1
|
Zhang Y, Ma Y, Liu Q, Du Y, Peng L, Zhou J, Zhao Z, Li C, Wang S. Single-cell transcriptome sequencing reveals tumor heterogeneity in family neuroblastoma. Front Immunol 2023; 14:1197773. [PMID: 37790931 PMCID: PMC10543897 DOI: 10.3389/fimmu.2023.1197773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 09/01/2023] [Indexed: 10/05/2023] Open
Abstract
Neuroblastoma(NB) is the most common extracranial solid tumor in childhood, and it is now believed that some patients with NB have an underlying genetic susceptibility, which may be one of the reasons for the multiplicity of NB patients within a family line. Even within the same family, the samples show great variation and can present as ganglioneuroblastoma or even benign ganglioneuroma. The genomics of NB is still unclear and more in-depth studies are needed to reveal its key components. We first performed single-cell RNA sequencing(sc-RNAseq) analysis on clinical specimens of two family neuroblastoma(FNB) and four sporadic NB cases. A complete transcriptional profile of FNB was constructed from 18,394 cells from FNB, and we found that SDHD may be genetically associated with FNB and identified a prognostic related CAF subtype in FNB: Fib-4. Single-cell flux estimation analysis (scFEA) results showed that malignant cells were associated with arginine spermine, oxaloacetate and hypoxanthine, and that malignant cells metabolize lactate at lower levels than T cells. Our study provides new resources and ideas for the development of the genomics of family NB, and the mechanisms of cell-to-cell interactions and communication and the metabolic landscape will provide new therapeutic targets.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Shan Wang
- Department of Pediatric Surgical Oncology Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| |
Collapse
|
2
|
Zhang HF, Delaidelli A, Javed S, Turgu B, Morrison T, Hughes CS, Yang X, Pachva M, Lizardo MM, Singh G, Hoffmann J, Huang YZ, Patel K, Shraim R, Kung SH, Morin GB, Aparicio S, Martinez D, Maris JM, Bosse KR, Williams KC, Sorensen PH. A MYCN-independent mechanism mediating secretome reprogramming and metastasis in MYCN-amplified neuroblastoma. SCIENCE ADVANCES 2023; 9:eadg6693. [PMID: 37611092 PMCID: PMC10446492 DOI: 10.1126/sciadv.adg6693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 07/21/2023] [Indexed: 08/25/2023]
Abstract
MYCN amplification (MNA) is a defining feature of high-risk neuroblastoma (NB) and predicts poor prognosis. However, whether genes within or in close proximity to the MYCN amplicon also contribute to MNA+ NB remains poorly understood. Here, we identify that GREB1, a transcription factor encoding gene neighboring the MYCN locus, is frequently coexpressed with MYCN and promotes cell survival in MNA+ NB. GREB1 controls gene expression independently of MYCN, among which we uncover myosin 1B (MYO1B) as being highly expressed in MNA+ NB and, using a chick chorioallantoic membrane (CAM) model, as a crucial regulator of invasion and metastasis. Global secretome and proteome profiling further delineates MYO1B in regulating secretome reprogramming in MNA+ NB cells, and the cytokine MIF as an important pro-invasive and pro-metastatic mediator of MYO1B activity. Together, we have identified a putative GREB1-MYO1B-MIF axis as an unconventional mechanism promoting aggressive behavior in MNA+ NB and independently of MYCN.
Collapse
Affiliation(s)
- Hai-Feng Zhang
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T1Z4, Canada
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, BC V5Z1L3, Canada
| | - Alberto Delaidelli
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T1Z4, Canada
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, BC V5Z1L3, Canada
| | - Sumreen Javed
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC, Canada
| | - Busra Turgu
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T1Z4, Canada
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, BC V5Z1L3, Canada
| | - Taylor Morrison
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, BC V5Z1L3, Canada
| | - Christopher S. Hughes
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T1Z4, Canada
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, BC V5Z1L3, Canada
| | - Xiaqiu Yang
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, BC V5Z1L3, Canada
| | - Manideep Pachva
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T1Z4, Canada
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, BC V5Z1L3, Canada
| | - Michael M. Lizardo
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T1Z4, Canada
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, BC V5Z1L3, Canada
| | - Gurdeep Singh
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, BC V5Z1L3, Canada
| | - Jennifer Hoffmann
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Yue Zhou Huang
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, BC V5Z1L3, Canada
| | - Khushbu Patel
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rawan Shraim
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | | | - Gregg B. Morin
- Canada’s Michael Smith Genome Sciences Centre, Vancouver, BC V5Z4S6, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T1Z4, Canada
| | - Samuel Aparicio
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T1Z4, Canada
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, BC V5Z1L3, Canada
| | - Daniel Martinez
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - John M. Maris
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kristopher R. Bosse
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Karla C. Williams
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC, Canada
| | - Poul H. Sorensen
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T1Z4, Canada
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, BC V5Z1L3, Canada
| |
Collapse
|
3
|
Garcia-Gerique L, García M, Garrido-Garcia A, Gómez-González S, Torrebadell M, Prada E, Pascual-Pasto G, Muñoz O, Perez-Jaume S, Lemos I, Salvador N, Vila-Ubach M, Doncel-Requena A, Suñol M, Carcaboso AM, Mora J, Lavarino C. MIF/CXCR4 signaling axis contributes to survival, invasion, and drug resistance of metastatic neuroblastoma cells in the bone marrow microenvironment. BMC Cancer 2022; 22:669. [PMID: 35715791 PMCID: PMC9206243 DOI: 10.1186/s12885-022-09725-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 05/18/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The bone marrow (BM) is the most common site of dissemination in patients with aggressive, metastatic neuroblastoma (NB). However, the molecular mechanisms underlying the aggressive behavior of NB cells in the BM niche are still greatly unknown. In the present study, we explored biological mechanisms that play a critical role in NB cell survival and progression in the BM and investigated potential therapeutic targets. METHODS Patient-derived bone marrow (BM) primary cultures were generated using fresh BM aspirates obtained from NB patients. NB cell lines were cultured in the presence of BM conditioned media containing cell-secreted factors, and under low oxygen levels (1% O2) to mimic specific features of the BM microenvironment of high-risk NB patients. The BM niche was explored using cytokine profiling assays, cell migration-invasion and viability assays, flow cytometry and analysis of RNA-sequencing data. Selective pharmacological inhibition of factors identified as potential mediators of NB progression within the BM niche was performed in vitro and in vivo. RESULTS We identified macrophage migration inhibitory factor (MIF) as a key inflammatory cytokine involved in BM infiltration. Cytokine profiling and RNA-sequencing data analysis revealed NB cells as the main source of MIF in the BM, suggesting a potential role of MIF in tumor invasion. Exposure of NB cells to BM-conditions increased NB cell-surface expression of the MIF receptor CXCR4, which was associated with increased cell viability, enhanced migration-invasion, and activation of PI3K/AKT and MAPK/ERK signaling pathways. Moreover, subcutaneous co-injection of NB and BM cells enhanced tumor engraftment in mice. MIF inhibition with 4-IPP impaired in vitro NB aggressiveness, and improved drug response while delayed NB growth, improving survival of the NB xenograft model. CONCLUSIONS Our findings suggest that BM infiltration by NB cells may be mediated, in part, by MIF-CXCR4 signaling. We demonstrate the antitumor efficacy of MIF targeting in vitro and in vivo that could represent a novel therapeutic target for patients with disseminated high-risk NB.
Collapse
Affiliation(s)
- Laura Garcia-Gerique
- Developmental Tumor Biology Laboratory, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Marta García
- Developmental Tumor Biology Laboratory, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Alícia Garrido-Garcia
- Developmental Tumor Biology Laboratory, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Soledad Gómez-González
- Developmental Tumor Biology Laboratory, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Montserrat Torrebadell
- Developmental Tumor Biology Laboratory, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Estela Prada
- Developmental Tumor Biology Laboratory, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Guillem Pascual-Pasto
- Developmental Tumor Biology Laboratory, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Oscar Muñoz
- Developmental Tumor Biology Laboratory, Institut de Recerca Sant Joan de Déu, Barcelona, Spain.,Department of Pathology, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Sara Perez-Jaume
- Developmental Tumor Biology Laboratory, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Isadora Lemos
- Developmental Tumor Biology Laboratory, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Noelia Salvador
- Developmental Tumor Biology Laboratory, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Monica Vila-Ubach
- Developmental Tumor Biology Laboratory, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Ana Doncel-Requena
- Developmental Tumor Biology Laboratory, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Mariona Suñol
- Department of Pathology, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Angel M Carcaboso
- Developmental Tumor Biology Laboratory, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Jaume Mora
- Developmental Tumor Biology Laboratory, Institut de Recerca Sant Joan de Déu, Barcelona, Spain.,Laboratory of Molecular Oncology, Pediatric Cancer Center Barcelona (PCCB), Hospital Sant Joan de Déu, Barcelona, Spain
| | - Cinzia Lavarino
- Developmental Tumor Biology Laboratory, Institut de Recerca Sant Joan de Déu, Barcelona, Spain. .,Laboratory of Molecular Oncology, Pediatric Cancer Center Barcelona (PCCB), Hospital Sant Joan de Déu, Barcelona, Spain.
| |
Collapse
|
4
|
Grundy M, Narendran A. The hepatocyte growth factor/mesenchymal epithelial transition factor axis in high-risk pediatric solid tumors and the anti-tumor activity of targeted therapeutic agents. Front Pediatr 2022; 10:910268. [PMID: 36034555 PMCID: PMC9399617 DOI: 10.3389/fped.2022.910268] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 07/15/2022] [Indexed: 01/04/2023] Open
Abstract
Clinical trials completed in the last two decades have contributed significantly to the improved overall survival of children with cancer. In spite of these advancements, disease relapse still remains a significant cause of death in this patient population. Often, increasing the intensity of current protocols is not feasible because of cumulative toxicity and development of drug resistance. Therefore, the identification and clinical validation of novel targets in high-risk and refractory childhood malignancies are essential to develop effective new generation treatment protocols. A number of recent studies have shown that the hepatocyte growth factor (HGF) and its receptor Mesenchymal epithelial transition factor (c-MET) influence the growth, survival, angiogenesis, and metastasis of cancer cells. Therefore, the c-MET receptor tyrosine kinase and HGF have been identified as potential targets for cancer therapeutics and recent years have seen a race to synthesize molecules to block their expression and function. In this review we aim to summarize the literature that explores the potential and biological rationale for targeting the HGF/c-MET pathway in common and high-risk pediatric solid tumors. We also discuss selected recent and ongoing clinical trials with these agents in relapsed pediatric tumors that may provide applicable future treatments for these patients.
Collapse
Affiliation(s)
- Megan Grundy
- Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Aru Narendran
- POETIC Laboratory for Preclinical and Drug Discovery Studies, Division of Pediatric Oncology, Alberta Children's Hospital, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
5
|
Sun F, Lu S, Zhen Z, Zhu J, Wang J, Huang J, Zhang Y, Li H, Cai R, Liu M, Wu L, Sun X, Zhang Y. The Efficacy and Safety of Apatinib in Refractory/Relapse Advanced Pediatric Solid Tumor: A Retrospective Study. Cancer Manag Res 2020; 12:6177-6185. [PMID: 32801866 PMCID: PMC7383017 DOI: 10.2147/cmar.s258689] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 07/02/2020] [Indexed: 11/23/2022] Open
Abstract
Background The prognosis of recurrent or refractory advanced childhood solid tumor patients is very poor and new therapeutic strategies are in urgent need. This study aimed to determine the efficacy and safety of apatinib in pediatric refractory/relapse advanced solid tumor patients. Patients and Methods The study retrospectively reviewed recurrent or refractory advanced pediatric solid tumor patients who were treated with apatinib, an oral small-molecule tyrosine kinase inhibitor (TKI) that targets vascular endothelial growth factor receptor-2 (VEGFR2), at the Sun Yat-sen University Cancer Center (China) from January 2016 to March 2019. Results Fifty-six patients were included in the safety evaluation and 49 patients were included in the efficacy evaluation. The objective responses rate (ORR) was 26.5% (95% CI 15–41): 0 CR (complete response) and 13 PR (partial response). Disease control rate (DCR) (CR+PR+SD) was 79.6% (95% CI 65–90). The median progression-free survival (PFS) was 4.0 months (95% CI 2.6–5.4). There was no significant difference for ORR or PFS between the A (apatinib monotherapy), A+MT (apatinib combined with oral metronomic therapy) and A+SC (apatinib combined with salvage combination chemotherapy) group (p>0.05). The most common grade 3 or 4 adverse events were neutropenia (9[16.1%]), thrombocytopenia (8[14.3%]), hand-foot syndrome (3[5.4%]), hypertension (3[5.4%]), anaemia (3[5.4%]) and mucositis (2[3.6%]). Hypertension was the most serious adverse event and one death that occurred was considered as drug-related. Conclusion Apatinib showed promising clinical activity in heavily treated recurrent or refractory advanced childhood solid tumor patients. However, it is necessary to pay special attention to monitoring blood pressure when using apatinib in children. Prospective randomized controlled clinical trial is warranted.
Collapse
Affiliation(s)
- Feifei Sun
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China.,Department of Pediatric Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | - Suying Lu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China.,Department of Pediatric Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | - Zijun Zhen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China.,Department of Pediatric Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | - Jia Zhu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China.,Department of Pediatric Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | - Juan Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China.,Department of Pediatric Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | - Junting Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China.,Department of Pediatric Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | - Yu Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China.,Department of Pathology, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | - Hui Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China.,Department of Imaging, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | - Ruiqing Cai
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China.,Department of Pediatric Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | - Meiling Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China.,Department of Pediatric Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | - Liuhong Wu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China.,Department of Pediatric Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | - Xiaofei Sun
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China.,Department of Pediatric Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| | - Yizhuo Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China.,Department of Pediatric Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, People's Republic of China
| |
Collapse
|
6
|
Emerging Role of the Macrophage Migration Inhibitory Factor Family of Cytokines in Neuroblastoma. Pathogenic Effectors and Novel Therapeutic Targets? Molecules 2020; 25:molecules25051194. [PMID: 32155795 PMCID: PMC7179464 DOI: 10.3390/molecules25051194] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/02/2020] [Accepted: 03/03/2020] [Indexed: 12/17/2022] Open
Abstract
Neuroblastoma (NB) is the most frequent extracranial pediatric tumor. Despite the current available multiple therapeutic options, the prognosis for high-risk NB patients remains unsatisfactory and makes the disease a clear unmet medical need. Thus, more tailored therapeutic approaches are warranted to improve both the quality of life and the survival of the patients. Macrophage migration inhibitory factor (MIF) is a pleiotropic cytokine that plays a key role in several diseases, including cancer. Preclinical and clinical studies in NB patients convergently indicate that MIF exerts pro-tumorigenic properties in NB. MIF is upregulated in NB tumor tissues and cell lines and it contributes to NB aggressiveness and immune-escape. To date, there are only a few data about the role of the second member of the MIF family, the MIF homolog d-dopachrome tautomerase (DDT), in NB. Here, we review the preclinical and clinical studies on the role of the MIF family of cytokines in NB and suggest that MIF and possibly DDT inhibitors may be promising novel prognostic and therapeutic targets in NB management.
Collapse
|
7
|
Cavalli E, Mazzon E, Mammana S, Basile MS, Lombardo SD, Mangano K, Bramanti P, Nicoletti F, Fagone P, Petralia MC. Overexpression of Macrophage Migration Inhibitory Factor and Its Homologue D-Dopachrome Tautomerase as Negative Prognostic Factor in Neuroblastoma. Brain Sci 2019; 9:brainsci9100284. [PMID: 31635049 PMCID: PMC6826588 DOI: 10.3390/brainsci9100284] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 10/01/2019] [Accepted: 10/18/2019] [Indexed: 01/09/2023] Open
Abstract
Neuroblastoma (NB) represents one of the most frequent pediatric solid tumors. Macrophage migration inhibitory factor (MIF) is a cytokine exerting multiple biological functions. More recently, a second member of the MIF family of cytokine has been identified, the D-dopachrome tautomerase (DDT), that exerts several overlapping functions with MIF. Growing evidence suggests a key role for MIF and DDT in the development of cancer. The aim of this study is to characterize the prognostic value of MIF and DDT in NB. We show that higher expression levels of MIF and DDT in Stage 4 NB samples are associated with a poorer prognosis, independently of the presence of MYCN amplification. Moreover, higher levels of MIF are mostly enriched by Th1 cells, while lower levels of MIF are associated with an increased proportion of B cells, Cytotoxic T cells, Dendritic cells and Natural Killer T cells. We also show that treatment with the histone deacetylase (HDAC) inhibitor, vorinostat, of the NB cell line, SH-SY5Y, determines a significant reduction in the expression of both MIF and DDT. Finally, MIF and DDT inhibition by short interfering RNA is able to revert vincristine sensitivity in vitro. Overall, our data suggest that MIF exert pro-tumorigenic properties in NB, likely by dampening antigen presentation and cytotoxic immune responses, and we propose the HDAC inhibitors as a potential therapeutic strategy for NB patients.
Collapse
Affiliation(s)
- Eugenio Cavalli
- IRCCS Centro Neurolesi Bonino Pulejo, C.da Casazza, 98124 Messina, Italy.
| | - Emanuela Mazzon
- IRCCS Centro Neurolesi Bonino Pulejo, C.da Casazza, 98124 Messina, Italy.
| | - Santa Mammana
- IRCCS Centro Neurolesi Bonino Pulejo, C.da Casazza, 98124 Messina, Italy.
| | - Maria Sofia Basile
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy.
| | - Salvo Danilo Lombardo
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy.
| | - Katia Mangano
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy.
| | - Placido Bramanti
- IRCCS Centro Neurolesi Bonino Pulejo, C.da Casazza, 98124 Messina, Italy.
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy.
| | - Paolo Fagone
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy.
| | | |
Collapse
|
8
|
Ssadh HA, Abdulmonem WA, Rasheed Z, Madar IH, Alhoderi J, Eldeen SKN, Alradhwan A, Alasmael N, Alkhamiss A, Fernández N. Knockdown of CD-74 in the Proliferative and Apoptotic Activity of Breast Cancer Cells. Open Access Maced J Med Sci 2019; 7:3169-3176. [PMID: 31949511 PMCID: PMC6953917 DOI: 10.3889/oamjms.2019.354] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 08/01/2019] [Accepted: 08/02/2019] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND The cluster of differentiation (CD) 74 is known for its immunological functions and its elevated level was reported in various cancer cells. AIM The aim of the present study was to investigate the expression and potential roles of CD74 in the proliferative and apoptotic activity of breast cancer. METHODS Expression of CD74, macrophage migration inhibitory factor (MIF) and CD44 was assayed in CAMA-1 and MDA-MB-231 cell lines using flow cytometry. CD74 was knocked down using CD74 siRNA-transfection in CAMA-1, and MDA-MB-231 cells and proliferation and apoptosis were determined in the transfected breast cancer cells. RESULTS The data showed that CD74, MIF and CD44 were expressed in breast cancer cell lines and were associated with cell proliferation and apoptosis. Correlation analysis revealed that CD74 was positively correlated and colocalised with MIF on the cell-surface of CAMA-1 and MDA-MB-231. The knockdown of CD74 significantly reduced CAMA-1 and MDA-MB-231 cell proliferation and increased the level of apoptotic cells. CONCLUSION We concluded that the interactions of CD74 with MIF and CD74 with CD44 could be a potential tumour marker for breast cancer cells. Moreover, the level of co-expression of MIF and CD74 or CD44 could be a surrogate marker for the efficacy of anti-angiogenic drugs, particularly in breast cancer tumours. In short, the study revealed the potential roles of CD74 in the proliferation and apoptosis of breast cancer which may serve as a potential therapeutic target for breast cancer.
Collapse
Affiliation(s)
- Hussain Al Ssadh
- School of Biological Sciences, University of Essex, Colchester, UK
| | - Waleed Al Abdulmonem
- Department of Pathology, College of Medicine, Qassim University, Qassim, Saudi Arabia
| | - Zafar Rasheed
- Department of Medical Biochemistry, College of Medicine, Qassim University, Saudi Arabia
| | - Inamul Hasan Madar
- Department of Biotechnology and Genetic Engineering, Bharathidasan University, Tiruchirappalli, India
| | - Jamila Alhoderi
- School of Biological Sciences, University of Essex, Colchester, UK
| | - Samah K Nasr Eldeen
- Clinical Laboratory Sciences, Inaya Medical College, Riyadh, Saudi Arabia.,Central Laboratories, Egyptian Ministry of Health, Tanta, Egypt
| | - Ali Alradhwan
- Biochemistry Department, College of Medicine, Imam Abdulrahman Bin Faisal University, Saudi Arabia
| | | | - Abdullah Alkhamiss
- Department of Pathology, College of Medicine, Qassim University, Qassim, Saudi Arabia
| | - Nelson Fernández
- School of Biological Sciences, University of Essex, Colchester, UK
| |
Collapse
|
9
|
Trivedi-Parmar V, Jorgensen WL. Advances and Insights for Small Molecule Inhibition of Macrophage Migration Inhibitory Factor. J Med Chem 2018; 61:8104-8119. [PMID: 29812929 DOI: 10.1021/acs.jmedchem.8b00589] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Macrophage migration inhibitory factor (MIF) is an upstream regulator of the immune response whose dysregulation is tied to a broad spectrum of inflammatory and proliferative disorders. As its complex signaling pathways and pleiotropic nature have been elucidated, it has become an attractive target for drug discovery. Remarkably, MIF is both a cytokine and an enzyme that functions as a keto-enol tautomerase. Strategies including in silico modeling, virtual screening, high-throughput screening, and screening of anti-inflammatory natural products have led to a large and diverse catalogue of MIF inhibitors as well as some understanding of the structure-activity relationships for compounds binding MIF's tautomerase active site. With possible clinical trials of some MIF inhibitors on the horizon, it is an opportune time to review the literature to seek trends, address inconsistencies, and identify promising new avenues of research.
Collapse
Affiliation(s)
- Vinay Trivedi-Parmar
- Department of Chemistry , Yale University , New Haven , Connecticut 06520-8107 , United States
| | - William L Jorgensen
- Department of Chemistry , Yale University , New Haven , Connecticut 06520-8107 , United States
| |
Collapse
|
10
|
Oxidized macrophage migration inhibitory factor is a potential new tissue marker and drug target in cancer. Oncotarget 2018; 7:73486-73496. [PMID: 27636991 PMCID: PMC5341993 DOI: 10.18632/oncotarget.11970] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 09/02/2016] [Indexed: 01/16/2023] Open
Abstract
Macrophage migration inhibitory factor (MIF) is a pleiotropic cytokine, which was shown to be upregulated in cancers and to exhibit tumor promoting properties. Unlike other cytokines, MIF is ubiquitously present in the circulation and tissue of healthy subjects. We recently described a previously unrecognized, disease-related isoform of MIF, designated oxMIF, which is present in the circulation of patients with different inflammatory diseases. In this article, we report that oxMIF is also linked to different solid tumors as it is specifically expressed in tumor tissue from patients with colorectal, pancreatic, ovarian and lung cancer. Furthermore, oxMIF can be specifically targeted by a subset of phage display-derived fully human, monoclonal anti-MIF antibodies (mAbs) that were shown to neutralize pro-tumorigenic activities of MIF in vivo. We further demonstrate that anti-oxMIF mAbs sensitize human cancer cell lines (LNCaP, PC3, A2780 and A2780ADR) to the action of cytotoxic drugs (mitoxantrone, cisplatin and doxorubicin) in vitro and in an A2780 xenograft mouse model of ovarian cancer. We conclude that oxMIF is the disease related isoform of MIF in solid tumors and a potential new diagnostic marker and drug target in cancer.
Collapse
|
11
|
D'Amato-Brito C, Cipriano D, Colin DJ, Germain S, Seimbille Y, Robert JH, Triponez F, Serre-Beinier V. Role of MIF/CD74 signaling pathway in the development of pleural mesothelioma. Oncotarget 2017; 7:11512-25. [PMID: 26883190 PMCID: PMC4905490 DOI: 10.18632/oncotarget.7314] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 01/24/2016] [Indexed: 11/25/2022] Open
Abstract
Macrophage migration inhibitory factor (MIF) is a pro-inflammatory cytokine implicated in acute and chronic inflammatory diseases. MIF is overexpressed in various tumors. It displays a number of functions that provide a direct link between the process of inflammation and tumor growth. Our group recently identified the MIF-receptor CD74 as an independent prognostic factor for overall survival in patients with malignant pleural mesothelioma. In the present study, we compared the levels of expression of MIF and CD74 in different human mesothelioma cell lines and investigated their physiopathological functions in vitro and in vivo. Human mesothelioma cells expressed more CD74 and secreted less MIF than non tumoral MeT5A cells, suggesting a higher sensitivity to MIF. In mesothelioma cells, high MIF levels were associated with a high multiplication rate of cells. In vitro, reduction of MIF or CD74 levels in both mesothelioma cell lines showed that the MIF/CD74 signaling pathway promoted tumor cell proliferation and protected MPM cells from apoptosis. Finally, mesothelioma cell lines expressing high CD74 levels had a low tumorigenic potential after xenogeneic implantation in athymic nude mice. All these data highlight the complexity of the MIF/CD74 signaling pathway in the development of mesothelioma.
Collapse
Affiliation(s)
- Cintia D'Amato-Brito
- Department of Thoracic and Endocrine Surgery, University Hospitals and University of Geneva, Geneva, Switzerland
| | - Davide Cipriano
- Department of Thoracic and Endocrine Surgery, University Hospitals and University of Geneva, Geneva, Switzerland
| | - Didier J Colin
- MicroPET/SPECT/CT Imaging Laboratory, Centre for BioMedical Imaging (CIBM), University Hospitals and University of Geneva, Geneva, Switzerland
| | - Stéphane Germain
- MicroPET/SPECT/CT Imaging Laboratory, Centre for BioMedical Imaging (CIBM), University Hospitals and University of Geneva, Geneva, Switzerland
| | - Yann Seimbille
- Cyclotron Unit, University Hospitals and University of Geneva, Geneva, Switzerland
| | - John H Robert
- Department of Thoracic and Endocrine Surgery, University Hospitals and University of Geneva, Geneva, Switzerland
| | - Frédéric Triponez
- Department of Thoracic and Endocrine Surgery, University Hospitals and University of Geneva, Geneva, Switzerland
| | - Véronique Serre-Beinier
- Department of Thoracic and Endocrine Surgery, University Hospitals and University of Geneva, Geneva, Switzerland
| |
Collapse
|
12
|
Maboni G, Davenport R, Sessford K, Baiker K, Jensen TK, Blanchard AM, Wattegedera S, Entrican G, Tötemeyer S. A Novel 3D Skin Explant Model to Study Anaerobic Bacterial Infection. Front Cell Infect Microbiol 2017; 7:404. [PMID: 28959685 PMCID: PMC5604072 DOI: 10.3389/fcimb.2017.00404] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 08/31/2017] [Indexed: 12/11/2022] Open
Abstract
Skin infection studies are often limited by financial and ethical constraints, and alternatives, such as monolayer cell culture, do not reflect many cellular processes limiting their application. For a more functional replacement, 3D skin culture models offer many advantages such as the maintenance of the tissue structure and the cell types present in the host environment. A 3D skin culture model can be set up using tissues acquired from surgical procedures or post slaughter, making it a cost effective and attractive alternative to animal experimentation. The majority of 3D culture models have been established for aerobic pathogens, but currently there are no models for anaerobic skin infections. Footrot is an anaerobic bacterial infection which affects the ovine interdigital skin causing a substantial animal welfare and financial impact worldwide. Dichelobacter nodosus is a Gram-negative anaerobic bacterium and the causative agent of footrot. The mechanism of infection and host immune response to D. nodosus is poorly understood. Here we present a novel 3D skin ex vivo model to study anaerobic bacterial infections using ovine skin explants infected with D. nodosus. Our results demonstrate that D. nodosus can invade the skin explant, and that altered expression of key inflammatory markers could be quantified in the culture media. The viability of explants was assessed by tissue integrity (histopathological features) and cell death (DNA fragmentation) over 76 h showing the model was stable for 28 h. D. nodosus was quantified in all infected skin explants by qPCR and the bacterium was visualized invading the epidermis by Fluorescent in situ Hybridization. Measurement of pro-inflammatory cytokines/chemokines in the culture media revealed that the explants released IL1β in response to bacteria. In contrast, levels of CXCL8 production were no different to mock-infected explants. The 3D skin model realistically simulates the interdigital skin and has demonstrated that D. nodosus invades the skin and triggered an early cellular inflammatory response to this bacterium. This novel model is the first of its kind for investigating an anaerobic bacterial infection.
Collapse
Affiliation(s)
- Grazieli Maboni
- School of Veterinary Medicine and Science, University of NottinghamNottingham, United Kingdom
| | - Rebecca Davenport
- School of Veterinary Medicine and Science, University of NottinghamNottingham, United Kingdom
| | - Kate Sessford
- School of Veterinary Medicine and Science, University of NottinghamNottingham, United Kingdom
| | - Kerstin Baiker
- School of Veterinary Medicine and Science, University of NottinghamNottingham, United Kingdom
| | - Tim K. Jensen
- National Veterinary Institute, Technical University of DenmarkCopenhagen, Denmark
| | - Adam M. Blanchard
- School of Veterinary Medicine and Science, University of NottinghamNottingham, United Kingdom
| | | | | | - Sabine Tötemeyer
- School of Veterinary Medicine and Science, University of NottinghamNottingham, United Kingdom
| |
Collapse
|
13
|
Alam S, Khan F. QSAR, docking, ADMET, and system pharmacology studies on tormentic acid derivatives for anticancer activity. J Biomol Struct Dyn 2017; 36:2373-2390. [PMID: 28705120 DOI: 10.1080/07391102.2017.1355846] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
To explore the anticancer compounds from tormentic acid derivatives, a quantitative structure-activity relationship (QSAR) model was developed by the multiple linear regression methods. The developed QSAR model yielded a high activity-descriptors relationship accuracy of 94% referred by regression coefficient (r2 = .94) and a high activity prediction accuracy of 91%. The QSAR study indicates that chemical descriptors, chiV5, T_T_Cl_7, T_2_T_4, SsCH3count, and Epsilon3 are significantly correlated with anticancer activity. This validated model was further been used for virtual screening and thus identification of new potential breast cancer inhibitors. Lipinski's rule of five, ADMET risk and synthetic accessibility are used to filter false positive hits. Filtered compounds were then docked to identify the possible target binding pocket, to obtain a set of aligned ligand poses and to prioritize the predicted active compounds. The scrutinized compounds, as well as their metabolites, were predicted and analyzed for different pharmacokinetics parameters such as absorption, distribution, metabolism, excretion, and toxicity. Finally, the top-ranked compound NB-12 was evaluated by system pharmacology approach. Later studied the metabolic networks, disease biomarker networks, pathway maps, drug-target networks and generate significant gene networks. The strategy applied in this research work may act as a framework for rational design of potential anticancer drugs.
Collapse
Affiliation(s)
- Sarfaraz Alam
- a Metabolic & Structural Biology Department , CSIR-Central Institute of Medicinal & Aromatic Plants , Lucknow , India.,b Academy of Scientific & Innovative Research (AcSIR), CSIR-CIMAP Campus , Lucknow , India
| | - Feroz Khan
- a Metabolic & Structural Biology Department , CSIR-Central Institute of Medicinal & Aromatic Plants , Lucknow , India.,b Academy of Scientific & Innovative Research (AcSIR), CSIR-CIMAP Campus , Lucknow , India.,c Skaggs School of Pharmacy & Pharmaceutical Sciences , University of California San Diego (UCSD) , San Diego , CA , USA
| |
Collapse
|
14
|
Macrophage migration inhibitory factor: A multifaceted cytokine implicated in multiple neurological diseases. Exp Neurol 2017; 301:83-91. [PMID: 28679106 DOI: 10.1016/j.expneurol.2017.06.021] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 06/06/2017] [Accepted: 06/21/2017] [Indexed: 12/12/2022]
Abstract
Macrophage migration inhibitory factor (MIF) is a conserved cytokine found as a homotrimer protein. It is found in a wide spectrum of cell types in the body including neuronal and non-neuronal cells. MIF is implicated in several biological processes; chemo-attraction, cytokine activity, and receptor binding, among other functions. More recently, a chaperone-like activity has been added to its repertoire. In this review, we focus on the implication of MIF in the central nervous system and peripheries, its role in neurological disorders, and the mechanisms by which MIF is regulated. Numerous studies have associated MIF with various disease settings. MIF plays an important role in advocating tumorigenic processes, Alzheimer's disease, and is also upregulated in autism-spectrum disorders and spinal cord injury where it contributes to the severity of the injured area. The protective effect of MIF has been reported in amyotrophic lateral sclerosis by its reduction of aggregated misfolded SOD1, subsequently reducing the severity of this disease. Interestingly, a protective as well as pathological role for MIF has been implicated in stroke and cerebral ischemia, as well as depression. Thus, the role of MIF in neurological disorders appears to be diverse with both beneficial and adversary effects. Furthermore, its modulation is rather complex and it is regulated by different proteins, either on a molecular or protein level. This complexity might be dependent on the pathophysiological context and/or cellular microenvironment. Hence, further clarification of its diverse roles in neurological pathologies is warranted to provide new mechanistic insights which may lead in the future to the development of therapeutic strategies based on MIF, to fight some of these neurological disorders.
Collapse
|
15
|
Wang C, Zhou X, Li W, Li M, Tu T, Ba X, Wu Y, Huang Z, Fan G, Zhou G, Wu S, Zhao J, Zhang J, Chen J. Macrophage migration inhibitory factor promotes osteosarcoma growth and lung metastasis through activating the RAS/MAPK pathway. Cancer Lett 2017. [PMID: 28642171 DOI: 10.1016/j.canlet.2017.06.011] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Emerging evidence suggests that the tumour microenvironment plays a critical role in osteosarcoma (OS) development. Thus, cytokine immunotherapy could be a novel strategy for OS treatment. In this study, we explored the role of macrophage migration inhibitory factor (MIF), an important cytokine in OS progression, and investigated the anti-tumour effects of targeting MIF in OS. The results showed that MIF significantly increased in the tissue and serum samples of OS patients and was associated with tumour size, pulmonary metastasis and the survival rate of OS patients. We verified a positive correlation between MIF and p-ERK1/2 in OS patients. The in vitro results indicated that MIF could activate the RAS/MAPK pathway in a time- and dose-dependent manner, thereby promoting cell proliferation and migration. Furthermore, shRNA targeting MIF significantly inhibited tumour growth and lung metastasis in a mouse xenograft model and orthotopic model of OS. Additionally, inhibition of MIF significantly enhanced the sensitivity of OS cells to cisplatin and doxorubicin. Our findings suggest that immunotherapy targeting MIF to block the RAS/MAPK kinase cascade may represent a feasible and promising approach for OS treatment.
Collapse
Affiliation(s)
- Chen Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Department of Orthopaedics, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210023, PR China
| | - Xing Zhou
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Department of Orthopaedics, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210023, PR China; Clinical School of Nanjing, Second Military Medical University, Nanjing 210002, PR China
| | - Wentao Li
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Department of Orthopaedics, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210023, PR China
| | - Mingyue Li
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Department of Orthopaedics, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210023, PR China
| | - Tingyue Tu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Department of Orthopaedics, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210023, PR China
| | - Ximing Ba
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Department of Orthopaedics, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210023, PR China
| | - Yinyu Wu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Department of Orthopaedics, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210023, PR China
| | - Zhen Huang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Department of Orthopaedics, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210023, PR China
| | - Gentao Fan
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Department of Orthopaedics, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210023, PR China; Clinical School of Nanjing, Second Military Medical University, Nanjing 210002, PR China
| | - Guangxin Zhou
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Department of Orthopaedics, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210023, PR China; Clinical School of Nanjing, Second Military Medical University, Nanjing 210002, PR China
| | - Sujia Wu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Department of Orthopaedics, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210023, PR China; Clinical School of Nanjing, Second Military Medical University, Nanjing 210002, PR China
| | - Jianning Zhao
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Department of Orthopaedics, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210023, PR China; Clinical School of Nanjing, Second Military Medical University, Nanjing 210002, PR China.
| | - Junfeng Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Department of Orthopaedics, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210023, PR China.
| | - Jiangning Chen
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Department of Orthopaedics, Jinling Hospital, School of Medicine, Nanjing University, Nanjing 210023, PR China.
| |
Collapse
|
16
|
Macrophage Migration Inhibitory Factor (MIF): Biological Activities and Relation with Cancer. Pathol Oncol Res 2016; 23:235-244. [DOI: 10.1007/s12253-016-0138-6] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 10/13/2016] [Indexed: 12/28/2022]
|
17
|
Kindt N, Journe F, Laurent G, Saussez S. Involvement of macrophage migration inhibitory factor in cancer and novel therapeutic targets. Oncol Lett 2016; 12:2247-2253. [PMID: 27698786 DOI: 10.3892/ol.2016.4929] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 05/16/2016] [Indexed: 12/18/2022] Open
Abstract
Macrophage migration inhibitory factor (MIF) was originally identified in 1966 by Bloom and Bennett as a pro-inflammatory cytokine involved in the inhibition of macrophage motility. Since then, studies have investigated the functional contribution of this pro-inflammatory cytokine in several immune diseases, including rheumatoid arthritis and lupus erythematous. Recently, MIF has been reported to be involved in a variety of neoplastic diseases. The present review discusses previous cancer research studies that have investigated the involvement of MIF in carcinogenesis, disease prognosis, tumor cell proliferation and invasion, and tumor-induced angiogenesis. Finally, potential therapeutic approaches based on the use of MIF antagonists and neutralizing antibodies are examined. The review concludes that MIF could be a good prognostic biomarker in several types of cancer, but also that the inhibition of MIF could represent a novel therapy against cancer.
Collapse
Affiliation(s)
- Nadège Kindt
- Laboratory of Anatomy and Cellular Biology, Faculty of Medicine and Pharmacy, University of Mons, Mons 7000, Belgium
| | - Fabrice Journe
- Laboratory of Anatomy and Cellular Biology, Faculty of Medicine and Pharmacy, University of Mons, Mons 7000, Belgium; Laboratory of Oncology and Experimental Surgery, Institut Jules Bordet, Free University of Brussels, Brussels 1000, Belgium
| | - Guy Laurent
- Laboratory of Histology, Faculty of Medicine and Pharmacy, University of Mons, Mons 7000, Belgium
| | - Sven Saussez
- Laboratory of Anatomy and Cellular Biology, Faculty of Medicine and Pharmacy, University of Mons, Mons 7000, Belgium; Department of Otorhinolaryngology, Faculty of Medicine, Free University of Brussels, Brussels 1000, Belgium
| |
Collapse
|
18
|
Zheng Y, Wang Q, Li T, Qian J, Lu Y, Li Y, Bi E, Reu F, Qin Y, Drazba J, Hsi E, Yang J, Cai Z, Yi Q. Role of Myeloma-Derived MIF in Myeloma Cell Adhesion to Bone Marrow and Chemotherapy Response. J Natl Cancer Inst 2016; 108:djw131. [PMID: 27381622 PMCID: PMC5241902 DOI: 10.1093/jnci/djw131] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 04/15/2016] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Multiple myeloma (MM) remains an incurable cancer characterized by accumulation of malignant plasma cells in the bone marrow (BM). The mechanism underlying MM homing to BM is poorly elucidated. METHODS The clinical significance of migration inhibitory factor (MIF) expression was examined by analyzing six independent gene expression profile databases of primary MM cells using the Student's t test and Kaplan-Meier test. Enzyme-linked immunosorbent assay was used to examine MIF expression. In vivo bioluminescent imaging was used to determine MM cell localization and treatment efficacy in human MM xenograft mouse models, with three to four mice per group. MM cell attachment to BM stromal cells (BMSCs) was monitored by cell adhesion assay. MIF regulation of the expression of adhesion molecules was determined by chromatin immunoprecipitation (ChIP) assay. Statistical tests were two-sided. RESULTS High levels of MIF were detected in MM BM (MIF level in BM plasma: healthy = 10.72 ± 5.788 ng/mL, n = 5; MM = 1811 ± 248.7 ng/mL, n = 10; P < .001) and associated with poor survival of patients (Kaplan-Meier test for MM OS: 87 MIF(high) patients, 86 MIF(low) patients, P = .02). Knocking down MIF impaired MM cell adhesion to BMSCs in vitro and led to formation of extramedullary tumors in SCID mice. MIF acted through surface receptor CXCR4 and adaptor COPS5 to regulate the expression of adhesion molecules ALCAM, ITGAV, and ITGB5 on MM cells. More importantly, MIF-deficient MM cells were sensitive to chemotherapy in vitro when cocultured with BMSCs and in vivo. MIF inhibitor 4-IPP sensitized MM cells to chemotherapy. CONCLUSIONS MIF is an important player and a novel therapeutic target in MM. Inhibiting MIF activity will sensitize MM cells to chemotherapy.
Collapse
MESH Headings
- Activated-Leukocyte Cell Adhesion Molecule/genetics
- Animals
- Antigens, Differentiation, B-Lymphocyte/genetics
- Antigens, Differentiation, B-Lymphocyte/metabolism
- Antineoplastic Agents, Alkylating/pharmacology
- Apoptosis/drug effects
- Autocrine Communication
- Bone Marrow/metabolism
- Bortezomib/pharmacology
- COP9 Signalosome Complex
- Cell Adhesion/drug effects
- Cell Adhesion/genetics
- Cell Line, Tumor
- Chemotaxis/genetics
- Coculture Techniques
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Female
- Gene Expression
- Gene Knockdown Techniques
- Heterografts
- Histocompatibility Antigens Class II/genetics
- Histocompatibility Antigens Class II/metabolism
- Humans
- Intracellular Signaling Peptides and Proteins/metabolism
- Intramolecular Oxidoreductases/antagonists & inhibitors
- Intramolecular Oxidoreductases/genetics
- Intramolecular Oxidoreductases/metabolism
- Macrophage Migration-Inhibitory Factors/antagonists & inhibitors
- Macrophage Migration-Inhibitory Factors/genetics
- Macrophage Migration-Inhibitory Factors/metabolism
- Melphalan/pharmacology
- Mesenchymal Stem Cells/metabolism
- Mice
- Mice, SCID
- Multiple Myeloma/drug therapy
- Multiple Myeloma/genetics
- Multiple Myeloma/metabolism
- Multiple Myeloma/pathology
- Neoplasm Transplantation
- Peptide Hydrolases/metabolism
- Plasma Cells/metabolism
- Pyrimidines/pharmacology
- RNA, Messenger/metabolism
- Receptors, CXCR4/genetics
- Receptors, CXCR4/metabolism
Collapse
Affiliation(s)
- Yuhuan Zheng
- Affiliations of authors: Department of Hematology and State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, West China Hospital, China (YZ); Department of Cancer Biology (YZ, QW, TL, JQ, YLu, YLi, EB, QY), Taussig Cancer Center (FR), Imaging Core Facility, Lerner Research Institute (JD), and Department of Pathology and Laboratory Medicine Institute (EH), Cleveland Clinic, Cleveland, OH; Department of Hematology, Zhejiang University, China (YLi, ZC); Department of Internal Medicine, Weiss Memory Hospital, Chicago, IL (YQ); Department of Lymphoma/Myeloma, MD Anderson Cancer Center, Houston, TX (JY)
| | - Qiang Wang
- Affiliations of authors: Department of Hematology and State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, West China Hospital, China (YZ); Department of Cancer Biology (YZ, QW, TL, JQ, YLu, YLi, EB, QY), Taussig Cancer Center (FR), Imaging Core Facility, Lerner Research Institute (JD), and Department of Pathology and Laboratory Medicine Institute (EH), Cleveland Clinic, Cleveland, OH; Department of Hematology, Zhejiang University, China (YLi, ZC); Department of Internal Medicine, Weiss Memory Hospital, Chicago, IL (YQ); Department of Lymphoma/Myeloma, MD Anderson Cancer Center, Houston, TX (JY)
| | - Tianshu Li
- Affiliations of authors: Department of Hematology and State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, West China Hospital, China (YZ); Department of Cancer Biology (YZ, QW, TL, JQ, YLu, YLi, EB, QY), Taussig Cancer Center (FR), Imaging Core Facility, Lerner Research Institute (JD), and Department of Pathology and Laboratory Medicine Institute (EH), Cleveland Clinic, Cleveland, OH; Department of Hematology, Zhejiang University, China (YLi, ZC); Department of Internal Medicine, Weiss Memory Hospital, Chicago, IL (YQ); Department of Lymphoma/Myeloma, MD Anderson Cancer Center, Houston, TX (JY)
| | - Jianfei Qian
- Affiliations of authors: Department of Hematology and State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, West China Hospital, China (YZ); Department of Cancer Biology (YZ, QW, TL, JQ, YLu, YLi, EB, QY), Taussig Cancer Center (FR), Imaging Core Facility, Lerner Research Institute (JD), and Department of Pathology and Laboratory Medicine Institute (EH), Cleveland Clinic, Cleveland, OH; Department of Hematology, Zhejiang University, China (YLi, ZC); Department of Internal Medicine, Weiss Memory Hospital, Chicago, IL (YQ); Department of Lymphoma/Myeloma, MD Anderson Cancer Center, Houston, TX (JY)
| | - Yong Lu
- Affiliations of authors: Department of Hematology and State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, West China Hospital, China (YZ); Department of Cancer Biology (YZ, QW, TL, JQ, YLu, YLi, EB, QY), Taussig Cancer Center (FR), Imaging Core Facility, Lerner Research Institute (JD), and Department of Pathology and Laboratory Medicine Institute (EH), Cleveland Clinic, Cleveland, OH; Department of Hematology, Zhejiang University, China (YLi, ZC); Department of Internal Medicine, Weiss Memory Hospital, Chicago, IL (YQ); Department of Lymphoma/Myeloma, MD Anderson Cancer Center, Houston, TX (JY)
| | - Yi Li
- Affiliations of authors: Department of Hematology and State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, West China Hospital, China (YZ); Department of Cancer Biology (YZ, QW, TL, JQ, YLu, YLi, EB, QY), Taussig Cancer Center (FR), Imaging Core Facility, Lerner Research Institute (JD), and Department of Pathology and Laboratory Medicine Institute (EH), Cleveland Clinic, Cleveland, OH; Department of Hematology, Zhejiang University, China (YLi, ZC); Department of Internal Medicine, Weiss Memory Hospital, Chicago, IL (YQ); Department of Lymphoma/Myeloma, MD Anderson Cancer Center, Houston, TX (JY)
| | - Enguang Bi
- Affiliations of authors: Department of Hematology and State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, West China Hospital, China (YZ); Department of Cancer Biology (YZ, QW, TL, JQ, YLu, YLi, EB, QY), Taussig Cancer Center (FR), Imaging Core Facility, Lerner Research Institute (JD), and Department of Pathology and Laboratory Medicine Institute (EH), Cleveland Clinic, Cleveland, OH; Department of Hematology, Zhejiang University, China (YLi, ZC); Department of Internal Medicine, Weiss Memory Hospital, Chicago, IL (YQ); Department of Lymphoma/Myeloma, MD Anderson Cancer Center, Houston, TX (JY)
| | - Frederic Reu
- Affiliations of authors: Department of Hematology and State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, West China Hospital, China (YZ); Department of Cancer Biology (YZ, QW, TL, JQ, YLu, YLi, EB, QY), Taussig Cancer Center (FR), Imaging Core Facility, Lerner Research Institute (JD), and Department of Pathology and Laboratory Medicine Institute (EH), Cleveland Clinic, Cleveland, OH; Department of Hematology, Zhejiang University, China (YLi, ZC); Department of Internal Medicine, Weiss Memory Hospital, Chicago, IL (YQ); Department of Lymphoma/Myeloma, MD Anderson Cancer Center, Houston, TX (JY)
| | - Yu Qin
- Affiliations of authors: Department of Hematology and State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, West China Hospital, China (YZ); Department of Cancer Biology (YZ, QW, TL, JQ, YLu, YLi, EB, QY), Taussig Cancer Center (FR), Imaging Core Facility, Lerner Research Institute (JD), and Department of Pathology and Laboratory Medicine Institute (EH), Cleveland Clinic, Cleveland, OH; Department of Hematology, Zhejiang University, China (YLi, ZC); Department of Internal Medicine, Weiss Memory Hospital, Chicago, IL (YQ); Department of Lymphoma/Myeloma, MD Anderson Cancer Center, Houston, TX (JY)
| | - Judy Drazba
- Affiliations of authors: Department of Hematology and State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, West China Hospital, China (YZ); Department of Cancer Biology (YZ, QW, TL, JQ, YLu, YLi, EB, QY), Taussig Cancer Center (FR), Imaging Core Facility, Lerner Research Institute (JD), and Department of Pathology and Laboratory Medicine Institute (EH), Cleveland Clinic, Cleveland, OH; Department of Hematology, Zhejiang University, China (YLi, ZC); Department of Internal Medicine, Weiss Memory Hospital, Chicago, IL (YQ); Department of Lymphoma/Myeloma, MD Anderson Cancer Center, Houston, TX (JY)
| | - Eric Hsi
- Affiliations of authors: Department of Hematology and State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, West China Hospital, China (YZ); Department of Cancer Biology (YZ, QW, TL, JQ, YLu, YLi, EB, QY), Taussig Cancer Center (FR), Imaging Core Facility, Lerner Research Institute (JD), and Department of Pathology and Laboratory Medicine Institute (EH), Cleveland Clinic, Cleveland, OH; Department of Hematology, Zhejiang University, China (YLi, ZC); Department of Internal Medicine, Weiss Memory Hospital, Chicago, IL (YQ); Department of Lymphoma/Myeloma, MD Anderson Cancer Center, Houston, TX (JY)
| | - Jing Yang
- Affiliations of authors: Department of Hematology and State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, West China Hospital, China (YZ); Department of Cancer Biology (YZ, QW, TL, JQ, YLu, YLi, EB, QY), Taussig Cancer Center (FR), Imaging Core Facility, Lerner Research Institute (JD), and Department of Pathology and Laboratory Medicine Institute (EH), Cleveland Clinic, Cleveland, OH; Department of Hematology, Zhejiang University, China (YLi, ZC); Department of Internal Medicine, Weiss Memory Hospital, Chicago, IL (YQ); Department of Lymphoma/Myeloma, MD Anderson Cancer Center, Houston, TX (JY)
| | - Zhen Cai
- Affiliations of authors: Department of Hematology and State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, West China Hospital, China (YZ); Department of Cancer Biology (YZ, QW, TL, JQ, YLu, YLi, EB, QY), Taussig Cancer Center (FR), Imaging Core Facility, Lerner Research Institute (JD), and Department of Pathology and Laboratory Medicine Institute (EH), Cleveland Clinic, Cleveland, OH; Department of Hematology, Zhejiang University, China (YLi, ZC); Department of Internal Medicine, Weiss Memory Hospital, Chicago, IL (YQ); Department of Lymphoma/Myeloma, MD Anderson Cancer Center, Houston, TX (JY)
| | - Qing Yi
- Affiliations of authors: Department of Hematology and State Key Laboratory of Biotherapy and Cancer Center, Sichuan University, West China Hospital, China (YZ); Department of Cancer Biology (YZ, QW, TL, JQ, YLu, YLi, EB, QY), Taussig Cancer Center (FR), Imaging Core Facility, Lerner Research Institute (JD), and Department of Pathology and Laboratory Medicine Institute (EH), Cleveland Clinic, Cleveland, OH; Department of Hematology, Zhejiang University, China (YLi, ZC); Department of Internal Medicine, Weiss Memory Hospital, Chicago, IL (YQ); Department of Lymphoma/Myeloma, MD Anderson Cancer Center, Houston, TX (JY)
| |
Collapse
|
19
|
Liu G, Xu Z, Hao D. MicroRNA‑451 inhibits neuroblastoma proliferation, invasion and migration by targeting macrophage migration inhibitory factor. Mol Med Rep 2016; 13:2253-60. [PMID: 26783235 DOI: 10.3892/mmr.2016.4770] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Accepted: 11/19/2015] [Indexed: 11/05/2022] Open
Abstract
Neuroblastoma (NB) is the most prevalent type of extracranial solid tumour in young children. To improve current understanding of the mechanisms, which modulate cancer cell proliferation, invasion and migration, investigations have focused on microRNAs (miRs), a class of small non‑coding RNAs, which post‑transcriptionally regulate gene expression during various crucial cell processes. The present study aimed to investigate the role of miR‑451 in NB. Human NB tissue and adjacent normal tissue were surgically removed, and the expression of miR‑451, and development and pathological characteristics of NB were investigated. The expression of miR‑451 was reduced in the NB tissue, compared with that in the adjacent tissue, and correlations between the reduction in miR‑451 and unfavourable variables included tumour size (P=0.0081), differentiation (P=0.0217), lymph node metastasis (P=0.0489), tumour‑node‑metastasis stage (0.0220) and distant metastases (P=0.0201). Transfection of the SK‑N‑SH and GI‑LA‑N NB cell lines with miR‑451 inhibited cell growth, invasion and migration. Furthermore, the present study demonstrated that macrophage migration inhibitory factor (MIF) was regulated directly by miR‑451 and was a critical mediator of the biological effects of miR‑451 in NB. The re‑expression of MIF markedly reversed the carcinogenic inhibitory property of miR‑451. These data provide a more detailed understanding of the essential role of miR‑451 in NB, which relies on regulation of the expression of MIF.
Collapse
Affiliation(s)
- Geng Liu
- Department of Hand Surgery, Hong‑Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710054, P.R. China
| | - Zhengwei Xu
- Department of Spine Surgery, Hong‑Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710054, P.R. China
| | - Dingjun Hao
- Department of Spine Surgery, Hong‑Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710054, P.R. China
| |
Collapse
|
20
|
HIF-α/MIF and NF-κB/IL-6 axes contribute to the recruitment of CD11b+Gr-1+ myeloid cells in hypoxic microenvironment of HNSCC. Neoplasia 2014; 16:168-79. [PMID: 24709424 DOI: 10.1593/neo.132034] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 01/14/2014] [Accepted: 01/27/2014] [Indexed: 02/05/2023]
Abstract
CD11b+Gr-1+ myeloid cells have gained much attention due to their roles in tumor immunity suppression as well as promotion of angiogenesis, invasion, and metastases. However, the mechanisms by which CD11b+Gr-1+ myeloid cells recruit to the tumor site have not been well clarified. In the present study, we showed that hypoxia could stimulate the migration of CD11b+Gr-1+ myeloid cells through increased production of macrophage migration inhibitory factor (MIF) and interleukin-6 (IL-6) by head and neck squamous cell carcinoma (HNSCC) cells. Hypoxia-inducible factor-1α (HIF-1α)- and HIF-2α-dependent MIF regulated chemotaxis, differentiation, and pro-angiogenic function of CD11b+Gr-1+ myeloid cells through binding to CD74/CXCR2, and CD74/CXCR4 complexes, and then activating p38/mitogen-activated protein kinase (MAPK) and phosphatidylinositide 3-kinases (PI3K)/AKT signaling pathways. Knockdown (KD) of HIF-1α and HIF-2α in HNSCC cells decreased MIF level but failed to inhibit the CD11b+Gr-1+ myeloid cell migration, because HIF-1α/2α KD enhanced nuclear factor κB (NF-κB) activity that increased IL-6 secretion. Simultaneously blocking NF-κB and HIF-1α/HIF-2α had better inhibitory effect on CD11b+Gr-1+ myeloid cell recruitment in the hypoxic zone than individually silencing HIF-1α/2α or NF-κB. In conclusion, the interaction between HIF-α/MIF and NF-κB/IL-6 axes plays an important role in the hypoxia-induced accumulation of CD11b+Gr-1+ myeloid cells and tumor growth in HNSCC.
Collapse
|
21
|
Abstract
Recent genomic and biological studies of neuroblastoma have shed light on the dramatic heterogeneity in the clinical behaviour of this disease, which spans from spontaneous regression or differentiation in some patients, to relentless disease progression in others, despite intensive multimodality therapy. This evidence also suggests several possible mechanisms to explain the phenomena of spontaneous regression in neuroblastomas, including neurotrophin deprivation, humoral or cellular immunity, loss of telomerase activity and alterations in epigenetic regulation. A better understanding of the mechanisms of spontaneous regression might help to identify optimal therapeutic approaches for patients with these tumours. Currently, the most druggable mechanism is the delayed activation of developmentally programmed cell death regulated by the tropomyosin receptor kinase A pathway. Indeed, targeted therapy aimed at inhibiting neurotrophin receptors might be used in lieu of conventional chemotherapy or radiation in infants with biologically favourable tumours that require treatment. Alternative approaches consist of breaking immune tolerance to tumour antigens or activating neurotrophin receptor pathways to induce neuronal differentiation. These approaches are likely to be most effective against biologically favourable tumours, but they might also provide insights into treatment of biologically unfavourable tumours. We describe the different mechanisms of spontaneous neuroblastoma regression and the consequent therapeutic approaches.
Collapse
Affiliation(s)
- Garrett M Brodeur
- Division of Oncology, The Children's Hospital of Philadelphia, 3501 Civic Center Boulevard, Philadelphia, PA 19104-4302, USA
| | - Rochelle Bagatell
- Division of Oncology, The Children's Hospital of Philadelphia, 3501 Civic Center Boulevard, Philadelphia, PA 19104-4302, USA
| |
Collapse
|
22
|
Bottino C, Dondero A, Bellora F, Moretta L, Locatelli F, Pistoia V, Moretta A, Castriconi R. Natural killer cells and neuroblastoma: tumor recognition, escape mechanisms, and possible novel immunotherapeutic approaches. Front Immunol 2014; 5:56. [PMID: 24575100 PMCID: PMC3921882 DOI: 10.3389/fimmu.2014.00056] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 01/30/2014] [Indexed: 12/19/2022] Open
Abstract
Neuroblastoma (NB) is the most common extra-cranial solid tumor of childhood and arises from developing sympathetic nervous system. Most primary tumors localize in the abdomen, the adrenal gland, or lumbar sympathetic ganglia. Amplification in tumor cells of MYCN, the major oncogenic driver, patients' age over 18 months, and the presence at diagnosis of a metastatic disease (stage IV, M) identify NB at high risk of treatment failure. Conventional therapies did not significantly improve the overall survival of these patients. Moreover, the limited landscape of somatic mutations detected in NB is hampering the development of novel pharmacological approaches. Major efforts aim to identify novel NB-associated surface molecules that activate immune responses and/or direct drugs to tumor cells and tumor-associated vessels. PVR (Poliovirus Receptor) and B7-H3 are promising targets, since they are expressed by most high-risk NB, are upregulated in tumor vasculature and are essential for tumor survival/invasiveness. PVR is a ligand of DNAM-1 activating receptor that triggers the cytolytic activity of natural killer (NK) cells against NB. In animal models, targeting of PVR with an attenuated oncolytic poliovirus induced tumor regression and elimination. Also B7-H3 was successfully targeted in preclinical studies and is now being tested in phase I/II clinical trials. B7-H3 down-regulates NK cytotoxicity, providing NB with a mechanism of escape from immune response. The immunosuppressive potential of NB can be enhanced by the release of soluble factors that impair NK cell function and/or recruitment. Among these, TGF-β1 modulates the cytotoxicity receptors and the chemokine receptor repertoire of NK cells. Here, we summarize the current knowledge on the main cell surface molecules and soluble mediators that modulate the function of NK cells in NB, considering the pros and cons that must be taken into account in the design of novel NK cell-based immunotherapeutic approaches.
Collapse
Affiliation(s)
- Cristina Bottino
- Dipartimento di Medicina Sperimentale, Università degli Studi di Genova , Genova , Italy ; Istituto Giannina Gaslini , Genova , Italy
| | - Alessandra Dondero
- Dipartimento di Medicina Sperimentale, Università degli Studi di Genova , Genova , Italy
| | - Francesca Bellora
- Dipartimento di Medicina Sperimentale, Università degli Studi di Genova , Genova , Italy
| | | | - Franco Locatelli
- Dipartimento di Onco-Ematologia Pediatrica, Ospedale Bambino Gesù , Roma , Italy ; Università di Pavia , Pavia , Italy
| | | | - Alessandro Moretta
- Dipartimento di Medicina Sperimentale, Università degli Studi di Genova , Genova , Italy ; Centro di Eccellenza per le Ricerche Biomediche, Università degli Studi di Genova , Genova , Italy
| | - Roberta Castriconi
- Dipartimento di Medicina Sperimentale, Università degli Studi di Genova , Genova , Italy ; Centro di Eccellenza per le Ricerche Biomediche, Università degli Studi di Genova , Genova , Italy
| |
Collapse
|
23
|
Wang D, Luo L, Chen W, Chen LZ, Zeng WT, Li W, Huang XH. Significance of the vascular endothelial growth factor and the macrophage migration inhibitory factor in the progression of hepatocellular carcinoma. Oncol Rep 2013; 31:1199-204. [PMID: 24366206 DOI: 10.3892/or.2013.2946] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 12/16/2013] [Indexed: 12/26/2022] Open
Abstract
The aim of the present study was to investigate the expression of vascular endothelial growth factor (VEGF) and macrophage migration inhibitory factor (MIF) in HCC progression and their correlation with clinicopathological factors as well as the relationship between their expression levels. The expression of serum VEGF and MIF was evaluated in 150 patients with HCC and in 30 normal volunteers by enzyme-linked immunosorbent assay (ELISA). VEGF and MIF expression levels were evaluated by immunohistochemistry on tissue microarrays containing 150 HCCs with paired adjacent non-cancer liver tissues. VEGF and MIF mRNA levels were determined by quantitative PCR in another 48 HCCs. The correlation of VEGF and MIF with clinicopathological factors was analyzed in HCC. Serum VEGF and MIF concentrations were higher in HCC patients than the levels in the controls. The expression levels of VEGF and MIF in the HCC tissues were both higher than those in the adjacent non-tumor liver tissues. Overexpression of VEGF and MIF was significantly associated with tumor size (P=0.027 and 0.022, respectively), intrahepatic metastasis (P=0.032 and 0.027, respectively), vascular invasion (P=0.044 and 0.039, respectively) and TNM stage (P=0.028 and 0.013, respectively). Furthermore, VEGF and MIF mRNA levels were higher in HCC compared to levels in the paired non-cancer liver tissues. VEGF and MIF mRNA levels were correlated with tumor stage and metastasis. The expression of VEGF was positively related with MIF expression in HCC. The expression of MIF and VEGF in HCC was markedly positively correlated, which suggests that MIF and VEGF play an important role in the progression of HCC. Both factors may concomitantly accelerate the progression of HCC.
Collapse
Affiliation(s)
- Dong Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, P.R. China
| | - Liang Luo
- Department of Medical Intensive Care Unit, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, P.R. China
| | - Wei Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, P.R. China
| | - Lian-Zhou Chen
- Department of General Surgical Laboratory, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, P.R. China
| | - Wen-Tao Zeng
- Department of General Surgical Laboratory, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, P.R. China
| | - Wen Li
- Department of General Surgical Laboratory, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, P.R. China
| | - Xiao-Hui Huang
- Department of General Surgical Laboratory, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, P.R. China
| |
Collapse
|
24
|
Choudhary S, Hegde P, Pruitt JR, Sielecki TM, Choudhary D, Scarpato K, Degraff DJ, Pilbeam CC, Taylor JA. Macrophage migratory inhibitory factor promotes bladder cancer progression via increasing proliferation and angiogenesis. Carcinogenesis 2013; 34:2891-9. [PMID: 23825153 DOI: 10.1093/carcin/bgt239] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Macrophage migratory inhibitory factor (MIF) is a proinflammatory cytokine shown to promote tumorigenesis. Using the N-butyl-N-(4-hydroxybutyl)-nitrosamine (BBN) model of bladder cancer, we previously showed that MIF knockout mice display decreased angiogenesis and invasion compared with wild-type. This study examines the role of MIF in bladder cancer via use of oral inhibitors of MIF. In vitro, high-grade bladder cancer cells were treated with recombinant human MIF +/- (rhMIF+/-) inhibitor. Measurements included cell counts, proliferation by (3)H-thymidine incorporation (TdR), extracellular signal-regulated kinase (ERK) phosphorylation by western blot analysis, messenger RNA (mRNA) expression by quantitative PCR and protein secretion by enzyme-linked immunosorbent assay. Treatment with rhMIF increased ERK phosphorylation, cell counts, TdR and mRNA expression and protein secretion of vascular endothelial growth factor, which were blocked by specific inhibitors of ERK and MIF. In vivo, 3-month-old male C57Bl/6 mice were given BBN for 22 and 16 weeks in study 1 and study 2, respectively. Mice (n = 8-10 per group) were gavaged with vehicle or doses of MIF inhibitors daily from weeks 16-22 in both studies. Average bladder weights, reflecting tumor mass, tumor stage/burden, mitotic rate and proliferation indices, and microvessel densities were reduced in inhibitor groups versus controls. In summary, MIF promotes bladder cancer via increasing cell proliferation and angiogenesis and oral inhibitors of MIF may prove useful in treatment of this disease.
Collapse
|
25
|
Abstract
Neuroblastoma is a solid tumour that arises from the developing sympathetic nervous system. Over the past decade, our understanding of this disease has advanced tremendously. The future challenge is to apply the knowledge gained to developing risk-based therapies and, ultimately, improving outcome. In this Review we discuss the key discoveries in the developmental biology, molecular genetics and immunology of neuroblastoma, as well as new translational tools for bringing these promising scientific advances into the clinic.
Collapse
Affiliation(s)
- Nai-Kong V. Cheung
- Department of Pediatrics, Memorial Sloan-Kettering Cancer Center, New York, NY 10065
| | - Michael A. Dyer
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN 38105
- Department of Ophthalmology, University of Tennessee Health Science Center, Memphis, TN 38163
- Howard Hughes Medical Institute, Chevy Chase, MD 20815
| |
Collapse
|
26
|
Babu SN, Chetal G, Kumar S. Macrophage migration inhibitory factor: a potential marker for cancer diagnosis and therapy. Asian Pac J Cancer Prev 2013; 13:1737-44. [PMID: 22901113 DOI: 10.7314/apjcp.2012.13.5.1737] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Macrophage migration inhibitory factor (MIF) is a pluripotent cytokine which plays roles in inflammation, immune responses and cancer development. It assists macrophages in carrying out functions like phagocytosis, adherence and motility. Of late, MIF is implicated in almost all stages of neoplasia and expression is a feature of most types of cancer. The presence of MIF in almost all tumors and all stages of cancer makes it an interesting candidate for cancer therapy. This review explores the roles of MIF in neoplasia.
Collapse
Affiliation(s)
- Spoorthy N Babu
- IGNOU-I2IT Centre of Excellence for Advanced Education and Research, Pune, Maharashtra, India
| | | | | |
Collapse
|
27
|
Huang Q, Zhao SL, Tian XY, Li B, Li Z. Increased co-expression of macrophage migration inhibitory factor and matrix metalloproteinase 9 is associated with tumor recurrence of meningioma. Int J Med Sci 2013; 10:276-85. [PMID: 23372434 PMCID: PMC3558716 DOI: 10.7150/ijms.5185] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Accepted: 12/27/2012] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND AND OBJECTIVE We detected the expression of MIF and matrix metalloproteinase 9 (MMP9) in meningiomas to determine whether they are valuable recurrence predictor for meningioma. METHODS 67 cases of meningiomas, including 57 benign tumors (WHO grade I) and 10 non-benign tumors (WHO grade II and III), were collected, and expression of MIF and MMP9 in tissue microarray was evaluated immunohistochemically. The correlations between immunostainings and clinicopathological parameters, as well as the follow-up data of patients, were analyzed statistically. RESULTS Increased expressions of both MIF (58.2%, 39/67) and MMP9 (55.2%, 37/67) were significantly associated with microvessel density (MVD) of tumor, but only dual high-expression of MIF and MMP9 was in relation to tumor invasion (P=0.016) and tumor recurrence (P=0.001). Based on univariate analysis, histological grade, tumor invasion and co-expression of MIF and MMP9 were significant predictors for recurrence. However, only histological grade and co-expression of MIF and MMP9 in tumor were independent recurrence factors with a hazard ratio of 49.033 (P=0.002) and 37.766 (P=0.002) in multivariate analysis. CONCLUSIONS Together with histological grade, increased co-expression of MIF and MMP9 in tumor might be a valuable predictor for recurrence, especially for benign meningiomas.
Collapse
Affiliation(s)
- Quan Huang
- Department of Neurosurgery, The First Affiliated Hospital, Sun Yat-sen University. 58 Zhongshan Road II, Guangzhou 510080, China
| | | | | | | | | |
Collapse
|
28
|
Abstract
During evolution, beta cells adapted to a sole aim: the production and stimulus-dependent secretion of insulin. This acquired specificity was accompanied by a loss of protection mechanisms predisposing beta cell to a high vulnerability. Among beta cell-damaging molecules, a new one has been identified recently: macrophage migration inhibitory factor (MIF). MIF was at first designated as a T-cell product that inhibits random movement of macrophages. Over the years, the number of functions attributed to this protein increased significantly, positioning MIF at the top of inflammatory cascade in the combat against infection and in immunoinflammatory and autoimmune diseases. This exceptionally versatile molecule regulates insulin secretion in physiological conditions, while in pathological states it alters beta cell function and induces their apoptosis or necrosis and affects beta cell neoplasia.
Collapse
|
29
|
Girard E, Strathdee C, Trueblood E, Quéva C. Macrophage migration inhibitory factor produced by the tumour stroma but not by tumour cells regulates angiogenesis in the B16-F10 melanoma model. Br J Cancer 2012; 107:1498-505. [PMID: 22955855 PMCID: PMC3493755 DOI: 10.1038/bjc.2012.392] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Background: Macrophage migration inhibitory factor (MIF) has been proposed as a link between inflammation and tumorigenesis. Despite its potentially broad influence in tumour biology and prevalent expression, the value of MIF as a therapeutic target in cancer remains unclear. We sought to validate MIF in tumour models by achieving a complete inhibition of its expression in tumour cells and in the tumour stroma. Methods: We used MIF shRNA-transduced B16-F10 melanoma cells implanted in wild-type and MIF−/− C57Bl6 mice to investigate the effect of loss of MIF on tumour growth. Cytokine detection and immunohistochemistry (IHC) were used to evaluate tumours ex vivo. Results: Macrophage migration inhibitory factor shRNA inhibited expression of MIF protein by B16-F10 melanoma cells in vitro and in vivo. In vitro, the loss of MIF in this cell line resulted in a decreased response to hypoxia as indicated by reduced expression of VEGF. In vivo the growth of B16-F10 tumours was inhibited by an average of 47% in the MIF−/− mice compared with wild-type but was unaffected by loss of MIF expression by the tumour cells. Immunohistochemistry analysis revealed that microvessel density was decreased in tumours implanted in the MIF−/− mice. Profiling of serum cytokines showed a decrease in pro-angiogenic cytokines in MIF−/− mice. Conclusion: We report that the absence of MIF in the host resulted in slower tumour growth, which was associated with reduced vascularity. While the major contribution of MIF appeared to be in the regulation of angiogenesis, tumour cell-derived MIF played a negligible role in this process.
Collapse
Affiliation(s)
- E Girard
- Hematology Oncology Research Area, Amgen Inc., Seattle, WA, USA
| | | | | | | |
Collapse
|
30
|
Morgenstern DA, Anderson J. Inflammation: what role in pediatric cancer? Pediatr Blood Cancer 2012; 58:659-64. [PMID: 22162439 DOI: 10.1002/pbc.24008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2011] [Accepted: 10/24/2011] [Indexed: 01/22/2023]
Abstract
There is growing evidence for the importance of chronic inflammation in the pathogenesis of adult cancers and for an ongoing role of the inflammatory response in tumor growth and metastasis. Here, we examine how these processes relate to pediatric malignancies. While it is unlikely that chronic inflammation plays a significant role in driving malignant progression in childhood tumors that typically have developmental origins, the inflammatory response does appear to play an important role in the development and progression of many types of childhood cancer. An enhanced understanding of these processes will be of critical importance in developing novel therapeutic strategies.
Collapse
Affiliation(s)
- Daniel A Morgenstern
- Molecular Haematology and Cancer Biology Unit, Institute of Child Health, London, UK.
| | | |
Collapse
|
31
|
Wang X, Chen T, Leng L, Fan J, Cao K, Duan Z, Zhang X, Shao C, Wu M, Tadmori I, Li T, Liang L, Sun D, Zheng S, Meinhardt A, Young W, Bucala R, Ren Y. MIF produced by bone marrow-derived macrophages contributes to teratoma progression after embryonic stem cell transplantation. Cancer Res 2012; 72:2867-78. [PMID: 22461508 DOI: 10.1158/0008-5472.can-11-3247] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Although stem cell therapy holds promise as a potential treatment in a number of diseases, the tumorigenicity of embryonic stem cells (ESC) and induced pluripotent stem cells remains a major obstacle. In vitro predifferentiation of ESCs can help prevent the risk of teratoma formation, yet proliferating neural progenitors can generate tumors, especially in the presence of immunosuppressive therapy. In this study, we investigated the effects of the microenvironment on stem cell growth and teratoma development using undifferentiated ESCs. Syngeneic ESC transplantation triggered an inflammatory response that involved the recruitment of bone marrow (BM)-derived macrophages. These macrophages differentiated into an M2 or angiogenic phenotype that expressed multiple angiogenic growth factors and proteinases, such as macrophage migration inhibitory factor (MIF), VEGF, and matrix metalloproteinase 9, creating a microenvironment that supported the initiation of teratoma development. Genetic deletion of MIF from the host but not from ESCs specifically reduced angiogenesis and teratoma growth, and MIF inhibition effectively reduced teratoma development after ESC transplantation. Together, our findings show that syngeneic ESC transplantation provokes an inflammatory response that involves the rapid recruitment and activation of BM-derived macrophages, which may be a crucial driving force in the initiation and progression of teratomas.
Collapse
Affiliation(s)
- Xi Wang
- W. M. Keck Center for Collaborative Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Pistoia V, Bianchi G, Borgonovo G, Raffaghello L. Cytokines in neuroblastoma: from pathogenesis to treatment. Immunotherapy 2012; 3:895-907. [PMID: 21751957 DOI: 10.2217/imt.11.80] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Cytokines released by cancer cells or by cells of the tumor microenvironment stimulate angiogenesis, act as autocrine or paracrine growth factors for malignant cells, promote tumor cell migration and metastasis or create an immunosuppressive microenvironment. These tumor-promoting effects of cytokines also apply to neuroblastoma (NB), a pediatric neuroectodermal malignancy with frequent metastatic presentation at diagnosis and poor prognosis. IL-6 and VEGF are the best characterized cytokines that stimulated tumor growth and metastasis, while others such as IFN-γ can exert anti-NB activity by inducing tumor cell apoptosis and inhibiting angiogenesis. On the other hand, cytokines are part of the anti-NB therapeutic armamentarium, as exemplified by IL-2 and granulocyte-macrophage colony stimulating factor that potentiate the activity of anti-NB antibodies. These recent results raise hope for more efficacious treatment of this ominous pediatric malignancy.
Collapse
Affiliation(s)
- Vito Pistoia
- Laboratory of Oncology, G Gaslini Institute, Largo G Gaslini 5, 16148 Genova, Italy.
| | | | | | | |
Collapse
|
33
|
Fimognari C, Turrini E, Ferruzzi L, Lenzi M, Hrelia P. Natural isothiocyanates: genotoxic potential versus chemoprevention. Mutat Res 2011; 750:107-131. [PMID: 22178957 DOI: 10.1016/j.mrrev.2011.12.001] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Revised: 12/01/2011] [Accepted: 12/02/2011] [Indexed: 12/12/2022]
Abstract
Isothiocyanates, occurring in many dietary cruciferous vegetables, show interesting chemopreventive activities against several chronic-degenerative diseases, including cancer, cardiovascular diseases, neurodegeneration, diabetes. The electrophilic carbon residue in the isothiocyanate moiety reacts with biological nucleophiles and modification of proteins is recognized as a key mechanism underlying the biological activity of isothiocyanates. The nuclear factor-erythroid-2-related factor 2 system, which orchestrates the expression of a wide array of antioxidant genes, plays a role in the protective effect of isothiocyanates against almost all the pathological conditions reported above. Recent emerging findings suggest a further common mechanism. Chronic inflammation plays a central role in many human diseases and isothiocyanates inhibit the activity of many inflammation components, suppress cyclooxygenase 2, and irreversibly inactivate the macrophage migration inhibitory factor. Due to their electrophilic reactivity, some isothiocyanates are able to form adducts with DNA and induce gene mutations and chromosomal aberrations. DNA damage has been demonstrated to be involved in the pathogenesis of various chronic-degenerative diseases of epidemiological relevance. Thus, the genotoxicity of the isothiocyanates should be carefully considered. In addition, the dose-response relationship for genotoxic compounds does not suggest evidence of a threshold. Thus, chemicals that are genotoxic pose a greater potential risk to humans than non-genotoxic compounds. Dietary consumption levels of isothiocyanates appear to be several orders of magnitude lower than the doses used in the genotoxicity studies and thus it is highly unlikely that such toxicities would occur in humans. However, the beneficial properties of isothiocyanates stimulated an increase of dietary supplements and functional foods with highly enriched isothiocyanate concentrations on the market. Whether such concentrations may exert a potential health risk cannot be excluded with certainty and an accurate evaluation of the toxicological profile of isothiocyanates should be prompted before any major increase in their consumption be recommended or their clinical use suggested.
Collapse
Affiliation(s)
- Carmela Fimognari
- Department of Pharmacology, University of Bologna, via Irnerio 48, 40126 Bologna, Italy.
| | - Eleonora Turrini
- Department of Pharmacology, University of Bologna, via Irnerio 48, 40126 Bologna, Italy
| | - Lorenzo Ferruzzi
- Department of Pharmacology, University of Bologna, via Irnerio 48, 40126 Bologna, Italy
| | - Monia Lenzi
- Department of Pharmacology, University of Bologna, via Irnerio 48, 40126 Bologna, Italy
| | - Patrizia Hrelia
- Department of Pharmacology, University of Bologna, via Irnerio 48, 40126 Bologna, Italy
| |
Collapse
|
34
|
Verissimo CS, Molenaar JJ, Fitzsimons CP, Vreugdenhil E. Neuroblastoma therapy: what is in the pipeline? Endocr Relat Cancer 2011; 18:R213-31. [PMID: 21971288 DOI: 10.1530/erc-11-0251] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Despite the expansion of knowledge about neuroblastoma (NB) in recent years, the therapeutic outcome for children with a high-risk NB has not significantly improved. Therefore, more effective therapies are needed. This might be achieved by aiming future efforts at recently proposed but not yet developed targets for NB therapy. In this review, we discuss the recently proposed molecular targets that are in clinical trials and, in particular, those that are not yet explored in the clinic. We focus on the selection of these molecular targets for which promising in vitro and in vivo results have been obtained by silencing/inhibiting them. In addition, these selected targets are involved at least in one of the NB tumorigenic processes: proliferation, anti-apoptosis, angiogenesis and/or metastasis. In particular, we will review a recently proposed target, the microtubule-associated proteins (MAPs) encoded by doublecortin-like kinase gene (DCLK1). DCLK1-derived MAPs are crucial for proliferation and survival of neuroblasts and are highly expressed not only in NB but also in other tumours such as gliomas. Additionally, we will discuss neuropeptide Y, its Y2 receptor and cathepsin L as examples of targets to decrease angiogenesis and metastasis of NB. Furthermore, we will review the micro-RNAs that have been proposed as therapeutic targets for NB. Detailed investigation of these not yet developed targets as well as exploration of multi-target approaches might be the key to a more effective NB therapy, i.e. increasing specificity, reducing toxicity and avoiding long-term side effects.
Collapse
Affiliation(s)
- Carla S Verissimo
- Division of Medical Pharmacology, Leiden/Amsterdam Center for Drug Research, Leiden University Medical Center, Gorlaeus Laboratories, The Netherlands
| | | | | | | |
Collapse
|
35
|
Truitt L, Freywald A. Dancing with the dead: Eph receptors and their kinase-null partners. Biochem Cell Biol 2011; 89:115-29. [PMID: 21455264 DOI: 10.1139/o10-145] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Eph receptor tyrosine kinases and their ligands, ephrins, are membrane proteins coordinating a wide range of biological functions both in developing embryos and in adult multicellular organisms. Numerous studies have implicated Eph receptors in the induction of opposing responses, including cell adhesion or repulsion, support or inhibition of cell proliferation and cell migration, and progression or suppression of multiple malignancies. Similar to other receptor tyrosine kinases, Eph receptors rely on their ability to catalyze tyrosine phosphorylation for signal transduction. Interestingly, however, Eph receptors also actively utilize three kinase-deficient receptor tyrosine kinases, EphB6, EphA10, and Ryk, in their signaling network. The accumulating evidence suggests that the unusual flexibility of the Eph family, allowing it to initiate antagonistic responses, might be partially explained by the influence of the kinase-dead participants and that the exact outcome of an Eph-mediated action is likely to be defined by the balance between the signaling of catalytically potent and catalytically null receptors. We discuss in this minireview the emerging functions of the kinase-dead EphB6, EphA10, and Ryk receptors both in normal biological responses and in malignancy, and analyze currently available information related to the molecular mechanisms of their action in the context of the Eph family.
Collapse
Affiliation(s)
- Luke Truitt
- Department of Chemistry and Biochemistry, University of Regina, Sasketchewan, Canada
| | | |
Collapse
|
36
|
Dumitru CA, Gholaman H, Trellakis S, Bruderek K, Dominas N, Gu X, Bankfalvi A, Whiteside TL, Lang S, Brandau S. Tumor-derived macrophage migration inhibitory factor modulates the biology of head and neck cancer cells via neutrophil activation. Int J Cancer 2011; 129:859-69. [DOI: 10.1002/ijc.25991] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2010] [Accepted: 01/27/2011] [Indexed: 11/09/2022]
|
37
|
Li YH, Chen MHC, Gong HY, Hu SY, Li YW, Lin GH, Lin CC, Liu W, Wu JL. Progranulin A-mediated MET signaling is essential for liver morphogenesis in zebrafish. J Biol Chem 2010; 285:41001-9. [PMID: 20961855 DOI: 10.1074/jbc.m110.138743] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The mechanism that regulates embryonic liver morphogenesis remains elusive. Progranulin (PGRN) is postulated to play a critical role in regulating pathological liver growth. Nevertheless, the exact regulatory mechanism of PGRN in relation to its functional role in embryonic liver development remains to be elucidated. In our study, the knockdown of progranulin A (GrnA), an orthologue of mammalian PGRN, using antisense morpholinos resulted in impaired liver morphogenesis in zebrafish (Danio rerio). The vital role of GrnA in hepatic outgrowth and not in liver bud formation was further confirmed using whole-mount in situ hybridization markers. In addition, a GrnA deficiency was also found to be associated with the deregulation of MET-related genes in the neonatal liver using a microarray analysis. In contrast, the decrease in liver size that was observed in grnA morphants was avoided when ectopic MET expression was produced by co-injecting met mRNA and grnA morpholinos. This phenomenon suggests that GrnA might play a role in liver growth regulation via MET signaling. Furthermore, our study has shown that GrnA positively modulates hepatic MET expression both in vivo and in vitro. Therefore, our data have indicated that GrnA plays a vital role in embryonic liver morphogenesis in zebrafish. As a result, a novel link between PGRN and MET signaling is proposed.
Collapse
Affiliation(s)
- Yen-Hsing Li
- Molecular and Biological Agricultural Sciences Program, Taiwan International Graduate Program, National Chung Hsing University and Academia Sinica, Taipei 115, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Cates JMM, Friedman DB, Seeley EH, Dupont WD, Schwartz HS, Holt GE, Caprioli RM, Young PP. Proteomic analysis of osteogenic sarcoma: association of tumour necrosis factor with poor prognosis. Int J Exp Pathol 2010; 91:335-49. [PMID: 20353421 DOI: 10.1111/j.1365-2613.2010.00711.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
A significant proportion of patients with osteogenic sarcoma die from lung metastasis within 5 years of diagnosis. Molecular signatures that predict pulmonary metastasis from primary osteogenic sarcoma and identify those patients at risk would be clinically useful as prognostic markers. Protein expression profiles of two clonally related murine osteogenic sarcoma cell lines with low (K12) and high (K7M2) metastatic potential were compared using two different proteomic technologies, two-dimensional difference gel electrophoresis and cell profiling by matrix-assisted laser desorption/ionization mass spectrometry. Interrogation of a molecular pathways network database suggested several additional candidate molecules that potentially predict metastatic potential of primary osteogenic sarcoma. Two such proteins, macrophage migration inhibitory factor and tumour necrosis factor were selected for further validation studies. Western blots confirmed increased expression of both cytokines in K7M2 cells compared to K12 cells. Levels of migration inhibitory factor and tumour necrosis factor were semi-quantitatively measured in human osteogenic sarcoma samples by immunohistochemistry and were correlated with clinicopathologic parameters and patient outcomes. Multivariate survival analysis demonstrated that tumour necrosis factor expression in chemotherapy naïve osteogenic sarcoma is an independent prognostic factor for overall and metastasis-free survival. No significant differences in adverse outcomes were observed based on macrophage migration inhibitory factor expression.
Collapse
Affiliation(s)
- Justin M M Cates
- Department of Pathology, Vanderbilt University Medical Center, Nashville, TN, USA.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Port M, Wang Y, Schmelz HU, Pottek T, Meineke V, Ruf C, Abend M. A gene signature of primary tumor identifies metastasized seminoma. Urol Oncol 2009; 29:764-73. [PMID: 19945308 DOI: 10.1016/j.urolonc.2009.08.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2009] [Revised: 08/09/2009] [Accepted: 08/10/2009] [Indexed: 10/20/2022]
Abstract
BACKGROUND The aim of this study was the prediction of metastatic status in seminoma based on examination of the primary tumor. METHODS Total RNA was isolated from metastasized seminoma (n = 10, T1N1-2M0), non-metastasized seminoma (n = 21, T1-3N0M0), and corresponding normal tissues. Pooled RNA from 10 biopsies of each tissue type was hybridized on whole genome microarrays for screening purposes. Ninety-two selected gene candidates were quantitatively examined using real-time quantitative polymerase chain reaction (RTQ-PCR). RESULTS Agreement in gene expression was 88% between the whole genome microarrays and RTQ-PCR. Metastasized seminoma showed 1,912 up-regulated and 2,179 down-regulated genes with ≥ 2-fold differences in gene expression compared non-metastasized seminoma. RTQ-PCR of selected genes showed that mean gene expression values were significantly reduced in metastasized compared with non-metastasized seminoma. The presence of metastases could be predicted based on an 85-gene expression signature by using logistic regression. Sensitivity and accuracy of the 10-fold cross-validation model were 77.8% and 84.2%, respectively. CONCLUSION A logistic regression model using an 85 gene expression signature allowed identification of metastasized seminoma from the primary tumor with a sensitivity of 77.8%.
Collapse
Affiliation(s)
- Matthias Port
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | | | | | | | | | | | | |
Collapse
|
40
|
Nutrient isothiocyanates covalently modify and inhibit the inflammatory cytokine macrophage migration inhibitory factor (MIF). Biochem J 2009; 423:315-21. [PMID: 19723024 DOI: 10.1042/bj20091170] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Dietary ITCs (isothiocyanates) prevent cancer and show other bioactivities in vivo. As electrophiles, ITCs may covalently modify cellular proteins. Using a novel proteomics screen, we identified MIF (macrophage migration inhibitory factor) as the principal target of nutrient ITCs in intact cells. ITCs covalently modify the N-terminal proline residue of MIF and extinguish its catalytic tautomerase activity. MIF deficiency does not prevent induction of Phase 2 gene expression, a hallmark of many cancer chemopreventives, including ITCs. Due to the emerging role of MIF in the control of malignant cell growth and its clear involvement in inflammation, inhibition of MIF by nutrient ITCs suggests therapeutic strategies for inflammatory diseases and cancer.
Collapse
|
41
|
Lim S, Choong LY, Kuan CP, Yunhao C, Lim YP. Regulation of macrophage inhibitory factor (MIF) by epidermal growth factor receptor (EGFR) in the MCF10AT model of breast cancer progression. J Proteome Res 2009; 8:4062-76. [PMID: 19530702 DOI: 10.1021/pr900430n] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Genetic aberration of EGFR is one of the major molecular characteristics of breast cancer. However, the molecular changes associated with EGFR signaling during different stages of breast cancer development have not been studied. In this study, complementary two-dimensional-DIGE and iTRAQ technologies were used to profile the expression level of proteins in 4 isogenic cell lines in the MCF10AT model of breast cancer progression following a time course of EGF stimulation. A total of 80 proteins (67 from iTRAQ, 15 from DIGE, 2 common in both) were identified to be up- or down-regulated by EGF treatment. Following EGF stimulation, the expression level of MIF, a cytokine that has been implicated in many human cancers, was decreased in MCF10A1 normal breast mammary epithelial cells, increased in MCF10AT1k preneoplastic and MCF10CA1h low grade breast cancer cells, but showed no obvious difference in the MCF10CA1a high grade cancer cells. The increase in MIF expression level following EGF treatment could also be observed in A431 cervical cancer cells. EGF-induced increases of MIF expression levels in CA1h breast cancer cells were abrogated when MEK, but not PIK3CA, was knocked down. In addition, silencing of MIF diminished the proliferation of EGF-stimulated CA1h cells when compared to control cells. Taken together, our data suggested an EGFR --> MEK --> MIF proliferative pathway that has never been reported previously and that this pathway "evolves" during disease progression as modeled by the MCF10AT system. Revelation of the novel relationship between MIF and EGF may contribute to an integrated understanding of the roles of these oncogenic factors during breast cancer development.
Collapse
Affiliation(s)
- Simin Lim
- Cancer Science Institute of Singapore, National University of Singapore, Department of Biological Sciences, Singapore
| | | | | | | | | |
Collapse
|
42
|
Piette C, Deprez M, Roger T, Noël A, Foidart JM, Munaut C. The dexamethasone-induced inhibition of proliferation, migration, and invasion in glioma cell lines is antagonized by macrophage migration inhibitory factor (MIF) and can be enhanced by specific MIF inhibitors. J Biol Chem 2009; 284:32483-92. [PMID: 19759012 DOI: 10.1074/jbc.m109.014589] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Glioblastomas (GBMs) are the most frequent and malignant brain tumors in adults. Glucocorticoids (GCs) are routinely used in the treatment of GBMs for their capacity to reduce the tumor-associated edema. Few in vitro studies have suggested that GCs inhibit the migration and invasion of GBM cells through the induction of MAPK phosphatase 1 (MKP-1). Macrophage migration inhibitory factor (MIF), an endogenous GC antagonist is up-regulated in GBMs. Recently, MIF has been involved in tumor growth and migration/invasion and specific MIF inhibitors have been developed on their capacity to block its enzymatic tautomerase activity site. In this study, we characterized several glioma cell lines for their MIF production. U373 MG cells were selected for their very low endogenous levels of MIF. We showed that dexamethasone inhibits the migration and invasion of U373 MG cells, through a glucocorticoid receptor (GR)- dependent inhibition of the ERK1/2 MAPK pathway. Oppositely, we found that exogenous MIF increases U373 MG migration and invasion through the stimulation of the ERK1/2 MAP kinase pathway and that this activation is CD74 independent. Finally, we used the Hs 683 glioma cells that are resistant to GCs and produce high levels of endogenous MIF, and showed that the specific MIF inhibitor ISO-1 could restore dexamethasone sensitivity in these cells. Collectively, our results indicate an intricate pathway between MIF expression and GC resistance. They suggest that MIF inhibitors could increase the response of GBMs to corticotherapy.
Collapse
Affiliation(s)
- Caroline Piette
- Laboratory of Tumor and Developmental Biology, GIGA-R, CHU, Liège, Belgium
| | | | | | | | | | | |
Collapse
|
43
|
Lugrin J, Ding XC, Le Roy D, Chanson AL, Sweep FCGJ, Calandra T, Roger T. Histone deacetylase inhibitors repress macrophage migration inhibitory factor (MIF) expression by targeting MIF gene transcription through a local chromatin deacetylation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2009; 1793:1749-58. [PMID: 19747950 DOI: 10.1016/j.bbamcr.2009.09.007] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2009] [Revised: 08/30/2009] [Accepted: 09/03/2009] [Indexed: 12/20/2022]
Abstract
The cytokine macrophage migration inhibitory factor plays a central role in inflammation, cell proliferation and tumorigenesis. Moreover, macrophage migration inhibitory factor levels correlate with tumor aggressiveness and metastatic potential. Histone deacetylase inhibitors are potent antitumor agents recently introduced in the clinic. Therefore, we hypothesized that macrophage migration inhibitory factor would represent a target of histone deacetylase inhibitors. Confirming our hypothesis, we report that histone deacetylase inhibitors of various chemical classes strongly inhibited macrophage migration inhibitory factor expression in a broad range of cell lines, in primary cells and in vivo. Nuclear run on, transient transfection with macrophage migration inhibitory factor promoter reporter constructs and transduction with macrophage migration inhibitory factor expressing adenovirus demonstrated that trichostatin A (a prototypical histone deacetylase inhibitor) inhibited endogenous, but not episomal, MIF gene transcription. Interestingly, trichostatin A induced a local and specific deacetylation of macrophage migration inhibitory factor promoter-associated H3 and H4 histones which did not affect chromatin accessibility but was associated with an impaired recruitment of RNA polymerase II and Sp1 and CREB transcription factors required for basal MIF gene transcription. Altogether, this study describes a new molecular mechanism by which histone deacetylase inhibitors inhibit MIF gene expression, and suggests that macrophage migration inhibitory factor inhibition by histone deacetylase inhibitors may contribute to the antitumorigenic effects of histone deacetylase inhibitors.
Collapse
Affiliation(s)
- Jérôme Lugrin
- Infectious Diseases Service, Department of Medicine, Centre Hospitalier Universitaire Vaudois and University of Lausanne, CH-1011 Lausanne, Switzerland
| | | | | | | | | | | | | |
Collapse
|
44
|
Bach JP, Deuster O, Balzer-Geldsetzer M, Meyer B, Dodel R, Bacher M. The role of macrophage inhibitory factor in tumorigenesis and central nervous system tumors. Cancer 2009; 115:2031-40. [PMID: 19326434 DOI: 10.1002/cncr.24245] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Macrophage migration inhibitory factor (MIF) has been described as a protein that plays an important role in both innate and acquired immunity. Further research has shown that MIF plays a particularly critical part in cell cycle regulation and therefore in tumorigenesis as well. Over the past few years, the significance of the role of MIF in a variety of both solid and hematologic tumors has been established. More recently, interest has increased in the role of MIF in the development of central nervous system (CNS) tumors, in which it appears to influence cell cycle control. In addition, MIF has been identified as an essential actor in metastasis and angiogenesis. Vascular growth factor concentration raises because of increased levels of MIF in brain tumors. Recently, the MIF receptor complex has been described, and it appears that this may be a suitable drug target for treatment of brain tumors. In light of these findings, the authors chose to conduct a systematic search for information regarding MIF that has been published within the past 15 years using the terms "inflammation," "glioblastoma," "brain tumor," "astrocytoma," "microglia," "glioblastoma," "immune system and brain tumors," "glioblastoma and MIF," and "brain tumor and MIF." The aim of this article was thus to present a detailed review of current knowledge regarding the role of MIF in CNS tumor pathophysiology.
Collapse
Affiliation(s)
- Jan-Philipp Bach
- Department of Neurology, Philipps-University Marburg, Marburg, Germany
| | | | | | | | | | | |
Collapse
|
45
|
Liu YH, Lin CY, Lin WC, Tang SW, Lai MK, Lin JY. Up-regulation of vascular endothelial growth factor-D expression in clear cell renal cell carcinoma by CD74: a critical role in cancer cell tumorigenesis. THE JOURNAL OF IMMUNOLOGY 2009; 181:6584-94. [PMID: 18941249 DOI: 10.4049/jimmunol.181.9.6584] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Elevation of CD74 is associated with a number of human cancers, including clear cell renal cell carcinoma (ccRCC). To understand the role of CD74 in the oncogenic process of ccRCC, we ectopically expressed CD74 in human embryonic kidney 293 cells (HEK/CD74) and evaluated its oncogenic potential. Through overexpression of CD74 in HEK293 and Caki-2 cells and down-regulation of CD74 in Caki-1 cells, we show that vascular endothelial growth factor-D (VEGF-D) expression is modified accordingly. A significant, positive correlation between CD74 and VEGF-D is found in human ccRCC tissues (Pearson's correlation, r = 0.65, p < 0.001). In HEK/CD74 xenograft mice, CD74 significantly induced the formation of tumor masses, increased tumor-induced angiogenesis, and promoted cancer cell metastasis. Blockage of VEGF-D expression by small interference RNA resulted in a decrease in cell proliferation, invasion, and cancer cell-induced HUVEC migration enhanced by CD74. Furthermore, we provide evidence that the intracellular signaling cascade responsible for VEGF-D up-regulation by CD74 is both PI3K/AKT- and MEK/ERK-dependent, both of which are associated with NF-kappaB nuclear translocation and DNA-binding activity. These results suggest that VEGF-D is crucial for CD74-induced human renal carcinoma cancer cell tumorigenesis.
Collapse
Affiliation(s)
- Yu-Huei Liu
- Graduate Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | | | | | | | | | | |
Collapse
|
46
|
Arscott WT, LaBauve AE, May V, Wesley UV. Suppression of neuroblastoma growth by dipeptidyl peptidase IV: relevance of chemokine regulation and caspase activation. Oncogene 2008; 28:479-91. [PMID: 18978811 PMCID: PMC2633428 DOI: 10.1038/onc.2008.402] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Imbalanced protease expression and activities may contribute to the development of cancers, including neuroblastoma (NB). NB is a fatal childhood cancer of the sympathetic nervous system that frequently overexpresses mitogenic peptides, chemokines and their receptors. Dipeptidyl peptidase IV (DPPIV), a cell surface serine protease, inactivates or degrades some of these bioactive peptides and chemokines, thereby regulating cell proliferation and survival. Our studies show that DPPIV is expressed in normal neural crest-derived structures, including superior cervical and dorsal root ganglion cells, sciatic nerve, and in adrenal glands, but its expression is greatly decreased or lost in cells derived from NB, their malignant counterpart. Restoration of DPPIV expression in NB cells led to their differentiation in association with increased expression of the neural marker MAP2 and decreased expression of chemokines, including stromal-derived factor 1 (SDF1) and its receptor CXCR4. Furthermore, DPPIV promoted apoptosis, and inhibited SDF1-mediated in vitro cell migration and angiogenic potential. These changes were accompanied by caspase activation and decreased levels of phospho-Akt and MMP9 activity, which are downstream effectors of SDF1-CXCR4 signaling. Importantly, DPPIV suppressed the tumorigenic potential of NB cells in a xenotransplantation mouse model. These data support a potential role for DPPIV in inhibiting NB growth and progression.
Collapse
Affiliation(s)
- W T Arscott
- Department of Microbiology and Molecular Genetics, Vermont Cancer Center, University of Vermont, Burlington, VT 05405, USA
| | | | | | | |
Collapse
|
47
|
Coleman AM, Rendon BE, Zhao M, Qian MW, Bucala R, Xin D, Mitchell RA. Cooperative regulation of non-small cell lung carcinoma angiogenic potential by macrophage migration inhibitory factor and its homolog, D-dopachrome tautomerase. THE JOURNAL OF IMMUNOLOGY 2008; 181:2330-7. [PMID: 18684922 DOI: 10.4049/jimmunol.181.4.2330] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Tumor-derived growth factors and cytokines stimulate neoangiogenesis from surrounding capillaries to support tumor growth. Recent studies have revealed that macrophage migration inhibitory factor (MIF) expression is increased in lung cancer, particularly non-small cell lung carcinomas (NSCLC). Because MIF has important autocrine effects on normal and transformed cells, we investigated whether autocrine MIF and its only known family member, D-dopachrome tautomerase (D-DT), promote the expression of proangiogenic factors CXCL8 and vascular endothelial growth factor in NSCLC cells. Our results demonstrate that the expression of CXCL8 and vascular endothelial growth factor are strongly reliant upon both the individual and cooperative activities of the two family members. CXCL8 transcriptional regulation by MIF and D-DT appears to involve a signaling pathway that includes the activation of JNK, c-jun phosphorylation, and subsequent AP-1 transcription factor activity. Importantly, HUVEC migration and tube formation induced by supernatants from lung adenocarcinoma cells lacking either or both MIF and D-DT are substantially reduced when compared with normal supernatants. Finally, we demonstrate that the cognate MIF receptor, CD74, is necessary for both MIF- and D-DT-induced JNK activation and CXCL8 expression, suggesting its potential involvement in angiogenic growth factor expression. This is the first demonstration of a biological role for D-DT, and its synergism with MIF suggests that the combined therapeutic targeting of both family members may enhance current anti-MIF-based therapies.
Collapse
Affiliation(s)
- Arlixer M Coleman
- Microbiology and Immunology Program, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Ceccarelli J, Delfino L, Zappia E, Castellani P, Borghi M, Ferrini S, Tosetti F, Rubartelli A. The redox state of the lung cancer microenvironment depends on the levels of thioredoxin expressed by tumor cells and affects tumor progression and response to prooxidants. Int J Cancer 2008; 123:1770-8. [PMID: 18661523 DOI: 10.1002/ijc.23709] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Here we report that human nonsmall cell lung carcinomas overexpress macrophage migration inhibitory factor (MIF) and thioredoxin (Trx), 2 oxidoreductases with cytokine function, and contain more abundant nonprotein thiols (glutathione and cysteine) than nonneoplastic lung tissues. Cell clones derived from the same lung carcinoma cell lines but expressing different levels of Trx and/or MIF displayed growth rates in vitro and in vivo correlating with Trx but not with MIF. Interestingly, the different clones generate extracellularly reduced nonprotein thiols, in amounts related to the Trx content and inhibited by inhibitors of Trx function. Each clone also showed distinct responses to the prooxidant compound arsenic trioxide. Cells with a strongly antioxidant and aggressive phenotype were more susceptible to the cytotoxic effect of the drug than cells expressing little Trx. The latter counteracted the oxidative stress by increasing Trx expression and thiol release. Together these results indicate that different human lung cancer cell lines have distinct redox properties defined by the levels of Trx and nonprotein thiols, the higher antioxidant phenotype correlating with the higher aggressiveness. Moreover, the redox phenotype dictates their response to prooxidant drugs and must be taken into account when therapeutic interventions with redox active substances are considered.
Collapse
Affiliation(s)
- Jenny Ceccarelli
- Cell Biology Unit, National Cancer Research Institute, Genoa, Italy
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Zhou Q, Yan X, Gershan J, Orentas RJ, Johnson BD. Expression of macrophage migration inhibitory factor by neuroblastoma leads to the inhibition of antitumor T cell reactivity in vivo. THE JOURNAL OF IMMUNOLOGY 2008; 181:1877-86. [PMID: 18641325 DOI: 10.4049/jimmunol.181.3.1877] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Neuroblastomas and many other solid tumors produce high amounts of macrophage migration inhibitory factor (MIF), which appears to play a role in tumor progression. We found that MIF expression in neuroblastoma inhibits T cell proliferation in vitro, raising the possibility that MIF promotes tumorigenesis, in part, by suppressing antitumor immunity. To examine whether tumor-derived MIF leads to suppression of T cell immunity in vivo, we generated MIF-deficient neuroblastoma cell lines using short hairpin small interfering RNAs (siRNA). The MIF knockdown (MIFKD) AGN2a neuroblastoma cells were more effectively rejected in immune-competent mice than control siRNA-transduced or wild-type AGN2a. However, the increased rejection of MIFKD AGN2a was not observed in T cell-depleted mice. MIFKD tumors had increased infiltration of CD8(+) and CD4(+) T cells, as well as increased numbers of macrophages, dendritic cells, and B cells. Immunization with MIFKD AGN2a cells significantly increased protection against tumor challenge as compared with immunization with wild-type AGN2a, and the increased protection correlated with elevated frequencies of tumor-reactive CD8(+) T cells in the lymphoid tissue of treated animals. Increased numbers of infiltrating tumor-reactive CD8(+) T cells were also observed at the site of tumor vaccination. In vitro, treatment of AGN2a-derived culture supernatants with neutralizing MIF-specific Ab failed to reverse T cell suppressive activity, suggesting that MIF is not directly responsible for the immune suppression in vivo. This supports a model whereby MIF expression in neuroblastoma initiates a pathway that leads to the suppression of T cell immunity in vivo.
Collapse
Affiliation(s)
- Qiang Zhou
- Department of Pediatrics, Medical College of Wisconsin, and the Children's Research Institute, Children's Hospital of Wisconsin, Milwaukee, WI 53226, USA
| | | | | | | | | |
Collapse
|
50
|
Krockenberger M, Dombrowski Y, Weidler C, Ossadnik M, Hönig A, Häusler S, Voigt H, Becker JC, Leng L, Steinle A, Weller M, Bucala R, Dietl J, Wischhusen J. Macrophage migration inhibitory factor contributes to the immune escape of ovarian cancer by down-regulating NKG2D. THE JOURNAL OF IMMUNOLOGY 2008; 180:7338-48. [PMID: 18490733 DOI: 10.4049/jimmunol.180.11.7338] [Citation(s) in RCA: 131] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The proinflammatory cytokine macrophage migration inhibitory factor (MIF) stimulates tumor cell proliferation, migration, and metastasis; promotes tumor angiogenesis; suppresses p53-mediated apoptosis; and inhibits antitumor immunity by largely unknown mechanisms. We here describe an overexpression of MIF in ovarian cancer that correlates with malignancy and the presence of ascites. Functionally, we find that MIF may contribute to the immune escape of ovarian carcinoma by transcriptionally down-regulating NKG2D in vitro and in vivo which impairs NK cell cytotoxicity toward tumor cells. Together with the additional tumorigenic properties of MIF, this finding provides a rationale for novel small-molecule inhibitors of MIF to be used for the treatment of MIF-secreting cancers.
Collapse
|