51
|
Csf1 Deficiency Dysregulates Glial Responses to Demyelination and Disturbs CNS White Matter Remyelination. Cells 2019; 9:cells9010099. [PMID: 31906095 PMCID: PMC7017166 DOI: 10.3390/cells9010099] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 12/23/2019] [Accepted: 12/28/2019] [Indexed: 12/30/2022] Open
Abstract
Remyelination, a highly efficient central nervous system (CNS) regenerative process, is performed by oligodendrocyte progenitor cells (OPCs), which are recruited to the demyelination sites and differentiate into mature oligodendrocytes to form a new myelin sheath. Microglia, the specialized CNS-resident phagocytes, were shown to support remyelination through secretion of factors stimulating OPC recruitment and differentiation, and their pharmacological depletion impaired remyelination. Macrophage colony-stimulating factor (Csf1) has been implicated in the control of recruitment and polarization of microglia/macrophages in injury-induced CNS inflammation. However, it remains unclear how Csf1 regulates a glial inflammatory response to demyelination as well as axonal survival and new myelin formation. Here, we have investigated the effects of the inherent Csf1 deficiency in a murine model of remyelination. We showed that remyelination was severely impaired in Csf1-/- mutant mice despite the fact that reduction in monocyte/microglia accumulation affects neither the number of OPCs recruited to the demyelinating lesion nor their differentiation. We identified a specific inflammatory gene expression signature and found aberrant astrocyte activation in Csf1-/- mice. We conclude that Csf1-dependent microglia activity is essential for supporting the equilibrium between microglia and astrocyte pro-inflammatory vs. regenerative activation, demyelinated axons integration and, ultimately, reconstruction of damaged white matter.
Collapse
|
52
|
Cheray M, Stratoulias V, Joseph B, Grabert K. The Rules of Engagement: Do Microglia Seal the Fate in the Inverse Relation of Glioma and Alzheimer's Disease? Front Cell Neurosci 2019; 13:522. [PMID: 31824268 PMCID: PMC6879422 DOI: 10.3389/fncel.2019.00522] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 11/07/2019] [Indexed: 12/30/2022] Open
Abstract
Microglia, the immune cells of the brain, play a major role in the maintenance of brain homeostasis and constantly screen the brain environment to detect any infection or damage. Once activated by a stimulus, microglial cells initiate an immune response followed by the resolution of brain inflammation. A failure or deviation in the housekeeping function of these guardian cells can lead to multiple diseases, including brain cancer and neurodegenerative diseases such as Alzheimer's disease (AD). A small number of studies have investigated the causal relation of both diseases, thereby revealing an inverse relationship where cancer patients have a reduced risk to develop AD and vice versa. In this review, we aim to shed light on the role of microglia in the fate to develop specifically glioma as one type of cancer or AD. We will examine the common and/or opposing genetic predisposition as well as associated pathways of these diseases to unravel a possible involvement of microglia in the occurrence of either disease. Lastly, a set of guidelines will be proposed for future research and diagnostics to clarify and improve the knowledge on the role of microglia in the decision toward one pathology or another.
Collapse
Affiliation(s)
- Mathilde Cheray
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Vassilis Stratoulias
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.,Neuroscience Center, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Bertrand Joseph
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Kathleen Grabert
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
53
|
BRCA-1 depletion impairs pro-inflammatory polarization and activation of RAW 264.7 macrophages in a NF-κB-dependent mechanism. Mol Cell Biochem 2019; 462:11-23. [DOI: 10.1007/s11010-019-03605-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 08/08/2019] [Indexed: 12/15/2022]
|
54
|
Zhang H, Zhang L, Tang Y, Wang C, Chen Y, Shu J, Zhang K. Systemic screening identifies GABRD, a subunit gene of GABAA receptor as a prognostic marker in adult IDH wild-type diffuse low-grade glioma. Biomed Pharmacother 2019; 118:109215. [PMID: 31545245 DOI: 10.1016/j.biopha.2019.109215] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 07/04/2019] [Accepted: 07/08/2019] [Indexed: 12/22/2022] Open
Abstract
Currently, no reliable prognostic biomarkers have been identified for adult IDH wild-type (WT) diffuse low grade glioma (LGG). With data from The Cancer Genome Atlas (TCGA)-LGG, we examined the prognostic value of GABAA receptor subunits in adult IDH WT LGG. Using 2016 WHO CNS tumor classification, we re-classified the TCGA-LGG and identified 95 IDH WT patients. Among 16 GABAA receptor subunit genes with RNA-seq data, eight genes showed significantly different expression in IDH WT LGG compared with IDH mutant (MT) cases. Among these genes, only GABRD expression was related to overall survival (OS) status. Preserved GABRD expression was independently associated with longer OS (HR: 0.799, 95%CI: 0.691-0.925, p = 0.003) in IDH WT LGG. GABRD expression showed a moderately negative correlation with tumor infiltration macrophage (TIM) and CSF1 expression. The methylation status of 34 CpG sites across GABRD gene was checked and only cg13916816 showed a moderately negative correlation with GABRD expression. In conclusion, GABRD expression might serve as a potential independent prognostic marker in patients with IDH WT LGG. Meanwhile, its expression was negatively correlated with the extent of TIM, which might help to explain the favorable survival outcome. Cg13916816 might be a critical CpG site influencing GABRD expression in IDH WT LGG.
Collapse
Affiliation(s)
- Hongwei Zhang
- Department of Anesthesiology, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610041, China
| | - Lixia Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610041, China
| | - Yumin Tang
- Department of Anesthesiology, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610041, China
| | - Chaoji Wang
- Department of Anesthesiology, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610041, China
| | - Yiding Chen
- Department of Anesthesiology, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610041, China
| | - Jinjun Shu
- Department of Anesthesiology, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610041, China
| | - Kexian Zhang
- Department of Anesthesiology, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
55
|
Zhong WQ, Li ZZ, Jiang H, Zou YP, Wang HT, Cai Y, Zhao Y, Zhao JH. Elevated ATF4 Expression in Odontogenic Keratocysts Epithelia: Potential Involvement in Tissue Hypoxia and Stromal M2 Macrophage Infiltration. J Histochem Cytochem 2019; 67:801-812. [PMID: 31424999 DOI: 10.1369/0022155419871550] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The aim of this study was to investigate the expression of the activating transcription factor 4 (ATF4) in odontogenic keratocysts (OKC), its association with hypoxia and M2-polarized macrophages infiltration, and its potential relationships with angiogenesis in OKC. The expression of ATF4, hypoxia-inducible factor 1α (HIF-1α), macrophage colony-stimulating factor (M-CSF), and receptor activator of nuclear factor κ-B ligand (RANKL) in OKC samples and normal oral mucosa (OM) was detected by immunohistochemistry. Meanwhile, microvessel density (MVD) was measured using antibody against CD31. M2-polarized macrophages were identified using double-staining for CD68+ and CD163+. The correlations of ATF4 with HIF-1α, M-CSF, and M2-polarized macrophages infiltration were determined by Spearman's rank correlation test and hierarchical clustering. Human immortalized oral epithelial cells (HIOECs) were used in in vitro experiments. Our data showed that the expression of HIF-1α, ATF4, and M-CSF was significantly upregulated in the epithelium of OKC when compared with the OM. The expression of ATF4 was positively correlated with that of HIF-1α, M-CSF, MVD, and M2-polarized macrophages infiltration. Elevated expression of ATF4 in the epithelial lining of OKC may facilitate the M2 macrophages infiltration in response to hypoxia, leading to the development of OKC.
Collapse
Affiliation(s)
- Wen-Qun Zhong
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China.,Department of Oral and Maxillofacial Surgery, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhi-Zheng Li
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Hao Jiang
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yan-Ping Zou
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Hai-Tao Wang
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yu Cai
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China.,Department of Oral and Maxillofacial Surgery, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yi Zhao
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China.,Department of Prosthodontics, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Ji-Hong Zhao
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China.,Department of Oral and Maxillofacial Surgery, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
56
|
Ocoxin Modulates Cancer Stem Cells and M2 Macrophage Polarization in Glioblastoma. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:9719730. [PMID: 31467641 PMCID: PMC6701394 DOI: 10.1155/2019/9719730] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 04/17/2019] [Accepted: 05/05/2019] [Indexed: 12/29/2022]
Abstract
Glioblastoma (GBM) is the most common and devastating primary brain tumor. The presence of cancer stem cells (CSCs) has been linked to their therapy resistance. Molecular and cellular components of the tumor microenvironment also play a fundamental role in the aggressiveness of these tumors. In particular, high levels of hypoxia and reactive oxygen species participate in several aspects of GBM biology. Moreover, GBM contains a large number of macrophages, which normally behave as immunosuppressive tumor-supportive cells. In fact, the presence of both, hypoxia and M2-like macrophages, correlates with malignancy and poor prognosis in gliomas. Antioxidant agents, as nutritional supplements, might have antitumor activity. Ocoxin® oral solution (OOS), in particular, has anti-inflammatory and antioxidant properties, as well as antitumor properties in several neoplasia, without known side effects. Here, we describe how OOS affects stem cell properties in certain GBMs, slowing down their tumor growth. In parallel, OOS has a direct effect on macrophage polarization in vitro and in vivo, inhibiting the protumoral features of M2 macrophages. Therefore, OOS could be a feasible candidate to be used in combination therapies during GBM treatment because it can target the highly resilient CSCs as well as their supportive immune microenvironment, without adding toxicity to conventional treatments.
Collapse
|
57
|
Ding AS, Routkevitch D, Jackson C, Lim M. Targeting Myeloid Cells in Combination Treatments for Glioma and Other Tumors. Front Immunol 2019; 10:1715. [PMID: 31396227 PMCID: PMC6664066 DOI: 10.3389/fimmu.2019.01715] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 07/09/2019] [Indexed: 02/06/2023] Open
Abstract
Myeloid cells constitute a significant part of the immune system in the context of cancer, exhibiting both immunostimulatory effects, through their role as antigen presenting cells, and immunosuppressive effects, through their polarization to myeloid-derived suppressor cells (MDSCs) and tumor-associated macrophages. While they are rarely sufficient to generate potent anti-tumor effects on their own, myeloid cells have the ability to interact with a variety of immune populations to aid in mounting an appropriate anti-tumor immune response. Therefore, myeloid therapies have gained momentum as a potential adjunct to current therapies such as immune checkpoint inhibitors (ICIs), dendritic cell vaccines, oncolytic viruses, and traditional chemoradiation to enhance therapeutic response. In this review, we outline critical pathways involved in the recruitment of the myeloid population to the tumor microenvironment and in their polarization to immunostimulatory or immunosuppressive phenotypes. We also emphasize existing strategies of modulating myeloid recruitment and polarization to improve anti-tumor immune responses. We then summarize current preclinical and clinical studies that highlight treatment outcomes of combining myeloid targeted therapies with other immune-based and traditional therapies. Despite promising results from reports of limited clinical trials thus far, there remain challenges in optimally harnessing the myeloid compartment as an adjunct to enhancing anti-tumor immune responses. Further large Phase II and ultimately Phase III clinical trials are needed to elucidate the treatment benefit of combination therapies in the fight against cancer.
Collapse
Affiliation(s)
| | | | | | - Michael Lim
- Department of Neurosurgery, Johns Hopkins School of Medicine, Baltimore, MD, United States
| |
Collapse
|
58
|
Linsenmann T, Jawork A, Westermaier T, Homola G, Monoranu CM, Vince GH, Kessler AF, Ernestus RI, Löhr M. Tumor growth under rhGM-CSF application in an orthotopic rodent glioma model. Oncol Lett 2019; 17:4843-4850. [PMID: 31186691 PMCID: PMC6507467 DOI: 10.3892/ol.2019.10179] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 02/14/2019] [Indexed: 12/14/2022] Open
Abstract
Regulation of the host immune response serves a pivotal role in the persistence and progression of malignant glioma. To date, cytotoxic cluster of differentiation (CD)-8+ T and natural killer cells are considered the main cellular components of host tumor control. The influence of macrophages in an orthotropic C6 tumor implantation model was investigated and the aim of the present study was to characterize the effects of systemic macrophage-activation on glioma growth by using the granulocyte macrophage colony stimulating factor (rhGM-CSF). A total of 20 male Sprague-Dawley rats were orthotopically implanted with C6 glioma spheroids and treated subcutaneously with 10 µg/kg rhGM-CSF every other day; 9 animals served as controls. Serial magnetic resonance imaging was performed on days 7, 14, 21, 28, 32 and 42 post-implantation to monitor tumor volume. Histological work-up included hematoxylin and eosin, CD68/ED-1 macrophage, CD8 T-cell and Ki-67 MIB1 proliferation staining in gliomas and spleen. Experimental C6-gliomas developed in 15/20 (75%) animals. In rhGM-CSF treated rats, tumors developed significantly later and reached a smaller size (median, 134 mm3) compared with the controls (median, 262 mm3). On day 14, solid tumors presented in 11/17 (65%) rhGM-CSF-treated animals; in control animals tumor growth was detected in 3/9 animals on day 7 and in all animals on day 14. The mean survival time was 35 days in the rhGM-CSF group and significantly longer when compared with the control group (24 days). Immunohistochemistry exhibited significantly more macrophages in tumors, particularly in the perivascular zone of the rhGM-CSF group when compared with untreated animals; intratumoral CD8+ counts were equal in both groups. A systemic stimulation of macrophages by rhGM-CSF resulted in significantly reduced and delayed tumor growth in the rodent C6 glioma model. The present data suggested a significant role of macrophages in host control of experimental gliomas on the innate immune response. Until now, the role of macrophages may have been underestimated in host glioma control.
Collapse
Affiliation(s)
- Thomas Linsenmann
- Department of Neurosurgery, Julius Maximilians University, Wuerzburg, D-97080 Wuerzburg, Germany
| | - Anna Jawork
- Department of Neurosurgery, Julius Maximilians University, Wuerzburg, D-97080 Wuerzburg, Germany
| | - Thomas Westermaier
- Department of Neurosurgery, Julius Maximilians University, Wuerzburg, D-97080 Wuerzburg, Germany
| | - György Homola
- Department of Neuroradiology, Julius Maximilians University, Wuerzburg, D-97080 Wuerzburg, Germany
| | - Camelia Maria Monoranu
- Department of Neuropathology, Julius Maximilians University, Wuerzburg, D-97080 Wuerzburg, Germany
| | - Giles Hamilton Vince
- Department of Neurosurgery, Clinical Centre of Aschaffenburg-Alzenau, D-63739 Aschaffenburg, Germany
| | | | - Ralf-Ingo Ernestus
- Department of Neurosurgery, Julius Maximilians University, Wuerzburg, D-97080 Wuerzburg, Germany
| | - Mario Löhr
- Department of Neurosurgery, Julius Maximilians University, Wuerzburg, D-97080 Wuerzburg, Germany
| |
Collapse
|
59
|
Waters MR, Gupta AS, Mockenhaupt K, Brown LN, Biswas DD, Kordula T. RelB acts as a molecular switch driving chronic inflammation in glioblastoma multiforme. Oncogenesis 2019; 8:37. [PMID: 31142741 PMCID: PMC6541631 DOI: 10.1038/s41389-019-0146-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 05/03/2019] [Accepted: 05/16/2019] [Indexed: 01/31/2023] Open
Abstract
Glioblastoma multiforme (GBM) is a primary brain tumor characterized by extensive necrosis and immunosuppressive inflammation. The mechanisms by which this inflammation develops and persists in GBM remain elusive. We identified two cytokines interleukin-1β (IL-1) and oncostatin M (OSM) that strongly negatively correlate with patient survival. We found that these cytokines activate RelB/p50 complexes by a canonical NF-κB pathway, which surprisingly drives expression of proinflammatory cytokines in GBM cells, but leads to their inhibition in non-transformed astrocytes. We discovered that one allele of the gene encoding deacetylase Sirtuin 1 (SIRT1), needed for repression of cytokine genes, is deleted in 80% of GBM tumors. Furthermore, RelB specifically interacts with a transcription factor Yin Yang 1 (YY1) in GBM cells and activates GBM-specific gene expression programs. As a result, GBM cells continuously secrete proinflammatory cytokines and factors attracting/activating glioma-associated microglia/macrophages and thus, promote a feedforward inflammatory loop.
Collapse
Affiliation(s)
- Michael R Waters
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth, University School of Medicine and the Massey Cancer Center, Richmond, VI, 23298, USA
| | - Angela S Gupta
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth, University School of Medicine and the Massey Cancer Center, Richmond, VI, 23298, USA
| | - Karli Mockenhaupt
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth, University School of Medicine and the Massey Cancer Center, Richmond, VI, 23298, USA
| | - LaShardai N Brown
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth, University School of Medicine and the Massey Cancer Center, Richmond, VI, 23298, USA
| | - Debolina D Biswas
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth, University School of Medicine and the Massey Cancer Center, Richmond, VI, 23298, USA
| | - Tomasz Kordula
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth, University School of Medicine and the Massey Cancer Center, Richmond, VI, 23298, USA.
| |
Collapse
|
60
|
Seledtsov VI, Malashchenko VV, Gazatova ND, Meniailo ME, Morozova EM, Seledtsova GV. Directs effects of granulocyte-macrophage colony stimulating factor (GM-CSF) on adaptive immunogenesis. Hum Vaccin Immunother 2019; 15:2903-2909. [PMID: 31063025 DOI: 10.1080/21645515.2019.1614396] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Background: We studied direct effects of human granulocyte-macrophage colony stimulating factor (GM-CSF) on phenotypical characteristics and cytokine-production of non-activated and activated human monocytes/macrophages (Mc/Mphs) and T cells.Methods: Purified Mc/Mphs were activated by bacterial lipopolysaccharide (LPS, 1 μg/ml) for 24 h, while T cells were activated by particles conjugated and antibodies (Abs) against human CD2, CD3, and CD28 for 48 h.Results: GM-CSF treatment (0.01-10 ng/ml) was shown to reduce percentages of CD197 (CCR7)-positive cells in non-activated Mph cultures, without affecting significantly CD14+ (LPS co-receptor), CD16+ (FcγRIII, low-affinity Fc-receptor), CD119+ (interferon-gamma receptor 1), and CD124+ (IL4 receptor α-subunit) cells. In addition, GM-CSF reduced relative numbers of CD197+ cells, as well as CD14+, CD16+, and CD119+ cells in activated Mph cultures without affecting CD124+ cell distribution. GM-CSF at the highest dose of 10 ng/ml enhanced TNF-α and IL-6 (but not IL-1β and IL-10) production in activated Mc/Mphs. In activated T cell cultures, GM-CSF at 0.1-1.0 ng/ml augmented CD38+ cell numbers in naïve СD45RA+/СD197+ and central memory СD45RA-/СD197+ cell subsets, with no effect on effector СD45RA-/СD197- and terminally differentiated effector СD45RA+/СD197- cells. GM-CSF at a low dose (0.01 ng/ml) down-regulated INF-γ production, while at a high dosage (10.0 ng/ml) up-regulated IL-2 and IL-4 production.Conclusion: In general, the results suggest that GM-CSF is able to facilitate the implication of both Mph and T cells in the adaptive immunogenesis.
Collapse
Affiliation(s)
| | - Vladimir V Malashchenko
- Center for Medical Biotechnologies, Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - Natalja D Gazatova
- Center for Medical Biotechnologies, Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - Maksim E Meniailo
- Center for Medical Biotechnologies, Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - Ekaterina M Morozova
- Center for Medical Biotechnologies, Immanuel Kant Baltic Federal University, Kaliningrad, Russia
| | - Galina V Seledtsova
- Laboratory for Cellular Biotechnologies, Scientific Research Institute for Fundamental and Clinical Immunology, Novosibirsk, Russia
| |
Collapse
|
61
|
Xie C, Lu D, Xu M, Qu Z, Zhang W, Wang H. Knockdown of RAD18 inhibits glioblastoma development. J Cell Physiol 2019; 234:21100-21112. [PMID: 31081138 DOI: 10.1002/jcp.28713] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 04/08/2019] [Accepted: 04/11/2019] [Indexed: 12/20/2022]
Abstract
This study aimed at investigating the role of RAD18 in the regulation of glioblastoma development as well as the underlying mechanisms. The human glioblastoma U251 and U87MG cells were transfected with siRNAs specifically targeting RAD18, and the effects of knockdown of RAD18 on the viability, apoptosis, migration, and invasion of U251 and U87MG cells were investigated. Transcriptome sequencing of the siRNA-RAD18-tranfected and siRNA-NC-transfected U251 cells was performed, followed by bioinformatic analyses for sequencing data. The results showed that knockdown of RAD18 significantly inhibited cell viability, promoted apoptosis, and suppressed migration and invasion of U251 and U87MG cells. Bioinformatic analyses of sequencing data identified 1,051 differentially expressed genes (DEGs) (369 up- and 682 downregulated genes) in the siRNA-RAD18-transfected U251 cells compared with siRNA-NC-transfected U251 cells. Eleven DEGs, including nerve growth factor (NGF), colony-stimulating factor 2 (CSF2), matrix metallopeptidase 1 (MMP1), platelet-derived growth factor receptor α (PDGFRA), and heme oxygenase 1 (HMOX1), were identified as the hub nodes in protein-protein interaction (PPI) network. Moreover, the aforementioned 11 hub genes were significantly enriched in PI3K-Akt signaling pathway and GO functions associated with the extracellular region. Notably, quantitative real-time polymerase chain reaction further confirmed that the expression levels of NGF, CSF2, HMOX1, and MMP1 were significantly downregulated, while that of PDGFRA was markedly upregulated in the siRNA-RAD18-transfected U251 cells than in the siRNA-NC cells. In conclusion, the knockdown of RAD18 may inhibit glioblastoma development by regulating the expression of the aforementioned key DEGs.
Collapse
Affiliation(s)
- Chen Xie
- Department of Neurosurgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Dejuan Lu
- Department of Endoscope, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Meng Xu
- Department of Neurosurgery, First People's Hospital of Heihe, Heihe, Heilongjiang, People's Republic of China
| | - Zhengyi Qu
- Department of Neurology, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Weiguang Zhang
- Department of Neurosurgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Hongwei Wang
- Department of Minimally Invasive Neurosurgery, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| |
Collapse
|
62
|
Almeida J, Costa J, Coelho P, Cea V, Galesio M, Noronha JP, Diniz MS, Prudêncio C, Soares R, Sala C, Fernandes R. Adipocyte proteome and secretome influence inflammatory and hormone pathways in glioma. Metab Brain Dis 2019; 34:141-152. [PMID: 30302719 DOI: 10.1007/s11011-018-0327-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 09/27/2018] [Indexed: 01/09/2023]
Abstract
Gliomas represent the most common primary malignant brain tumors in adults, with an extremely poor prognosis. Among several risk factors, lifestyle was also recently identified as a major risk factor for the development of primary glioma. In the present study, we explore the relationship between obesity and glioma in a cellular model. Thus, we have study the influence of adipocytes secretome on glioma cell line GL261. Using the 3T3-L1 adipocyte cell line, and its conditioned medium (adipokines-enriched medium), we showed that adipocyte-released factors relate with glioma angiogenic, growth, hormones and metabolic behavior by MALDI-TOF-MS and proteomic array analysis. In a first view, STI1, hnRNPs and PGK1 are under expressed on CGl. Similarly, both carbonic anhydrase and aldose reductase are even suppressed in glioma cells that grown under adipokines-enriched environment. Contrariwise, RFC1, KIF5C, ANXA2, N-RAP and RACK1 are overexpressed in GL261 cell the in the presence of the adipokines-enriched medium. We further identified the factors that are released by adipocyte cells, and revealed that several pro-inflammatory and angiogenic factors, such as IL-6, IL-11, LIF, PAI-1, TNF-α, endocan, HGF, VEGF IGF-I, were secreted to the medium into a high extent, whereas TIMP-1 and SerpinE1 were under expressed on CGl. This study discloses an interesting in vitro model for the study of glioma biology under a "obesity" environment, that can be explored for the understanding of cancer cells biology, for the search of biomarkers, prognostic markers and therapeutic approaches.
Collapse
Affiliation(s)
- J Almeida
- School of Health, Polytechnic of Porto, Porto, Portugal
- Department of Functional Biology and Health Sciences, University of Vigo, Vigo, Spain
| | - J Costa
- School of Health, Polytechnic of Porto, Porto, Portugal
| | - P Coelho
- School of Health, Polytechnic of Porto, Porto, Portugal
- Unit of Metabolism, Nutrition and Endocrinology, i3S, University of Porto, Porto, Portugal
| | - V Cea
- CNR Neuroscience Institute Milan, and Department of Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - M Galesio
- REQUIMTE, Department of Chemistry, Faculty of Sciences and Technology, Centre for Fine Chemistry and Biotechnology, NOVA University, Fort Lauderdale, FL, USA
| | - J P Noronha
- REQUIMTE, Department of Chemistry, Faculty of Sciences and Technology, Centre for Fine Chemistry and Biotechnology, NOVA University, Fort Lauderdale, FL, USA
| | - M S Diniz
- REQUIMTE, Department of Chemistry, Faculty of Sciences and Technology, Centre for Fine Chemistry and Biotechnology, NOVA University, Fort Lauderdale, FL, USA
| | - C Prudêncio
- School of Health, Polytechnic of Porto, Porto, Portugal
- Department of Functional Biology and Health Sciences, University of Vigo, Vigo, Spain
| | - R Soares
- Unit of Metabolism, Nutrition and Endocrinology, i3S, University of Porto, Porto, Portugal
- Department of Biomedicine, Unit of Biochemistry, Faculty of Medicine, University of Porto, Porto, Portugal
| | - C Sala
- CNR Neuroscience Institute Milan, and Department of Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Rúben Fernandes
- School of Health, Polytechnic of Porto, Porto, Portugal.
- Unit of Metabolism, Nutrition and Endocrinology, i3S, University of Porto, Porto, Portugal.
| |
Collapse
|
63
|
Pons V, Rivest S. New Therapeutic Avenues of mCSF for Brain Diseases and Injuries. Front Cell Neurosci 2018; 12:499. [PMID: 30618643 PMCID: PMC6306462 DOI: 10.3389/fncel.2018.00499] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 12/03/2018] [Indexed: 01/08/2023] Open
Abstract
Macrophage colony-stimulating factor (mCSF) is a cytokine known to promote the recruitment of macrophages inducing the release of CCL2, a chemokine mobilizing monocytes to sites of inflammation. Additionally, it induces microglia/macrophage proliferation and the polarization of these cells towards a M2-like phenotype, impairing their ability to release pro-inflammatory factors and toxic mediators, while favoring the release of mediators promoting tissue repair. Another important player is the mCSF receptor CSFR1, which is highly expressed in monocytes, macrophages and microglia. Here, we discuss the new interesting therapeutic avenue of the mCSF/CSFR1 axis on brain diseases. More specifically, mCSF cascade might stimulate the survival/proliferation of oligodendrocytes, enhance the immune response as well as modulate the release of growth factors and the phagocytic activity of immune cells to remove myelin debris and toxic proteins from the brain.
Collapse
Affiliation(s)
- Vincent Pons
- Neuroscience Laboratory, Centre Hospitalier Universitaire (CHU) de Québec Research Center and Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec, QC, Canada
| | - Serge Rivest
- Neuroscience Laboratory, Centre Hospitalier Universitaire (CHU) de Québec Research Center and Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec, QC, Canada
| |
Collapse
|
64
|
Sun L, Liang H, Yu W, Jin X. Increased invasive phenotype of CSF-1R expression in glioma cells via the ERK1/2 signaling pathway. Cancer Gene Ther 2018; 26:136-144. [DOI: 10.1038/s41417-018-0053-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 09/29/2018] [Accepted: 10/06/2018] [Indexed: 12/23/2022]
|
65
|
Liu Z, Kuang W, Zhou Q, Zhang Y. TGF-β1 secreted by M2 phenotype macrophages enhances the stemness and migration of glioma cells via the SMAD2/3 signalling pathway. Int J Mol Med 2018; 42:3395-3403. [PMID: 30320350 PMCID: PMC6202079 DOI: 10.3892/ijmm.2018.3923] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 09/28/2018] [Indexed: 12/11/2022] Open
Abstract
The positive correlation between the number of M2 phenotype TAMs (M2-TAMs) and tumour development suggests a supportive role of M2-TAMs in glioma progression. In the present study, the molecular link between glioma cells and M2-TAMs was investigated and it was demonstrated that transforming growth factor-β1 (TGF-β1) secreted by M2-TAMs is key in facilitating the stemness and migration of glioma cells. Cluster of differentiation (CD)133 and CD44, markers for the M2 phenotype, were assessed by western blotting. A sphere formation assay and trans-well assay were applied to test the stemness and migration abilities of glioma cells following co-cultured with M2-TAMs. Stemness markers CD133 and CD44, epithelial-mesenchymal transition-associated markers and mothers against decapentaplegic homolog (SMAD)2/3 and sex determining region Y-box 4/2 (SOX4/2) levels were also evaluated by western blotting. A xenograft tumor mouse model was used to demonstrate the tumor forming ability of glioma cells. The results showed that the U251 glioma cells co-cultured with M2-TAMs exhibited high level of sphere formation, stemness and migration ability. Recombinant TGF-β1 protein treatment was able to achieve the same effects on U251 cells, whereas a TGF-β pathway inhibitor reversed the stemness and migration abilities of the glioma cells induced by M2-TAMs. It was also demonstrated that TGF-β1 secreted by M2-TAMs upregulated the phosphorylation of SMAD2/3 and the expression of SOX4/2 in glioma cells. In a mouse xenograft model, solid tumours formed by U251 cells co-cultured with M2-TAMs or pre-treated with TGF-β1 were larger in size and had a higher growth rate. Taken together, results of the present study demonstrated that M2-TAMs promoted the stemness and migration abilities of glioma cells by secreting TGF-β1, which activated the SMAD2/3 pathway and induced the expression of SOX4 and SOX2. These results highlight the mechanism by which M2-TAMs and glioma interact and demonstrate potential therapeutic strategies for glioma treatment.
Collapse
Affiliation(s)
- Zhengzheng Liu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Weilu Kuang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Qin Zhou
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Yingying Zhang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
66
|
Szepesi Z, Manouchehrian O, Bachiller S, Deierborg T. Bidirectional Microglia-Neuron Communication in Health and Disease. Front Cell Neurosci 2018; 12:323. [PMID: 30319362 PMCID: PMC6170615 DOI: 10.3389/fncel.2018.00323] [Citation(s) in RCA: 306] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 09/06/2018] [Indexed: 12/12/2022] Open
Abstract
Microglia are ramified cells that exhibit highly motile processes, which continuously survey the brain parenchyma and react to any insult to the CNS homeostasis. Although microglia have long been recognized as a crucial player in generating and maintaining inflammatory responses in the CNS, now it has become clear, that their function are much more diverse, particularly in the healthy brain. The innate immune response and phagocytosis represent only a little segment of microglia functional repertoire that also includes maintenance of biochemical homeostasis, neuronal circuit maturation during development and experience-dependent remodeling of neuronal circuits in the adult brain. Being equipped by numerous receptors and cell surface molecules microglia can perform bidirectional interactions with other cell types in the CNS. There is accumulating evidence showing that neurons inform microglia about their status and thus are capable of controlling microglial activation and motility while microglia also modulate neuronal activities. This review addresses the topic: how microglia communicate with other cell types in the brain, including fractalkine signaling, secreted soluble factors and extracellular vesicles. We summarize the current state of knowledge of physiological role and function of microglia during brain development and in the mature brain and further highlight microglial contribution to brain pathologies such as Alzheimer’s and Parkinson’s disease, brain ischemia, traumatic brain injury, brain tumor as well as neuropsychiatric diseases (depression, bipolar disorder, and schizophrenia).
Collapse
Affiliation(s)
- Zsuzsanna Szepesi
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Oscar Manouchehrian
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Sara Bachiller
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Tomas Deierborg
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
67
|
D'Alessandro G, Limatola C, Catalano M. Functional Roles of the Ca2+-activated K+ Channel, KCa3.1, in Brain Tumors. Curr Neuropharmacol 2018; 16:636-643. [PMID: 28707595 PMCID: PMC5997864 DOI: 10.2174/0929867324666170713103621] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 06/22/2017] [Accepted: 07/12/2017] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Glioblastoma is the most aggressive and deadly brain tumor, with low disease-free period even after surgery and combined radio and chemotherapies. Among the factors contributing to the devastating effect of this tumor in the brain are the elevated proliferation and invasion rate, and the ability to induce a local immunosuppressive environment. The intermediateconductance Ca2+-activated K+ channel KCa3.1 is expressed in glioblastoma cells and in tumorinfiltrating cells. METHODS We first describe the researches related to the role of KCa3.1 channels in the invasion of brain tumor cells and the regulation of cell cycle. In the second part we review the involvement of KCa3.1 channel in tumor-associated microglia cell behaviour. RESULTS In tumor cells, the functional expression of KCa3.1 channels is important to substain cell invasion and proliferation. In tumor infiltrating cells, KCa3.1 channel activity is required to regulate their activation state. Interfering with KCa3.1 activity can be an adjuvant therapeutic approach in addition to classic chemotherapy and radiotherapy, to counteract tumor growth and prolong patient's survival. CONCLUSION In this mini-review we discuss the evidence of the functional roles of KCa3.1 channels in glioblastoma biology.
Collapse
Affiliation(s)
- Giuseppina D'Alessandro
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy.,IRCCS Neuromed, Pozzilli, Italy
| | - Cristina Limatola
- IRCCS Neuromed, Pozzilli, Italy.,Department of Physiology and Pharmacology, Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Myriam Catalano
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy.,IRCCS Neuromed, Pozzilli, Italy
| |
Collapse
|
68
|
Morisse MC, Jouannet S, Dominguez-Villar M, Sanson M, Idbaih A. Interactions between tumor-associated macrophages and tumor cells in glioblastoma: unraveling promising targeted therapies. Expert Rev Neurother 2018; 18:729-737. [PMID: 30099909 DOI: 10.1080/14737175.2018.1510321] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Glioblastoma (GBM) is the deadliest primary malignant central nervous system (CNS) tumor with a median overall survival of 15 months despite a very intensive therapeutic regimen including maximal safe surgery, radiotherapy, and chemotherapy. Therefore, GBM treatment still raises major biological and therapeutic challenges. Areas covered: One of the hallmarks of the GBM is its tumor microenvironment including tumor-associated macrophages (TAM). TAM, accounting for approximately 30% of the GBM bulk cell population, may explain, at least in part, the immunosuppressive features of GBMs. The TAM are active and highly plastic immune cells and include two major ontogenetically different cell populations: (i) microglia and, (ii) monocytes-derived macrophages (MDM). TAM recruited to the tumor bulk can be reprogramed by GBM cells resulting in an ineffective anti-tumor response. Interestingly, interactions between TAM and GBM cells promote tumor oncogenesis (i.e. tumor cells proliferation and migration/invasion). This review aims to explore TAM targeting in GBM as a promising therapeutic option in the near future. Expert Commentary: A better understanding of TAM-GBM interactions and dynamics will certainly uncover new anti-GBM therapeutic avenues.
Collapse
Affiliation(s)
- Mony Chenda Morisse
- a Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, AP-HP , Hôpitaux Universitaires Pitié Salpêtrière - Charles Foix , Service de Neurologie 2-Mazarin, Paris , France.,b Department of Medical Oncology , CHU Sud , Amiens , France
| | - Stéphanie Jouannet
- a Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, AP-HP , Hôpitaux Universitaires Pitié Salpêtrière - Charles Foix , Service de Neurologie 2-Mazarin, Paris , France
| | | | - Marc Sanson
- a Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, AP-HP , Hôpitaux Universitaires Pitié Salpêtrière - Charles Foix , Service de Neurologie 2-Mazarin, Paris , France
| | - Ahmed Idbaih
- a Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, AP-HP , Hôpitaux Universitaires Pitié Salpêtrière - Charles Foix , Service de Neurologie 2-Mazarin, Paris , France
| |
Collapse
|
69
|
Matias D, Balça-Silva J, da Graça GC, Wanjiru CM, Macharia LW, Nascimento CP, Roque NR, Coelho-Aguiar JM, Pereira CM, Dos Santos MF, Pessoa LS, Lima FRS, Schanaider A, Ferrer VP, Moura-Neto V. Microglia/Astrocytes-Glioblastoma Crosstalk: Crucial Molecular Mechanisms and Microenvironmental Factors. Front Cell Neurosci 2018; 12:235. [PMID: 30123112 PMCID: PMC6086063 DOI: 10.3389/fncel.2018.00235] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Accepted: 07/16/2018] [Indexed: 12/11/2022] Open
Abstract
In recent years, the functions of glial cells, namely, astrocytes and microglia, have gained prominence in several diseases of the central nervous system, especially in glioblastoma (GB), the most malignant primary brain tumor that leads to poor clinical outcomes. Studies showed that microglial cells or astrocytes play a critical role in promoting GB growth. Based on the recent findings, the complex network of the interaction between microglial/astrocytes cells and GB may constitute a potential therapeutic target to overcome tumor malignancy. In the present review, we summarize the most important mechanisms and functions of the molecular factors involved in the microglia or astrocytes-GB interactions, which is particularly the alterations that occur in the cell's extracellular matrix and the cytoskeleton. We overview the cytokines, chemokines, neurotrophic, morphogenic, metabolic factors, and non-coding RNAs actions crucial to these interactions. We have also discussed the most recent studies regarding the mechanisms of transportation and communication between microglial/astrocytes - GB cells, namely through the ABC transporters or by extracellular vesicles. Lastly, we highlight the therapeutic challenges and improvements regarding the crosstalk between these glial cells and GB.
Collapse
Affiliation(s)
- Diana Matias
- Instituto Estadual do Cérebro Paulo Niemeyer - Secretaria de Estado de Saúde, Rio de Janeiro, Brazil.,Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Joana Balça-Silva
- Instituto Estadual do Cérebro Paulo Niemeyer - Secretaria de Estado de Saúde, Rio de Janeiro, Brazil.,Center for Neuroscience and Cell Biology and Institute for Biomedical Imaging and Life Sciences Consortium, University of Coimbra, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Grazielle C da Graça
- Instituto Estadual do Cérebro Paulo Niemeyer - Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
| | - Caroline M Wanjiru
- Instituto Estadual do Cérebro Paulo Niemeyer - Secretaria de Estado de Saúde, Rio de Janeiro, Brazil.,Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lucy W Macharia
- Instituto Estadual do Cérebro Paulo Niemeyer - Secretaria de Estado de Saúde, Rio de Janeiro, Brazil.,Programa de Pós-Graduação em Anatomia Patológica, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carla Pires Nascimento
- Instituto Estadual do Cérebro Paulo Niemeyer - Secretaria de Estado de Saúde, Rio de Janeiro, Brazil.,Programa de Pós-Graduação em Anatomia Patológica, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Natalia R Roque
- Instituto Estadual do Cérebro Paulo Niemeyer - Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
| | - Juliana M Coelho-Aguiar
- Instituto Estadual do Cérebro Paulo Niemeyer - Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
| | | | - Marcos F Dos Santos
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luciana S Pessoa
- Instituto Estadual do Cérebro Paulo Niemeyer - Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
| | - Flavia R S Lima
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alberto Schanaider
- Centro de Cirurgia Experimental do Departamento de Cirurgia da Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Valéria P Ferrer
- Instituto Estadual do Cérebro Paulo Niemeyer - Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
| | | | - Vivaldo Moura-Neto
- Instituto Estadual do Cérebro Paulo Niemeyer - Secretaria de Estado de Saúde, Rio de Janeiro, Brazil.,Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Universidade do Grande Rio (Unigranrio), Duque de Caxias, Brazil
| |
Collapse
|
70
|
Barberi T, Martin A, Suresh R, Barakat DJ, Harris-Bookman S, Drake CG, Lim M, Friedman AD. Absence of host NF-κB p50 induces murine glioblastoma tumor regression, increases survival, and decreases T-cell induction of tumor-associated macrophage M2 polarization. Cancer Immunol Immunother 2018; 67:1491-1503. [PMID: 30030559 DOI: 10.1007/s00262-018-2184-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 06/07/2018] [Indexed: 12/31/2022]
Abstract
High-grade gliomas harbor abundant myeloid cells that suppress anti-tumor immunity and support tumor growth. Targeting transcription factors, such as NF-κB p50, that mediate suppressive myeloid M2 polarization may prove therapeutic. GL261-Luc glioblastoma cells were inoculated into wild-type and p50-/- mice, followed by analysis of tumor growth, survival, tumor myeloid cells, and T cells. The absence of host p50 slows tumor growth and enables regression in 30% of recipients, leading to prolonged survival. Tumors developing in p50-/- mice possess a greater concentration of tumor-infiltrating myeloid cells (TIMs) than those in wild-type mice. TIMs are predominantly F4/80hi macrophages which, along with tumor-associated microglia, express increased pro-inflammatory M1 and reduced immune-suppressive M2 markers. In p50-/- mice, total tumor CD4 T cells are threefold more abundant, whereas CD8 T-cell numbers are unchanged, and both produce increased IFNγ and Granzyme B. Naïve splenic p50-/- CD8 T cells manifest increased activation, whereas naïve p50-/- and WT CD4 T cells show similar Th1, Th2, and Th17 polarization. Antibody targeting CD4, but not CD8, fully obviates the p50-/- survival advantage. Combined CD4 and CD8 T-cell depletion reverses myeloid M2 polarization in wild-type hosts, without affecting myeloid M1 polarization in p50-/- hosts. Finally, gliomas grow similarly in p50(f/f) and p50(f/f);Lysozyme-Cre mice, the latter having reduced p50 specifically in myeloid cells and tumor microglia. Thus, high-grade glioma T cells play a key role in directing M2 polarization of tumor myeloid cells, and reducing NF-κB p50 in both tumor myeloid cells and T cells may contribute to glioma therapy.
Collapse
Affiliation(s)
- Theresa Barberi
- Division of Pediatric Oncology, Department of Oncology, Johns Hopkins University, CRB I, Rm 253, 1650 Orleans St., Baltimore, MD, 21231, USA
| | - Allison Martin
- Division of Pediatric Oncology, Department of Oncology, Johns Hopkins University, CRB I, Rm 253, 1650 Orleans St., Baltimore, MD, 21231, USA
| | - Rahul Suresh
- Division of Pediatric Oncology, Department of Oncology, Johns Hopkins University, CRB I, Rm 253, 1650 Orleans St., Baltimore, MD, 21231, USA
| | - David J Barakat
- Division of Pediatric Oncology, Department of Oncology, Johns Hopkins University, CRB I, Rm 253, 1650 Orleans St., Baltimore, MD, 21231, USA
| | - Sarah Harris-Bookman
- Division of Pediatric Oncology, Department of Oncology, Johns Hopkins University, CRB I, Rm 253, 1650 Orleans St., Baltimore, MD, 21231, USA
| | - Charles G Drake
- Division of Pediatric Oncology, Department of Oncology, Johns Hopkins University, CRB I, Rm 253, 1650 Orleans St., Baltimore, MD, 21231, USA
| | - Michael Lim
- Division of Pediatric Oncology, Department of Oncology, Johns Hopkins University, CRB I, Rm 253, 1650 Orleans St., Baltimore, MD, 21231, USA
| | - Alan D Friedman
- Division of Pediatric Oncology, Department of Oncology, Johns Hopkins University, CRB I, Rm 253, 1650 Orleans St., Baltimore, MD, 21231, USA.
| |
Collapse
|
71
|
Zhang Y, Feng J, Fu H, Liu C, Yu Z, Sun Y, She X, Li P, Zhao C, Liu Y, Liu T, Liu Q, Liu Q, Li G, Wu M. Coagulation Factor X Regulated by CASC2c Recruited Macrophages and Induced M2 Polarization in Glioblastoma Multiforme. Front Immunol 2018; 9:1557. [PMID: 30034397 PMCID: PMC6043648 DOI: 10.3389/fimmu.2018.01557] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Accepted: 06/25/2018] [Indexed: 12/18/2022] Open
Abstract
Tumor-associated macrophages (TAMs) constitute a major component of inflammatory cells in the glioblastoma multiforme (GBM) tumor microenvironment. TAMs have been implicated in GBM angiogenesis, invasion, local tumor recurrence, and immunosuppression. Coagulation factor X (FX) is a vitamin K-dependent plasma protein that plays a role in the regulation of blood coagulation. In this study, we first found that FX was highly expressed and positively correlated with TAM density in human GBM. FX exhibited a potent chemotactic capacity to recruit macrophages and promoted macrophages toward M2 subtype polarization, accelerating GBM growth. FX bound to extracellular signal-related kinase (ERK)1/2 and inhibited p-ERK1/2 in GBM cells. FX was secreted in the tumor microenvironment and increased the phosphorylation and activation of ERK1/2 and AKT in macrophages, which may have been responsible for the M2 subtype macrophage polarization. Moreover, although the lncRNA CASC2c has been verified to function as a miR-101 competing endogenous RNA (ceRNA) to promote miR-101 target genes in GBM cells, we first confirmed that CASC2c did not function as a miR-338-3p ceRNA to promote FX expression, and that FX was a target gene of miR-338-3p. CASC2c interacted with and reciprocally repressed miR-338-3p. Both CASC2c and miR-388-3p bound to FX and commonly inhibited its expression and secretion. CASC2c repressed M2 subtype macrophage polarization. Taken together, our findings revealed a novel mechanism highlighting CASC2c and FX as potential therapeutic targets to improve GBM patients by altering the GBM microenvironment.
Collapse
Affiliation(s)
- Yan Zhang
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| | - Jianbo Feng
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| | - Haijuan Fu
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| | - Changhong Liu
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| | - Zhibin Yu
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| | - Yingnan Sun
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, China
| | - Xiaoling She
- The Second Xiangya Hospital, Central South University, Changsha, China
| | - Peiyao Li
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| | - Chunhua Zhao
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| | - Yang Liu
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| | - Tao Liu
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| | - Qiang Liu
- The Third Xiangya Hospital, Central South University, Changsha, China
| | - Qing Liu
- The Xiangya Hospital, Central South University, Changsha, China
| | - Guiyuan Li
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| | - Minghua Wu
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| |
Collapse
|
72
|
Walentynowicz KA, Ochocka N, Pasierbinska M, Wojnicki K, Stepniak K, Mieczkowski J, Ciechomska IA, Kaminska B. In Search for Reliable Markers of Glioma-Induced Polarization of Microglia. Front Immunol 2018; 9:1329. [PMID: 29963047 PMCID: PMC6013650 DOI: 10.3389/fimmu.2018.01329] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 05/28/2018] [Indexed: 12/24/2022] Open
Abstract
Immune cells accumulating in the microenvironment of malignant tumors are tumor educated and contribute to its growth, progression, and evasion of antitumor immune responses. Glioblastoma (GBM), the common and most malignant primary brain tumor in adults, shows considerable accumulation of resident microglia and peripheral macrophages, and their polarization into tumor-supporting cells. There are controversies regarding a functional phenotype of glioma-associated microglia/macrophages (GAMs) due to a lack of consistent markers. Previous categorization of GAM polarization toward the M2 phenotype has been found inaccurate because of oversimplification of highly complex and heterogeneous responses. In this study, we characterized functional responses and gene expression in mouse and human microglial cultures exposed to fresh conditioned media [glioma-conditioned medium (GCM)] from human U87 and LN18 glioma cells. Functional analyses revealed mutual communication reflected by strong stimulation of glioma invasion by microglial cells and increased microglial phagocytosis after GCM treatment. To define transcriptomic markers of GCM-activated microglia, we performed selected and global gene expression analyses of stimulated microglial cells. We found activated pathways associated with immune evasion and TGF signaling. We performed computational comparison of the expression patterns of GAMs from human GBMs and rodent experimental gliomas to select genes consistently changed in different datasets. The analyses of marker genes in GAMs from different experimental models and clinical samples revealed only a small set of common genes, which reflects variegated responses in clinical and experimental settings. Tgm2 and Gpnmb were the only two genes common in the analyzed data sets. We discuss potential sources of the observed differences and stress a great need for definitive elucidation of a functional state of GAMs.
Collapse
Affiliation(s)
- Kacper A Walentynowicz
- Laboratory of Molecular Neurobiology, Neurobiology Center, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Natalia Ochocka
- Laboratory of Molecular Neurobiology, Neurobiology Center, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Maria Pasierbinska
- Laboratory of Molecular Neurobiology, Neurobiology Center, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland.,Glia Sp. z o.o, Warsaw, Poland
| | - Kamil Wojnicki
- Laboratory of Molecular Neurobiology, Neurobiology Center, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Karolina Stepniak
- Laboratory of Molecular Neurobiology, Neurobiology Center, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Jakub Mieczkowski
- Laboratory of Molecular Neurobiology, Neurobiology Center, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Iwona A Ciechomska
- Laboratory of Molecular Neurobiology, Neurobiology Center, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Bozena Kaminska
- Laboratory of Molecular Neurobiology, Neurobiology Center, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
73
|
Zeng J, Liu Z, Sun S, Xie J, Cao L, Lv P, Nie S, Zhang B, Xie B, Peng S, Jiang B. Tumor-associated macrophages recruited by periostin in intrahepatic cholangiocarcinoma stem cells. Oncol Lett 2018; 15:8681-8686. [PMID: 29805605 DOI: 10.3892/ol.2018.8372] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 11/03/2017] [Indexed: 12/26/2022] Open
Abstract
Periostin (POSTN) secreted by intrahepatic cholangiocarcinoma stem cells (ICSCs) serves important roles in promoting tumor progression. The present study aimed to investigate POSTN-recruited tumor-associated macrophages (TAMs) in intrahepatic cholangiocarcinoma (ICC). A total of 50 cases were used to investigate the distribution of ICSCs and TAMs in ICC. HCCC-9810 cells were sorted by cluster of differentiation (CD)44, the expression of POSTN of CD44+ (cancer stem cells) and CD44- cells (non-cancer stem cells), and medium were evaluated by western blot analysis. HCCC-9810 cells and THP-1 macrophages were used to detect the effects of POSTN on recruiting TAMs in vitro. The present study revealed that CD44+ cells in ICC tissues and the HCCC-9810 cell line were associated with high POSTN secretion levels. Furthermore, POSTN was associated with TAM density in primary ICC tissues. Additionally, POSTN increased the migration of TAMs derived from THP-1 cells. These findings suggested that POSTN secreted by ICSCs may serve important functions in TAM recruitment, and it may be a potential curative strategy to target the tumor microenvironment in ICC.
Collapse
Affiliation(s)
- Jiehong Zeng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Hunan Normal University-Hunan Provincial People's Hospital, Changsha, Hunan 410006, P.R. China.,Department of General Surgery, Yiyang Central Hospital, Yiyang, Hunan 413000, P.R. China
| | - Zhengkai Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Hunan Normal University-Hunan Provincial People's Hospital, Changsha, Hunan 410006, P.R. China
| | - Shuwen Sun
- Department of Pharmacy, Yiyang Central Hospital, Yiyang, Hunan 413000, P.R. China
| | - Jianhong Xie
- Department of General Surgery, Yiyang Central Hospital, Yiyang, Hunan 413000, P.R. China
| | - Li Cao
- Department of General Surgery, Yiyang Central Hospital, Yiyang, Hunan 413000, P.R. China
| | - Pin Lv
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Hunan Normal University-Hunan Provincial People's Hospital, Changsha, Hunan 410006, P.R. China
| | - Shengdan Nie
- Intistute of Clinical Medical Research, The First Affiliated Hospital of Hunan Normal University-Hunan Provincial People's Hospital, Changsha, Hunan 410006, P.R. China
| | - Bao Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Hunan Normal University-Hunan Provincial People's Hospital, Changsha, Hunan 410006, P.R. China
| | - Bowen Xie
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Hunan Normal University-Hunan Provincial People's Hospital, Changsha, Hunan 410006, P.R. China
| | - Siyuan Peng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Hunan Normal University-Hunan Provincial People's Hospital, Changsha, Hunan 410006, P.R. China
| | - Bo Jiang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Hunan Normal University-Hunan Provincial People's Hospital, Changsha, Hunan 410006, P.R. China
| |
Collapse
|
74
|
Hohensee I, Chuang HN, Grottke A, Werner S, Schulte A, Horn S, Lamszus K, Bartkowiak K, Witzel I, Westphal M, Matschke J, Glatzel M, Jücker M, Pukrop T, Pantel K, Wikman H. PTEN mediates the cross talk between breast and glial cells in brain metastases leading to rapid disease progression. Oncotarget 2018; 8:6155-6168. [PMID: 28008153 PMCID: PMC5351620 DOI: 10.18632/oncotarget.14047] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 12/13/2016] [Indexed: 12/31/2022] Open
Abstract
Despite improvement of therapeutic treatments for breast cancer, the development of brain metastases has become a major limitation to life expectancy for many patients. Brain metastases show very commonly alterations in EGFR and HER2 driven pathways, of which PTEN is an important regulator. Here, we analyzed PTEN expression in 111 tissue samples of breast cancer brain metastases (BCBM). Loss of PTEN was found in a substantial proportion of BCBM samples (48.6%) and was significantly associated with triple-negative breast cancer (67.5%, p = 0.001) and a shorter survival time after surgical resection of brain metastases (p = 0.048). Overexpression of PTEN in brain-seeking MDA-MB-231 BR cells in vitro reduced activation of the AKT pathway, notably by suppression of Akt1 kinase activity. Furthermore, the migration of MDA-MB-231 BR cells in vitro was promoted by co-culturing with both astrocytes and microglial cells. Interestingly, when PTEN was overexpressed the migration was significantly inhibited. Moreover, in an ex vivo organotypic brain slice model, PTEN overexpression reduced invasion of tumor cells. This was accompanied by reduced astrocyte activation that was mediated by autocrine and paracrine activation of GM-CSF/ CSF2RA and AKT/ PTEN pathways. In conclusion, loss of PTEN is frequently detected in triple-negative BCBM patients and associated with poor prognosis. The findings of our functional studies suggest that PTEN loss promotes a feedback loop between tumor cells and glial cells, which might contribute to disease progression.
Collapse
Affiliation(s)
- Ina Hohensee
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Han-Ning Chuang
- Department of Hematology/Oncology, University Medical Center Göttingen, 37099 Göttingen, Germany
| | - Astrid Grottke
- Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Stefan Werner
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Alexander Schulte
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Stefan Horn
- Bone Marrow Transplantation Unit, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Katrin Lamszus
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Kai Bartkowiak
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Isabell Witzel
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Manfred Westphal
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Jakob Matschke
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Markus Glatzel
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Manfred Jücker
- Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Tobias Pukrop
- Department of Hematology/Oncology, University Medical Center Göttingen, 37099 Göttingen, Germany.,Department of Medicine III, University Medical Center Regensburg, 93053 Regensburg, Germany
| | - Klaus Pantel
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| | - Harriet Wikman
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, 20246, Germany
| |
Collapse
|
75
|
Qi L, Yu H, Zhang Y, Zhao D, Lv P, Zhong Y, Xu Y. IL-10 secreted by M2 macrophage promoted tumorigenesis through interaction with JAK2 in glioma. Oncotarget 2018; 7:71673-71685. [PMID: 27765933 PMCID: PMC5342110 DOI: 10.18632/oncotarget.12317] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 09/22/2016] [Indexed: 12/21/2022] Open
Abstract
M2 tumor-associated macrophage has been found to play a supportive role in the progression of glioma. The underlying mechanism, nevertheless, has been largely unknown. In our study, to investigate how M2 macrophage played role in glioma, firstly we've analyzed the clinicopathological significance of M2 macrophage existence on clinical tissues of glioma using detection of CD163 expression with immunohistochemistry. Then, we've artificially induced M2 macrophage from human monocyte cell line THP-1, followed by co-culture with glioma cell lines in vitro. It was found that M2 macrophage was shown to be markedly distributed in glioma relative to paired normal control; and high prevalence of M2 macrophage was significantly associated with poorer overall survival and tumor progression. Moreover, M2 macrophage was found to be able to promote the growth in vitro and tumorigenesis in vivo in xenografted mice model. Mechanistically, it is IL-10 from M2 macrophage that was shown to promote proliferation, dependent on activation of JAK2/STAT3 pathway. Further, IL-10 was found to be able to interact with JAK2 in glioma cells. Taking together, we for the first time found that IL-10 from M2 macrophage promoted proliferation of glioma through interaction with JAK2; thereby activating the JAK2/STAT3 pathway, indicative of IL-10 could be used as a therapeutic target in the curing of glioma.
Collapse
Affiliation(s)
- Ling Qi
- The Department of Pathology, Jilin Medical University, Jilin 132013, PR China
| | - Hongquan Yu
- The Department of Neurosurgery, First Affiliated Hospital of Jilin University, Jilin 130021, PR China
| | - Yu Zhang
- The Department of Neurosurgery, First Affiliated Hospital of Jilin University, Jilin 130021, PR China
| | - Donghai Zhao
- The Department of Pathology, Jilin Medical University, Jilin 132013, PR China
| | - Peng Lv
- The Department of Pathology, Jilin Medical University, Jilin 132013, PR China
| | - Yue Zhong
- The Department of Science and Technology, Jilin Medical University, Jilin 132013, PR China
| | - Ye Xu
- The Medical Research Laboratory, Jilin Medical University, Jilin 132013, PR China
| |
Collapse
|
76
|
Wang Q, He Z, Huang M, Liu T, Wang Y, Xu H, Duan H, Ma P, Zhang L, Zamvil SS, Hidalgo J, Zhang Z, O'Rourke DM, Dahmane N, Brem S, Mou Y, Gong Y, Fan Y. Vascular niche IL-6 induces alternative macrophage activation in glioblastoma through HIF-2α. Nat Commun 2018; 9:559. [PMID: 29422647 PMCID: PMC5805734 DOI: 10.1038/s41467-018-03050-0] [Citation(s) in RCA: 180] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 01/16/2018] [Indexed: 12/25/2022] Open
Abstract
Spatiotemporal regulation of tumor immunity remains largely unexplored. Here we identify a vascular niche that controls alternative macrophage activation in glioblastoma (GBM). We show that tumor-promoting macrophages are spatially proximate to GBM-associated endothelial cells (ECs), permissive for angiocrine-induced macrophage polarization. We identify ECs as one of the major sources for interleukin-6 (IL-6) expression in GBM microenvironment. Furthermore, we reveal that colony-stimulating factor-1 and angiocrine IL-6 induce robust arginase-1 expression and macrophage alternative activation, mediated through peroxisome proliferator-activated receptor-γ-dependent transcriptional activation of hypoxia-inducible factor-2α. Finally, utilizing a genetic murine GBM model, we show that EC-specific knockout of IL-6 inhibits macrophage alternative activation and improves survival in the GBM-bearing mice. These findings illustrate a vascular niche-dependent mechanism for alternative macrophage activation and cancer progression, and suggest that targeting endothelial IL-6 may offer a selective and efficient therapeutic strategy for GBM, and possibly other solid malignant tumors.
Collapse
Affiliation(s)
- Qirui Wang
- Department of Radiation Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- School of Traditional Chinese Medicine, Southern Medical University, 510515, Guangzhou, China
| | - Zhenqiang He
- Department of Radiation Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Neurosurgery, Sun Yat-sen University Cancer Center, 510060, Guangzhou, China
| | - Menggui Huang
- Department of Radiation Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Tianrun Liu
- Department of Radiation Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Otorhinolaryngology, Division of Head and Neck Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, 510655, Guangzhou, China
| | - Yanling Wang
- Department of Radiation Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Haineng Xu
- Department of Radiation Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Hao Duan
- Department of Radiation Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Department of Neurosurgery, Sun Yat-sen University Cancer Center, 510060, Guangzhou, China
| | - Peihong Ma
- Department of Radiation Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Lin Zhang
- Department of Obstetrics and Gynecology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Scott S Zamvil
- Department of Neurology and Program in Immunology, University of California at San Francisco, San Francisco, CA, 94158, USA
| | - Juan Hidalgo
- Department of Cellular Biology, Physiology, and Immunology, Autonomous University of Barcelona, 08193, Barcelona, Spain
| | - Zhenfeng Zhang
- Department of Radiology The, Second Affiliated Hospital of Guangzhou Medical University, 510260, Guangzhou, China
| | - Donald M O'Rourke
- Department of Neurosurgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Nadia Dahmane
- Department of Neurosurgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Steven Brem
- Department of Neurosurgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Yonggao Mou
- Department of Neurosurgery, Sun Yat-sen University Cancer Center, 510060, Guangzhou, China
| | - Yanqing Gong
- Department of Medicine, Division of Human Genetics and Translational Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Yi Fan
- Department of Radiation Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.
- Department of Neurosurgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.
| |
Collapse
|
77
|
Roesch S, Rapp C, Dettling S, Herold-Mende C. When Immune Cells Turn Bad-Tumor-Associated Microglia/Macrophages in Glioma. Int J Mol Sci 2018; 19:ijms19020436. [PMID: 29389898 PMCID: PMC5855658 DOI: 10.3390/ijms19020436] [Citation(s) in RCA: 216] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 12/29/2017] [Accepted: 01/29/2018] [Indexed: 12/31/2022] Open
Abstract
As a substantial part of the brain tumor microenvironment (TME), glioma-associated microglia/macrophages (GAMs) have an emerging role in tumor progression and in controlling anti-tumor immune responses. We review challenges and improvements of cell models and highlight the contribution of this highly plastic cell population to an immunosuppressive TME, besides their well-known functional role regarding glioma cell invasion and angiogenesis. Finally, we summarize first therapeutic interventions to target GAMs and their effect on the immunobiology of gliomas, focusing on their interaction with T cells.
Collapse
Affiliation(s)
- Saskia Roesch
- Division of Experimental Neurosurgery, Department of Neurosurgery, University Hospital Heidelberg, INF400, 69120 Heidelberg, Germany.
| | - Carmen Rapp
- Division of Experimental Neurosurgery, Department of Neurosurgery, University Hospital Heidelberg, INF400, 69120 Heidelberg, Germany.
| | - Steffen Dettling
- Division of Experimental Neurosurgery, Department of Neurosurgery, University Hospital Heidelberg, INF400, 69120 Heidelberg, Germany.
| | - Christel Herold-Mende
- Division of Experimental Neurosurgery, Department of Neurosurgery, University Hospital Heidelberg, INF400, 69120 Heidelberg, Germany.
| |
Collapse
|
78
|
McFarland BC, Marks MP, Rowse AL, Fehling SC, Gerigk M, Qin H, Benveniste EN. Loss of SOCS3 in myeloid cells prolongs survival in a syngeneic model of glioma. Oncotarget 2018; 7:20621-35. [PMID: 26967393 PMCID: PMC4991480 DOI: 10.18632/oncotarget.7992] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 02/16/2016] [Indexed: 12/22/2022] Open
Abstract
In glioma, microglia and macrophages are the largest population of tumor-infiltrating cells, referred to as glioma associated macrophages (GAMs). Herein, we sought to determine the role of Suppressor of Cytokine Signaling 3 (SOCS3), a negative regulator of Signal Transducer and Activator of Transcription 3 (STAT3), in GAM functionality in glioma. We utilized a conditional model in which SOCS3 deletion is restricted to the myeloid cell population. We found that SOCS3-deficient bone marrow-derived macrophages display enhanced and prolonged expression of pro-inflammatory M1 cytokines when exposed to glioma tumor cell conditioned medium in vitro. Moreover, we found that deletion of SOCS3 in the myeloid cell population delays intracranial tumor growth and increases survival of mice bearing orthotopic glioma tumors in vivo. Although intracranial tumors from mice with SOCS3-deficient myeloid cells appear histologically similar to control mice, we observed that loss of SOCS3 in myeloid cells results in decreased M2 polarized macrophage infiltration in the tumors. Furthermore, loss of SOCS3 in myeloid cells results in increased CD8+ T-cell and decreased regulatory T-cell infiltration in the tumors. These findings demonstrate a beneficial effect of M1 polarized macrophages on suppressing glioma tumor growth, and highlight the importance of immune cells in the tumor microenvironment.
Collapse
Affiliation(s)
- Braden C McFarland
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Margaret P Marks
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Amber L Rowse
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Samuel C Fehling
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Magda Gerigk
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hongwei Qin
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Etty N Benveniste
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
79
|
Liu H, Lin C, Shen Z, Zhang H, He H, Li H, Qin J, Qin X, Xu J, Sun Y. Decreased expression of granulocyte-macrophage colony-stimulating factor is associated with adverse clinical outcome in patients with gastric cancer undergoing gastrectomy. Oncol Lett 2017; 14:4701-4707. [PMID: 28943964 PMCID: PMC5594243 DOI: 10.3892/ol.2017.6738] [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: 12/22/2015] [Accepted: 05/26/2017] [Indexed: 11/05/2022] Open
Abstract
Previous studies have revealed the clinical significance of tumor-associated macrophages (TAMs) in gastric cancer, whereas the role of the cytokines that orchestrate TAM polarization in gastric cancer remains elusive. The present study aimed to evaluate the prognostic value of granulocyte-macrophage colony-stimulating factor (GM-CSF) expression in patients with gastric cancer. Intratumoral GM-CSF expression was investigated by immunohistochemical staining in 408 retrospectively enrolled patients. Kaplan-Meier analysis and Cox regression models were used to evaluate the prognostic value of GM-CSF expression. Predictive nomograms were generated to predict the overall survival and disease-free survival rates of the patients. Decreased intratumoral GM-CSF expression was identified, and indicated a poorer clinical outcome for patients with gastric cancer, particularly in advanced stages. Intratumoral GM-CSF expression may provide an additional risk stratification for the prognosis of patients with gastric cancer based on the Tumor-Node-Metastasis (TNM) staging system. Cox multivariate analysis identified GM-CSF expression as an independent prognostic factor for overall survival and disease-free survival time. The generated nomograms performed well in predicting the 3-and 5-year clinical outcome of patients with gastric cancer. In conclusion, GM-CSF is a potential independent prognostic indicator for patients with gastric cancer, which may be integrated with TNM staging systems to improve the predictive accuracy for clinical outcome, particularly in advanced tumors.
Collapse
Affiliation(s)
- Hao Liu
- Department of General Surgery, Zhongshan Hospital, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P.R. China
| | - Chao Lin
- Department of General Surgery, Zhongshan Hospital, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P.R. China
| | - Zhenbin Shen
- Department of General Surgery, Zhongshan Hospital, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P.R. China
| | - Heng Zhang
- Department of General Surgery, Zhongshan Hospital, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P.R. China
| | - Hongyong He
- Department of General Surgery, Zhongshan Hospital, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P.R. China
| | - He Li
- Department of General Surgery, Zhongshan Hospital, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P.R. China
| | - Jing Qin
- Department of General Surgery, Zhongshan Hospital, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P.R. China
| | - Xinyu Qin
- Department of General Surgery, Zhongshan Hospital, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P.R. China
| | - Jiejie Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P.R. China
| | - Yihong Sun
- Department of General Surgery, Zhongshan Hospital, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P.R. China
| |
Collapse
|
80
|
Komohara Y, Kawauchi R, Makiyama E, Mikami K, Horlad H, Fujiwara Y, Kida T, Takeya M, Niidome T. Selective depletion of cultured macrophages by magnetite nanoparticles modified with gelatin. Exp Ther Med 2017; 14:1640-1646. [PMID: 28810630 DOI: 10.3892/etm.2017.4640] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 05/05/2017] [Indexed: 12/16/2022] Open
Abstract
Previous studies have indicated pro-tumor functions of macrophages in tumor progression in different types of malignant tumors. The detailed mechanisms of cell-cell interaction between macrophages and tumor cells have been investigated by means of in vitro co-culture experiments. The present study developed magnetite nanoparticles modified with gelatin that are specifically engulfed by macrophages and investigated methods to deplete these macrophages in co-culture experiments using a magnet. T98G glioma cell line and human monocyte-derived macrophages were mixed and co-cultured for 2 days. The T98G cells were isolated by depletion of the macrophages using the magnetite nanoparticles. mRNA expression of a number of pro-tumor molecules in the isolated T98G cells, with or without co-culture with macrophages, was then evaluated. The mRNA expression levels of chemokine (CC motif) ligand 2, interleukin-6 and macrophage-colony stimulating factor receptor (M-CSFR) were significantly upregulated in T98G cells by co-culture with macrophages (P<0.01). M-CSFR protein expression was also increased by co-culture with macrophages. The conditioned medium of co-cultured cells increased M-CSFR expression in T98G cells. Magnetite nanoparticles may be a novel tool not only for investigating the unique activation status of tumor cells in co-culture conditions, but also for targeting pro-tumor macrophages in tumor tissues.
Collapse
Affiliation(s)
- Yoshihiro Komohara
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Ryuta Kawauchi
- Department of Applied Chemistry and Biochemistry, Graduate School of Science and Technology, Kumamoto University, Kumamoto 860-8555, Japan
| | - Erika Makiyama
- Department of Applied Chemistry and Biochemistry, Graduate School of Science and Technology, Kumamoto University, Kumamoto 860-8555, Japan
| | - Kazuki Mikami
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Hasita Horlad
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Yukio Fujiwara
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Tetsuya Kida
- Magnesium Research Center, Kumamoto University, Kumamoto 860-8555, Japan
| | - Motohiro Takeya
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Takuro Niidome
- Department of Applied Chemistry and Biochemistry, Graduate School of Science and Technology, Kumamoto University, Kumamoto 860-8555, Japan
| |
Collapse
|
81
|
Poon CC, Sarkar S, Yong VW, Kelly JJP. Glioblastoma-associated microglia and macrophages: targets for therapies to improve prognosis. Brain 2017; 140:1548-1560. [PMID: 28334886 DOI: 10.1093/brain/aww355] [Citation(s) in RCA: 152] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 11/20/2016] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma is the most common and most malignant primary adult human brain tumour. Diagnosis of glioblastoma carries a dismal prognosis. Treatment resistance and tumour recurrence are the result of both cancer cell proliferation and their interaction with the tumour microenvironment. A large proportion of the tumour microenvironment consists of an inflammatory infiltrate predominated by microglia and macrophages, which are thought to be subverted by glioblastoma cells for tumour growth. Thus, glioblastoma-associated microglia and macrophages are logical therapeutic targets. Their emerging roles in glioblastoma progression are reflected in the burgeoning research into therapeutics directed at their modification or elimination. Here, we review the biology of glioblastoma-associated microglia and macrophages, and model systems used to study these cells in vitro and in vivo. We discuss translation of results using these model systems and review recent advances in immunotherapies targeting microglia and macrophages in glioblastoma. Significant challenges remain but medications that affect glioblastoma-associated microglia and macrophages hold considerable promise to improve the prognosis for patients with this disease.
Collapse
Affiliation(s)
- Candice C Poon
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - Susobhan Sarkar
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - V Wee Yong
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - John J P Kelly
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
82
|
Lan X, Han X, Li Q, Yang QW, Wang J. Modulators of microglial activation and polarization after intracerebral haemorrhage. Nat Rev Neurol 2017; 13:420-433. [PMID: 28524175 PMCID: PMC5575938 DOI: 10.1038/nrneurol.2017.69] [Citation(s) in RCA: 572] [Impact Index Per Article: 71.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Intracerebral haemorrhage (ICH) is the most lethal subtype of stroke but currently lacks effective treatment. Microglia are among the first non-neuronal cells on the scene during the innate immune response to ICH. Microglia respond to acute brain injury by becoming activated and developing classic M1-like (proinflammatory) or alternative M2-like (anti-inflammatory) phenotypes. This polarization implies as yet unrecognized actions of microglia in ICH pathology and recovery, perhaps involving microglial production of proinflammatory or anti-inflammatory cytokines and chemokines. Furthermore, alternatively activated M2-like microglia might promote phagocytosis of red blood cells and tissue debris, a major contribution to haematoma clearance. Interactions between microglia and other cells modulate microglial activation and function, and are also important in ICH pathology. This Review summarizes key studies on modulators of microglial activation and polarization after ICH, including M1-like and M2-like microglial phenotype markers, transcription factors and key signalling pathways. Microglial phagocytosis, haematoma resolution, and the potential crosstalk between microglia and T lymphocytes, neurons, astrocytes, and oligodendrocytes in the ICH brain are described. Finally, the clinical and translational implications of microglial polarization in ICH are presented, including the evidence that therapeutic approaches aimed at modulating microglial function might mitigate ICH injury and improve brain repair.
Collapse
Affiliation(s)
- Xi Lan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross Building 370B, Baltimore, Maryland 21205, USA
| | - Xiaoning Han
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross Building 370B, Baltimore, Maryland 21205, USA
| | - Qian Li
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross Building 370B, Baltimore, Maryland 21205, USA
| | - Qing-Wu Yang
- Department of Neurology, Xinqiao Hospital, Third Military Medical University, 183 Xinqiao Main Street, Shapingba District, Chongqing 400037, China
| | - Jian Wang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross Building 370B, Baltimore, Maryland 21205, USA
| |
Collapse
|
83
|
Kumar R, de Mooij T, Peterson TE, Kaptzan T, Johnson AJ, Daniels DJ, Parney IF. Modulating glioma-mediated myeloid-derived suppressor cell development with sulforaphane. PLoS One 2017; 12:e0179012. [PMID: 28666020 PMCID: PMC5493295 DOI: 10.1371/journal.pone.0179012] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 05/23/2017] [Indexed: 11/18/2022] Open
Abstract
Glioblastoma is the most common primary tumor of the brain and has few long-term survivors. The local and systemic immunosuppressive environment created by glioblastoma allows it to evade immunosurveillance. Myeloid-derived suppressor cells (MDSCs) are a critical component of this immunosuppression. Understanding mechanisms of MDSC formation and function are key to developing effective immunotherapies. In this study, we developed a novel model to reliably generate human MDSCs from healthy-donor CD14+ monocytes by culture in human glioma-conditioned media. Monocytic MDSC frequency was assessed by flow cytometry and confocal microscopy. The resulting MDSCs robustly inhibited T cell proliferation. A cytokine array identified multiple components of the GCM potentially contributing to MDSC generation, including Monocyte Chemoattractive Protein-1, interleukin-6, interleukin-8, and Macrophage Migration Inhibitory Factor (MIF). Of these, Macrophage Migration Inhibitory Factor is a particularly attractive therapeutic target as sulforaphane, a naturally occurring MIF inhibitor derived from broccoli sprouts, has excellent oral bioavailability. Sulforaphane inhibits the transformation of normal monocytes to MDSCs by glioma-conditioned media in vitro at pharmacologically relevant concentrations that are non-toxic to normal leukocytes. This is associated with a corresponding increase in mature dendritic cells. Interestingly, sulforaphane treatment had similar pro-inflammatory effects on normal monocytes in fresh media but specifically increased immature dendritic cells. Thus, we have used a simple in vitro model system to identify a novel contributor to glioblastoma immunosuppression for which a natural inhibitor exists that increases mature dendritic cell development at the expense of myeloid-derived suppressor cells when normal monocytes are exposed to glioma conditioned media.
Collapse
Affiliation(s)
- Ravi Kumar
- Department of Neurological Surgery, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Tristan de Mooij
- Department of Neurological Surgery, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Timothy E. Peterson
- Department of Neurological Surgery, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Tatiana Kaptzan
- Department of Neurological Surgery, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Aaron J. Johnson
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - David J. Daniels
- Department of Neurological Surgery, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Ian F. Parney
- Department of Neurological Surgery, Mayo Clinic, Rochester, Minnesota, United States of America
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, United States of America
| |
Collapse
|
84
|
Shetty GA, Hattiangady B, Upadhya D, Bates A, Attaluri S, Shuai B, Kodali M, Shetty AK. Chronic Oxidative Stress, Mitochondrial Dysfunction, Nrf2 Activation and Inflammation in the Hippocampus Accompany Heightened Systemic Inflammation and Oxidative Stress in an Animal Model of Gulf War Illness. Front Mol Neurosci 2017; 10:182. [PMID: 28659758 PMCID: PMC5469946 DOI: 10.3389/fnmol.2017.00182] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 05/26/2017] [Indexed: 01/21/2023] Open
Abstract
Memory and mood dysfunction are the key symptoms of Gulf war illness (GWI), a lingering multi-symptom ailment afflicting >200,000 veterans who served in the Persian Gulf War-1. Research probing the source of the disease has demonstrated that concomitant exposures to anti-nerve gas agent pyridostigmine bromide (PB), pesticides, and war-related stress are among the chief causes of GWI. Indeed, exposures to GWI-related chemicals (GWIR-Cs) and mild stress in animal models cause memory and mood impairments alongside reduced neurogenesis and chronic low-level inflammation in the hippocampus. In the current study, we examined whether exposure to GWIR-Cs and stress causes chronic changes in the expression of genes related to increased oxidative stress, mitochondrial dysfunction, and inflammation in the hippocampus. We also investigated whether GWI is linked with chronically increased activation of Nrf2 (a master regulator of antioxidant response) in the hippocampus, and inflammation and enhanced oxidative stress at the systemic level. Adult male rats were exposed daily to low-doses of PB and pesticides (DEET and permethrin), in combination with 5 min of restraint stress for 4 weeks. Analysis of the hippocampus performed 6 months after the exposure revealed increased expression of many genes related to oxidative stress response and/or antioxidant activity (Hmox1, Sepp1, and Srxn1), reactive oxygen species metabolism (Fmo2, Sod2, and Ucp2) and oxygen transport (Ift172 and Slc38a1). Furthermore, multiple genes relevant to mitochondrial respiration (Atp6a1, Cox6a1, Cox7a2L, Ndufs7, Ndufv1, Lhpp, Slc25a10, and Ucp1) and neuroinflammation (Nfkb1, Bcl6, Csf2, IL6, Mapk1, Mapk3, Ngf, N-pac, and Prkaca) were up-regulated, alongside 73–88% reduction in the expression of anti-inflammatory genes IL4 and IL10, and nuclear translocation and increased expression of Nrf2 protein. These hippocampal changes were associated with elevated levels of pro-inflammatory cytokines and chemokines (Tnfa, IL1b, IL1a, Tgfb, and Fgf2) and lipid peroxidation byproduct malondialdehyde in the serum, suggesting the presence of an incessant systemic inflammation and elevated oxidative stress. These results imply that chronic oxidative stress, inflammation, and mitochondrial dysfunction in the hippocampus, and heightened systemic inflammation and oxidative stress likely underlie the persistent memory and mood dysfunction observed in GWI.
Collapse
Affiliation(s)
- Geetha A Shetty
- Research Service, Olin E. Teague Veterans' Medical Center, Central Texas Veterans Health Care System, TempleTX, United States.,Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine, Temple and College StationTX, United States.,Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College StationTX, United States
| | - Bharathi Hattiangady
- Research Service, Olin E. Teague Veterans' Medical Center, Central Texas Veterans Health Care System, TempleTX, United States.,Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine, Temple and College StationTX, United States.,Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College StationTX, United States
| | - Dinesh Upadhya
- Research Service, Olin E. Teague Veterans' Medical Center, Central Texas Veterans Health Care System, TempleTX, United States.,Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine, Temple and College StationTX, United States.,Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College StationTX, United States
| | - Adrian Bates
- Research Service, Olin E. Teague Veterans' Medical Center, Central Texas Veterans Health Care System, TempleTX, United States.,Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine, Temple and College StationTX, United States.,Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College StationTX, United States
| | - Sahithi Attaluri
- Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine, Temple and College StationTX, United States.,Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College StationTX, United States
| | - Bing Shuai
- Research Service, Olin E. Teague Veterans' Medical Center, Central Texas Veterans Health Care System, TempleTX, United States.,Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine, Temple and College StationTX, United States.,Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College StationTX, United States
| | - Maheedhar Kodali
- Research Service, Olin E. Teague Veterans' Medical Center, Central Texas Veterans Health Care System, TempleTX, United States.,Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine, Temple and College StationTX, United States.,Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College StationTX, United States
| | - Ashok K Shetty
- Research Service, Olin E. Teague Veterans' Medical Center, Central Texas Veterans Health Care System, TempleTX, United States.,Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine, Temple and College StationTX, United States.,Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College StationTX, United States
| |
Collapse
|
85
|
Schiffer D, Mellai M, Bovio E, Annovazzi L. The neuropathological basis to the functional role of microglia/macrophages in gliomas. Neurol Sci 2017; 38:1571-1577. [PMID: 28593528 DOI: 10.1007/s10072-017-3002-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 05/19/2017] [Indexed: 12/19/2022]
Abstract
The paper wants to be a tracking shot of the main recent acquisitions on the function and significance of microglia/macrophages in gliomas. The observations have been principally carried out on in vitro cultures and on tumor transplants in animals. Contrary to what is deduced from microglia in non-neoplastic pathologic conditions of central nervous system (CNS), most conclusions indicate that microglia acts favoring tumor proliferation through an immunosuppression induced by glioma cells. By immunohistochemistry, different microglia phenotypes are recognized in gliomas, from ramified microglia to frank macrophagic aspect. One wonders whether the functional conclusions drawn from many microglia studies, but not in conditions of human pathology, apply to all the phenotypes recognizable in them. It is difficult to verify in human pathology a prognostic significance of microglia. Only CD163-positive microglia/macrophages inversely correlate with glioma patients' survival, whereas the total number of microglia does not change with the malignancy grade.
Collapse
Affiliation(s)
- Davide Schiffer
- Research Center, Policlinico di Monza Foundation, Via Pietro Micca 29, 13100, Vercelli, Italy.
| | - Marta Mellai
- Research Center, Policlinico di Monza Foundation, Via Pietro Micca 29, 13100, Vercelli, Italy
| | - Enrica Bovio
- Research Center, Policlinico di Monza Foundation, Via Pietro Micca 29, 13100, Vercelli, Italy
| | - Laura Annovazzi
- Research Center, Policlinico di Monza Foundation, Via Pietro Micca 29, 13100, Vercelli, Italy
| |
Collapse
|
86
|
Ahn SH, Ahn JH, Ryu DR, Lee J, Cho MS, Choi YH. Effect of Necrosis on the miRNA-mRNA Regulatory Network in CRT-MG Human Astroglioma Cells. Cancer Res Treat 2017; 50:382-397. [PMID: 28546527 PMCID: PMC5912152 DOI: 10.4143/crt.2016.551] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 04/25/2017] [Indexed: 12/14/2022] Open
Abstract
Purpose Glioblastoma multiforme (GBM) is the most common adult primary intracranial tumor. The remarkable features of GBM include central necrosis. MicroRNAs (miRNAs) have been considered as diagnostic/prognostic biomarkers for many cancers, including glioblastoma. However, the effect of necrosis on the miRNA expression profile and predicted miRNA-mRNA regulatory information remain unclear. The purpose of this study is to examine the effect of necrotic cells on the modulation of miRNA and mRNA expression profiles and miRNA-mRNA network in CRT-MG cells. Materials and Methods We used human astroglioma cells, CRT-MG, treated with necrotic CRT-MG cells to examine the effect of necrosis on the modulation of miRNA and mRNA by next-generation sequencing. For preparation of necrotic cells, CRT-MGcellswere frozen and thawed through cycle of liquid nitrogen–water bath. The putative miRNA-mRNA regulatory relationshipwas inferred through target information, using miRDB. Results The necrotic cells induced dysregulation of 106 miRNAs and 887 mRNAs. Among them, 11 miRNAs that had a negative correlation value of p < 0.05 by the hypergeometric test were screened, and their target mRNAs were analyzed by Gene Ontology enrichment analysis. Using the Kyoto Encyclopedia of Genes and Genomes database, we also found several necrotic cell treatment-activated pathways that were modulated by relevant gene targets of differentially expressed miRNAs. Conclusion Our result demonstrated that dysregulation of miRNA and mRNA expression profiles occurs when GBM cells are exposed to necrotic cells, suggesting that several miRNAs may have the potential to be used as biomarkers for predicting GBM progression and pathogenesis.
Collapse
Affiliation(s)
- So-Hee Ahn
- Department of Physiology, Ewha Womans University School of Medicine,Seoul, Korea.,Tissue Injury Defense Research Center, Ewha Womans University School of Medicine,Seoul, Korea
| | - Jung-Hyuck Ahn
- Tissue Injury Defense Research Center, Ewha Womans University School of Medicine,Seoul, Korea.,Department of Biochemistry, Ewha Womans University School of Medicine,Seoul, Korea
| | - Dong-Ryeol Ryu
- Tissue Injury Defense Research Center, Ewha Womans University School of Medicine,Seoul, Korea.,Department of Internal medicine, Ewha Womans University School of Medicine,Seoul, Korea
| | - Jisoo Lee
- Department of Internal medicine, Ewha Womans University School of Medicine,Seoul, Korea
| | - Min-Sun Cho
- Department of Pathology, Ewha Womans University School of Medicine,Seoul, Korea
| | - Youn-Hee Choi
- Department of Physiology, Ewha Womans University School of Medicine,Seoul, Korea.,Tissue Injury Defense Research Center, Ewha Womans University School of Medicine,Seoul, Korea
| |
Collapse
|
87
|
Immune microenvironment of gliomas. J Transl Med 2017; 97:498-518. [PMID: 28287634 DOI: 10.1038/labinvest.2017.19] [Citation(s) in RCA: 377] [Impact Index Per Article: 47.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 01/16/2017] [Accepted: 01/19/2017] [Indexed: 12/13/2022] Open
Abstract
High-grade gliomas are rapidly progressing tumors of the central nervous system (CNS) with a very poor prognosis despite extensive resection combined with radiation and/or chemotherapy. Histopathological and flow cytometry analyses of human and rodent experimental gliomas revealed heterogeneity of a tumor and its niche, composed of reactive astrocytes, endothelial cells, and numerous immune cells. Infiltrating immune cells consist of CNS resident (microglia) and peripheral macrophages, granulocytes, myeloid-derived suppressor cells (MDSCs), and T lymphocytes. Intratumoral density of glioma-associated microglia/macrophages (GAMs) and MDSCs is the highest in malignant gliomas and inversely correlates with patient survival. Although GAMs have a few innate immune functions intact, their ability to be stimulated via TLRs, secrete cytokines, and upregulate co-stimulatory molecules is not sufficient to initiate antitumor immune responses. Moreover, tumor-reprogrammed GAMs release immunosuppressive cytokines and chemokines shaping antitumor responses. Both GAMs and MDSCs have ability to attract T regulatory lymphocytes to the tumor, but MDSCs inhibit cytotoxic responses mediated by natural killer cells, and block the activation of tumor-reactive CD4+ T helper cells and cytotoxic CD8+ T cells. The presence of regulatory T cells may further contribute to the lack of effective immune activation against malignant gliomas. We review the immunological aspects of glioma microenvironment, in particular composition and various roles of the immune cells infiltrating malignant human gliomas and experimental rodent gliomas. We describe tumor-derived signals and mechanisms driving myeloid cell accumulation and reprogramming. Although, understanding the complexity of cell-cell interactions in glioma microenvironment is far from being achieved, recent studies demonstrated several glioma-derived factors that trigger migration, accumulation, and reprogramming of immune cells. Identification of these factors may facilitate development of immunotherapy for gliomas as immunomodulatory and immune evasion mechanisms employed by malignant gliomas pose an appalling challenge to brain tumor immunotherapy.
Collapse
|
88
|
Anti-colony-stimulating factor therapies for inflammatory and autoimmune diseases. Nat Rev Drug Discov 2016; 16:53-70. [PMID: 28031576 DOI: 10.1038/nrd.2016.231] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
89
|
Zanotto-Filho A, Gonçalves RM, Klafke K, de Souza PO, Dillenburg FC, Carro L, Gelain DP, Moreira JCF. Inflammatory landscape of human brain tumors reveals an NFκB dependent cytokine pathway associated with mesenchymal glioblastoma. Cancer Lett 2016; 390:176-187. [PMID: 28007636 DOI: 10.1016/j.canlet.2016.12.015] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 12/08/2016] [Accepted: 12/12/2016] [Indexed: 12/14/2022]
Abstract
The tumor microenvironment is being increasingly recognized as a key factor in cancer aggressiveness. In this study, we characterized the inflammatory gene signatures altered in glioma cell lines and tumor specimens of differing histological and molecular subtypes. The results showed that glioblastoma multiforme (GBM) shows upregulation of a subset of inflammatory genes when compared to astrocytomas and oligodendrogliomas. With molecular subtypes of GBM, the expression of inflammatory genes is heterogeneous, being enriched in mesenchymal and downregulated in Proneural/GCIMP. Other inflammation-associated processes such as tumor-associated macrophage (TAM) signatures are upregulated in mesenchymal, and a subset of 33 mesenchymal-enriched inflammatory and TAM markers showed correlation with poor survival. We found that various GBM tumor-upregulated genes such as IL6, IL8 and CCL2 are also actively expressed in glioma cell lines, playing differential and cooperative roles in promoting proliferation, invasion, angiogenesis and macrophage polarization in vitro. These genes can be stimulated by pathways typically altered in GBM, including the EGFR, PDGFR, MEK1/2-ERK1/2, PI3K/Akt and NFκB cascades. Taken together, the results presented herein depict some inflammatory pathways altered in gliomas and highlight potentially relevant targets to therapy improvement.
Collapse
Affiliation(s)
- Alfeu Zanotto-Filho
- Departamento de Farmacologia, Centro de Ciências Biológicas (CCB), Universidade Federal de Santa Catarina (UFSC), Florianópolis, SC, Brazil; Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil.
| | - Rosângela Mayer Gonçalves
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Karina Klafke
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Priscila Oliveira de Souza
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Fabiane Cristine Dillenburg
- Instituto de Informática, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Luigi Carro
- Instituto de Informática, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Daniel Pens Gelain
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - José Cláudio Fonseca Moreira
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| |
Collapse
|
90
|
Motaln H, Koren A, Gruden K, Ramšak Ž, Schichor C, Lah TT. Heterogeneous glioblastoma cell cross-talk promotes phenotype alterations and enhanced drug resistance. Oncotarget 2016; 6:40998-1017. [PMID: 26517510 PMCID: PMC4747385 DOI: 10.18632/oncotarget.5701] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 09/16/2015] [Indexed: 01/30/2023] Open
Abstract
Glioblastoma multiforme is the most lethal of brain cancer, and it comprises a heterogeneous mixture of functionally distinct cancer cells that affect tumor progression. We examined the U87, U251, and U373 malignant cell lines as in vitro models to determine the impact of cellular cross-talk on their phenotypic alterations in co-cultures. These cells were also studied at the transcriptome level, to define the mechanisms of their observed mutually affected genomic stability, proliferation, invasion and resistance to temozolomide. This is the first direct demonstration of the neural and mesenchymal molecular fingerprints of U87 and U373 cells, respectively. U87-cell conditioned medium lowered the genomic stability of U373 (U251) cells, without affecting cell proliferation. In contrast, upon exposure of U87 cells to U373 (U251) conditioned medium, U87 cells showed increased genomic stability, decreased proliferation rates and increased invasion, due to a plethora of produced cytokines identified in the co-culture media. This cross talk altered the expression 264 genes in U87 cells that are associated with proliferation, inflammation, migration, and adhesion, and 221 genes in U373 cells that are associated with apoptosis, the cell cycle, cell differentiation and migration. Indirect and direct co-culturing of U87 and U373 cells showed mutually opposite effects on temozolomide resistance. In conclusion, definition of transcriptional alterations of distinct glioblastoma cells upon co-culturing provides better understanding of the mechanisms of glioblastoma heterogeneity, which will provide the basis for more informed glioma treatment in the future.
Collapse
Affiliation(s)
- Helena Motaln
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia
| | - Ana Koren
- Laboratory for Clinical Immunology and Molecular Genetics, University Clinic Golnik, Golnik, Slovenia
| | - Kristina Gruden
- Department of Biotechnology and Systems Biology, National Institute of Biology, Ljubljana, Slovenia
| | - Živa Ramšak
- Department of Biotechnology and Systems Biology, National Institute of Biology, Ljubljana, Slovenia
| | - Christian Schichor
- Department of Neurosurgery, Klinikum Großhadern, Ludwig-Maximilians-Universität, Munich, Germany
| | - Tamara T Lah
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia.,Department of Biochemistry, Faculty of Chemistry and Chemical Engineering, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
91
|
Down-regulation of IKKβ expression in glioma-infiltrating microglia/macrophages is associated with defective inflammatory/immune gene responses in glioblastoma. Oncotarget 2016; 6:33077-90. [PMID: 26427514 PMCID: PMC4741750 DOI: 10.18632/oncotarget.5310] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 09/15/2015] [Indexed: 11/25/2022] Open
Abstract
Glioblastoma (GBM) is an aggressive malignancy associated with profound host immunosuppression. Microglia and macrophages infiltrating GBM acquire the pro-tumorigenic, M2 phenotype and support tumor invasion, proliferation, survival, angiogenesis and block immune responses both locally and systematically. Mechanisms responsible for immunological deficits in GBM patients are poorly understood. We analyzed immune/inflammatory gene expression in five datasets of low and high grade gliomas, and performed Gene Ontology and signaling pathway analyses to identify defective transcriptional responses. The expression of many immune/inflammatory response and TLR signaling pathway genes was reduced in high grade gliomas compared to low grade gliomas. In particular, we found the reduced expression of the IKBKB, a gene coding for IKKβ, which phosphorylates IκB proteins and represents a convergence point for most signal transduction pathways leading to NFκB activation. The reduced IKBKB expression and IKKβ levels in GBM tissues were demonstrated by qPCR, Western blotting and immunohistochemistry. The IKKβ expression was down-regulated in microglia/macrophages infiltrating glioblastoma. NFκB activation, prominent in microglia/macrophages infiltrating low grade gliomas, was reduced in microglia/macrophages in glioblastoma tissues. Down-regulation of IKBKB expression and NFκB signaling in microglia/macrophages infiltrating glioblastoma correlates with defective expression of immune/inflammatory genes and M2 polarization that may result in the global impairment of anti-tumor immune responses in glioblastoma.
Collapse
|
92
|
Michelson N, Rincon-Torroella J, Quiñones-Hinojosa A, Greenfield JP. Exploring the role of inflammation in the malignant transformation of low-grade gliomas. J Neuroimmunol 2016; 297:132-40. [DOI: 10.1016/j.jneuroim.2016.05.019] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 05/09/2016] [Accepted: 05/23/2016] [Indexed: 01/14/2023]
|
93
|
Takeya M, Komohara Y. Role of tumor-associated macrophages in human malignancies: friend or foe? Pathol Int 2016; 66:491-505. [PMID: 27444136 DOI: 10.1111/pin.12440] [Citation(s) in RCA: 134] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 05/29/2016] [Accepted: 06/24/2016] [Indexed: 12/11/2022]
Abstract
Tumor-associated macrophages (TAMs) play a pivotal role in tumor growth in human malignancies. Published studies have analyzed the relationship between TAM infiltration and the prognosis of patients for many human tumors. Most studies reported a positive correlation between TAM density and a poor prognosis. Studies focusing on macrophage phenotypes emphasized the protumor role of M2 anti-inflammatory macrophages in many types of human tumors. However, TAMs influence tumor progression in various ways that depend on differences in tumor sites, histology, and microenvironments. In this review, we summarize the function of TAMs in various human malignancies by reviewing the data provided in studies of TAMs in human malignancies.
Collapse
Affiliation(s)
- Motohiro Takeya
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yoshihiro Komohara
- Department of Cell Pathology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
94
|
Mignogna C, Signorelli F, Vismara MFM, Zeppa P, Camastra C, Barni T, Donato G, Di Vito A. A reappraisal of macrophage polarization in glioblastoma: Histopathological and immunohistochemical findings and review of the literature. Pathol Res Pract 2016; 212:491-9. [DOI: 10.1016/j.prp.2016.02.020] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 01/28/2016] [Accepted: 02/23/2016] [Indexed: 12/22/2022]
|
95
|
The role of microglia and macrophages in glioma maintenance and progression. Nat Neurosci 2016; 19:20-7. [PMID: 26713745 DOI: 10.1038/nn.4185] [Citation(s) in RCA: 1141] [Impact Index Per Article: 126.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 06/23/2015] [Indexed: 11/08/2022]
Abstract
There is a growing recognition that gliomas are complex tumors composed of neoplastic and non-neoplastic cells, which each individually contribute to cancer formation, progression and response to treatment. The majority of the non-neoplastic cells are tumor-associated macrophages (TAMs), either of peripheral origin or representing brain-intrinsic microglia, that create a supportive stroma for neoplastic cell expansion and invasion. TAMs are recruited to the glioma environment, have immune functions, and can release a wide array of growth factors and cytokines in response to those factors produced by cancer cells. In this manner, TAMs facilitate tumor proliferation, survival and migration. Through such iterative interactions, a unique tumor ecosystem is established, which offers new opportunities for therapeutic targeting.
Collapse
|
96
|
Ghoochani A, Schwarz MA, Yakubov E, Engelhorn T, Doerfler A, Buchfelder M, Bucala R, Savaskan NE, Eyüpoglu IY. MIF-CD74 signaling impedes microglial M1 polarization and facilitates brain tumorigenesis. Oncogene 2016; 35:6246-6261. [PMID: 27157615 DOI: 10.1038/onc.2016.160] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 12/17/2015] [Accepted: 02/23/2016] [Indexed: 12/28/2022]
Abstract
Microglial cells in the brain tumor microenvironment are associated with enhanced glioma malignancy. They persist in an immunosuppressive M2 state at the peritumoral site and promote the growth of gliomas. Here, we investigated the underlying factors contributing to the abolished immune surveillance. We show that brain tumors escape pro-inflammatory M1 conversion of microglia via CD74 activation through the secretion of the cytokine macrophage migration inhibitory factor (MIF), which results in a M2 shift of microglial cells. Interruption of this glioma-microglial interaction through an antibody-neutralizing approach or small interfering RNA (siRNA)-mediated inhibition prolongs survival time in glioma-implanted mice by reinstating the microglial pro-inflammatory M1 function. We show that MIF-CD74 signaling inhibits interferon (IFN)-γ secretion in microglia through phosphorylation of microglial ERK1/2 (extracellular signal-regulated protein kinases 1 and 2). The inhibition of MIF signaling or its receptor CD74 promotes IFN-γ release and amplifies tumor death either through pharmacological inhibition or through siRNA-mediated knockdown. The reinstated IFN-γ secretion leads both to direct inhibition of glioma growth as well as inducing a M2 to M1 shift in glioma-associated microglia. Our data reveal that interference with the MIF signaling pathway represents a viable therapeutic option for the restoration of IFN-γ-driven immune surveillance.
Collapse
Affiliation(s)
- A Ghoochani
- Department of Neurosurgery, Universitätsklinikum Erlangen, Medical Faculty of the Friedrich Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - M A Schwarz
- Department of Neurosurgery, Universitätsklinikum Erlangen, Medical Faculty of the Friedrich Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - E Yakubov
- Department of Neurosurgery, Universitätsklinikum Erlangen, Medical Faculty of the Friedrich Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - T Engelhorn
- Department of Neuroradiology, Universitätsklinikum Erlangen, Medical Faculty of the Friedrich Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - A Doerfler
- Department of Neuroradiology, Universitätsklinikum Erlangen, Medical Faculty of the Friedrich Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - M Buchfelder
- Department of Neurosurgery, Universitätsklinikum Erlangen, Medical Faculty of the Friedrich Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - R Bucala
- Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - N E Savaskan
- Department of Neurosurgery, Universitätsklinikum Erlangen, Medical Faculty of the Friedrich Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany.,BiMECON Ent., Kurfürstenstrasse 21, Berlin, Germany
| | - I Y Eyüpoglu
- Department of Neurosurgery, Universitätsklinikum Erlangen, Medical Faculty of the Friedrich Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
97
|
Salacz ME, Kast RE, Saki N, Brüning A, Karpel-Massler G, Halatsch ME. Toward a noncytotoxic glioblastoma therapy: blocking MCP-1 with the MTZ Regimen. Onco Targets Ther 2016; 9:2535-45. [PMID: 27175087 PMCID: PMC4854261 DOI: 10.2147/ott.s100407] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
To improve the prognosis of glioblastoma, we developed an adjuvant treatment directed to a neglected aspect of glioblastoma growth, the contribution of nonmalignant monocyte lineage cells (MLCs) (monocyte, macrophage, microglia, dendritic cells) that infiltrated a main tumor mass. These nonmalignant cells contribute to glioblastoma growth and tumor homeostasis. MLCs comprise of approximately 10%-30% of glioblastoma by volume. After integration into the tumor mass, these become polarized toward an M2 immunosuppressive, pro-angiogenic phenotype that promotes continued tumor growth. Glioblastoma cells initiate and promote this process by synthesizing 13 kDa MCP-1 that attracts circulating monocytes to the tumor. Infiltrating monocytes, after polarizing toward an M2 phenotype, synthesize more MCP-1, forming an amplification loop. Three noncytotoxic drugs, an antibiotic - minocycline, an antihypertensive drug - telmisartan, and a bisphosphonate - zoledronic acid, have ancillary attributes of MCP-1 synthesis inhibition and could be re-purposed, singly or in combination, to inhibit or reverse MLC-mediated immunosuppression, angiogenesis, and other growth-enhancing aspects. Minocycline, telmisartan, and zoledronic acid - the MTZ Regimen - have low-toxicity profiles and could be added to standard radiotherapy and temozolomide. Re-purposing older drugs has advantages of established safety and low drug cost. Four core observations support this approach: 1) malignant glioblastoma cells require a reciprocal trophic relationship with nonmalignant macrophages or microglia to thrive; 2) glioblastoma cells secrete MCP-1 to start the cycle, attracting MLCs, which subsequently also secrete MCP-1 perpetuating the recruitment cycle; 3) increasing cytokine levels in the tumor environment generate further immunosuppression and tumor growth; and 4) MTZ regimen may impede MCP-1-driven processes, thereby interfering with glioblastoma growth.
Collapse
Affiliation(s)
- Michael E Salacz
- Department of Internal Medicine, University of Kansas, Kansas City, KS, USA; Department of Neurosurgery, University of Kansas, Kansas City, KS, USA
| | | | - Najmaldin Saki
- Health Research Institute, Research Center of Thalassemia and Hemoglobinopathy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ansgar Brüning
- Molecular Biology Laboratory, University Hospital Munich, Munich, Germany
| | | | | |
Collapse
|
98
|
Ellert-Miklaszewska A, Wisniewski P, Kijewska M, Gajdanowicz P, Pszczolkowska D, Przanowski P, Dabrowski M, Maleszewska M, Kaminska B. Tumour-processed osteopontin and lactadherin drive the protumorigenic reprogramming of microglia and glioma progression. Oncogene 2016; 35:6366-6377. [DOI: 10.1038/onc.2016.55] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 01/17/2016] [Accepted: 02/02/2016] [Indexed: 12/17/2022]
|
99
|
De I, Steffen MD, Clark PA, Patros CJ, Sokn E, Bishop SM, Litscher S, Maklakova VI, Kuo JS, Rodriguez FJ, Collier LS. CSF1 Overexpression Promotes High-Grade Glioma Formation without Impacting the Polarization Status of Glioma-Associated Microglia and Macrophages. Cancer Res 2016; 76:2552-60. [PMID: 27013192 DOI: 10.1158/0008-5472.can-15-2386] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 02/22/2016] [Indexed: 12/11/2022]
Abstract
Current therapies for high-grade gliomas extend survival only modestly. The glioma microenvironment, including glioma-associated microglia/macrophages (GAM), is a potential therapeutic target. The microglia/macrophage cytokine CSF1 and its receptor CSF1R are overexpressed in human high-grade gliomas. To determine whether the other known CSF1R ligand IL34 is expressed in gliomas, we examined expression array data of human high-grade gliomas and performed RT-PCR on glioblastoma sphere-forming cell lines (GSC). Expression microarray analyses indicated that CSF1, but not IL34, is frequently overexpressed in human tumors. We found that while GSCs did express CSF1, most GSC lines did not express detectable levels of IL34 mRNA. We therefore studied the impact of modulating CSF1 levels on gliomagenesis in the context of the GFAP-V12Ha-ras-IRESLacZ (Ras*) model. Csf1 deficiency deterred glioma formation in the Ras* model, whereas CSF1 transgenic overexpression decreased the survival of Ras* mice and promoted the formation of high-grade gliomas. Conversely, CSF1 overexpression increased GAM density, but did not impact GAM polarization state. Regardless of CSF1 expression status, most GAMs were negative for the M2 polarization markers ARG1 and CD206; when present, ARG1(+) and CD206(+) cells were found in regions of peripheral immune cell invasion. Therefore, our findings indicate that CSF1 signaling is oncogenic during gliomagenesis through a mechanism distinct from modulating GAM polarization status. Cancer Res; 76(9); 2552-60. ©2016 AACR.
Collapse
Affiliation(s)
- Ishani De
- School of Pharmacy, Carbone Cancer Center and the Molecular and Cellular Pharmacology Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin
| | - Megan D Steffen
- School of Pharmacy, Carbone Cancer Center and the Molecular and Cellular Pharmacology Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin
| | - Paul A Clark
- Department of Neurological Surgery and Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin
| | - Clayton J Patros
- School of Pharmacy, Carbone Cancer Center and the Molecular and Cellular Pharmacology Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin
| | - Emily Sokn
- School of Pharmacy, Carbone Cancer Center and the Molecular and Cellular Pharmacology Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin
| | - Stephanie M Bishop
- School of Pharmacy, Carbone Cancer Center and the Molecular and Cellular Pharmacology Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin
| | - Suzanne Litscher
- School of Pharmacy, Carbone Cancer Center and the Molecular and Cellular Pharmacology Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin
| | - Vilena I Maklakova
- School of Pharmacy, Carbone Cancer Center and the Molecular and Cellular Pharmacology Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin
| | - John S Kuo
- Department of Neurological Surgery and Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin
| | - Fausto J Rodriguez
- Division of Neuropathology, Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Lara S Collier
- School of Pharmacy, Carbone Cancer Center and the Molecular and Cellular Pharmacology Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin.
| |
Collapse
|
100
|
Domingues P, González-Tablas M, Otero Á, Pascual D, Miranda D, Ruiz L, Sousa P, Ciudad J, Gonçalves JM, Lopes MC, Orfao A, Tabernero MD. Tumor infiltrating immune cells in gliomas and meningiomas. Brain Behav Immun 2016. [PMID: 26216710 DOI: 10.1016/j.bbi.2015.07.019] [Citation(s) in RCA: 198] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Tumor-infiltrating immune cells are part of a complex microenvironment that promotes and/or regulates tumor development and growth. Depending on the type of cells and their functional interactions, immune cells may play a key role in suppressing the tumor or in providing support for tumor growth, with relevant effects on patient behavior. In recent years, important advances have been achieved in the characterization of immune cell infiltrates in central nervous system (CNS) tumors, but their role in tumorigenesis and patient behavior still remain poorly understood. Overall, these studies have shown significant but variable levels of infiltration of CNS tumors by macrophage/microglial cells (TAM) and to a less extent also lymphocytes (particularly T-cells and NK cells, and less frequently also B-cells). Of note, TAM infiltrate gliomas at moderate numbers where they frequently show an immune suppressive phenotype and functional behavior; in contrast, infiltration by TAM may be very pronounced in meningiomas, particularly in cases that carry isolated monosomy 22, where the immune infiltrates also contain greater numbers of cytotoxic T and NK-cells associated with an enhanced anti-tumoral immune response. In line with this, the presence of regulatory T cells, is usually limited to a small fraction of all meningiomas, while frequently found in gliomas. Despite these differences between gliomas and meningiomas, both tumors show heterogeneous levels of infiltration by immune cells with variable functionality. In this review we summarize current knowledge about tumor-infiltrating immune cells in the two most common types of CNS tumors-gliomas and meningiomas-, as well as the role that such immune cells may play in the tumor microenvironment in controlling and/or promoting tumor development, growth and control.
Collapse
Affiliation(s)
- Patrícia Domingues
- Centre for Neurosciences and Cell Biology and Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; Centre for Cancer Research (CIC-IBMCC; CSIC/USAL; IBSAL) and Department of Medicine, University of Salamanca, Salamanca, Spain
| | - María González-Tablas
- Centre for Cancer Research (CIC-IBMCC; CSIC/USAL; IBSAL) and Department of Medicine, University of Salamanca, Salamanca, Spain
| | - Álvaro Otero
- Neurosurgery Service of the University Hospital of Salamanca, Salamanca, Spain
| | - Daniel Pascual
- Neurosurgery Service of the University Hospital of Salamanca, Salamanca, Spain
| | - David Miranda
- Neurosurgery Service of the University Hospital of Salamanca, Salamanca, Spain
| | - Laura Ruiz
- Neurosurgery Service of the University Hospital of Salamanca, Salamanca, Spain
| | - Pablo Sousa
- Neurosurgery Service of the University Hospital of Salamanca, Salamanca, Spain
| | - Juana Ciudad
- Centre for Cancer Research (CIC-IBMCC; CSIC/USAL; IBSAL) and Department of Medicine, University of Salamanca, Salamanca, Spain
| | | | - María Celeste Lopes
- Centre for Neurosciences and Cell Biology and Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Alberto Orfao
- Centre for Cancer Research (CIC-IBMCC; CSIC/USAL; IBSAL) and Department of Medicine, University of Salamanca, Salamanca, Spain
| | - María Dolores Tabernero
- Centre for Cancer Research (CIC-IBMCC; CSIC/USAL; IBSAL) and Department of Medicine, University of Salamanca, Salamanca, Spain; Neurosurgery Service of the University Hospital of Salamanca, Salamanca, Spain; Instituto de Estudios de Ciencias de la salud de Castilla y León (IECSCYL-IBSAL) and Research Unit of the University Hospital of Salamanca, Salamanca, Spain.
| |
Collapse
|