducing Egr2 expression. BMC Cancer 2020; 20:966. [PMID: 33023539 PMCID: PMC7542386 DOI: 10.1186/s12885-020-07474-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 09/29/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Nuclear factor of activated T cells 2 (NFAT2) has been reported to regulate the development and malignancy of few tumors. In this study, we aimed to explore the effect of NFAT2 expression on cell fate of HepG2 cell and its potential mechanisms. METHODS Firstly, the pcDNA3.1-NFAT2 plasmid was transfected into HepG2 cells to construct NFAT2 overexpressed HepG2 cells. Then, the chemical count kit-8 cell viability assay, Annexin V-FITC apoptosis detection, EdU labeling proliferation detection, transwell and wound healing experiments were performed. The expression of Egr2 and FasL, and the phosphorylation of AKT and ERK, after ionomycin and PMA co-stimulation, was detected, while the Ca2+ mobilization stimulated by K+ solution was determined. At last, the mRNA and protein expression of NFAT2, Egr2, FasL, COX-2 and c-myc in carcinoma and adjacent tissues was investigated. RESULTS The NFAT2 overexpression suppressed the cell viability, invasion and migration capabilities, and promoted apoptosis of HepG2 cells. NFAT2 overexpression induced the expression of Egr2 and FasL and suppressed the phosphorylation of AKT and ERK. The sensitivity and Ca2+ mobilization of HepG2 cells was also inhibited by NFAT2 overexpression. Compared with adjacent tissues, the carcinoma tissues expressed less NFAT2, Egr2, FasL and more COX-2 and c-myc. CONCLUSION The current study firstly suggested that NFAT2 suppressed the aggression and malignancy of HepG2 cells through inducing the expression of Egr2. The absence of NFAT2 and Egr2 in carcinoma tissues reminded us that NFAT2 may be a promising therapeutic target for hepatocellular carcinoma treatment.
Collapse
Affiliation(s)
- Jian Wang
- Hepatobiliary Surgery Department, Tianjin First Center Hospital, Tianjin Clinical Research Center for Organ Transplantation, Key Laboratory for Critical Care Medicine of the Ministry of Health, No. 24 Fukang Road, Nankai District, Tianjin, 300192, PR China
| | - Yamin Zhang
- Hepatobiliary Surgery Department, Tianjin First Center Hospital, Tianjin Clinical Research Center for Organ Transplantation, Key Laboratory for Critical Care Medicine of the Ministry of Health, No. 24 Fukang Road, Nankai District, Tianjin, 300192, PR China.
| | - Lei Liu
- Department of Transplantation Center, Tianjin First Center Hospital, Tianjin Clinical Research Center for Organ Transplantation, Key Laboratory for Critical Care Medicine of the Ministry of Health, Tianjin, 300192, PR China
| | - Zilin Cui
- Hepatobiliary Surgery Department, Tianjin First Center Hospital, Tianjin Clinical Research Center for Organ Transplantation, Key Laboratory for Critical Care Medicine of the Ministry of Health, No. 24 Fukang Road, Nankai District, Tianjin, 300192, PR China
| | - Rui Shi
- Hepatobiliary Surgery Department, Tianjin First Center Hospital, Tianjin Clinical Research Center for Organ Transplantation, Key Laboratory for Critical Care Medicine of the Ministry of Health, No. 24 Fukang Road, Nankai District, Tianjin, 300192, PR China
| | - Jiancun Hou
- Hepatobiliary Surgery Department, Tianjin First Center Hospital, Tianjin Clinical Research Center for Organ Transplantation, Key Laboratory for Critical Care Medicine of the Ministry of Health, No. 24 Fukang Road, Nankai District, Tianjin, 300192, PR China
| | - Zirong Liu
- Hepatobiliary Surgery Department, Tianjin First Center Hospital, Tianjin Clinical Research Center for Organ Transplantation, Key Laboratory for Critical Care Medicine of the Ministry of Health, No. 24 Fukang Road, Nankai District, Tianjin, 300192, PR China
| | - Long Yang
- Hepatobiliary Surgery Department, Tianjin First Center Hospital, Tianjin Clinical Research Center for Organ Transplantation, Key Laboratory for Critical Care Medicine of the Ministry of Health, No. 24 Fukang Road, Nankai District, Tianjin, 300192, PR China
| | - Lianjiang Wang
- Hepatobiliary Surgery Department, Tianjin First Center Hospital, Tianjin Clinical Research Center for Organ Transplantation, Key Laboratory for Critical Care Medicine of the Ministry of Health, No. 24 Fukang Road, Nankai District, Tianjin, 300192, PR China
| | - Yang Li
- Hepatobiliary Surgery Department, Tianjin First Center Hospital, Tianjin Clinical Research Center for Organ Transplantation, Key Laboratory for Critical Care Medicine of the Ministry of Health, No. 24 Fukang Road, Nankai District, Tianjin, 300192, PR China
| |
Collapse
|
35
|
Zakaria MA, Rajab NF, Chua EW, Selvarajah GT, Masre SF. The Roles of Tissue Rigidity and Its Underlying Mechanisms in Promoting Tumor Growth. Cancer Invest 2020; 38:445-462. [PMID: 32713210 DOI: 10.1080/07357907.2020.1802474] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Tissues become more rigid during tumorigenesis and have been identified as a driving factor for tumor growth. Here, we highlight the concept of tissue rigidity, contributing factors that increase tissue rigidity, and mechanisms that promote tumor growth initiated by increased tissue rigidity. Various factors lead to increased tissue rigidity, promoting tumor growth by activating focal adhesion kinase (FAK) and Rho-associated kinase (ROCK). Consequently, result in recruitment of cancer-associated fibroblasts (CAFs), epithelial-mesenchymal transition (EMT) and tumor protection from immunosurveillance. We also discussed the rationale for targeting tumor tissue rigidity and its potential for cancer treatment.
Collapse
Affiliation(s)
- Muhammad Asyaari Zakaria
- Faculty of Health Sciences, Biomedical Science Programme, Centre for Toxicology & Health Risk Studies, Universiti Kebangsaan Malaysia (UKM), Kuala Lumpur, Malaysia
| | - Nor Fadilah Rajab
- Faculty of Health Sciences, Centre for Healthy Ageing and Wellness, Universiti Kebangsaan Malaysia (UKM), Kuala Lumpur, Malaysia
| | - Eng Wee Chua
- Faculty of Pharmacy, Universiti Kebangsaan Malaysia (UKM), Kuala Lumpur, Malaysia
| | - Gayathri Thevi Selvarajah
- Faculty of Veterinary Medicine, Department of Veterinary Clinical Studies, Universiti Putra Malaysia (UPM), Serdang, Malaysia
| | - Siti Fathiah Masre
- Faculty of Health Sciences, Biomedical Science Programme, Centre for Toxicology & Health Risk Studies, Universiti Kebangsaan Malaysia (UKM), Kuala Lumpur, Malaysia
| |
Collapse
|
36
|
O'Meara T, Marczyk M, Qing T, Yaghoobi V, Blenman K, Cole K, Pelekanou V, Rimm DL, Pusztai L. Immunological Differences Between Immune-Rich Estrogen Receptor-Positive and Immune-Rich Triple-Negative Breast Cancers. JCO Precis Oncol 2020; 4:1900350. [PMID: 32923897 PMCID: PMC7446500 DOI: 10.1200/po.19.00350] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/26/2020] [Indexed: 12/20/2022] Open
Abstract
PURPOSE A subset of estrogen receptor–positive (ER-positive) breast cancer (BC) contains high levels of tumor-infiltrating lymphocytes (TILs), similar to triple-negative BC (TNBC). The majority of immuno-oncology trials target TNBCs because of the greater proportion of TIL-rich TNBCs. The extent to which the immune microenvironments of immune-rich ER-positive BC and TNBC differ is unknown. PATIENTS AND METHODS RNA sequencing data from The Cancer Genome Atlas (TCGA; n = 697 ER-positive BCs; n = 191 TNBCs) were used for discovery; microarray expression data from Molecular Taxonomy of Breast Cancer International Consortium (METABRIC; n = 1,186 ER-positive BCs; n = 297 TNBCs) was used for validation. Patients in the top 25th percentile of a previously published total TIL metagene score distribution were considered immune rich. We compared expression of immune cell markers, immune function metagenes, and immuno-oncology therapeutic targets among immune-rich subtypes. RESULTS Relative fractions of resting mast cells (TCGA Padj = .009; METABRIC Padj = 4.09E-15), CD8+ T cells (TCGA Padj = .015; METABRIC Padj = 0.390), and M2-like macrophages (TCGA Padj= 4.68E-05; METABRIC Padj = .435) were higher in immune-rich ER-positive BCs, but M0-like macrophages (TCGA Padj = 0.015; METABRIC Padj = .004) and M1-like macrophages (TCGA Padj = 9.39E-08; METABRIC Padj = 6.24E-11) were higher in immune-rich TNBCs. Ninety-one immune-related genes (eg, CXCL14, CSF3R, TGF-B3, LRRC32/GARP, TGFB-R2) and a transforming growth factor β (TGF-β) response metagene were significantly overexpressed in immune-rich ER-positive BCs, whereas 41 immune-related genes (eg, IFNG, PD-L1, CTLA4, MAGEA4) were overexpressed in immune-rich TNBCs in both discovery and validation data sets. TGF-β pathway member genes correlated negatively with expression of immune activation markers (IFNG, granzyme-B, perforin) and positively with M2-like macrophages (IL4, IL10, and MMP9) and regulatory T-cell (FOXP3) markers in both subtypes. CONCLUSION Different immunotherapy strategies may be optimal in immune-rich ER-positive BC and TNBC. Drugs targeting the TGF-β pathway and M2-like macrophages are promising strategies in immune-rich ER-positive BCs to augment antitumor immunity.
Collapse
Affiliation(s)
- Tess O'Meara
- Department of Medical Oncology, Yale School of Medicine, New Haven, CT
| | - Michal Marczyk
- Department of Medical Oncology, Yale School of Medicine, New Haven, CT.,Data Mining Division, Silesian University of Technology, Gliwice, Poland
| | - Tao Qing
- Department of Medical Oncology, Yale School of Medicine, New Haven, CT
| | - Vesal Yaghoobi
- Department of Pathology, Yale School of Medicine, New Haven, CT
| | - Kim Blenman
- Department of Medical Oncology, Yale School of Medicine, New Haven, CT
| | - Kimberly Cole
- Department of Pathology, Yale School of Medicine, New Haven, CT
| | - Vasiliki Pelekanou
- Department of Pathology, Yale School of Medicine, New Haven, CT.,Sanofi, Oncology and Translational Medicine, Bridgewater Township, NJ
| | - David L Rimm
- Department of Pathology, Yale School of Medicine, New Haven, CT
| | - Lajos Pusztai
- Department of Medical Oncology, Yale School of Medicine, New Haven, CT
| |
Collapse
|
37
|
Chiang KH, Shieh JM, Shen CJ, Chang TW, Wu PT, Hsu JY, Tsai JP, Chang WC, Chen BK. Epidermal growth factor-induced COX-2 regulates metastasis of head and neck squamous cell carcinoma through upregulation of angiopoietin-like 4. Cancer Sci 2020; 111:2004-2015. [PMID: 32227417 PMCID: PMC7293094 DOI: 10.1111/cas.14400] [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: 09/22/2019] [Revised: 03/18/2020] [Accepted: 03/20/2020] [Indexed: 02/06/2023] Open
Abstract
Epidermal growth factor receptor (EGFR) expression and activation are the major causes of metastasis in cancers such as head and neck squamous cell carcinoma (HNSCC). However, the reciprocal effect of EGF‐induced COX‐2 and angiopoietin‐like 4 (ANGPTL4) on HNSCC metastasis remains unclear. In this study, we revealed that the expression of ANGPTL4 is essential for COX‐2‐derived prostaglandin E2 (PGE2)‐induced tumor cell metastasis. We showed that EGF‐induced ANGPTL4 expression was dramatically inhibited with the depletion and inactivation of COX‐2 by knockdown of COX‐2 and celecoxib treatment, respectively. Prostaglandin E2 induced ANGPTL4 expression in a time‐ and dose‐dependent manners in various HNSCC cell lines through the ERK pathway. In addition, the depletion of ANGPTL4 and MMP1 significantly impeded the PGE2‐induced transendothelial invasion ability of HNSCC cells and the binding of tumor cells to endothelial cells. The induction of molecules involved in the regulation of epithelial‐mesenchymal transition was also dependent on ANGPTL4 expression in PGE2‐treated cells. The depletion of ANGPTL4 further blocked PGE2‐primed tumor cell metastatic seeding of lungs. These results indicate that the EGF‐activated PGE2/ANGPTL4 axis enhanced HNSCC metastasis. The concurrent expression of COX‐2 and ANGPTL4 in HNSCC tumor specimens provides insight into potential therapeutic targets for the treatment of EGFR‐associated HNSCC metastasis.
Collapse
Affiliation(s)
- Kuo-Hwa Chiang
- Department of Internal Medicine, Chi-Mei Medical Center, Tainan, Taiwan
| | - Jiunn-Min Shieh
- Department of Internal Medicine, Chi-Mei Medical Center, Tainan, Taiwan
| | - Chih-Jie Shen
- Graduate Institute of Medical Science, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ting-Wei Chang
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Pei-Ting Wu
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Jinn-Yuan Hsu
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jhih-Peng Tsai
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Wen-Chang Chang
- Graduate Institute of Medical Science, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ben-Kuen Chen
- Graduate Institute of Medical Science, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
38
|
Messex JK, Byrd CJ, Liou GY. Signaling of Macrophages that Contours the Tumor Microenvironment for Promoting Cancer Development. Cells 2020; 9:cells9040919. [PMID: 32283687 PMCID: PMC7226754 DOI: 10.3390/cells9040919] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/05/2020] [Accepted: 04/06/2020] [Indexed: 12/12/2022] Open
Abstract
The immune response is critical in the maintenance of an organism’s health. The immune response can be broken down into two groups. The innate response, which is fast-acting and rids the body of most foreign material before infection occurs, and the adaptive response, a more specific defense against pathogen composed mostly of antibody production and killer cells. Linking the two responses via cytokine and chemokine secretion are macrophages, motile phagocytic cells that ingest and present foreign material playing a role in the innate and adaptive immune response. Although macrophages are necessary for the survival of an organism, studies have also shown macrophages play a more sinister role in the initiation, progression, and metastasis in tumorous cells. In this comprehensive review, we show how macrophages induce such a response through abnormal cellular signaling and creating a cellular microenvironment conducive for tumor growth and metastasis, as well as the future outlook of this field.
Collapse
Affiliation(s)
- Justin K. Messex
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA; (J.K.M.); (C.J.B.)
| | - Crystal J. Byrd
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA; (J.K.M.); (C.J.B.)
- Department of Biological Sciences, Clark Atlanta University, Atlanta, GA 30314, USA
| | - Geou-Yarh Liou
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA; (J.K.M.); (C.J.B.)
- Department of Biological Sciences, Clark Atlanta University, Atlanta, GA 30314, USA
- Correspondence: ; Tel.: +1-(404)-880-6981; Fax: +1-(404)-880-6756
| |
Collapse
|
39
|
Pattern of expression of immune- and stroma-associated genes in blood of mice with experimental B16 melanoma. UKRAINIAN BIOCHEMICAL JOURNAL 2020. [DOI: 10.15407/ubj92.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
40
|
Augimeri G, Plastina P, Gionfriddo G, Rovito D, Giordano C, Fazio A, Barone I, Catalano S, Andò S, Bonofiglio D, Meijerink J, Witkamp R. N-Eicosapentaenoyl Dopamine, A Conjugate of Dopamine and Eicosapentaenoic Acid (EPA), Exerts Anti-inflammatory Properties in Mouse and Human Macrophages. Nutrients 2019; 11:nu11092247. [PMID: 31540502 PMCID: PMC6769480 DOI: 10.3390/nu11092247] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 09/04/2019] [Accepted: 09/16/2019] [Indexed: 12/28/2022] Open
Abstract
A large body of evidence suggests that dietary n-3 polyunsaturated fatty acids (PUFAs), including eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), contribute to a reduced inflammatory tone thereby lowering the risk for several chronic and degenerative diseases. Different mechanisms have been proposed to explain these anti-inflammatory effects, including those involving endocannabinoids and endocannabinoid-like molecules. In this context, fatty acid amides (FAAs), conjugates of fatty acids with amines or amino acids, are an emerging class of compounds. Dopamine conjugates of DHA (N-docosahexaenoyl dopamine, DHDA) and EPA (N-eicosapentaenoyl dopamine, EPDA) have previously been shown to induce autophagy, apoptosis, and cell death in different tumor lines. Additionally, DHDA has displayed anti-inflammatory properties in vitro. Here, we tested the immune-modulatory properties of EPDA in mouse RAW 264.7 and human THP-1 macrophages stimulated with lipopolysaccharide (LPS). EPDA suppressed the production of monocyte chemoattractant protein-1 (MCP-1), and interleukin-6 (IL-6) in both cell lines, and nitric oxide (NO), and macrophage-inflammatory protein-3α (MIP3A) in RAW 264.7 macrophages. At a transcriptional level, EPDA attenuated cyclooxygenase-2 (COX-2) expression in both cell lines and that of MCP-1, IL-6, and interleukin-1β (IL-1β) in THP-1 macrophages. Although further research is needed to reveal whether EPDA is an endogenous metabolite, our data suggest that this EPA-derived conjugate possesses interesting immune-modulating properties.
Collapse
Affiliation(s)
- Giuseppina Augimeri
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (G.A.); (G.G.); (D.R.); (C.G.); (A.F.); (I.B.); (S.C.); (S.A.)
| | - Pierluigi Plastina
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (G.A.); (G.G.); (D.R.); (C.G.); (A.F.); (I.B.); (S.C.); (S.A.)
| | - Giulia Gionfriddo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (G.A.); (G.G.); (D.R.); (C.G.); (A.F.); (I.B.); (S.C.); (S.A.)
| | - Daniela Rovito
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (G.A.); (G.G.); (D.R.); (C.G.); (A.F.); (I.B.); (S.C.); (S.A.)
| | - Cinzia Giordano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (G.A.); (G.G.); (D.R.); (C.G.); (A.F.); (I.B.); (S.C.); (S.A.)
| | - Alessia Fazio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (G.A.); (G.G.); (D.R.); (C.G.); (A.F.); (I.B.); (S.C.); (S.A.)
| | - Ines Barone
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (G.A.); (G.G.); (D.R.); (C.G.); (A.F.); (I.B.); (S.C.); (S.A.)
| | - Stefania Catalano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (G.A.); (G.G.); (D.R.); (C.G.); (A.F.); (I.B.); (S.C.); (S.A.)
| | - Sebastiano Andò
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (G.A.); (G.G.); (D.R.); (C.G.); (A.F.); (I.B.); (S.C.); (S.A.)
| | - Daniela Bonofiglio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende (CS), Italy; (G.A.); (G.G.); (D.R.); (C.G.); (A.F.); (I.B.); (S.C.); (S.A.)
- Correspondence: (D.B.); (R.W.); Tel.: +39-0984-496208 (D.B.); +31-0317-485136 (R.W.)
| | - Jocelijn Meijerink
- Division of Human Nutrition and Health, Wageningen University, 6700 AA Wageningen, The Netherlands;
| | - Renger Witkamp
- Division of Human Nutrition and Health, Wageningen University, 6700 AA Wageningen, The Netherlands;
- Correspondence: (D.B.); (R.W.); Tel.: +39-0984-496208 (D.B.); +31-0317-485136 (R.W.)
| |
Collapse
|
41
|
Barriga V, Kuol N, Nurgali K, Apostolopoulos V. The Complex Interaction between the Tumor Micro-Environment and Immune Checkpoints in Breast Cancer. Cancers (Basel) 2019; 11:cancers11081205. [PMID: 31430935 PMCID: PMC6721629 DOI: 10.3390/cancers11081205] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 08/08/2019] [Accepted: 08/12/2019] [Indexed: 02/07/2023] Open
Abstract
The progression of breast cancer and its association with clinical outcome and treatment remain largely unexplored. Accumulating data has highlighted the interaction between cells of the immune system and the tumor microenvironment in cancer progression, and although studies have identified multiple facets of cancer progression within the development of the tumor microenvironment (TME) and its constituents, there is lack of research into the associations between breast cancer subtype and staging. Current literature has provided insight into the cells and pathways associated with breast cancer progression through expression analysis. However, there is lack of co-expression studies between immune pathways and cells of the TME that form pro-tumorigenic relationships contributing to immune-evasion. We focus on the immune checkpoint and TME elements that influence cancer progression, particularly studies in molecular subtypes of breast cancer.
Collapse
Affiliation(s)
- Vanessa Barriga
- College of Health and Biomedicine, Victoria University, Melbourne 3030, Australia
- Institute for Health and Sport, Victoria University, Melbourne 3030, Australia
| | - Nyanbol Kuol
- Institute for Health and Sport, Victoria University, Melbourne 3030, Australia
| | - Kulmira Nurgali
- Institute for Health and Sport, Victoria University, Melbourne 3030, Australia
| | | |
Collapse
|
42
|
Pham TH, Bak Y, Kwon T, Kwon SB, Oh JW, Park JH, Choi YK, Hong JT, Yoon DY. Interleukin-32θ inhibits tumor-promoting effects of macrophage-secreted CCL18 in breast cancer. Cell Commun Signal 2019; 17:53. [PMID: 31126309 PMCID: PMC6534939 DOI: 10.1186/s12964-019-0374-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Accepted: 05/20/2019] [Indexed: 12/24/2022] Open
Abstract
Background Tumor-associated macrophages can promote breast cancer metastasis by secreting cytokines and growth factors. Interleukin (IL)-32θ, a newly identified IL-32 isoform, was previously shown to down-regulate various proinflammatory factors of macrophages. Here, we report the presence of IL-32θ in breast cancer tissues and evaluate its effects on macrophage-regulated breast cancer metastasis. Methods RT-qPCR was used to analyze the mRNA expression of IL-32θ, Chemokine (C-C motif) ligand 18 (CCL18) in breast cancer tissues. In vitro cell-based experiments using IL-32θ-expressing MDA-MB-231 cells were conducted to examine the effects of IL-32θ on metastasis and its molecular signaling. In vivo xenograft, immunohistochemistry, and optical imaging models were generated to support in vitro and clinical findings. Results The clinical data displayed opposite expression patterns of CCL18 and IL-32θ mRNA in macrophage-infiltrated breast tumor tissues compared with those in the other tissues tested. In MDA-MB-231 cells, IL-32θ overexpression attenuated migration, invasion, tumor-promoting factors, and increased epithelial markers levels upon treatment with conditioned media from THP-1-derived macrophages. Additionally, IL-32θ expression in a xenograft model led to a remarkable decrease in tumor size and macrophage-stimulated tumor promotion. This inhibition was mediated through a direct interaction with protein kinase C-δ (PKCδ), subsequently eliminating the downstream factors STAT3 and NF-κB. Blocking CCL18 during co-culture of macrophages and breast cancer cells reduced the levels of breast cancer progression-related factors and PKCδ downstream signaling suggesting CCL18 as the main macrophage-secreted factors triggering the signaling pathway inhibited by IL-32θ. Conclusions Our findings demonstrate a novel role of IL-32θ as an intracellular modulator to suppress macrophage-promoted breast cancer progression by targeting CCL18-dependent signaling. Electronic supplementary material The online version of this article (10.1186/s12964-019-0374-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Thu-Huyen Pham
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Jayang-dong, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Yesol Bak
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Jayang-dong, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Taeho Kwon
- Primate Resource Center, Division of Bioinfrastructure, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup, Jeollabuk-do, Republic of Korea
| | - Sae-Bom Kwon
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Jayang-dong, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Jae-Wook Oh
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul, Republic of Korea
| | - Jong-Hyung Park
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Yang-Kyu Choi
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul, Republic of Korea
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk, 28160, Republic of Korea.
| | - Do-Young Yoon
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Jayang-dong, Gwangjin-gu, Seoul, 05029, Republic of Korea.
| |
Collapse
|
43
|
Abstract
The tumor immune landscape gained considerable interest based on the knowledge that genetic aberrations in cancer cells alone are insufficient for tumor development. Macrophages are basically supporting all hallmarks of cancer and owing to their tremendous plasticity they may exert a whole spectrum of anti-tumor and pro-tumor activities. As part of the innate immune response, macrophages are armed to attack tumor cells, alone or in concert with distinct T cell subsets. However, in the tumor microenvironment, they sense nutrient and oxygen gradients, receive multiple signals, and respond to this incoming information with a phenotype shift. Often, their functional output repertoire is shifted to become tumor-supportive. Incoming and outgoing signals are chemically heterogeneous but also comprise lipid mediators. Here, we review the current understanding whereby arachidonate metabolites derived from the cyclooxygenase and lipoxygenase pathways shape the macrophage phenotype in a tumor setting. We discuss these findings in the context of cyclooxygenase-2 (COX-2) and microsomal prostaglandin E synthase-1 (mPGES-1) expression and concomitant prostaglandin E2 (PGE2) formation. We elaborate the multiple actions of this lipid in affecting macrophage biology, which are sensors for and generators of this lipid. Moreover, we summarize properties of 5-lipoxygenases (ALOX5) and 15-lipoxygenases (ALOX15, ALOX15B) in macrophages and clarify how these enzymes add to the role of macrophages in a dynamically changing tumor environment. This review will illustrate the potential routes how COX-2/mPGES-1 and ALOX5/-15 in macrophages contribute to the development and progression of a tumor.
Collapse
Affiliation(s)
- Andreas Weigert
- Institute of Biochemistry I/Pathobiochemistry, Faculty of Medicine, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Elisabeth Strack
- Institute of Biochemistry I/Pathobiochemistry, Faculty of Medicine, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Ryan G Snodgrass
- Institute of Biochemistry I/Pathobiochemistry, Faculty of Medicine, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Bernhard Brüne
- Institute of Biochemistry I/Pathobiochemistry, Faculty of Medicine, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany. .,German Cancer Consortium (DKTK), Partner Site Frankfurt, Frankfurt, Germany.
| |
Collapse
|
44
|
TNF-α derived from M2 tumor-associated macrophages promotes epithelial-mesenchymal transition and cancer stemness through the Wnt/β-catenin pathway in SMMC-7721 hepatocellular carcinoma cells. Exp Cell Res 2019; 378:41-50. [PMID: 30844387 DOI: 10.1016/j.yexcr.2019.03.005] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 03/01/2019] [Accepted: 03/03/2019] [Indexed: 02/08/2023]
Abstract
M2-polarized tumor-associated macrophages (M2-TAMs) infiltrating the tumor microenvironment contribute to hepatocellular carcinoma (HCC) progression. It was reported that cancer cells undergoing EMT will acquire stemness characteristics. Here, the HCC SMMC-7721 cell line was co-cultured with M2-TAMs polarized from THP-1 cells in vitro. In in vivo studies, we used nude mice subcutaneous tumor model to test whether the growth of the tumor was affected by M2-TAMs. Subsequently, EMT, stemness and Wnt/β-catenin pathway related markers were detected in cells and subcutaneous tumor tissues. TNF-α was also assessed in both the co-culture system supernatants and in nude mice serum. We found that SMMC-7721 underwent EMT and acquired stemness after co-culture with M2-TAMs, and resulted in larger tumor size following subcutaneous injection of SMMC-7721 suspended in M2-TAMs supernatants compared with SMMC-7721 alone. Enzyme linked immunosorbent assay showed that TNF-α expression was elevated in supernatants of M2-TAMs and positively correlated with tumor size in the serum of nude mice. Furthermore, we found that the Wnt/β-catenin pathway was a downstream target of TNF-α and that the Wnt/β-catenin inhibitor ICG-001 partially reversed EMT and attenuated cancer stemness. Our results indicate that TNF-α derived from M2-TAMs promote EMT and cancer stemness cells via the Wnt/β-catenin pathway.
Collapse
|
45
|
Wallace TR, Tarullo SE, Crump LS, Lyons TR. Studies of postpartum mammary gland involution reveal novel pro-metastatic mechanisms. ACTA ACUST UNITED AC 2019; 5. [PMID: 30847405 PMCID: PMC6400586 DOI: 10.20517/2394-4722.2019.01] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Postpartum involution is the process by which the lactating mammary gland returns to the pre-pregnant state after weaning. Expression of tumor-promotional collagen, upregulation of matrix metalloproteinases, infiltration of M2 macrophages, and remodeling of blood and lymphatic vasculature are all characteristics shared by the involuting mammary gland and breast tumor microenvironment. The tumor promotional nature of the involuting mammary gland is perhaps best evidenced by cases of postpartum breast cancer (PPBC), or those cases diagnosed within 10 years of most recent childbirth. Women with PPBC experience more aggressive disease and higher risk of metastasis than nulliparous patients and those diagnosed outside the postpartum window. Semaphorin 7a (SEMA7A), cyclooxygenase-2 (COX-2), and collagen are all expressed in the involuting mammary gland and, together, predict for decreased metastasis free survival in breast cancer. Studies investigating the role of these proteins in involution have been important for understanding their contributions to PPBC. Postpartum involution thus represents a valuable model for the identification of novel molecular drivers of PPBC and classical cancer hallmarks. In this review, we will highlight the similarities between involution and cancer in the mammary gland, and further define the contribution of SEMA7A/COX-2/collagen interplay to postpartum involution and breast tumor progression and metastasis.
Collapse
Affiliation(s)
- Taylor R Wallace
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.,Young Women's Breast Cancer Translational Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Sarah E Tarullo
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.,Young Women's Breast Cancer Translational Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Lyndsey S Crump
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.,Young Women's Breast Cancer Translational Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Traci R Lyons
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.,Young Women's Breast Cancer Translational Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.,University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.,University of Colorado Gates Center for Regenerative Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
46
|
Tandon I, Sharma NK. Macrophage Flipping from Foe to Friend: A Matter of Interest in Breast Carcinoma Heterogeneity Driving Drug Resistance. Curr Cancer Drug Targets 2019; 19:189-198. [DOI: 10.2174/1568009618666180628102247] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 05/09/2018] [Accepted: 06/13/2018] [Indexed: 02/06/2023]
Abstract
Tumor heterogeneity within various cancer types including breast carcinoma is pivotal in the manifestations of tumor hallmarks. Tumor heterogeneity is seen as a common landscape where intra-tumoral components including cellular and non-cellular factors create an interface with outside environment that leads to the unique identity of a specific cancer type. Among various contributors to tumor heterogeneity, cellular heterogeneity immensely plays a role in drug resistance and relapse of cancer. Within cellular heterogeneity of tumor, tumor-associated macrophages (TAMs) are the pro-tumor type of immune cells that promote growth, metastasis and drug resistance in breast carcinoma and other cancer types. Revealing the molecular aspects of TAMs can provide a breakthrough to remove therapeutics blockade to existing drugs and this understanding in future will pave the way for a new class of cancer immunotherapeutic. This review addresses current understanding of the role of TAMs in breast carcinoma hallmarks and clarifies the current scenario of pre-clinical drugs directed to tame pro-cancer TAMs.
Collapse
Affiliation(s)
- Ishita Tandon
- Cancer and Translational Research Lab, Dr. D. Y. Patil Biotechnology & Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Pune, Maharashtra 411033, India
| | - Nilesh Kumar Sharma
- Cancer and Translational Research Lab, Dr. D. Y. Patil Biotechnology & Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Pune, Maharashtra 411033, India
| |
Collapse
|
47
|
Pelekanou V, Villarroel-Espindola F, Schalper KA, Pusztai L, Rimm DL. CD68, CD163, and matrix metalloproteinase 9 (MMP-9) co-localization in breast tumor microenvironment predicts survival differently in ER-positive and -negative cancers. Breast Cancer Res 2018; 20:154. [PMID: 30558648 PMCID: PMC6298021 DOI: 10.1186/s13058-018-1076-x] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 11/07/2018] [Indexed: 12/11/2022] Open
Abstract
Background The role of tumor-associated macrophages (TAMs) in the cancer immune landscape and their potential as treatment targets or modulators of response to treatment are gaining increasing interest. TAMs display high molecular and functional complexity. Therefore their objective assessment as breast cancer biomarkers is critical. The aims of this study were to objectively determine the in situ expression and significance of TAM biomarkers (CD68, CD163, and MMP-9) in breast cancer and to identify subclasses of patients who could benefit from TAM-targeting therapies. Methods We measured CD68, CD163, and MMP-9 protein expression in formalin-fixed paraffin-embedded tissues of breast carcinomas represented in tissue microarray format using multiplexed quantitative immunofluorescence (QIF) in two independent Yale cohorts: cohort A—n = 398, estrogen receptor–positive (ER+) and ER− cases—and the triple-negative breast cancer (TNBC)-only cohort B (n = 160). Associations between macrophage markers, ER status, and survival were assessed. Protein expression measured by QIF was compared with mRNA expression data from the METABRIC study. Results All three macrophage markers were co-expressed, displaying higher expression in ER− cancers. High pan-macrophage marker CD68 correlated with poorer overall survival (OS) only in ER− cases of cohort A (P = 0.02). High expression of CD163 protein in TAMs was associated with improved OS in ER− cases (cohort A, P = 0.03 and TNBC cohort B, P = 0.04, respectively) but not in ER+ cancers. MMP-9 protein was not individually associated with OS. High expression of MMP-9 in the CD68+/CD163+ TAMs was associated with worse OS in ER+ tumors (P <0.001) but not in ER− cancers. In the METABRIC dataset, mRNA levels followed the co-expression pattern observed in QIF but did not always show the same trend regarding OS. Conclusions Macrophage activity markers correlate with survival differently in ER+ and ER− cancers. The association between high co-expression and co-localization of MMP-9/CD163/CD68 and poor survival in ER+ cancers suggests that these cancers may be candidates for macrophage-targeted therapies. Electronic supplementary material The online version of this article (10.1186/s13058-018-1076-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Vasiliki Pelekanou
- Department of Pathology, Yale School of Medicine, 310 Cedar Street, P.O. Box 208023, New Haven, CT, 06520, USA. .,Sanofi US Services Inc., Bridgewater Township, USA.
| | - Franz Villarroel-Espindola
- Department of Pathology, Yale School of Medicine, 310 Cedar Street, P.O. Box 208023, New Haven, CT, 06520, USA
| | - Kurt A Schalper
- Department of Pathology, Yale School of Medicine, 310 Cedar Street, P.O. Box 208023, New Haven, CT, 06520, USA
| | - Lajos Pusztai
- Department of Medical Oncology, Yale School of Medicine, 330 Cedar Street, New Haven, 06520, CT, USA
| | - David L Rimm
- Department of Pathology, Yale School of Medicine, 310 Cedar Street, P.O. Box 208023, New Haven, CT, 06520, USA
| |
Collapse
|
48
|
Guo S, Deng CX. Effect of Stromal Cells in Tumor Microenvironment on Metastasis Initiation. Int J Biol Sci 2018; 14:2083-2093. [PMID: 30585271 PMCID: PMC6299363 DOI: 10.7150/ijbs.25720] [Citation(s) in RCA: 190] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Accepted: 06/02/2018] [Indexed: 12/11/2022] Open
Abstract
The cellular environment where tumor cells reside is called the tumor microenvironment (TME), which consists of borders, blood vessels, lymph vessels, extracellular matrix (ECM), stromal cells, immune/inflammatory cells, secreted proteins, RNAs and small organelles. By dynamically interacting with tumor cells, stromal cells participate in all stages of tumor initiation, progression, metastasis, recurrence and drug response, and consequently, affect the fate of patients. During the processes of tumor evolution and metastasis initiation, stromal cells in TME also experience some changes and play roles in both the suppression and promotion of metastasis, while the overall function of stromal cells is beneficial for cancer cell survival and movement. In this review, we examine the effects of stromal cells in TME on metastasis initiation, including angiogenesis, epithelial-mesenchymal transition (EMT) and invasion. We also highlight functions of proteins, RNAs and small organelles secreted by stromal cells in their influences on multiple stages of tumor metastasis.
Collapse
Affiliation(s)
| | - Chu-Xia Deng
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| |
Collapse
|
49
|
Hashemi Goradel N, Najafi M, Salehi E, Farhood B, Mortezaee K. Cyclooxygenase-2 in cancer: A review. J Cell Physiol 2018; 234:5683-5699. [PMID: 30341914 DOI: 10.1002/jcp.27411] [Citation(s) in RCA: 459] [Impact Index Per Article: 76.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Accepted: 08/22/2018] [Indexed: 12/17/2022]
Abstract
Cyclooxygenase-2 (COX-2) is frequently expressed in many types of cancers exerting a pleiotropic and multifaceted role in genesis or promotion of carcinogenesis and cancer cell resistance to chemo- and radiotherapy. COX-2 is released by cancer-associated fibroblasts (CAFs), macrophage type 2 (M2) cells, and cancer cells to the tumor microenvironment (TME). COX-2 induces cancer stem cell (CSC)-like activity, and promotes apoptotic resistance, proliferation, angiogenesis, inflammation, invasion, and metastasis of cancer cells. COX-2 mediated hypoxia within the TME along with its positive interactions with YAP1 and antiapoptotic mediators are all in favor of cancer cell resistance to chemotherapeutic drugs. COX-2 exerts most of the functions through its metabolite prostaglandin E2. In some and limited situations, COX-2 may act as an antitumor enzyme. Multiple signals are contributed to the functions of COX-2 on cancer cells or its regulation. Members of mitogen-activated protein kinase (MAPK) family, epidermal growth factor receptor (EGFR), and nuclear factor-κβ are main upstream modulators for COX-2 in cancer cells. COX-2 also has interactions with a number of hormones within the body. Inhibition of COX-2 provides a high possibility to exert therapeutic outcomes in cancer. Administration of COX-2 inhibitors in a preoperative setting could reduce the risk of metastasis in cancer patients. COX-2 inhibition also sensitizes cancer cells to treatments like radio- and chemotherapy. Chemotherapeutic agents adversely induce COX-2 activity. Therefore, choosing an appropriate chemotherapy drugs along with adjustment of the type and does for COX-2 inhibitors based on the type of cancer would be an effective adjuvant strategy for targeting cancer.
Collapse
Affiliation(s)
- Nasser Hashemi Goradel
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Eniseh Salehi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Farhood
- Departments of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| |
Collapse
|
50
|
MiRNA-206 suppresses PGE2-induced colorectal cancer cell proliferation, migration, and invasion by targetting TM4SF1. Biosci Rep 2018; 38:BSR20180664. [PMID: 30135139 PMCID: PMC6146292 DOI: 10.1042/bsr20180664] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 08/10/2018] [Accepted: 08/20/2018] [Indexed: 12/21/2022] Open
Abstract
MiRNA (miR)-206 plays a tumor suppressor role in various cancer types. Here, we investigated whether miR-206 is involved in prostaglandin E2 (PGE2)-induced epithelial–mesenchymal transition (EMT) in colorectal cancer (CRC) cells through the targetting of transmembrane 4 L six family member 1 (TM4SF1). The effect of PGE2 on growth and apoptosis of CRC cells was evaluated using the MTT assay and flow cytometry analysis, respectively. TM4SF1 and miR-206 expression levels were determined with quantitative polymerase chain reaction (qRT-PCR) in CRC tissues and cell lines. The concentration of PGE2 in the serum of CRC patients and healthy controls was measured with an ELISA kit. A miR-206 or TM4SF1 construct was transfected into cells with PGE2. Transwell migration and invasion assays were used to examine cell migration and invasion properties. Additionally, a luciferase assay was performed to determine whether TM4SF1 was directly targetted by miR-206. We found that miR-206 was down-regulated and TM4SF1 was up-regulated in human CRC tissues and cell lines. Moreover, miR-206 was negatively correlated with TM4SF1 expression. Bioinformatics analysis and a luciferase reporter assay revealed that miR-206 directly targetted the 3′-untranslated region (UTR) of TM4SF1, and TM4SF1 expression was reduced by miR-206 overexpression at both the mRNA and protein levels. Additionally, PGE2 significantly suppressed the expression of miR-206 and increased the expression of TM4SF1 in CRC cells. PGE2 induction led to enhanced CRC cell proliferation, migration, and invasion. Moreover, the overexpression of miR-206 decreased CRC cell proliferation, migration, and invasion compared with control group in PGE2-induced cells, and these effects could be recovered by the overexpression of TM4SF1. Overexpression of miR-206 also suppressed the expression of β-catenin, VEGF, MMP-9, Snail, and Vimentin and enhanced E-cadherin expression in PGE2-induced cells. These results could be reversed by the overexpression of TM4SF1. At last, up-regulation of miR-206 suppressed expression of p-AKT and p-ERK by targetting TM4SF1 in PGE2-induced cells. Our results provide further evidence that miR-206 has a protective effect on PGE2-induced colon carcinogenesis.
Collapse
|