1
|
Zhao Y, Tang X, Lei T, Fu D, Zhang H. Lipocalin-2 promotes breast cancer brain metastasis by enhancing tumor invasion and modulating brain microenvironment. Front Oncol 2024; 14:1448089. [PMID: 39188682 PMCID: PMC11345181 DOI: 10.3389/fonc.2024.1448089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 07/29/2024] [Indexed: 08/28/2024] Open
Abstract
Breast cancer is the leading cancer diagnosed in women globally, with brain metastasis emerging as a major cause of death, particularly in human epidermal growth factor receptor 2 positive and triple-negative breast cancer subtypes. Comprehensive understanding of the molecular foundations of central nervous system metastases is imperative for the evolution of efficacious treatment strategies. Lipocalin-2 (LCN2), a secreted iron transport protein with multiple functions, has been linked to the progression of breast cancer brain metastasis (BCBM). In primary tumors, LCN2 promotes the proliferation and angiogenesis of breast cancer cells, triggers the epithelial-mesenchymal transition, interacts with matrix metalloproteinase-9, thereby facilitating the reorganization of the extracellular matrix and enhancing cancer cell invasion and migration. In brain microenvironment, LCN2 undermines the blood-brain barrier and facilitates tumor seeding in the brain by modulating the behavior of key cellular components. In summary, this review meticulously examines the fuel role of LCN2 in BCBM cascade, and investigates the potential mechanisms involved. It highlights the potential of LCN2 as both a therapeutic target and biomarker, indicating that interventions targeting LCN2 may offer improved outcomes for patients afflicted with BCBM.
Collapse
Affiliation(s)
- Yang Zhao
- Department of Microbiology and Immunology, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Xiaogen Tang
- Department of Microbiology and Immunology, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Tingting Lei
- Department of Microbiology and Immunology, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Dongwei Fu
- Department of Oncology, The Affiliated Shunde Hospital of Jinan University, Foshan, Guangdong, China
| | - Hongyi Zhang
- Department of Microbiology and Immunology, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| |
Collapse
|
2
|
Shao N, Qiu H, Liu J, Xiao D, Zhao J, Chen C, Wan J, Guo M, Liang G, Zhao X, Xu L. Targeting lipid metabolism of macrophages: A new strategy for tumor therapy. J Adv Res 2024:S2090-1232(24)00071-7. [PMID: 38373649 DOI: 10.1016/j.jare.2024.02.009] [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: 09/13/2023] [Revised: 01/16/2024] [Accepted: 02/15/2024] [Indexed: 02/21/2024] Open
Abstract
BACKGROUND Lipid metabolism has been implicated in a variety of normal cellular processes and strongly related to the development of multiple diseases, including tumor. Tumor-associated macrophage (TAM) has emerged as a crucial regulator in tumorigenesis and promising target for tumor treatment. AIM OF REVIEW A thorough understanding of TAM lipid metabolism and its value in tumorigenesis may provide new ideas for TAM-based anti-tumor therapy. Key scientific concepts of review: TAMs can be divided into two main types, M1-like TAMs and M2-like TAMs, which play anti-tumor and pro-tumor functions in tumor occurrence and development, respectively. Accumulating evidence has shown that lipid metabolic reprogramming, including fatty acid uptake and utilization, cholesterol expulsion, controls the polarization of TAMs and affects the tumorgenesis. These advances in uncovering the intricacies of lipid metabolism and TAMs have yielded new insights on tumor development and treatment. In this review, we aim to provide an update on the current understanding of the lipid metabolic reprogramming made by TAMs to adapt to the harsh tumor microenvironment (TME). In particular, we emphasize that there is complex lipid metabolism connections between TAMs and distinct tumors, which influences TAM to bias from M1 to M2 phenotype in tumor progression, and ultimately promotes tumor occurrence and development. Finally, we discuss the existing issues on therapeutic strategies by reprogramming TAMs based on lipid metabolism regulation (or increasing the ratio of M1/M2-like TAMs) that could be applied in the future to clinical tumor treatment.
Collapse
Affiliation(s)
- Nan Shao
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Hui Qiu
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Jing Liu
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Daimin Xiao
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Juanjuan Zhao
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Chao Chen
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Jiajia Wan
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Mengmeng Guo
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Guiyou Liang
- Department of Cardiovascular Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang 550031, China.
| | - Xu Zhao
- School of Medicine, Guizhou University, Guizhou, Guiyang 550025, China; Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou 563000, China.
| | - Lin Xu
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou 563000, China; Department of Immunology, Zunyi Medical University, Zunyi, Guizhou 563000, China; Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, Guizhou 563000, China.
| |
Collapse
|
3
|
Bao Y, Yan Z, Shi N, Tian X, Li J, Li T, Cheng X, Lv J. LCN2: Versatile players in breast cancer. Biomed Pharmacother 2024; 171:116091. [PMID: 38171248 DOI: 10.1016/j.biopha.2023.116091] [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: 10/07/2023] [Revised: 12/06/2023] [Accepted: 12/26/2023] [Indexed: 01/05/2024] Open
Abstract
Lipocalin 2 (LCN2) is a secreted glycoprotein that is produced by immune cells, including neutrophils and macrophages. It serves various functions such as transporting hydrophobic ligands across the cellular membrane, regulating immune responses, keeping iron balance, and fostering epithelial cell differentiation. LCN2 plays a crucial role in several physiological processes. LCN2 expression is upregulated in a variety of human diseases and cancers. High levels of LCN2 are specifically linked to breast cancer (BC) cell proliferation, apoptosis, invasion, migration, angiogenesis, immune regulation, chemotherapy resistance, and prognosis. As a result, LCN2 has gained attention as a potential therapeutic target for BC. This article offered an in-depth review of the advancement of LCN2 in the context of BC occurrence and development.
Collapse
Affiliation(s)
- Yuxiang Bao
- Department of General Surgery, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563099, China
| | - Zhongliang Yan
- Department of General Surgery, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563099, China
| | - Nianmei Shi
- The First Clinical Institute, Zunyi Medical University, Zunyi, Guizhou 563006, China
| | - Xiaoyan Tian
- The First Clinical Institute, Zunyi Medical University, Zunyi, Guizhou 563006, China
| | - Jiayang Li
- Office of Drug Clinical Trial Institution, the Affiliated Hospital of Zunyi Medical University, Zunyi 563099, China
| | - Taolang Li
- Department of General Surgery, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563099, China
| | - Xiaoming Cheng
- Department of General Surgery, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563099, China.
| | - Junyuan Lv
- Department of General Surgery, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563099, China; Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563000, China.
| |
Collapse
|
4
|
Živalj M, Van Ginderachter JA, Stijlemans B. Lipocalin-2: A Nurturer of Tumor Progression and a Novel Candidate for Targeted Cancer Therapy. Cancers (Basel) 2023; 15:5159. [PMID: 37958332 PMCID: PMC10648573 DOI: 10.3390/cancers15215159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 11/15/2023] Open
Abstract
Within the tumor microenvironment (TME) exists a complex signaling network between cancer cells and stromal cells, which determines the fate of tumor progression. Hence, interfering with this signaling network forms the basis for cancer therapy. Yet, many types of cancer, in particular, solid tumors, are refractory to the currently used treatments, so there is an urgent need for novel molecular targets that could improve current anti-cancer therapeutic strategies. Lipocalin-2 (Lcn-2), a secreted siderophore-binding glycoprotein that regulates iron homeostasis, is highly upregulated in various cancer types. Due to its pleiotropic role in the crosstalk between cancer cells and stromal cells, favoring tumor progression, it could be considered as a novel biomarker for prognostic and therapeutic purposes. However, the exact signaling route by which Lcn-2 promotes tumorigenesis remains unknown, and Lcn-2-targeting moieties are largely uninvestigated. This review will (i) provide an overview on the role of Lcn-2 in orchestrating the TME at the level of iron homeostasis, macrophage polarization, extracellular matrix remodeling, and cell migration and survival, and (ii) discuss the potential of Lcn-2 as a promising novel drug target that should be pursued in future translational research.
Collapse
Affiliation(s)
- Maida Živalj
- Brussels Center for Immunology, Vrije Universiteit Brussel, 1050 Brussels, Belgium
- Myeloid Cell Immunology Laboratory, VIB Center for Inflammation Research, 1050 Brussels, Belgium
| | - Jo A. Van Ginderachter
- Brussels Center for Immunology, Vrije Universiteit Brussel, 1050 Brussels, Belgium
- Myeloid Cell Immunology Laboratory, VIB Center for Inflammation Research, 1050 Brussels, Belgium
| | - Benoit Stijlemans
- Brussels Center for Immunology, Vrije Universiteit Brussel, 1050 Brussels, Belgium
- Myeloid Cell Immunology Laboratory, VIB Center for Inflammation Research, 1050 Brussels, Belgium
| |
Collapse
|
5
|
Shao Y, Wang Y, Su R, Pu W, Chen S, Fu L, Yu H, Qiu Y. Dual identity of tumor-associated macrophage in regulated cell death and oncotherapy. Heliyon 2023; 9:e17582. [PMID: 37449180 PMCID: PMC10336529 DOI: 10.1016/j.heliyon.2023.e17582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 04/25/2023] [Accepted: 05/31/2023] [Indexed: 07/18/2023] Open
Abstract
Tumor-associated macrophage (TAM) affects the intrinsic properties of tumor cells and the tumor microenvironment (TME), which can stimulate tumor cell proliferation, migration, and genetic instability, and macrophage diversity includes the diversity of tumors with different functional characteristics. Macrophages are now a central drug target in various diseases, especially in the TME, which, as "tumor promoters" and "immunosuppressors", have different responsibilities during tumor development and accompany by significant dynamic alterations in various subpopulations. Remodelling immunosuppression of TME and promotion of pre-existing antitumor immune responses is critical by altering TAM polarization, which is relevant to the efficacy of immunotherapy, and uncovering the exact mechanism of action of TAMs and identifying their specific targets is vital to optimizing current immunotherapies. Hence, this review aims to reveal the triadic interactions of macrophages with programmed death and oncotherapy, and to integrate certain relationships in cancer treatment.
Collapse
Affiliation(s)
- Yingying Shao
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Yu Wang
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Ranran Su
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Weiling Pu
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Sibao Chen
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology (Incubation), Shenzhen, China
- Department of Applied Biology and Chemical Technology, Research Center for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hong Kong, China
| | - Leilei Fu
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Haiyang Yu
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Yuling Qiu
- School of Pharmacy, Tianjin Medical University, Tianjin, China
| |
Collapse
|
6
|
Hänggi K, Ruffell B. Cell death, therapeutics, and the immune response in cancer. Trends Cancer 2023; 9:381-396. [PMID: 36841748 PMCID: PMC10121860 DOI: 10.1016/j.trecan.2023.02.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/19/2023] [Accepted: 02/03/2023] [Indexed: 02/27/2023]
Abstract
Induction of cell death is inexorably linked with cancer therapy, but this can also initiate wound-healing processes that have been linked to cancer progression and therapeutic resistance. Here we describe the contribution of apoptosis and the lytic cell death pathways in the response to therapy (including chemotherapy and immunotherapy). We also discuss how necroptosis, pyroptosis, and ferroptosis function to promote tumor immunogenicity, along with emerging findings that these same forms of death can paradoxically contribute to immune suppression and tumor progression. Understanding the duality of cell death in cancer may allow for the development of therapeutics that shift the balance towards regression.
Collapse
Affiliation(s)
- Kay Hänggi
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Brian Ruffell
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA; Department of Breast Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.
| |
Collapse
|
7
|
Kazakova A, Sudarskikh T, Kovalev O, Kzhyshkowska J, Larionova I. Interaction of tumor‑associated macrophages with stromal and immune components in solid tumors: Research progress (Review). Int J Oncol 2023; 62:32. [PMID: 36660926 PMCID: PMC9851132 DOI: 10.3892/ijo.2023.5480] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 12/07/2022] [Indexed: 01/18/2023] Open
Abstract
Tumor‑associated macrophages (TAMs) are crucial cells of the tumor microenvironment (TME), which belong to the innate immune system and regulate primary tumor growth, immunosuppression, angiogenesis, extracellular matrix remodeling and metastasis. The review discusses current knowledge of essential cell‑cell interactions of TAMs within the TME of solid tumors. It summarizes the mechanisms of stromal cell (including cancer‑associated fibroblasts and endothelial cells)‑mediated monocyte recruitment and regulation of differentiation, as well as pro‑tumor and antitumor polarization of TAMs. Additionally, it focuses on the perivascular TAM subpopulations that regulate angiogenesis and lymphangiogenesis. It describes the possible mechanisms of reciprocal interactions of TAMs with other immune cells responsible for immunosuppression. Finally, it highlights the perspectives for novel therapeutic approaches to use combined cellular targets that include TAMs and other stromal and immune cells in the TME. The collected data demonstrated the importance of understanding cell‑cell interactions in the TME to prevent distant metastasis and reduce the risk of tumor recurrence.
Collapse
Affiliation(s)
- Anna Kazakova
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, Tomsk 634050, Russian Federation
| | - Tatiana Sudarskikh
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, Tomsk 634050, Russian Federation
| | - Oleg Kovalev
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 634009, Russian Federation
| | - Julia Kzhyshkowska
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, Tomsk 634050, Russian Federation
- Institute of Transfusion Medicine and Immunology, Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, D-68167 Mannheim, Germany
| | - Irina Larionova
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, Tomsk 634050, Russian Federation
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 634009, Russian Federation
| |
Collapse
|
8
|
Indino S, Borzi C, Moscheni C, Sartori P, De Cecco L, Bernardo G, Le Noci V, Arnaboldi F, Triulzi T, Sozzi G, Tagliabue E, Sfondrini L, Gagliano N, Moro M, Sommariva M. The Educational Program of Macrophages toward a Hyperprogressive Disease-Related Phenotype Is Orchestrated by Tumor-Derived Extracellular Vesicles. Int J Mol Sci 2022; 23:ijms232415802. [PMID: 36555441 PMCID: PMC9779478 DOI: 10.3390/ijms232415802] [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: 09/07/2022] [Revised: 12/03/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022] Open
Abstract
Hyperprogressive disease (HPD), an aggressive acceleration of tumor growth, was observed in a group of cancer patients treated with anti-PD1/PDL1 antibodies. The presence of a peculiar macrophage subset in the tumor microenvironment is reported to be a sort of "immunological prerequisite" for HPD development. These macrophages possess a unique phenotype that it is not clear how they acquire. We hypothesized that certain malignant cells may promote the induction of an "HPD-related" phenotype in macrophages. Bone-marrow-derived macrophages were exposed to the conditioned medium of five non-small cell lung cancer cell lines. Macrophage phenotype was analyzed by microarray gene expression profile and real-time PCR. We found that human NSCLC cell lines, reported as undergoing HPD-like tumor growth in immunodeficient mice, polarized macrophages towards a peculiar pro-inflammatory phenotype sharing both M1 and M2 features. Lipid-based factors contained in cancer cell-conditioned medium induced the over-expression of several pro-inflammatory cytokines and the activation of innate immune receptor signaling pathways. We also determined that tumor-derived Extracellular Vesicles represent the main components involved in the observed macrophage re-education program. The present study might represent the starting point for the future development of diagnostic tools to identify potential hyperprogressors.
Collapse
Affiliation(s)
- Serena Indino
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Via Mangiagalli 31, 20133 Milan, Italy
| | - Cristina Borzi
- Tumor Genomics Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Claudia Moscheni
- Dipartimento di Scienze Biomediche e Cliniche, Università degli Studi di Milano, Via G. B. Grassi, 74, L.I.T.A. Vialba, 20157 Milan, Italy
| | - Patrizia Sartori
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Via Mangiagalli 31, 20133 Milan, Italy
| | - Loris De Cecco
- Molecular Mechanisms Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy
| | - Giancarla Bernardo
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Via Mangiagalli 31, 20133 Milan, Italy
| | - Valentino Le Noci
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Via Mangiagalli 31, 20133 Milan, Italy
| | - Francesca Arnaboldi
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Via Mangiagalli 31, 20133 Milan, Italy
| | - Tiziana Triulzi
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy
| | - Gabriella Sozzi
- Tumor Genomics Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Elda Tagliabue
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy
| | - Lucia Sfondrini
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Via Mangiagalli 31, 20133 Milan, Italy
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy
| | - Nicoletta Gagliano
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Via Mangiagalli 31, 20133 Milan, Italy
| | - Massimo Moro
- Tumor Genomics Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133 Milan, Italy
| | - Michele Sommariva
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Via Mangiagalli 31, 20133 Milan, Italy
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy
- Correspondence: ; Tel.: +39-0250315401
| |
Collapse
|
9
|
Tsai HH, Hsieh YC, Lin JS, Kuo ZT, Ho CY, Chen CH, Chang CF. Functional Investigation of Meningeal Lymphatic System in Experimental Intracerebral Hemorrhage. Stroke 2022; 53:987-998. [DOI: 10.1161/strokeaha.121.037834] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background:
Promotion of hematoma resolution in a timely manner reduces intracerebral hemorrhage (ICH) brain injury induced by toxic blood components and subsequent neuroinflammation. The meningeal lymphatic system is responsible for clearance of macromolecules and pathogenic substances from the central nervous system; however, its role in intraparenchymal hematoma clearance and ICH outcomes is unknown. In the present study, we aimed to understand the contribution of the meningeal lymphatic system to ICH pathologies and to test whether pharmacological enhancement of meningeal lymphatic function promotes hematoma resolution and brain recovery after ICH.
Methods:
Immunofluorescence of whole-mount meninges was used to measure complexity and coverage level of meningeal lymphatic vasculature following ICH induction. Fluorescent microbeads and PKH-26-labeled erythrocytes were used to evaluate drainage function of the meningeal lymphatic system. Visudyne treatment, deep cervical lymph node ligation, and VEGF (vascular endothelial growth factor)-C injection were performed to manipulate meningeal lymphatic function. Neurobehavioral performance and hematoma volume were assayed by the cylinder test and histological measurements. Iron deposition, residual erythrocytes, neuronal loss, and astrogliosis were assessed by immunohistochemistry and antibody-based fluorescence staining.
Results:
Meningeal lymphangiogenesis and enhanced lymphatic drainage occurred during the late phase of ICH. Ablation and blockage of meningeal lymphatic vessels impeded hematoma clearance, whereas pharmacological enhancement of their function reduced hematoma volume, improved behavioral performance, and reduced brain residual erythrocytes, iron deposition, neuronal loss, and astroglial activation.
Conclusions:
Early enhancement of meningeal lymphatic function is beneficial for ICH recovery. Targeting the meningeal lymphatic system is therefore a potential therapeutic approach for treating ICH.
Collapse
Affiliation(s)
- Hsin-Hsi Tsai
- Department of Neurology, National Taiwan University Hospital Bei-Hu Branch, Taipei (H.-H.T.)
- Department of Neurology, National Taiwan University Hospital, Taipei (H.-H.T.)
| | - Yung-Chia Hsieh
- School of Medicine, National Taiwan University College of Medicine, Taipei. (Y.-C.H.)
- Department and Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei. (Y.-C.H., J.S.L., Z.-T.K., C.-Y.H., C.-H.C., C.-F.C.)
| | - Jhih Syuan Lin
- Department and Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei. (Y.-C.H., J.S.L., Z.-T.K., C.-Y.H., C.-H.C., C.-F.C.)
| | - Zi-Ting Kuo
- Department and Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei. (Y.-C.H., J.S.L., Z.-T.K., C.-Y.H., C.-H.C., C.-F.C.)
| | - Chi-Ying Ho
- Department and Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei. (Y.-C.H., J.S.L., Z.-T.K., C.-Y.H., C.-H.C., C.-F.C.)
| | - Chih-Hung Chen
- Department and Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei. (Y.-C.H., J.S.L., Z.-T.K., C.-Y.H., C.-H.C., C.-F.C.)
| | - Che-Feng Chang
- Department and Graduate Institute of Physiology, National Taiwan University College of Medicine, Taipei. (Y.-C.H., J.S.L., Z.-T.K., C.-Y.H., C.-H.C., C.-F.C.)
| |
Collapse
|
10
|
Cao S, Schnelzer A, Hannemann N, Schett G, Soulat D, Bozec A. The Transcription Factor FRA-1/AP-1 Controls Lipocalin-2 Expression and Inflammation in Sepsis Model. Front Immunol 2021; 12:701675. [PMID: 34712224 PMCID: PMC8546226 DOI: 10.3389/fimmu.2021.701675] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 09/24/2021] [Indexed: 12/29/2022] Open
Abstract
Sepsis is a life-threatening condition characterized by excessive inflammation in its early phase. This is followed by an aberrant resolution phase associated to a prolonged period of immune suppression that can ultimately lead to multiple organ dysfunctions. This immunosuppression can be mediated by the functional reprogramming of gene transcription in monocytes/macrophages in response to prolonged lipopolysaccharide (LPS) exposure. Surprisingly, there is no report on the role of AP-1 transcription factors in this reprogramming process. Herein, we used the endotoxin tolerance model on murine bone marrow-derived macrophages in which tolerant cells stimulated twice with LPS were compared to naïve cells stimulated once. Out of all AP-1 transcription factors tested, Fosl1 gene stood out because of its unique regulation in tolerized cells. Moreover, we could correlate FRA-1 expression to the expression of an essential anti-inflammatory molecule involved in sepsis response, Lipocalin 2 aka NGAL. Identical results were obtained in human PBMC following the endotoxin tolerance model. When using FRA-1 deficient macrophages, we could confirm that FRA-1 regulates NGAL expression during the tolerant state. Interestingly, ChIP-seq and ChIP-qPCR revealed the binding of FRA-1 on Lcn2 promoter after LPS stimulation in these cells. Finally, we used an in vivo septic model of consecutive injection of LPS, in which the second stimulation is performed before the resolution of inflammation, in wild type and FRA-1 deficient mice. NGAL secretion was elevated in lung, spleen and serum of wild type tolerant mice, whereas it was significantly lower in tolerant FRA-1 deficient mice. Moreover, an increased inflammatory state likely dependent of the low level of NGAL was observed in these FRA-1 deficient mice. This was characterized by an increase of neutrophil infiltration in lung and an increase of apoptotic follicular cells in spleen. This suggests that FRA-1 expression supports resolution of inflammation in this model. Collectively, our data indicate that FRA-1 is involved in myeloid cell tolerance responses by mediating the functional reprogramming of Lcn2 transcription in response to prolonged LPS exposure. In conclusion, FRA-1 may have a protective role in the tolerance response of sepsis through the regulation of NGAL, leading to resolution of inflammation.
Collapse
Affiliation(s)
- Shan Cao
- Department of Internal Medicine 3, Rheumatology and Immunology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Anne Schnelzer
- Department of Internal Medicine 3, Rheumatology and Immunology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Nicole Hannemann
- Department of Internal Medicine 3, Rheumatology and Immunology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany.,Institute of Regenerative Medicine and Biotherapies (IRMB), University of Montpellier, INSERM U1183, Montpellier, France
| | - Georg Schett
- Department of Internal Medicine 3, Rheumatology and Immunology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Didier Soulat
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Aline Bozec
- Department of Internal Medicine 3, Rheumatology and Immunology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
11
|
Villodre ES, Hu X, Larson R, Finetti P, Gomez K, Balema W, Stecklein SR, Santiago‐Sanchez G, Krishnamurthy S, Song J, Su X, Ueno NT, Tripathy D, Van Laere S, Bertucci F, Vivas‐Mejía P, Woodward WA, Debeb BG. Lipocalin 2 promotes inflammatory breast cancer tumorigenesis and skin invasion. Mol Oncol 2021; 15:2752-2765. [PMID: 34342930 PMCID: PMC8486564 DOI: 10.1002/1878-0261.13074] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 07/21/2021] [Accepted: 08/02/2021] [Indexed: 12/28/2022] Open
Abstract
Inflammatory breast cancer (IBC) is an aggressive form of primary breast cancer characterized by rapid onset and high risk of metastasis and poor clinical outcomes. The biological basis for the aggressiveness of IBC is still not well understood and no IBC-specific targeted therapies exist. In this study, we report that lipocalin 2 (LCN2), a small secreted glycoprotein belonging to the lipocalin superfamily, is expressed at significantly higher levels in IBC vs non-IBC tumors, independently of molecular subtype. LCN2 levels were also significantly higher in IBC cell lines and in their culture media than in non-IBC cell lines. High expression was associated with poor-prognosis features and shorter overall survival in IBC patients. Depletion of LCN2 in IBC cell lines reduced colony formation, migration, and cancer stem cell populations in vitro and inhibited tumor growth, skin invasion, and brain metastasis in mouse models of IBC. Analysis of our proteomics data showed reduced expression of proteins involved in cell cycle and DNA repair in LCN2-silenced IBC cells. Our findings support that LCN2 promotes IBC tumor aggressiveness and offer a new potential therapeutic target for IBC.
Collapse
Affiliation(s)
- Emilly S. Villodre
- Department of Breast Medical OncologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
- MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research ProgramThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Xiaoding Hu
- Department of Breast Medical OncologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
- MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research ProgramThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Richard Larson
- MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research ProgramThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
- Department of Radiation OncologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Pascal Finetti
- Laboratory of Predictive OncologyAix‐Marseille UniversityInsermCNRSInstitut Paoli‐CalmettesCRCMMarseilleFrance
| | - Kristen Gomez
- Department of Biological SciencesThe University of Texas at BrownsvilleTXUSA
| | - Wintana Balema
- MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research ProgramThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
- Department of Radiation OncologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Shane R. Stecklein
- MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research ProgramThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
- Department of Radiation OncologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Ginette Santiago‐Sanchez
- Department Biochemistry and Cancer CenterUniversity of Puerto Rico Medical Sciences CampusSan Juan, Puerto Rico
| | - Savitri Krishnamurthy
- MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research ProgramThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
- Department of PathologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Juhee Song
- Department of BiostatisticsThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Xiaoping Su
- Department of Bioinformatics and Computational BiologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Naoto T. Ueno
- Department of Breast Medical OncologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
- MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research ProgramThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Debu Tripathy
- Department of Breast Medical OncologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
- MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research ProgramThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Steven Van Laere
- Center for Oncological Research (CORE)Integrated Personalized and Precision Oncology Network (IPPON)University of AntwerpBelgium
| | - François Bertucci
- Laboratory of Predictive OncologyAix‐Marseille UniversityInsermCNRSInstitut Paoli‐CalmettesCRCMMarseilleFrance
| | - Pablo Vivas‐Mejía
- Department Biochemistry and Cancer CenterUniversity of Puerto Rico Medical Sciences CampusSan Juan, Puerto Rico
| | - Wendy A. Woodward
- MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research ProgramThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
- Department of Radiation OncologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Bisrat G. Debeb
- Department of Breast Medical OncologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
- MD Anderson Morgan Welch Inflammatory Breast Cancer Clinic and Research ProgramThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| |
Collapse
|
12
|
Tong N, He Z, Ma Y, Wang Z, Huang Z, Cao H, Xu L, Zou Y, Wang W, Yi C, Yin Z, Wang Q. Tumor Associated Macrophages, as the Dominant Immune Cells, Are an Indispensable Target for Immunologically Cold Tumor-Glioma Therapy? Front Cell Dev Biol 2021; 9:706286. [PMID: 34368156 PMCID: PMC8337013 DOI: 10.3389/fcell.2021.706286] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 06/28/2021] [Indexed: 12/12/2022] Open
Abstract
Tumor microenvironment (TME) is the cornerstone of the occurrence, development, invasion and diffusion of the malignant central nerve system (CNS) tumor, glioma. As the largest number of inflammatory cells in glioma TME, tumor associated macrophages (TAMs) and their secreted factors are indispensable to the progression of glioma, which is a well-known immunologically “cold” tumor, including the growth of tumor cells, invasion, migration, angiogenesis, cancer immunosuppression and metabolism. TAMs intimately interface with the treatment failure and poor prognosis of glioma patients, and their density increases with increasing glioma grade. Recently, great progress has been made in TAM-targeting for anti-tumor therapy. According to TAMs’ function in tumorigenesis and progression, the major anti-tumor treatment strategies targeting TAMs are to hinder macrophage recruitment in TME, reduce TAMs viability or remodel TAMs phenotype from M2 to M1. Different approaches offer unique and effective anti-tumor effect by regulating the phagocytosis, polarization and pro-tumor behaviors of macrophages in the therapy of glioma. The present review summarizes the significant characteristics and related mechanisms of TAMs and addresses the related research progress on targeting TAMs in glioma.
Collapse
Affiliation(s)
- Ni Tong
- Department of Pharmacy, Zhujiang Hospital of Southern Medical University, Guangzhou, China.,Department of Molecular Biology, State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Zhenqiang He
- State Key Laboratory of Oncology in South China, Department of Neurosurgery/Neuro-Oncology, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yujie Ma
- Department of Molecular Biology, State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Zheng Wang
- Breast Surgery Department, Nanyang Central Hospital, Nanyang, China
| | - Ziming Huang
- Department of Molecular Biology, State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Haihong Cao
- Department of Pharmacy, Zhujiang Hospital of Southern Medical University, Guangzhou, China.,Department of Molecular Biology, State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Lanyang Xu
- Department of Pharmacy, Zhujiang Hospital of Southern Medical University, Guangzhou, China.,Department of Molecular Biology, State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yuheng Zou
- Department of Pharmacy, Zhujiang Hospital of Southern Medical University, Guangzhou, China.,Department of Molecular Biology, State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Wanyu Wang
- Department of Pharmacy, Zhujiang Hospital of Southern Medical University, Guangzhou, China.,Department of Molecular Biology, State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Chenpeng Yi
- Department of Pharmacy, Zhujiang Hospital of Southern Medical University, Guangzhou, China.,Department of Molecular Biology, State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Zhixin Yin
- School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Qirui Wang
- Department of Pharmacy, Zhujiang Hospital of Southern Medical University, Guangzhou, China.,Department of Molecular Biology, State Administration of Traditional Chinese Medicine of the People's Republic of China, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
13
|
Zheng H, Siddharth S, Parida S, Wu X, Sharma D. Tumor Microenvironment: Key Players in Triple Negative Breast Cancer Immunomodulation. Cancers (Basel) 2021; 13:cancers13133357. [PMID: 34283088 PMCID: PMC8269090 DOI: 10.3390/cancers13133357] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 06/29/2021] [Accepted: 07/01/2021] [Indexed: 12/16/2022] Open
Abstract
Simple Summary The tumor microenvironment (TME) is a complicated network composed of various cells, signaling molecules, and extra cellular matrix. TME plays a crucial role in triple negative breast cancer (TNBC) immunomodulation and tumor progression, paradoxically, acting as an immunosuppressive as well as immunoreactive factor. Research regarding tumor immune microenvironment has contributed to a better understanding of TNBC subtype classification. Shall we treat patients precisely according to specific subtype classification? Moving beyond traditional chemotherapy, multiple clinical trials have recently implied the potential benefits of immunotherapy combined with chemotherapy. In this review, we aimed to elucidate the paradoxical role of TME in TNBC immunomodulation, summarize the subtype classification methods for TNBC, and explore the synergistic mechanism of chemotherapy plus immunotherapy. Our study may provide a new direction for the development of combined treatment strategies for TNBC. Abstract Triple negative breast cancer (TNBC) is a heterogeneous disease and is highly related to immunomodulation. As we know, the most effective approach to treat TNBC so far is still chemotherapy. Chemotherapy can induce immunogenic cell death, release of damage-associated molecular patterns (DAMPs), and tumor microenvironment (TME) remodeling; therefore, it will be interesting to investigate the relationship between chemotherapy-induced TME changes and TNBC immunomodulation. In this review, we focus on the immunosuppressive and immunoreactive role of TME in TNBC immunomodulation and the contribution of TME constituents to TNBC subtype classification. Further, we also discuss the role of chemotherapy-induced TME remodeling in modulating TNBC immune response and tumor progression with emphasis on DAMPs-associated molecules including high mobility group box1 (HMGB1), exosomes, and sphingosine-1-phosphate receptor 1 (S1PR1), which may provide us with new clues to explore effective combined treatment options for TNBC.
Collapse
Affiliation(s)
- Hongmei Zheng
- Hubei Provincial Clinical Research Center for Breast Cancer, Department of Breast Surgery, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430079, China
- The Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA; (S.S.); (S.P.); (D.S.)
- Correspondence: (H.Z.); (X.W.)
| | - Sumit Siddharth
- The Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA; (S.S.); (S.P.); (D.S.)
| | - Sheetal Parida
- The Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA; (S.S.); (S.P.); (D.S.)
| | - Xinhong Wu
- Hubei Provincial Clinical Research Center for Breast Cancer, Department of Breast Surgery, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430079, China
- Correspondence: (H.Z.); (X.W.)
| | - Dipali Sharma
- The Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA; (S.S.); (S.P.); (D.S.)
| |
Collapse
|
14
|
Delprat V, Michiels C. A bi-directional dialog between vascular cells and monocytes/macrophages regulates tumor progression. Cancer Metastasis Rev 2021; 40:477-500. [PMID: 33783686 PMCID: PMC8213675 DOI: 10.1007/s10555-021-09958-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 02/23/2021] [Indexed: 02/06/2023]
Abstract
Cancer progression largely depends on tumor blood vessels as well on immune cell infiltration. In various tumors, vascular cells, namely endothelial cells (ECs) and pericytes, strongly regulate leukocyte infiltration into tumors and immune cell activation, hence the immune response to cancers. Recently, a lot of compelling studies unraveled the molecular mechanisms by which tumor vascular cells regulate monocyte and tumor-associated macrophage (TAM) recruitment and phenotype, and consequently tumor progression. Reciprocally, TAMs and monocytes strongly modulate tumor blood vessel and tumor lymphatic vessel formation by exerting pro-angiogenic and lymphangiogenic effects, respectively. Finally, the interaction between monocytes/TAMs and vascular cells is also impacting several steps of the spread of cancer cells throughout the body, a process called metastasis. In this review, the impact of the bi-directional dialog between blood vascular cells and monocytes/TAMs in the regulation of tumor progression is discussed. All together, these data led to the design of combinations of anti-angiogenic and immunotherapy targeting TAMs/monocyte whose effects are briefly discussed in the last part of this review.
Collapse
Affiliation(s)
- Victor Delprat
- Biochemistry and Cellular Biology Research Unit (URBC), Namur Research Institute for Life Sciences (NARILIS), University of Namur (UNamur), 61 Rue de Bruxelles, B-5000, Namur, Belgium
| | - Carine Michiels
- Biochemistry and Cellular Biology Research Unit (URBC), Namur Research Institute for Life Sciences (NARILIS), University of Namur (UNamur), 61 Rue de Bruxelles, B-5000, Namur, Belgium.
| |
Collapse
|
15
|
Iron-Bound Lipocalin-2 Protects Renal Cell Carcinoma from Ferroptosis. Metabolites 2021; 11:metabo11050329. [PMID: 34069743 PMCID: PMC8161288 DOI: 10.3390/metabo11050329] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/12/2021] [Accepted: 05/17/2021] [Indexed: 02/08/2023] Open
Abstract
While the importance of the iron-load of lipocalin-2 (Lcn-2) in promoting tumor progression is widely appreciated, underlying molecular mechanisms largely remain elusive. Considering its role as an iron-transporter, we aimed at clarifying iron-loaded, holo-Lcn-2 (hLcn-2)-dependent signaling pathways in affecting renal cancer cell viability. Applying RNA sequencing analysis in renal CAKI1 tumor cells to explore highly upregulated molecular signatures in response to hLcn-2, we identified a cluster of genes (SLC7A11, GCLM, GLS), which are implicated in regulating ferroptosis. Indeed, hLcn-2-stimulated cells are protected from erastin-induced ferroptosis. We also noticed a rapid increase in reactive oxygen species (ROS) with subsequent activation of the antioxidant Nrf2 pathway. However, knocking down Nrf2 by siRNA was not sufficient to induce erastin-dependent ferroptotic cell death in hLcn-2-stimulated tumor cells. In contrast, preventing oxidative stress through N-acetyl-l-cysteine (NAC) supplementation was still able to induce erastin-dependent ferroptotic cell death in hLcn-2-stimulated tumor cells. Besides an oxidative stress response, we noticed activation of the integrated stress response (ISR), shown by enhanced phosphorylation of eIF-2α and induction of ATF4 after hLcn-2 addition. ATF4 knockdown as well as inhibition of the ISR sensitized hLcn-2-treated renal tumor cells to ferroptosis, thus linking the ISR to pro-tumor characteristics of hLcn-2. Our study provides mechanistic details to better understand tumor pro-survival pathways initiated by iron-loaded Lcn-2.
Collapse
|
16
|
He M, He Q, Cai X, Chen Z, Lao S, Deng H, Liu X, Zheng Y, Liu X, Liu J, Xie Z, Yao M, Liang W, He J. Role of lymphatic endothelial cells in the tumor microenvironment-a narrative review of recent advances. Transl Lung Cancer Res 2021; 10:2252-2277. [PMID: 34164274 PMCID: PMC8182726 DOI: 10.21037/tlcr-21-40] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background As lymphatic vessel is a major route for solid tumor metastasis, they are considered an essential part of tumor drainage conduits. Apart from forming the walls of lymphatic vessels, lymphatic endothelial cells (LECs) have been found to play multiple other roles in the tumor microenvironment, calling for a more in-depth review. We hope that this review may help researchers gain a detailed understanding of this fast-developing field and shed some light upon future research. Methods To achieve an informative review of recent advance, we carefully searched the Medline database for English literature that are openly published from the January 1995 to December 2020 and covered the topic of LEC or lymphangiogenesis in tumor progression and therapies. Two different authors independently examined the literature abstracts to exclude possible unqualified ones, and 310 papers with full texts were finally retrieved. Results In this paper, we discussed the structural and molecular basis of tumor-associated LECs, together with their roles in tumor metastasis and drug therapy. We then focused on their impacts on tumor cells, tumor stroma, and anti-tumor immunity, and the molecular and cellular mechanisms involved. Special emphasis on lung cancer and possible therapeutic targets based on LECs were also discussed. Conclusions LECs can play a much more complex role than simply forming conduits for tumor cell dissemination. Therapies targeting tumor-associated lymphatics for lung cancer and other tumors are promising, but more research is needed to clarify the mechanisms involved.
Collapse
Affiliation(s)
- Miao He
- Department of Thoracic Surgery, China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qihua He
- Department of Thoracic Surgery, China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Oncology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiuyu Cai
- Department of VIP Region, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Zisheng Chen
- Department of Thoracic Surgery, China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Respiratory Medicine, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, China
| | - Shen Lao
- Department of Thoracic Surgery, China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hongsheng Deng
- Department of Thoracic Surgery, China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiwen Liu
- Department of Thoracic Surgery, China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yongmei Zheng
- Department of Thoracic Surgery, China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaoyan Liu
- Department of Thoracic Surgery, China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jun Liu
- Department of Thoracic Surgery, China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhanhong Xie
- Department of Thoracic Surgery, China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Respiratory Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Maojin Yao
- Department of Thoracic Surgery, China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wenhua Liang
- Department of Thoracic Surgery, China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,The First People Hospital of Zhaoqing, Zhaoqing, China
| | - Jianxing He
- Department of Thoracic Surgery, China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
17
|
Iron-Bound Lipocalin-2 from Tumor-Associated Macrophages Drives Breast Cancer Progression Independent of Ferroportin. Metabolites 2021; 11:metabo11030180. [PMID: 33808732 PMCID: PMC8003561 DOI: 10.3390/metabo11030180] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 03/12/2021] [Accepted: 03/16/2021] [Indexed: 01/12/2023] Open
Abstract
Macrophages supply iron to the breast tumor microenvironment by enforced secretion of lipocalin-2 (Lcn-2)-bound iron as well as the increased expression of the iron exporter ferroportin (FPN). We aimed at identifying the contribution of each pathway in supplying iron for the growing tumor, thereby fostering tumor progression. Analyzing the expression profiles of Lcn-2 and FPN using the spontaneous polyoma-middle-T oncogene (PyMT) breast cancer model as well as mining publicly available TCGA (The Cancer Genome Atlas) and GEO Series(GSE) datasets from the Gene Expression Omnibus database (GEO), we found no association between tumor parameters and Lcn-2 or FPN. However, stromal/macrophage-expression of Lcn-2 correlated with tumor onset, lung metastases, and recurrence, whereas FPN did not. While the total iron amount in wildtype and Lcn-2-/- PyMT tumors showed no difference, we observed that tumor-associated macrophages from Lcn-2-/- compared to wildtype tumors stored more iron. In contrast, Lcn-2-/- tumor cells accumulated less iron than their wildtype counterparts, translating into a low migratory and proliferative capacity of Lcn-2-/- tumor cells in a 3D tumor spheroid model in vitro. Our data suggest a pivotal role of Lcn-2 in tumor iron-management, affecting tumor growth. This study underscores the role of iron for tumor progression and the need for a better understanding of iron-targeted therapy approaches.
Collapse
|
18
|
Hamada K, Ferguson LB, Mayfield RD, Krishnan HR, Maienschein-Cline M, Lasek AW. Binge-like ethanol drinking activates anaplastic lymphoma kinase signaling and increases the expression of STAT3 target genes in the mouse hippocampus and prefrontal cortex. GENES, BRAIN, AND BEHAVIOR 2021; 20:e12729. [PMID: 33641239 PMCID: PMC8944393 DOI: 10.1111/gbb.12729] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/04/2021] [Accepted: 02/26/2021] [Indexed: 02/03/2023]
Abstract
Alcohol use disorder (AUD) has a complex pathogenesis, making it a difficult disorder to treat. Identifying relevant signaling pathways in the brain may be useful for finding new pharmacological targets to treat AUD. The receptor tyrosine kinase anaplastic lymphoma kinase (ALK) activates the transcription factor STAT3 in response to ethanol in cell lines. Here, we show ALK activation and upregulation of known STAT3 target genes (Socs3, Gfap and Tnfrsf1a) in the prefrontal cortex (PFC) and ventral hippocampus (HPC) of mice after 4 days of binge-like ethanol drinking. Mice treated with the STAT3 inhibitor stattic drank less ethanol than vehicle-treated mice, demonstrating the behavioral importance of STAT3. To identify novel ethanol-induced target genes downstream of the ALK and STAT3 pathway, we analyzed the NIH LINCS L1000 database for gene signature overlap between ALK inhibitor (alectinib and NVP-TAE684) and STAT3 inhibitor (niclosamide) treatments on cell lines. These genes were then compared with differentially expressed genes in the PFC of mice after binge-like drinking. We found 95 unique gene candidates, out of which 57 had STAT3 binding motifs in their promoters. We further showed by qPCR that expression of the putative STAT3 genes Nr1h2, Smarcc1, Smarca4 and Gpnmb were increased in either the PFC or HPC after binge-like drinking. Together, these results indicate activation of the ALK-STAT3 signaling pathway in the brain after binge-like ethanol consumption, identify putative novel ethanol-responsive STAT3 target genes, and suggest that STAT3 inhibition may be a potential method to reduce binge drinking in humans.
Collapse
Affiliation(s)
- Kana Hamada
- Graduate Program in Neuroscience, University of Illinois at Chicago, Chicago, IL 60612 USA
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - Laura B. Ferguson
- Waggoner Center for Alcohol Addiction Research and Department of Neuroscience, University of Texas at Austin, Austin, TX 78712 USA
- Department of Neurology, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA
| | - R. Dayne Mayfield
- Waggoner Center for Alcohol Addiction Research and Department of Neuroscience, University of Texas at Austin, Austin, TX 78712 USA
| | - Harish R. Krishnan
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL 60612 USA
| | | | - Amy W. Lasek
- Center for Alcohol Research in Epigenetics, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL 60612 USA
| |
Collapse
|
19
|
Liang W, Ferrara N. Iron Metabolism in the Tumor Microenvironment: Contributions of Innate Immune Cells. Front Immunol 2021; 11:626812. [PMID: 33679721 PMCID: PMC7928394 DOI: 10.3389/fimmu.2020.626812] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 12/30/2020] [Indexed: 12/21/2022] Open
Abstract
Cells of the innate immune system are a major component of the tumor microenvironment. They play complex and multifaceted roles in the regulation of cancer initiation, growth, metastasis and responses to therapeutics. Innate immune cells like neutrophils and macrophages are recruited to cancerous tissues by chemotactic molecules released by cancer cells and cancer-associated stromal cells. Once they reach the tumor, they can be instructed by a network of proteins, nucleic acids and metabolites to exert protumoral or antitumoral functions. Altered iron metabolism is a feature of cancer. Epidemiological studies suggest that increased presence of iron and/or iron binding proteins is associated with increased risks of cancer development. It has been shown that iron metabolism is involved in shaping the immune landscapes in inflammatory/infectious diseases and cancer-associated inflammation. In this article, we will dissect the contribution of macrophages and neutrophils to dysregulated iron metabolism in malignant cells and its impact on cancer growth and metastasis. The mechanisms involved in regulating the actions of macrophages and neutrophils will also be discussed. Moreover, we will examine the effects of iron metabolism on the phenotypes of innate immune cells. Both iron chelating and overloading agents are being explored in cancer treatment. This review highlights alternative strategies for management of iron content in cancer cells by targeting the iron donation and modulation properties of macrophages and neutrophils in the tumor microenvironment.
Collapse
Affiliation(s)
- Wei Liang
- Oncology, BioDuro LLC, San Diego, CA, United States
| | - Napoleone Ferrara
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
| |
Collapse
|
20
|
Shen L, Zhou Y, He H, Chen W, Lenahan C, Li X, Deng Y, Shao A, Huang J. Crosstalk between Macrophages, T Cells, and Iron Metabolism in Tumor Microenvironment. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8865791. [PMID: 33628389 PMCID: PMC7889336 DOI: 10.1155/2021/8865791] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 09/28/2020] [Accepted: 01/12/2021] [Indexed: 02/06/2023]
Abstract
Leukocytes, including macrophages and T cells, represent key players in the human immune system, which plays a considerable role in the development and progression of tumors by immune surveillance or immune escape. Boosting the recruitment of leukocytes into the tumor microenvironment and promoting their antitumor responses have been hot areas of research in recent years. Although immunotherapy has manifested a certain level of success in some malignancies, the overall effectiveness is far from satisfactory. Iron is an essential trace element required in multiple, normal cellular processes, such as DNA synthesis and repair, cellular respiration, metabolism, and signaling, while dysregulated iron metabolism has been declared one of the metabolic hallmarks of malignant cancer cells. Furthermore, iron is implicated in the modulation of innate and adaptive immune responses, and elucidating the targeted regulation of iron metabolism may have the potential to benefit antitumor immunity and cancer treatment. In the present review, we briefly summarize the roles of leukocytes and iron metabolism in tumorigenesis, as well as their crosstalk in the tumor microenvironment. The combination of immunotherapy with targeted regulation of iron and iron-dependent regulated cell death (ferroptosis) may be a focus of future research.
Collapse
Affiliation(s)
- Lesang Shen
- Department of Breast Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Hangzhou 310009, China
| | - Yunxiang Zhou
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Haifei He
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Wuzhen Chen
- Department of Breast Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Hangzhou 310009, China
| | - Cameron Lenahan
- Burrell College of Osteopathic Medicine, Las Cruces, NM, USA
| | - Xiaoyi Li
- Department of Nuclear Medicine and PET-CT Center, The Second Affiliated Hospital, Zhejiang University, Hangzhou 310009, China
| | - Yongchuan Deng
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Jian Huang
- Department of Breast Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Hangzhou 310009, China
| |
Collapse
|
21
|
Xiang Y, Miao H. Lipid Metabolism in Tumor-Associated Macrophages. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1316:87-101. [PMID: 33740245 DOI: 10.1007/978-981-33-6785-2_6] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Macrophages are essential components of the immune system in tumors. It can be recruited and educated to two mainly polarized subpopulations (M1-like and M2-like) of tumor-associated macrophages (TAMs) to display anti-tumor or protumor function during the tumor occurrence and progression. Reprogramming of metabolism, especially lipid metabolism, is a typical characteristic of TAMs polarization, which was confirmed recently as a vital target for tumor therapy. However, the relationship between TAMs and lipid metabolism is still obscure in the past decade. In this review, we will first introduce the historical aspects of TAMs, and then discuss the correlation of main lipids (triglycerides, cholesterol, and phospholipids) to TAMs activation and summarize the mechanisms by which lipid metabolism mediated tumor escape the immunological surveillance as well as currently available drugs targeting these mechanisms. We hope that this chapter will give a better understanding of lipid metabolism in TAMs for those who are interested in this field, and lay a foundation to develop novel strategies for tumor therapy by targeting lipid metabolism.
Collapse
Affiliation(s)
- Yuancai Xiang
- Department of Biochemistry and Molecular Biology, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China
| | - Hongming Miao
- Department of Biochemistry and Molecular Biology, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China.
| |
Collapse
|
22
|
Hsu MY, Mina E, Roetto A, Porporato PE. Iron: An Essential Element of Cancer Metabolism. Cells 2020; 9:cells9122591. [PMID: 33287315 PMCID: PMC7761773 DOI: 10.3390/cells9122591] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 11/24/2020] [Accepted: 11/30/2020] [Indexed: 02/06/2023] Open
Abstract
Cancer cells undergo considerable metabolic changes to foster uncontrolled proliferation in a hostile environment characterized by nutrient deprivation, poor vascularization and immune infiltration. While metabolic reprogramming has been recognized as a hallmark of cancer, the role of micronutrients in shaping these adaptations remains scarcely investigated. In particular, the broad electron-transferring abilities of iron make it a versatile cofactor that is involved in a myriad of biochemical reactions vital to cellular homeostasis, including cell respiration and DNA replication. In cancer patients, systemic iron metabolism is commonly altered. Moreover, cancer cells deploy diverse mechanisms to increase iron bioavailability to fuel tumor growth. Although iron itself can readily participate in redox reactions enabling vital processes, its reactivity also gives rise to reactive oxygen species (ROS). Hence, cancer cells further rely on antioxidant mechanisms to withstand such stress. The present review provides an overview of the common alterations of iron metabolism occurring in cancer and the mechanisms through which iron promotes tumor growth.
Collapse
Affiliation(s)
- Myriam Y. Hsu
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Turin, Italy; (M.Y.H.); (E.M.)
| | - Erica Mina
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Turin, Italy; (M.Y.H.); (E.M.)
| | - Antonella Roetto
- Department of Clinical and Biological Science, University of Turin, AOU San Luigi Gonzaga, 10043 Orbassano, Italy
- Correspondence: (A.R.); (P.E.P.)
| | - Paolo E. Porporato
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Turin, Italy; (M.Y.H.); (E.M.)
- Correspondence: (A.R.); (P.E.P.)
| |
Collapse
|
23
|
Bhusal A, Rahman MH, Lee WH, Lee IK, Suk K. Satellite glia as a critical component of diabetic neuropathy: Role of lipocalin-2 and pyruvate dehydrogenase kinase-2 axis in the dorsal root ganglion. Glia 2020; 69:971-996. [PMID: 33251681 DOI: 10.1002/glia.23942] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 11/18/2020] [Accepted: 11/18/2020] [Indexed: 12/12/2022]
Abstract
Diabetic peripheral neuropathy (DPN) is a common complication of uncontrolled diabetes. The pathogenesis of DPN is associated with chronic inflammation in dorsal root ganglion (DRG), eventually causing structural and functional changes. Studies on DPN have primarily focused on neuronal component, and there is limited knowledge about the role of satellite glial cells (SGCs), although they completely enclose neuronal soma in DRG. Lipocalin-2 (LCN2) is a pro-inflammatory acute-phase protein found in high levels in diverse neuroinflammatory and metabolic disorders. In diabetic DRG, the expression of LCN2 was increased exclusively in the SGCs. This upregulation of LCN2 in SGCs correlated with increased inflammatory responses in DRG and sciatic nerve. Furthermore, diabetes-induced inflammation and morphological changes in DRG, as well as sciatic nerve, were attenuated in Lcn2 knockout (KO) mice. Lcn2 gene ablation also ameliorated neuropathy phenotype as determined by nerve conduction velocity and intraepidermal nerve fiber density. Mechanistically, studies using specific gene KO mice, adenovirus-mediated gene overexpression strategy, and primary cultures of DRG SGCs and neurons have demonstrated that LCN2 enhances the expression of mitochondrial gate-keeping regulator pyruvate dehydrogenase kinase-2 (PDK2) through PPARβ/δ, thereby inhibiting pyruvate dehydrogenase activity and increasing production of glycolytic end product lactic acid in DRG SGCs and neurons of diabetic mice. Collectively, our findings reveal a crucial role of glial LCN2-PPARβ/δ-PDK2-lactic acid axis in progression of DPN. Our results establish a link between pro-inflammatory LCN2 and glycolytic PDK2 in DRG SGCs and neurons and propose a novel glia-based mechanism and drug target for therapy of DPN. MAIN POINTS: Diabetes upregulates LCN2 in satellite glia, which in turn increases pyruvate dehydrogenase kinase-2 (PDK2) expression and lactic acid production in dorsal root ganglia (DRG). Glial LCN2-PDK2-lactic acid axis in DRG plays a crucial role in the pathogenesis of diabetic neuropathy.
Collapse
Affiliation(s)
- Anup Bhusal
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Md Habibur Rahman
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,Brain Science and Engineering Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Won-Ha Lee
- School of Life Sciences, Brain Korea 21 Plus/Kyungpook National University Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
| | - In-Kyu Lee
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea.,Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea
| | - Kyoungho Suk
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu, Republic of Korea.,Brain Science and Engineering Institute, Kyungpook National University, Daegu, Republic of Korea
| |
Collapse
|
24
|
Koliaraki V, Henriques A, Prados A, Kollias G. Unfolding innate mechanisms in the cancer microenvironment: The emerging role of the mesenchyme. J Exp Med 2020; 217:133714. [PMID: 32044979 PMCID: PMC7144533 DOI: 10.1084/jem.20190457] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 12/09/2019] [Accepted: 01/22/2020] [Indexed: 12/13/2022] Open
Abstract
Innate mechanisms in the tumor stroma play a crucial role both in the initial rejection of tumors and in cancer promotion. Here, we provide a concise overview of the innate system in cancer and recent advances in the field, including the activation and functions of innate immune cells and the emerging innate properties and modulatory roles of the fibroblastic mesenchyme. Novel insights into the diverse identities and functions of the innate immune and mesenchymal cells in the microenvironment of tumors should lead to improved anticancer therapies.
Collapse
Affiliation(s)
- Vasiliki Koliaraki
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece
| | - Ana Henriques
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece.,Institute for Bioinnovation, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece
| | - Alejandro Prados
- Institute for Bioinnovation, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece
| | - George Kollias
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece.,Institute for Bioinnovation, Biomedical Sciences Research Center "Alexander Fleming", Vari, Greece.,Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
25
|
Shibabaw T, Teferi B, Molla MD, Ayelign B. Inflammation Mediated Hepcidin-Ferroportin Pathway and Its Therapeutic Window in Breast Cancer. BREAST CANCER-TARGETS AND THERAPY 2020; 12:165-180. [PMID: 33116818 PMCID: PMC7585830 DOI: 10.2147/bctt.s276404] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 10/12/2020] [Indexed: 12/16/2022]
Abstract
Experimental and clinical data strongly support that iron is an essential element which plays a big role in cancer biology. Thus, hepcidin (Hp) and ferroportin (Fpn) are molecules that regulate and maintain the metabolism of iron. A peptide hormone hepcidin limits recycled and stored iron fluxes in macrophage and hepatic hepatocyte, respectively, to the blood stream by promoting degradation of the only iron exporter, Fpn, in the target cells. Moreover, the inflammatory microenvironment of breast cancer and altered hepcidin/ferroportin pathway is intimately linked. Breast cancer exhibits an iron seeking phenotype that is accomplished by tumor-associated macrophage (TAM). Because macrophages contribute to breast cancer growth and progression, this review will discuss TAM with an emphasis on describing how TAM (M2Ф phenotypic) interacts with their surrounding microenvironment and results in dysregulated Hp/Fpn and pathologic accumulation of iron as a hallmark of its malignant condition. Moreover, the underlying stroma or tumor microenvironment releases significant inflammatory cytokines like IL-6 and bone morphogenetic proteins like BMP-2 and 6 leading in aberrant Hp/Fpn pathways in breast cancer. Inflammation is primarily associated with the high intracellular iron levels, deregulated hepcidin/ferroportin pathway, and its upstream signaling in breast cancer. Subsequently, scholars have been reported that reducing iron level and manipulating the signaling molecules involved in iron metabolism can be used as a promising strategy of tumor chemotherapy. Here, we review the key molecular aspects of iron metabolism and its regulatory mechanisms of the hepcidin/ferroportin pathways and its current therapeutic strategies in breast cancer.
Collapse
Affiliation(s)
- Tewodros Shibabaw
- Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Banchamlak Teferi
- Department of Clinical Pharmacy, School of Pharmacy, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Meseret Derbew Molla
- Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Birhanu Ayelign
- Department of Immunology and Molecular Biology, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
26
|
Mertens C, Kuchler L, Sola A, Guiteras R, Grein S, Brüne B, von Knethen A, Jung M. Macrophage-Derived Iron-Bound Lipocalin-2 Correlates with Renal Recovery Markers Following Sepsis-Induced Kidney Damage. Int J Mol Sci 2020; 21:ijms21207527. [PMID: 33065981 PMCID: PMC7589935 DOI: 10.3390/ijms21207527] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 10/01/2020] [Accepted: 10/09/2020] [Indexed: 12/22/2022] Open
Abstract
During the course of sepsis in critically ill patients, kidney dysfunction and damage are among the first events of a complex scenario toward multi-organ failure and patient death. Acute kidney injury triggers the release of lipocalin-2 (Lcn-2), which is involved in both renal injury and recovery. Taking into account that Lcn-2 binds and transports iron with high affinity, we aimed at clarifying if Lcn-2 fulfills different biological functions according to its iron-loading status and its cellular source during sepsis-induced kidney failure. We assessed Lcn-2 levels both in serum and in the supernatant of short-term cultured renal macrophages (MΦ) as well as renal tubular epithelial cells (TEC) isolated from either Sham-operated or cecal ligation and puncture (CLP)-treated septic mice. Total kidney iron content was analyzed by Perls’ staining, while Lcn-2-bound iron in the supernatants of short-term cultured cells was determined by atomic absorption spectroscopy. Lcn-2 protein in serum was rapidly up-regulated at 6 h after sepsis induction and subsequently increased up to 48 h. Lcn-2-levels in the supernatant of TEC peaked at 24 h and were low at 48 h with no change in its iron-loading. In contrast, in renal MΦ Lcn-2 was low at 24 h, but increased at 48 h, where it mainly appeared in its iron-bound form. Whereas TEC-secreted, iron-free Lcn-2 was associated with renal injury, increased MΦ-released iron-bound Lcn-2 was linked to renal recovery. Therefore, we hypothesized that both the cellular source of Lcn-2 as well as its iron-load crucially adds to its biological function during sepsis-induced renal injury.
Collapse
Affiliation(s)
- Christina Mertens
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt am Main, Germany; (C.M.); (L.K.); (B.B.); (A.v.K.)
| | - Laura Kuchler
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt am Main, Germany; (C.M.); (L.K.); (B.B.); (A.v.K.)
| | - Anna Sola
- Department of Experimental Nephrology, IDIBELL, 08908 L’Hospitalet del Llobregat, Barcelona, Spain; (A.S.); (R.G.)
| | - Roser Guiteras
- Department of Experimental Nephrology, IDIBELL, 08908 L’Hospitalet del Llobregat, Barcelona, Spain; (A.S.); (R.G.)
| | - Stephan Grein
- Department of Mathematics, Temple University, Philadelphia, PA 19122, USA;
| | - Bernhard Brüne
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt am Main, Germany; (C.M.); (L.K.); (B.B.); (A.v.K.)
- Project Group Translational Medicine & Pharmacology TMP, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, 60590 Frankfurt am Main, Germany
| | - Andreas von Knethen
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt am Main, Germany; (C.M.); (L.K.); (B.B.); (A.v.K.)
- Project Group Translational Medicine & Pharmacology TMP, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, 60590 Frankfurt am Main, Germany
- Department of Anesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Goethe-University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Michaela Jung
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt am Main, Germany; (C.M.); (L.K.); (B.B.); (A.v.K.)
- Correspondence:
| |
Collapse
|
27
|
Torti SV, Torti FM. Iron and Cancer: 2020 Vision. Cancer Res 2020; 80:5435-5448. [PMID: 32928919 DOI: 10.1158/0008-5472.can-20-2017] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 08/06/2020] [Accepted: 09/08/2020] [Indexed: 12/18/2022]
Abstract
New and provocative insights into the relationships between iron and cancer have been uncovered in recent years. These include delineation of connections that link cellular iron to DNA repair, genomic integrity, and oncogenic signaling as well as the discovery of ferroptosis, a novel iron-dependent form of cell death. In parallel, new molecules and pathways that regulate iron influx, intracellular iron trafficking, and egress in normal cells, and their perturbations in cancer have been discovered. In addition, insights into the unique properties of iron handling in tumor-initiating cells (cancer stem cells), novel contributions of the tumor microenvironment to the uptake and regulation of iron in cancer cells, and new therapeutic modalities that leverage the iron dependence of cancer have emerged.
Collapse
Affiliation(s)
- Suzy V Torti
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, Connecticut.
| | - Frank M Torti
- Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut
| |
Collapse
|
28
|
Shi Z, Yin Y, Li C, Ding H, Mu N, Wang Y, Jin S, Ma H, Liu M, Zhou J. Lipocalin-2-induced proliferative endoplasmic reticulum stress participates in Kawasaki disease-related pulmonary arterial abnormalities. SCIENCE CHINA-LIFE SCIENCES 2020; 64:1000-1012. [PMID: 32915407 DOI: 10.1007/s11427-019-1772-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 06/30/2020] [Indexed: 11/30/2022]
Abstract
Clinical cases have reported pulmonary arterial structural and functional abnormalities in patients with Kawasaki disease (KD); however, the underlying mechanisms are unclear. In this study, a KD rat model was established via the intraperitoneal injection of Lactobacillus casei cell wall extract (LCWE). The results showed that pulmonary arterial functional and structural abnormalities were observed in KD rats. Furthermore, proliferative endoplasmic reticulum stress (ER stress) was observed in the pulmonary arteries of KD rats. Notably, the level of lipocalin-2 (Lcn 2), a trigger factor of inflammation, was remarkably elevated in the plasma and lung tissues of KD rats; increased Lcn 2 levels following LCWE stimulation may result from polymorphonuclear neutrophils (PMNs). Correspondingly, in cultured pulmonary artery smooth muscle cells (PASMCs), Lcn 2 markedly augmented the cleavage and nuclear localization of activating transcription factor-6 (ATF6), upregulated the transcription of glucose regulated protein 78 (GRP78) and neurite outgrowth inhibitor (NOGO), and promoted PASMCs proliferation. However, proapoptotic C/EBP homologous protein (CHOP) and caspase 12 levels were not elevated. Treatment with 4-phenyl butyric acid (4-PBA, a specific inhibitor of ER stress) inhibited PASMCs proliferation induced by Lcn 2 and attenuated pulmonary arterial abnormalities and right ventricular hypertrophy and reduced right ventricular systolic pressure in KD rats. In conclusion, Lcn 2 remarkably facilitates proliferative ER stress in PASMCs, which probably accounts for KD-related pulmonary arterial abnormalities.
Collapse
Affiliation(s)
- Zhaoling Shi
- Department of Pediatrics, Second Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, 712000, China.,Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fourth Military Medical University, Xi'an, 710032, China
| | - Yue Yin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fourth Military Medical University, Xi'an, 710032, China
| | - Chen Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fourth Military Medical University, Xi'an, 710032, China
| | - Hui Ding
- Department of Pediatrics, Second Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, 712000, China
| | - Nan Mu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fourth Military Medical University, Xi'an, 710032, China
| | - Yishi Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fourth Military Medical University, Xi'an, 710032, China
| | - Shanshan Jin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fourth Military Medical University, Xi'an, 710032, China
| | - Heng Ma
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fourth Military Medical University, Xi'an, 710032, China.
| | - Manling Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fourth Military Medical University, Xi'an, 710032, China.
| | - Jie Zhou
- Department of Endocrinology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
29
|
Chi Y, Remsik J, Kiseliovas V, Derderian C, Sener U, Alghader M, Saadeh F, Nikishina K, Bale T, Iacobuzio-Donahue C, Thomas T, Pe'er D, Mazutis L, Boire A. Cancer cells deploy lipocalin-2 to collect limiting iron in leptomeningeal metastasis. Science 2020; 369:276-282. [PMID: 32675368 DOI: 10.1126/science.aaz2193] [Citation(s) in RCA: 144] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 06/01/2020] [Indexed: 12/21/2022]
Abstract
The tumor microenvironment plays a critical regulatory role in cancer progression, especially in central nervous system metastases. Cancer cells within the cerebrospinal fluid (CSF)-filled leptomeninges face substantial microenvironmental challenges, including inflammation and sparse micronutrients. To investigate the mechanism by which cancer cells in these leptomeningeal metastases (LM) overcome these constraints, we subjected CSF from five patients with LM to single-cell RNA sequencing. We found that cancer cells, but not macrophages, within the CSF express the iron-binding protein lipocalin-2 (LCN2) and its receptor SCL22A17. These macrophages generate inflammatory cytokines that induce cancer cell LCN2 expression but do not generate LCN2 themselves. In mouse models of LM, cancer cell growth is supported by the LCN2/SLC22A17 system and is inhibited by iron chelation therapy. Thus, cancer cells appear to survive in the CSF by outcompeting macrophages for iron.
Collapse
Affiliation(s)
- Yudan Chi
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jan Remsik
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Vaidotas Kiseliovas
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Camille Derderian
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ugur Sener
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Majdi Alghader
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Fadi Saadeh
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Katie Nikishina
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Tejus Bale
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Christine Iacobuzio-Donahue
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.,Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Tiffany Thomas
- Department of Pathology and Cell Biology, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Dana Pe'er
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.,Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Linas Mazutis
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.,Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Adrienne Boire
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA. .,Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.,Brain Tumor Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
30
|
Munir H, Mazzaglia C, Shields JD. Stromal regulation of tumor-associated lymphatics. Adv Drug Deliv Rev 2020; 161-162:75-89. [PMID: 32783989 DOI: 10.1016/j.addr.2020.08.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 07/27/2020] [Accepted: 08/06/2020] [Indexed: 02/08/2023]
Abstract
Recent advances have identified a growing array of roles played by lymphatics in the tumor microenvironment, from providing a route of metastasis to immune modulation. The tumor microenvironment represents an exceptionally complex, dynamic niche comprised of a diverse mixture of cancer cells and normal host cells termed the stroma. This review discusses our current understanding of stromal elements and how they regulate lymphatic growth and functional properties in the tumor context.
Collapse
Affiliation(s)
- Hafsa Munir
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Box 197 Cambridge Biomedical Campus, Cambridge, CB2 0XZ
| | - Corrado Mazzaglia
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Box 197 Cambridge Biomedical Campus, Cambridge, CB2 0XZ
| | - Jacqueline D Shields
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Box 197 Cambridge Biomedical Campus, Cambridge, CB2 0XZ.
| |
Collapse
|
31
|
Malone MK, Smrekar K, Park S, Blakely B, Walter A, Nasta N, Park J, Considine M, Danilova LV, Pandey NB, Fertig EJ, Popel AS, Jin K. Cytokines secreted by stromal cells in TNBC microenvironment as potential targets for cancer therapy. Cancer Biol Ther 2020; 21:560-569. [PMID: 32213106 PMCID: PMC7515526 DOI: 10.1080/15384047.2020.1739484] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 12/08/2019] [Accepted: 02/27/2020] [Indexed: 02/07/2023] Open
Abstract
In triple-negative breast cancer (TNBC), the lack of therapeutic markers and effective targeted therapies result in an incurable metastatic disease associated with a poor prognosis. Crosstalks within the tumor microenvironment (TME), including those between cancer and stromal cells, affect the tumor heterogeneity, growth, and metastasis. Previously, we have demonstrated that IL-6, IL-8, and CCL5 play a significant role in TNBC growth and metastasis. In this study, we performed a systematic analysis of cytokine factors secreted from four stromal components (fibroblasts, macrophages, lymphatic endothelial cells, and blood microvascular endothelial cells) induced by four TNBC cell types. Through bioinformatic analysis, we selected putative candidates of secreted factors from stromal cells, which are involved in EMT activity, cell proliferation, metabolism, and matrisome pathways. Among the candidates, LCN2, GM-CSF, CST3, IL-6, IL-8, and CHI3L1 are ranked highly. Significantly, Lipocalin-2 (LCN2) is upregulated in the crosstalk of stromal cells and four different TNBC cells. We validated the increase of LCN2 secreted from four stromal cells induced by TNBC cells. Using a specific LCN2 antibody, we observed the inhibition of TNBC cell growth and migration. Taken together, these results propose secreted factors as molecular targets to treat TNBC progression via crosstalk with stromal components.
Collapse
Affiliation(s)
- Marie K. Malone
- Department of Pharmaceutical Science, Albany College of Pharmacy and Health Science, Albany, NY, USA
| | - Karly Smrekar
- Department of Pharmaceutical Science, Albany College of Pharmacy and Health Science, Albany, NY, USA
| | - Sunju Park
- Department of Pharmaceutical Science, Albany College of Pharmacy and Health Science, Albany, NY, USA
| | - Brianna Blakely
- Department of Pharmaceutical Science, Albany College of Pharmacy and Health Science, Albany, NY, USA
| | - Alec Walter
- Department of Pharmaceutical Science, Albany College of Pharmacy and Health Science, Albany, NY, USA
| | - Nicholas Nasta
- Department of Pharmaceutical Science, Albany College of Pharmacy and Health Science, Albany, NY, USA
| | - Jay Park
- Department of Pharmaceutical Science, Albany College of Pharmacy and Health Science, Albany, NY, USA
| | - Michael Considine
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ludmila V. Danilova
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Niranjan B. Pandey
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elana J. Fertig
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Applied Mathematics and Statistics, Johns Hopkins University Whiting School of Engineering, Baltimore, MD, USA
| | - Aleksander S. Popel
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kideok Jin
- Department of Pharmaceutical Science, Albany College of Pharmacy and Health Science, Albany, NY, USA
| |
Collapse
|
32
|
Mirabile A, Rivoltini L, Daveri E, Vernieri C, Mele R, Porcu L, Lazzari C, Bulotta A, Viganò MG, Cascinu S, Gregorc V. Metabolism and Immune Modulation in Patients with Solid Tumors: Systematic Review of Preclinical and Clinical Evidence. Cancers (Basel) 2020; 12:E1153. [PMID: 32375310 PMCID: PMC7281426 DOI: 10.3390/cancers12051153] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 04/22/2020] [Accepted: 04/27/2020] [Indexed: 02/07/2023] Open
Abstract
Several immunotherapy agents are the standard of care of many solid malignancies. Nevertheless, the majority of patients do not benefit from the currently available immunotherapies. It is therefore of paramount importance to identify the prognostic and predictive factors of tumor response/resistance and to design effective therapeutic strategies to overcome primary resistance and improve the efficacy of immunotherapy. The aim of this review is to underline the influence of the tumor and host metabolism on the antitumor immune response and to discuss possible strategies to improve the efficacy of available treatments by targeting the specific metabolic pathways in tumors or immune cells and by modifying patients' nutritional statuses. A systematic search of the Medline and EMBASE databases was carried out to identify scientific papers published until February 2020, which reported original research articles on the influence of tumor or host metabolism on antitumor immune response. The literature data showed the key role of glycolysis and mitochondrial oxidative phosphorylation, arginine, tryptophan, glutamine, lipid metabolism and microbiome on immune cell function. Moreover, specific nutritional behaviors, such as a low dietary intake of vitamin C, low glycemic index and alpha-linolenic acid, eicosapentenoic acid, docosahexaenoic acid, ornithine ketoglutarate, tryptophan and probiotic supplementation were associated with the potential clinical benefits from the currently available immunotherapies.
Collapse
Affiliation(s)
- Aurora Mirabile
- Department of Medical Oncology, Scientific Institute San Raffaele Hospital, Via Olgettina, 60, 20132 Milan, Italy; (C.L.); (A.B.); (M.G.V.); (S.C.); (V.G.)
| | - Licia Rivoltini
- Immunotherapy of Human Tumors, IRCCS National Cancer Institute (INT) and University of Milan, Via Venezian 1, 20133 Milan, Italy; (L.R.); (E.D.)
| | - Elena Daveri
- Immunotherapy of Human Tumors, IRCCS National Cancer Institute (INT) and University of Milan, Via Venezian 1, 20133 Milan, Italy; (L.R.); (E.D.)
| | - Claudio Vernieri
- Medical Oncology Department, IRCCS IRCCS National Cancer Institute (INT) and University of Milan, Via Venezian 1, 20133 Milan, Italy;
- IFOM, the FIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Roberto Mele
- Nutritionist biologist, Hospital Health Direction, Scientific Institute San Raffaele Hospital, Via Olgettina 60, 20132 Milan, Italy;
| | - Luca Porcu
- Methodological Research Unit, Institute of Pharmacological Research Mario Negri, Via Mario Negri 2, 20156 Milan, Italy;
| | - Chiara Lazzari
- Department of Medical Oncology, Scientific Institute San Raffaele Hospital, Via Olgettina, 60, 20132 Milan, Italy; (C.L.); (A.B.); (M.G.V.); (S.C.); (V.G.)
| | - Alessandra Bulotta
- Department of Medical Oncology, Scientific Institute San Raffaele Hospital, Via Olgettina, 60, 20132 Milan, Italy; (C.L.); (A.B.); (M.G.V.); (S.C.); (V.G.)
| | - Maria Grazia Viganò
- Department of Medical Oncology, Scientific Institute San Raffaele Hospital, Via Olgettina, 60, 20132 Milan, Italy; (C.L.); (A.B.); (M.G.V.); (S.C.); (V.G.)
| | - Stefano Cascinu
- Department of Medical Oncology, Scientific Institute San Raffaele Hospital, Via Olgettina, 60, 20132 Milan, Italy; (C.L.); (A.B.); (M.G.V.); (S.C.); (V.G.)
| | - Vanesa Gregorc
- Department of Medical Oncology, Scientific Institute San Raffaele Hospital, Via Olgettina, 60, 20132 Milan, Italy; (C.L.); (A.B.); (M.G.V.); (S.C.); (V.G.)
| |
Collapse
|
33
|
Brown RAM, Richardson KL, Kabir TD, Trinder D, Ganss R, Leedman PJ. Altered Iron Metabolism and Impact in Cancer Biology, Metastasis, and Immunology. Front Oncol 2020; 10:476. [PMID: 32328462 PMCID: PMC7160331 DOI: 10.3389/fonc.2020.00476] [Citation(s) in RCA: 136] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 03/17/2020] [Indexed: 12/12/2022] Open
Abstract
Iron is an essential nutrient that plays a complex role in cancer biology. Iron metabolism must be tightly controlled within cells. Whilst fundamental to many cellular processes and required for cell survival, excess labile iron is toxic to cells. Increased iron metabolism is associated with malignant transformation, cancer progression, drug resistance and immune evasion. Depleting intracellular iron stores, either with the use of iron chelating agents or mimicking endogenous regulation mechanisms, such as microRNAs, present attractive therapeutic opportunities, some of which are currently under clinical investigation. Alternatively, iron overload can result in a form of regulated cell death, ferroptosis, which can be activated in cancer cells presenting an alternative anti-cancer strategy. This review focuses on alterations in iron metabolism that enable cancer cells to meet metabolic demands required during different stages of tumorigenesis in relation to metastasis and immune response. The strength of current evidence is considered, gaps in knowledge are highlighted and controversies relating to the role of iron and therapeutic targeting potential are discussed. The key question we address within this review is whether iron modulation represents a useful approach for treating metastatic disease and whether it could be employed in combination with existing targeted drugs and immune-based therapies to enhance their efficacy.
Collapse
Affiliation(s)
- Rikki A. M. Brown
- Queen Elizabeth II Medical Centre, Harry Perkins Institute of Medical Research, Perth, WA, Australia
- UWA Centre for Medical Research, University of Western Australia, Perth, WA, Australia
- UWA Medical School, University of Western Australia, Perth, WA, Australia
| | - Kirsty L. Richardson
- Queen Elizabeth II Medical Centre, Harry Perkins Institute of Medical Research, Perth, WA, Australia
- UWA Centre for Medical Research, University of Western Australia, Perth, WA, Australia
| | - Tasnuva D. Kabir
- Queen Elizabeth II Medical Centre, Harry Perkins Institute of Medical Research, Perth, WA, Australia
- UWA Centre for Medical Research, University of Western Australia, Perth, WA, Australia
| | - Debbie Trinder
- Queen Elizabeth II Medical Centre, Harry Perkins Institute of Medical Research, Perth, WA, Australia
- UWA Centre for Medical Research, University of Western Australia, Perth, WA, Australia
- UWA Medical School, University of Western Australia, Perth, WA, Australia
| | - Ruth Ganss
- Queen Elizabeth II Medical Centre, Harry Perkins Institute of Medical Research, Perth, WA, Australia
- UWA Centre for Medical Research, University of Western Australia, Perth, WA, Australia
| | - Peter J. Leedman
- Queen Elizabeth II Medical Centre, Harry Perkins Institute of Medical Research, Perth, WA, Australia
- UWA Centre for Medical Research, University of Western Australia, Perth, WA, Australia
- UWA Medical School, University of Western Australia, Perth, WA, Australia
| |
Collapse
|
34
|
Urbschat A, Thiemens AK, Mertens C, Rehwald C, Meier JK, Baer PC, Jung M. Macrophage-secreted Lipocalin-2 Promotes Regeneration of Injured Primary Murine Renal Tubular Epithelial Cells. Int J Mol Sci 2020; 21:ijms21062038. [PMID: 32188161 PMCID: PMC7139578 DOI: 10.3390/ijms21062038] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/12/2020] [Accepted: 03/13/2020] [Indexed: 12/12/2022] Open
Abstract
Lipocalin-2 (Lcn-2) is rapidly upregulated in macrophages after renal tubular injury and acts as renoprotective and pro-regenerative agent. Lcn-2 possesses the ability to bind and transport iron with high affinity. Therefore, the present study focuses on the decisive role of the Lcn-2 iron-load for its pro-regenerative function. Primary mouse tubular epithelial cells were isolated from kidney tissue of wildtype mice and incubated with 5μM Cisplatin for 24h to induce injury. Bone marrow-derived macrophages of wildtype and Lcn-2-/- mice were isolated and polarized with IL-10 towards an anti-inflammatory, iron-release phenotype. Their supernatants as well as recombinant iron-loaded holo-Lcn-2 was used for stimulation of Cisplatin-injured tubular epithelial cells. Incubation of tubular epithelial cells with wildtype supernatants resulted in less damage and induced cellular proliferation, whereas in absence of Lcn-2 no protective effect was observed. Epithelial integrity as well as cellular proliferation showed a clear protection upon rescue experiments applying holo-Lcn-2. Notably, we detected a positive correlation between total iron amounts in tubular epithelial cells and cellular proliferation, which, in turn, reinforced the assumed link between availability of Lcn-2-bound iron and recovery. We hypothesize that macrophage-released Lcn-2-bound iron is provided to tubular epithelial cells during toxic cell damage, whereby injury is limited and recovery is favored.
Collapse
Affiliation(s)
- Anja Urbschat
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark;
| | - Anne-Kathrin Thiemens
- Division of Nephrology, Department of Internal Medicine III, Goethe-University Frankfurt, 60323 Frankfurt am Main, Germany; (A.-K.T.); (P.C.B.)
| | - Christina Mertens
- Institute of Biochemistry I, Goethe-University Frankfurt, Faculty of Medicine, 60323 Frankfurt am Main, Germany; (C.M.); (C.R.)
| | - Claudia Rehwald
- Institute of Biochemistry I, Goethe-University Frankfurt, Faculty of Medicine, 60323 Frankfurt am Main, Germany; (C.M.); (C.R.)
| | - Julia K. Meier
- Institute of Biochemistry I, Goethe-University Frankfurt, Faculty of Medicine, 60323 Frankfurt am Main, Germany; (C.M.); (C.R.)
| | - Patrick C. Baer
- Division of Nephrology, Department of Internal Medicine III, Goethe-University Frankfurt, 60323 Frankfurt am Main, Germany; (A.-K.T.); (P.C.B.)
| | - Michaela Jung
- Institute of Biochemistry I, Goethe-University Frankfurt, Faculty of Medicine, 60323 Frankfurt am Main, Germany; (C.M.); (C.R.)
- Correspondence:
| |
Collapse
|
35
|
Zhang L, Dong Y, Wang Y, Hu W, Dong S, Chen Y. Sphingosine-1-phosphate (S1P) receptors: Promising drug targets for treating bone-related diseases. J Cell Mol Med 2020; 24:4389-4401. [PMID: 32155312 PMCID: PMC7176849 DOI: 10.1111/jcmm.15155] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/22/2020] [Accepted: 02/01/2020] [Indexed: 12/20/2022] Open
Abstract
Sphingosine-1-phosphate (S1P) is a natural bioactive lipid molecule and a common first or second messenger in the cardiovascular and immune systems. By binding with its receptors, S1P can serve as mediator of signalling during cell migration, differentiation, proliferation and apoptosis. Although the predominant role of S1P in bone regeneration has been noted in many studies, this role is not as well-known as its roles in the cardiovascular and immune systems. In this review, we summarize previous research on the role of S1P receptors (S1PRs) in osteoblasts and osteoclasts. In addition, S1P is regarded as a bridge between bone resorption and formation, which brings hope to patients with bone-related diseases. Finally, we discuss S1P and its receptors as therapeutic targets for treating osteoporosis, inflammatory osteolysis and bone metastasis based on the biological effects of S1P in osteoclastic/osteoblastic cells, immune cells and tumour cells.
Collapse
Affiliation(s)
- Lincheng Zhang
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing, China.,Battalion One of Basic Medical Sciences, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yutong Dong
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing, China.,Battalion One of Basic Medical Sciences, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yiran Wang
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing, China
| | - Wenhui Hu
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing, China
| | - Shiwu Dong
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing, China.,State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), Chongqing, China.,Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yueqi Chen
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing, China.,Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
36
|
Jiang MJ, Gu DN, Dai JJ, Huang Q, Tian L. Dark Side of Cytotoxic Therapy: Chemoradiation-Induced Cell Death and Tumor Repopulation. Trends Cancer 2020; 6:419-431. [PMID: 32348737 DOI: 10.1016/j.trecan.2020.01.018] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 01/25/2020] [Accepted: 01/27/2020] [Indexed: 12/20/2022]
Abstract
Accelerated tumor repopulation following chemoradiation is often observed in the clinic, but the underlying mechanisms remain unclear. In recent years, dying cells caused by chemoradiation have attracted much attention, and they may manifest diverse forms of cell death and release complex factors and thus orchestrate tumor repopulation cascades. Dying cells potentiate the survival of residual living tumor cells, remodel the tumor microenvironment, boost cell proliferation, and accelerate cancer cell metastasis. Moreover, dying cells also mediate the side effects of chemoradiation. These findings suggest more caution when weighing the benefits of cytotoxic therapy and the need to accordingly develop new strategies for cancer treatment.
Collapse
Affiliation(s)
- Ming-Jie Jiang
- Institute of Translational Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China; Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Dian-Na Gu
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China; Department of Chemoradiotherapy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Juan-Juan Dai
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Qian Huang
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China; Cancer Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Ling Tian
- Institute of Translational Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China; Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China; Department of Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China.
| |
Collapse
|
37
|
Schnetz M, Meier JK, Rehwald C, Mertens C, Urbschat A, Tomat E, Akam EA, Baer P, Roos FC, Brüne B, Jung M. The Disturbed Iron Phenotype of Tumor Cells and Macrophages in Renal Cell Carcinoma Influences Tumor Growth. Cancers (Basel) 2020; 12:cancers12030530. [PMID: 32106629 PMCID: PMC7139531 DOI: 10.3390/cancers12030530] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 02/21/2020] [Accepted: 02/23/2020] [Indexed: 12/27/2022] Open
Abstract
Accumulating evidence suggests that iron homeostasis is disturbed in tumors. We aimed at clarifying the distribution of iron in renal cell carcinoma (RCC). Considering the pivotal role of macrophages for iron homeostasis and their association with poor clinical outcome, we investigated the role of macrophage-secreted iron for tumor progression by applying a novel chelation approach. We applied flow cytometry and multiplex-immunohistochemistry to detect iron-dependent markers and analyzed iron distribution with atomic absorption spectrometry in patients diagnosed with RCC. We further analyzed the functional significance of iron by applying a novel extracellular chelator using RCC cell lines as well as patient-derived primary cells. The expression of iron-regulated genes was significantly elevated in tumors compared to adjacent healthy tissue. Iron retention was detected in tumor cells, whereas tumor-associated macrophages showed an iron-release phenotype accompanied by enhanced expression of ferroportin. We found increased iron amounts in extracellular fluids, which in turn stimulated tumor cell proliferation and migration. In vitro, macrophage-derived iron showed pro-tumor functions, whereas application of an extracellular chelator blocked these effects. Our study provides new insights in iron distribution and iron-handling in RCC. Chelators that specifically scavenge iron in the extracellular space confirmed the importance of macrophage-secreted iron in promoting tumor growth.
Collapse
Affiliation(s)
- Matthias Schnetz
- Institute of Biochemistry I, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (M.S.); (J.K.M.); (C.R.); (C.M.); (B.B.)
| | - Julia K. Meier
- Institute of Biochemistry I, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (M.S.); (J.K.M.); (C.R.); (C.M.); (B.B.)
| | - Claudia Rehwald
- Institute of Biochemistry I, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (M.S.); (J.K.M.); (C.R.); (C.M.); (B.B.)
| | - Christina Mertens
- Institute of Biochemistry I, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (M.S.); (J.K.M.); (C.R.); (C.M.); (B.B.)
| | - Anja Urbschat
- Institute for Biomedicine, Aarhus University, C. F. Møllers Allé 6, 8000 Aarhus, Denmark;
| | - Elisa Tomat
- Department of Chemistry and Biochemistry, University of Arizona, 1306 E. University Blvd., Tucson, AZ 85721-0041, USA; (E.T.); (E.A.A.)
| | - Eman A. Akam
- Department of Chemistry and Biochemistry, University of Arizona, 1306 E. University Blvd., Tucson, AZ 85721-0041, USA; (E.T.); (E.A.A.)
| | - Patrick Baer
- Division of Nephrology, Department of Internal Medicine III, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany;
| | - Frederik C. Roos
- Clinic of Urology, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany;
| | - Bernhard Brüne
- Institute of Biochemistry I, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (M.S.); (J.K.M.); (C.R.); (C.M.); (B.B.)
- German Cancer Consortium (DKTK), partner site Frankfurt/Mainz, 60590 Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe-University Frankfurt, 60596 Frankfurt am Main, Germany
- Project Group Translational Medicine and Pharmacology TMP, Fraunhofer Institute for Molecular Biology and Applied Ecology, 60596 Frankfurt am Main, Germany
| | - Michaela Jung
- Institute of Biochemistry I, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (M.S.); (J.K.M.); (C.R.); (C.M.); (B.B.)
- Correspondence: ; Tel.: +49-69-6301-6931; Fax: +49-69-6301-4203
| |
Collapse
|
38
|
Wang Y, Wang H, Zhang C, Zhang C, Yang H, Gao R, Tong Z. Plasma Hsa-miR-92a-3p in correlation with lipocalin-2 is associated with sepsis-induced coagulopathy. BMC Infect Dis 2020; 20:155. [PMID: 32075600 PMCID: PMC7031893 DOI: 10.1186/s12879-020-4853-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 02/06/2020] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Sepsis is a life-threatening situation, and it can be rendered more severe by coagulopathy. We here examine a novel plasma biomarker for sepsis-induced coagulopathy. METHODS A total of 116 patients diagnosed with sepsis were recruited and divided into two groups by whether they also had coagulopathy. Plasma samples were collected on arrival at the intensive care unit. Fifteen sepsis-alone and 15 sepsis-induced coagulopathy plasma samples were mixed and sent for microRNA sequencing. Differently expressed microRNAs were then validated by quantitative reverse transcriptase polymerase chain reaction in 52 sepsis-alone and 34 sepsis-induced coagulopathy patients; plasma lipocalin-2 was measured as well. RESULTS Four microRNAs were selected from microRNA sequencing. Only hsa-mir-92a-3p was differently expressed in the validation set. Its level of expression was significantly lower in sepsis-induced coagulopathy group. Hsa-mir-92a-3p had an area under a receiver operating characteristic curve of 0.660 (95% confidence interval, 0.537, 0.782). The plasma Hsa-mir-92a-3p level was related to activated partial thromboplastin time, prothrombin activity, and plasma lipocalin-2 level. A binary logistic model showed an association between hsa-mir-92a-3p and fibrinogen with SIC. CONCLUSIONS The utility of hsa-mir-92a-3p as a biomarker for sepsis-induced coagulopathy needs more verification, and the regulatory mechanism of hsa-mir-92a-3p in coagulation disorder and its potency as a therapeutic target must be confirmed.
Collapse
Affiliation(s)
- Yishan Wang
- Department of Respiratory and Critical Care Medicine, Beijing Engineering Research Center of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing Institute of Respiratory Medicine, NO. 8, Gong Ti South Road, Chao-Yang District, Beijing, 100020, China
| | - Huijuan Wang
- Department of Respiratory and Critical Care Medicine, Beijing Engineering Research Center of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing Institute of Respiratory Medicine, NO. 8, Gong Ti South Road, Chao-Yang District, Beijing, 100020, China
| | - Chunfang Zhang
- Department of Anesthesiology, Pain Medicine and Critical Care Medicine, Aviation General Hospital of China Medical University and Beijing Institute of Translational Medicine, Chinese Academy of Sciences, Beijing, 100012, China
| | - Chao Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Engineering Research Center of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing Institute of Respiratory Medicine, NO. 8, Gong Ti South Road, Chao-Yang District, Beijing, 100020, China
| | - Huqin Yang
- Department of Respiratory and Critical Care Medicine, Beijing Engineering Research Center of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing Institute of Respiratory Medicine, NO. 8, Gong Ti South Road, Chao-Yang District, Beijing, 100020, China
| | - Ruiyue Gao
- Department of Respiratory and Critical Care Medicine, Beijing Engineering Research Center of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing Institute of Respiratory Medicine, NO. 8, Gong Ti South Road, Chao-Yang District, Beijing, 100020, China
| | - Zhaohui Tong
- Department of Respiratory and Critical Care Medicine, Beijing Engineering Research Center of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing Institute of Respiratory Medicine, NO. 8, Gong Ti South Road, Chao-Yang District, Beijing, 100020, China.
| |
Collapse
|
39
|
Implication and role of neutrophil gelatinase-associated lipocalin in cancer: lipocalin-2 as a potential novel emerging comprehensive therapeutic target for a variety of cancer types. Mol Biol Rep 2020; 47:2327-2346. [PMID: 31970626 DOI: 10.1007/s11033-020-05261-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 01/16/2020] [Indexed: 12/18/2022]
Abstract
Cancer is a leading cause of mortalities worldwide. Over the past few decades, exploration of molecular mechanisms behind cancer initiation and progression has been of great interest in the viewpoint of both basic and clinical scientists. It is generally believed that identification of key molecules implicated in cancer pathology not only improves our understanding of the disease, but also could result in introduction of novel therapeutic strategies. Neutrophil gelatinase-associated lipocalin (NGAL)/lipocalin-2 (LCN2) is a member of lipocalin superfamily with a variety of functions. Although the main function of LCN2 is still unknown, many studies confirmed its significant role in the initiation, progression, and metastasis of various types of cancer. Furthermore, aberrant expression of LCN2 is also concerned with the chemo- and radio-resistant phenotypes of tumors. Here, we will review the contribution of known functions of LCN2 to the pathophysiology of cancer. We also highlight how the deregulated expression of LCN2 is associated with a variety of fatal types of cancer for which there are no effective therapeutic modalities. The unique and multiple functions of LCN2 and its widespread expression in different types of cancer prompted us to suggest LCN2 could be considered either as a valuable diagnostic and prognostic biomarker or as a potential novel therapeutic target.
Collapse
|
40
|
Cancer progression is mediated by proline catabolism in non-small cell lung cancer. Oncogene 2020; 39:2358-2376. [PMID: 31911619 DOI: 10.1038/s41388-019-1151-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 12/05/2019] [Accepted: 12/23/2019] [Indexed: 12/15/2022]
Abstract
Dysregulated metabolism contributes to cancer initiation and progression, but the key drivers of these pathways are just being discovered. Here, we report a critical role for proline catabolism in non-small cell lung cancer (NSCLC). Proline dehydrogenase (PRODH) is activated to reduce proline levels by the chromatin remodeling factor lymphoid-specific helicase (LSH), an epigenetic driver of NSCLC. PRODH promotes NSCLC tumorigenesis by inducing epithelial to mesenchymal transition (EMT) and IKKα-dependent inflammatory genes, including CXCL1, LCN2, and IL17C. Consistently, proline addition promotes the expression of these inflammatory genes, as well as EMT, tumor cell proliferation, and migration in vitro and tumor growth in vivo, while the depletion or inhibition of PRODH blocks these phenotypes. In summary, we reveal an essential metabolic pathway amenable to targeting in NSCLC.
Collapse
|
41
|
Abstract
Cancer metabolism is a well-known target of cancer therapeutics. Classically, cancer metabolism has been studied in terms of the dependence of cancer cells on crucial metabolites, such as glucose and glutamine. But, the accumulating data show that iron metabolism in tumor microenvironment is also an important factor in preserving the survival of cancer cells. Cancer cells have a distinct phenotype of iron metabolism, which secures the much-needed iron for these metabolically active cells. In order to use this iron efficiently, cancer cells need to increase their iron supply and decrease iron loss. As recent research suggests, this is not only done by modifying the expression of iron-related proteins in cancer cells, but also by interaction of cancer cells with other cells from the tumor milieu. Tumor microenvironment is a dynamic environment characterized with intricate relationship between cancer cells, tumor-associated macrophages, fibroblasts, and other cells. Some of the mechanistic aspects of this relationship have been elucidated, while others are yet to be identified. In any case, identifying the details of the iron phenotype of the cells in tumor microenvironment presents with a new therapeutic opportunity to treat this deadly disease.
Collapse
|
42
|
Rehwald C, Schnetz M, Urbschat A, Mertens C, Meier JK, Bauer R, Baer P, Winslow S, Roos FC, Zwicker K, Huard A, Weigert A, Brüne B, Jung M. The iron load of lipocalin-2 (LCN-2) defines its pro-tumour function in clear-cell renal cell carcinoma. Br J Cancer 2019; 122:421-433. [PMID: 31772326 PMCID: PMC7000824 DOI: 10.1038/s41416-019-0655-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 10/16/2019] [Accepted: 11/05/2019] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND We aimed at clarifying the role of lipocalin-2 (LCN-2) in clear-cell renal cell carcinoma (ccRCC). Since LCN-2 was recently identified as a novel iron transporter, we explored its iron load as a decisive factor in conferring its biological function. METHODS LCN-2 expression was analysed at the mRNA and protein level by using immunohistochemistry, RNAscope® and qRT-PCR in patients diagnosed with clear-cell renal cell carcinoma compared with adjacent healthy tissue. We measured LCN-2-bound iron by atomic absorption spectrometry from patient-derived samples and applied functional assays by using ccRCC cell lines, primary cells, and 3D tumour spheroids to verify the role of the LCN-2 iron load in tumour progression. RESULTS LCN-2 was associated with poor patient survival and LCN-2 mRNA clustered in high- and low-expressing ccRCC patients. LCN-2 protein was found overexpressed in tumour compared with adjacent healthy tissue, whereby LCN-2 was iron loaded. In vitro, the iron load determines the biological function of LCN-2. Iron-loaded LCN-2 showed pro-tumour functions, whereas iron-free LCN-2 produced adverse effects. CONCLUSIONS We provide new insights into the pro-tumour function of LCN-2. LCN-2 donates iron to cells to promote migration and matrix adhesion. Since the iron load of LCN-2 determines its pro-tumour characteristics, targeting either its iron load or its receptor interaction might represent new therapeutic options.
Collapse
Affiliation(s)
- Claudia Rehwald
- Institute of Biochemistry I, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Matthias Schnetz
- Institute of Biochemistry I, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Anja Urbschat
- Institute for Biomedicine, Aarhus University, C. F. Møllers Allé 6, 8000, Aarhus, Denmark.,Clinic of Urology and Pediatric Urology, Philipps-University Marburg, Baldingerstraße, 35043, Marburg, Germany
| | - Christina Mertens
- Institute of Biochemistry I, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Julia K Meier
- Institute of Biochemistry I, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Rebekka Bauer
- Institute of Biochemistry I, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Patrick Baer
- Division of Nephrology, Department of Internal Medicine III, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Sofia Winslow
- Institute of Biochemistry I, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Frederik C Roos
- Clinic of Urology, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt, Germany
| | - Klaus Zwicker
- Institute of Biochemistry I, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Arnaud Huard
- Institute of Biochemistry I, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Andreas Weigert
- Institute of Biochemistry I, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Bernhard Brüne
- Institute of Biochemistry I, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany.,German Cancer Consortium (DKTK), partner site Frankfurt/Mainz, Germany.,Frankfurt Cancer Institute, Goethe-University Frankfurt, 60596, Frankfurt, Germany.,Project Group Translational Medicine and Pharmacology TMP, Fraunhofer Institute for Molecular Biology and Applied Ecology, 60596, Frankfurt, Germany
| | - Michaela Jung
- Institute of Biochemistry I, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany.
| |
Collapse
|
43
|
Dvoretskiy S, Garg K, Munroe M, Pincu Y, Mahmassani ZS, Coombs C, Blackwell B, Garcia G, Waterstradt G, Lee I, Drnevich J, Rhodes JS, Boppart MD. The impact of skeletal muscle contraction on CD146 +Lin - pericytes. Am J Physiol Cell Physiol 2019; 317:C1011-C1024. [PMID: 31433691 PMCID: PMC6879875 DOI: 10.1152/ajpcell.00156.2019] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 08/15/2019] [Accepted: 08/19/2019] [Indexed: 12/14/2022]
Abstract
Unaccustomed resistance exercise can initiate skeletal muscle remodeling and adaptive mechanisms that can confer protection from damage and enhanced strength with subsequent stimulation. The myofiber may provide the primary origin for adaptation, yet multiple mononuclear cell types within the surrounding connective tissue may also contribute. The purpose of this study was to evaluate the acute response of muscle-resident interstitial cells to contraction initiated by electrical stimulation (e-stim) and subsequently determine the contribution of pericytes to remodeling as a result of training. Mice were subjected to bilateral e-stim or sham treatment. Following a single session of e-stim, NG2+CD45-CD31- (NG2+Lin-) pericyte, CD146+Lin- pericyte, and PDGFRα+ fibroadipogenic progenitor cell quantity and function were evaluated via multiplex flow cytometry and targeted quantitative PCR. Relative quantity was not significantly altered 24 h postcontraction, yet unique gene signatures were observed for each cell population at 3 h postcontraction. CD146+Lin- pericytes appeared to be most responsive to contraction, and upregulation of genes related to immunomodulation and extracellular matrix remodeling was observed via RNA sequencing. Intramuscular injection of CD146+Lin- pericytes did not significantly increase myofiber size yet enhanced ECM remodeling and angiogenesis in response to repeated bouts of e-stim for 4 wk. The results from this study provide the first evidence that CD146+Lin- pericytes are responsive to skeletal muscle contraction and may contribute to the beneficial outcomes associated with exercise.
Collapse
Affiliation(s)
- Svyatoslav Dvoretskiy
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, Illinois
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Koyal Garg
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Michael Munroe
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, Illinois
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Yair Pincu
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, Illinois
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Ziad S Mahmassani
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, Illinois
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Charlotte Coombs
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Brent Blackwell
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, Illinois
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Gabriela Garcia
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, Illinois
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Garret Waterstradt
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, Illinois
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Isaac Lee
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, Illinois
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Jenny Drnevich
- Roy J. Carver Biotechnology Center, High Performance Biological Computing, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Justin S Rhodes
- Department of Psychology and Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Marni D Boppart
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, Illinois
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| |
Collapse
|
44
|
Yin M, Shen J, Yu S, Fei J, Zhu X, Zhao J, Zhai L, Sadhukhan A, Zhou J. Tumor-Associated Macrophages (TAMs): A Critical Activator In Ovarian Cancer Metastasis. Onco Targets Ther 2019; 12:8687-8699. [PMID: 31695427 PMCID: PMC6814357 DOI: 10.2147/ott.s216355] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 09/24/2019] [Indexed: 12/13/2022] Open
Abstract
Tumor-associated macrophages (TAMs) that appear in every stage of cancer progression are usually tumor-promoting cells and are present abundantly in the tumor-associated microenvironment. In ovarian cancer, the overall and intratumoral M1/M2 ratio is a relatively efficient TAM parameter for predicting the prognosis of patients, especially for serous tissue type cancer. TAMs exhibit immunological checkpoint modulators, such as the B7 family and programmed death-ligand 1 (PD-L1), and play a key role in the development, metastasis and invasion of ovarian cancer, but the underlying mechanism is barely understood. Ovarian cancer is a severe gynecological malignancy with high mortality. Ovarian cancer-associated death can primarily be attributed to cancer metastasis. The majority of patients are diagnosed with wide dissemination in the peritoneum and omentum, limiting the effectiveness of surgery and chemotherapy. In addition, unlike other well-documented cancers, metastasis through vasculature is not a usual dissemination pathway in ovarian cancer. This review sheds light on TAMs and the main process and mechanism of ovarian cancer metastasis.
Collapse
Affiliation(s)
- Meichen Yin
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
| | - Jiayu Shen
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
| | - Shuqian Yu
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
| | - Jing Fei
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
| | - Xiaoqing Zhu
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
| | - Jiayao Zhao
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
| | - Lingyun Zhai
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
| | - Annapurna Sadhukhan
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
| | - Jianwei Zhou
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
| |
Collapse
|
45
|
Su W, Han HH, Wang Y, Zhang B, Zhou B, Cheng Y, Rumandla A, Gurrapu S, Chakraborty G, Su J, Yang G, Liang X, Wang G, Rosen N, Scher HI, Ouerfelli O, Giancotti FG. The Polycomb Repressor Complex 1 Drives Double-Negative Prostate Cancer Metastasis by Coordinating Stemness and Immune Suppression. Cancer Cell 2019; 36:139-155.e10. [PMID: 31327655 PMCID: PMC7210785 DOI: 10.1016/j.ccell.2019.06.009] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 04/28/2019] [Accepted: 06/17/2019] [Indexed: 12/14/2022]
Abstract
The mechanisms that enable immune evasion at metastatic sites are poorly understood. We show that the Polycomb Repressor Complex 1 (PRC1) drives colonization of the bones and visceral organs in double-negative prostate cancer (DNPC). In vivo genetic screening identifies CCL2 as the top prometastatic gene induced by PRC1. CCL2 governs self-renewal and induces the recruitment of M2-like tumor-associated macrophages and regulatory T cells, thus coordinating metastasis initiation with immune suppression and neoangiogenesis. A catalytic inhibitor of PRC1 cooperates with immune checkpoint therapy to reverse these processes and suppress metastasis in genetically engineered mouse transplantation models of DNPC. These results reveal that PRC1 coordinates stemness with immune evasion and neoangiogenesis and point to the potential clinical utility of targeting PRC1 in DNPC.
Collapse
MESH Headings
- Adenocarcinoma/drug therapy
- Adenocarcinoma/immunology
- Adenocarcinoma/metabolism
- Adenocarcinoma/secondary
- Animals
- Antineoplastic Agents, Immunological/pharmacology
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Cell Movement/drug effects
- Cell Self Renewal/drug effects
- Chemokine CCL2/genetics
- Chemokine CCL2/metabolism
- Enzyme Inhibitors/pharmacology
- Gene Expression Regulation, Neoplastic
- Humans
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Macrophages/immunology
- Macrophages/metabolism
- Male
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Mice, Knockout
- Mice, Nude
- Mice, SCID
- Neoplasm Metastasis
- Neoplastic Stem Cells/immunology
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- PC-3 Cells
- Polycomb Repressive Complex 1/antagonists & inhibitors
- Polycomb Repressive Complex 1/genetics
- Polycomb Repressive Complex 1/metabolism
- Prostatic Neoplasms/drug therapy
- Prostatic Neoplasms/immunology
- Prostatic Neoplasms/metabolism
- Prostatic Neoplasms/pathology
- Receptors, Androgen/deficiency
- Receptors, Androgen/genetics
- Receptors, CCR4/genetics
- Receptors, CCR4/metabolism
- Signal Transduction
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Tumor Escape/drug effects
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Wenjing Su
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY 10065, USA
| | - Hyun Ho Han
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Unit 1906, PO Box 301429, Houston, TX 77054/77030-1429, USA; Department of Urology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Yan Wang
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Unit 1906, PO Box 301429, Houston, TX 77054/77030-1429, USA
| | - Boyu Zhang
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Unit 1906, PO Box 301429, Houston, TX 77054/77030-1429, USA
| | - Bing Zhou
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yuanming Cheng
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY 10065, USA
| | - Alekya Rumandla
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Unit 1906, PO Box 301429, Houston, TX 77054/77030-1429, USA
| | - Sreeharsha Gurrapu
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Unit 1906, PO Box 301429, Houston, TX 77054/77030-1429, USA
| | - Goutam Chakraborty
- Department of Medicine, Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY 10065, USA
| | - Jie Su
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY 10065, USA
| | - Guangli Yang
- Organic Synthesis Core Facility, Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY 10065, USA
| | - Xin Liang
- Department of Genitourinary Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Guocan Wang
- Department of Genitourinary Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Neal Rosen
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY 10065, USA
| | - Howard I Scher
- Genitourinary Oncology Service, Department of Medicine, MSKCC, Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Ouathek Ouerfelli
- Organic Synthesis Core Facility, Memorial Sloan Kettering Cancer Center (MSKCC), New York, NY 10065, USA
| | - Filippo G Giancotti
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Unit 1906, PO Box 301429, Houston, TX 77054/77030-1429, USA; Department of Genitourinary Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA.
| |
Collapse
|
46
|
Mora J, Mertens C, Meier JK, Fuhrmann DC, Brüne B, Jung M. Strategies to Interfere with Tumor Metabolism through the Interplay of Innate and Adaptive Immunity. Cells 2019; 8:cells8050445. [PMID: 31083487 PMCID: PMC6563030 DOI: 10.3390/cells8050445] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 05/04/2019] [Accepted: 05/07/2019] [Indexed: 01/19/2023] Open
Abstract
The inflammatory tumor microenvironment is an important regulator of carcinogenesis. Tumor-infiltrating immune cells promote each step of tumor development, exerting crucial functions from initiation, early neovascularization, to metastasis. During tumor outgrowth, tumor-associated immune cells, including myeloid cells and lymphocytes, acquire a tumor-supportive, anti-inflammatory phenotype due to their interaction with tumor cells. Microenvironmental cues such as inflammation and hypoxia are mainly responsible for creating a tumor-supportive niche. Moreover, it is becoming apparent that the availability of iron within the tumor not only affects tumor growth and survival, but also the polarization of infiltrating immune cells. The interaction of tumor cells and infiltrating immune cells is multifaceted and complex, finally leading to different activation phenotypes of infiltrating immune cells regarding their functional heterogeneity and plasticity. In recent years, it was discovered that these phenotypes are mainly implicated in defining tumor outcome. Here, we discuss the role of the metabolic activation of both tumor cells and infiltrating immune cells in order to adapt their metabolism during tumor growth. Additionally, we address the role of iron availability and the hypoxic conditioning of the tumor with regard to tumor growth and we describe the relevance of therapeutic strategies to target such metabolic characteristics.
Collapse
Affiliation(s)
- Javier Mora
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany.
| | - Christina Mertens
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany.
| | - Julia K Meier
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany.
| | - Dominik C Fuhrmann
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany.
| | - Bernhard Brüne
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany.
- German Cancer Consortium (DKTK), Partner Site Frankfurt, Germany.
- Project Group Translational Medicine and Pharmacology TMP, Fraunhofer Institute for Molecular Biology and Applied Ecology, 60596 Frankfurt, Germany.
| | - Michaela Jung
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany.
| |
Collapse
|
47
|
Secretomes from metastatic breast cancer cells, enriched for a prognostically unfavorable LCN2 axis, induce anti-inflammatory MSC actions and a tumor-supportive premetastatic lung. Oncotarget 2019; 10:3027-3039. [PMID: 31105883 PMCID: PMC6508963 DOI: 10.18632/oncotarget.26903] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 04/14/2019] [Indexed: 12/27/2022] Open
Abstract
Cancer metastasis is responsible for the clear majority of cancer-related deaths. Survival and expansion of cancer cells at secondary sites requires that these premetastatic microenvironments be primed by primary tumor cells and their secreted factors. Efforts to date have been limited by immune-deficient in vivo models and/or the need for finely-tuned analysis time points that reduce contributions from early-disseminating cancer cells. In this regard, we developed a tumor cell-free syngeneic breast cancer model for characterizing tumor cell secretome-mediated reprogramming of premetastatic tissues. We demonstrate that secretomes from metastatic breast cancer cells differentially regulate the lung and brain, promoting a tumor-supportive lung microenvironment with both elevated CD73 expression and decreased TNFα expression. Using in vitro models of CD73-positive mesenchymal stem cells (MSCs) and macrophages/monocytes, we tested whether MSCs can mediate anti-inflammatory effects of metastatic breast cancer cells. Notably, conditioned media from metastatic Py230 cells reprogrammed the secretomes of MSCs toward an anti-inflammatory state. Mining transcriptome data from Py8119 and Py230 cells revealed a lipocalin 2 (LCN2) axis that is selectively expressed in the metastatic Py230 cells, predicts poor breast cancer patient survival and is elevated in circulating serum of mice chronically treated with conditioned media from Py230 cells. Taken together, these results establish the utility of an immune-competent tumor cell-free model for characterizing the mechanisms of breast cancer cell priming of the premetastatic niche, demonstrate that MSCs can mediate the anti-inflammatory effects of metastatic breast cancer cells and substantiate LCN2 as a promising therapeutic target for blocking breast cancer progression.
Collapse
|
48
|
Cote B, Rao D, Alany RG, Kwon GS, Alani AW. Lymphatic changes in cancer and drug delivery to the lymphatics in solid tumors. Adv Drug Deliv Rev 2019; 144:16-34. [PMID: 31461662 DOI: 10.1016/j.addr.2019.08.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 08/05/2019] [Accepted: 08/23/2019] [Indexed: 02/08/2023]
Abstract
Although many solid tumors use the lymphatic system to metastasize, there are few treatment options that directly target cancer present in the lymphatic system, and those that do are highly invasive, uncomfortable, and/or have limitations. In this review we provide a brief overview of lymphatic function and anatomy, discusses changes that befall the lymphatics in cancer and the mechanisms by which these changes occur, and highlight limitations of lymphatic drug delivery. We then go on to summarize relevant techniques and new research for targeting cancer populations in the lymphatics and enhancing drug delivery intralymphatically, including intralymphatic injections, isolated limb perfusion, passive nano drug delivery systems, and actively targeted nanomedicine.
Collapse
|
49
|
Jung M, Mertens C, Tomat E, Brüne B. Iron as a Central Player and Promising Target in Cancer Progression. Int J Mol Sci 2019; 20:ijms20020273. [PMID: 30641920 PMCID: PMC6359419 DOI: 10.3390/ijms20020273] [Citation(s) in RCA: 167] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 01/08/2019] [Accepted: 01/09/2019] [Indexed: 02/07/2023] Open
Abstract
Iron is an essential element for virtually all organisms. On the one hand, it facilitates cell proliferation and growth. On the other hand, iron may be detrimental due to its redox abilities, thereby contributing to free radical formation, which in turn may provoke oxidative stress and DNA damage. Iron also plays a crucial role in tumor progression and metastasis due to its major function in tumor cell survival and reprogramming of the tumor microenvironment. Therefore, pathways of iron acquisition, export, and storage are often perturbed in cancers, suggesting that targeting iron metabolic pathways might represent opportunities towards innovative approaches in cancer treatment. Recent evidence points to a crucial role of tumor-associated macrophages (TAMs) as a source of iron within the tumor microenvironment, implying that specifically targeting the TAM iron pool might add to the efficacy of tumor therapy. Here, we provide a brief summary of tumor cell iron metabolism and updated molecular mechanisms that regulate cellular and systemic iron homeostasis with regard to the development of cancer. Since iron adds to shaping major hallmarks of cancer, we emphasize innovative therapeutic strategies to address the iron pool of tumor cells or cells of the tumor microenvironment for the treatment of cancer.
Collapse
Affiliation(s)
- Michaela Jung
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany.
| | - Christina Mertens
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany.
| | - Elisa Tomat
- Department of Chemistry and Biochemistry, University of Arizona, 1306 E. University Blvd., Tucson, AZ 85721-0041, USA.
| | - Bernhard Brüne
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany.
- German Cancer Consortium (DKTK), Partner Site Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany.
- Project Group Translational Medicine and Pharmacology TMP, Fraunhofer Institute for Molecular Biology and Applied Ecology, 60596 Frankfurt, Germany.
| |
Collapse
|
50
|
Vela D. Iron Metabolism in Prostate Cancer; From Basic Science to New Therapeutic Strategies. Front Oncol 2018; 8:547. [PMID: 30538952 PMCID: PMC6277552 DOI: 10.3389/fonc.2018.00547] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Accepted: 11/05/2018] [Indexed: 01/09/2023] Open
Abstract
An increasing amount of research has recently strengthened the case for the existence of iron dysmetabolism in prostate cancer. It is characterized with a wide array of differential expression of iron-related proteins compared to normal cells. These proteins control iron entry, cellular iron distribution but also iron exit from prostate cells. Iron dysmetabolism is not an exclusive feature of prostate cancer cells, but it is observed in other cells of the tumor microenvironment. Disrupting the machinery that secures iron for prostate cancer cells can retard tumor growth and its invasive potential. This review unveils the current understanding of the ways that prostate cancer cells secure iron in the tumor milieu and how can we exploit this knowledge for therapeutic purposes.
Collapse
Affiliation(s)
- Driton Vela
- Department of Physiology, University of Prishtina, Prishtina, Kosovo
| |
Collapse
|