1
|
Cheng T, Mao M, Liu Y, Xie L, Shi F, Liu H, Li X. The potential therapeutic effect of human umbilical cord mesenchymal stem cell-derived exosomes in bronchopulmonary dysplasia. Life Sci 2024; 357:123047. [PMID: 39260518 DOI: 10.1016/j.lfs.2024.123047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 08/25/2024] [Accepted: 09/07/2024] [Indexed: 09/13/2024]
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic lung disease of preterm infants, with its incidence rising due to improved survival rates of these infants. BPD results from a combination of prenatal and postnatal factors, such as mechanical ventilation, oxygen toxicity, and infections, all of which significantly impact the prognosis and growth of affected infants. Current treatment options for BPD are largely supportive and do not address the underlying pathology. Exosomes are cell-derived bilayer-enclosed membrane structures enclosing proteins, lipids, RNAs, growth factors, cytokines and metabolites. They have become recognized as crucial regulators of intercellular communication in various physiological and pathological processes. Previous studies have revealed the therapeutic potential of human umbilical cord mesenchymal stem cells-derived exosomes (HUCMSCs-Exos) in promoting tissue repair and regeneration. Therefore, HUCMSCs-Exos maybe a promising and effective therapeutic modality for BPD. In this review, we firstly provide a comprehensive overview of BPD, including its etiology and the mechanisms of lung injury. Then we detail the isolation, characterization, and contents of HUCMSCs-Exos, and discuss their potential mechanisms of HUCMSCs-Exos in BPD treatment. Additionally, we summarize current clinical trials and discuss the challenges in translating these findings from bench to bedside. This review aims to lay the groundwork for future clinical applications of HUCMSCs-Exos in treating BPD.
Collapse
Affiliation(s)
- Tianyu Cheng
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China; NHC Key Laboratory of Chronobiology (Sichuan University), Chengdu, China; The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, China; Sichuan Birth Defects Clinical Research Center, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Min Mao
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China; NHC Key Laboratory of Chronobiology (Sichuan University), Chengdu, China; The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, China; Sichuan Birth Defects Clinical Research Center, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yang Liu
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China; NHC Key Laboratory of Chronobiology (Sichuan University), Chengdu, China; The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, China; Sichuan Birth Defects Clinical Research Center, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Liang Xie
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China; NHC Key Laboratory of Chronobiology (Sichuan University), Chengdu, China; The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, China; Sichuan Birth Defects Clinical Research Center, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Fang Shi
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China; NHC Key Laboratory of Chronobiology (Sichuan University), Chengdu, China; The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, China; Sichuan Birth Defects Clinical Research Center, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Hanmin Liu
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China; NHC Key Laboratory of Chronobiology (Sichuan University), Chengdu, China; The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, China; Sichuan Birth Defects Clinical Research Center, West China Second University Hospital, Sichuan University, Chengdu, China.
| | - Xin Li
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, China; Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China; NHC Key Laboratory of Chronobiology (Sichuan University), Chengdu, China; The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu, China; Sichuan Birth Defects Clinical Research Center, West China Second University Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
2
|
Valdez CN, Sánchez-Zuno GA, Bucala R, Tran TT. Macrophage Migration Inhibitory Factor (MIF) and D-Dopachrome Tautomerase (DDT): Pathways to Tumorigenesis and Therapeutic Opportunities. Int J Mol Sci 2024; 25:4849. [PMID: 38732068 PMCID: PMC11084905 DOI: 10.3390/ijms25094849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Discovered as inflammatory cytokines, MIF and DDT exhibit widespread expression and have emerged as critical mediators in the response to infection, inflammation, and more recently, in cancer. In this comprehensive review, we provide details on their structures, binding partners, regulatory mechanisms, and roles in cancer. We also elaborate on their significant impact in driving tumorigenesis across various cancer types, supported by extensive in vitro, in vivo, bioinformatic, and clinical studies. To date, only a limited number of clinical trials have explored MIF as a therapeutic target in cancer patients, and DDT has not been evaluated. The ongoing pursuit of optimal strategies for targeting MIF and DDT highlights their potential as promising antitumor candidates. Dual inhibition of MIF and DDT may allow for the most effective suppression of canonical and non-canonical signaling pathways, warranting further investigations and clinical exploration.
Collapse
Affiliation(s)
- Caroline Naomi Valdez
- School of Medicine, Yale University, 333 Cedar St., New Haven, CT 06510, USA; (C.N.V.); (R.B.)
| | - Gabriela Athziri Sánchez-Zuno
- Section of Rheumatology, Allergy and Immunology, Department of Internal Medicine, Yale University, 333 Cedar St., New Haven, CT 06510, USA;
| | - Richard Bucala
- School of Medicine, Yale University, 333 Cedar St., New Haven, CT 06510, USA; (C.N.V.); (R.B.)
- Section of Rheumatology, Allergy and Immunology, Department of Internal Medicine, Yale University, 333 Cedar St., New Haven, CT 06510, USA;
- Yale Cancer Center, Yale University, 333 Cedar St., New Haven, CT 06510, USA
| | - Thuy T. Tran
- School of Medicine, Yale University, 333 Cedar St., New Haven, CT 06510, USA; (C.N.V.); (R.B.)
- Yale Cancer Center, Yale University, 333 Cedar St., New Haven, CT 06510, USA
- Section of Medical Oncology, Department of Internal Medicine, Yale University, 333 Cedar St., New Haven, CT 06510, USA
| |
Collapse
|
3
|
Shi K, Li Y, Yang L, Zhang Z, Guo D, Zhang J, Lu Y. Profiling transcriptional heterogeneity of epithelium, fibroblasts, and immune cells in esophageal squamous cell carcinoma by single-cell RNA sequencing. FASEB J 2022; 36:e22620. [PMID: 36260317 DOI: 10.1096/fj.202200898r] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 08/31/2022] [Accepted: 10/10/2022] [Indexed: 11/11/2022]
Abstract
Esophageal squamous cell carcinoma (ESCC) is one of the most aggressive malignancies with complex tumor microenvironment (TME) which has been proven to be associated with therapeutic failure or resistance. A deeper understanding of the complex TME and cellular heterogeneity is urgently needed in ESCC. Here, we generated single-cell RNA sequencing (scRNA-seq) of 25 796 immune and 8197 non-immune cells from three primary tumor and paired normal samples in ESCC patients. The results revealed intratumoral and intertumoral epithelium heterogeneity and tremendously differences in tumor and normal epithelium. The infiltration of myofibroblasts, one subtype of fibroblasts, might play important roles in the progression of ESCC. We also found that some differentially expressed genes and markers in epithelium and fibroblast subtypes showed prognostic values for ESCC. Diverse cell subtypes of T cells and myeloid cells were identified, including tumor-enriched HAVCR2+ CD4+ T cells with significantly exhausted signature. The epithelium and myeloid cells had more frequent cell-cell communication compared with epithelium and T cells. Taken together, this study provided in-depth insights into the cellular heterogeneity of TME in ESCC and highlighted potential therapeutic targets including for immunotherapy.
Collapse
Affiliation(s)
- Ke Shi
- Department of Biochemistry and Medical Genetics, Henan Medical College, Zhengzhou, China
| | - Yueqi Li
- Department of Clinical Medicine, School of Medicine, Zhengzhou University, Zhengzhou, China
| | - Liang Yang
- Department of Biochemistry and Medical Genetics, Henan Medical College, Zhengzhou, China
| | - Zichao Zhang
- Linyi Health School of Shandong Province, Linyi, China.,Department of Thoracic Surgery, Linyi People's Hospital, Linyi, China
| | - Dan Guo
- Department of Biochemistry and Medical Genetics, Henan Medical College, Zhengzhou, China
| | - Jinzhong Zhang
- Department of Biochemistry and Medical Genetics, Henan Medical College, Zhengzhou, China
| | - Yucheng Lu
- Biobank of Linyi People's Hospital, Linyi, China.,Central Laboratory of Linyi People's Hospital, Linyi, China
| |
Collapse
|
4
|
Thiele M, Donnelly SC, Mitchell RA. OxMIF: a druggable isoform of macrophage migration inhibitory factor in cancer and inflammatory diseases. J Immunother Cancer 2022; 10:e005475. [PMID: 36180072 PMCID: PMC9528626 DOI: 10.1136/jitc-2022-005475] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/12/2022] [Indexed: 11/04/2022] Open
Abstract
Macrophage migration inhibitory factor (MIF) is a proinflammatory cytokine with a pleiotropic spectrum of biological functions implicated in the pathogenesis of cancer and inflammatory diseases. MIF is constitutively present in several cell types and non-lymphoid tissues and is secreted after acute stress or inflammation. MIF triggers the release of proinflammatory cytokines, overrides the anti-inflammatory effects of glucocorticoids, and exerts chemokine function, resulting in increased migration and recruitment of leukocytes into inflamed tissue. Despite this, MIF is a challenging target for therapeutic intervention because of its ubiquitous nature and presence in the circulation and tissue of healthy individuals. Oxidized MIF (oxMIF) is an immunologically distinct disease-related structural isoform found in the plasma and tissues of patients with inflammatory diseases and in solid tumor tissues. MIF converts to oxMIF in an oxidizing, inflammatory environment. This review discusses the biology and activity of MIF and the potential for autoimmune disease and cancer modification by targeting oxMIF. Anti-oxMIF antibodies reduce cancer cell invasion/migration, angiogenesis, proinflammatory cytokine production, and ERK and AKT activation. Anti-oxMIF antibodies also elicit apoptosis and alter immune cell function and/or migration. When co-administered with a glucocorticoid, anti-oxMIF antibodies produced a synergistic response in inflammatory models. Anti-oxMIF antibodies therefore counterregulate biological activities attributed to MIF. oxMIF expression has been observed in inflammatory diseases (eg, sepsis, psoriasis, asthma, inflammatory bowel disease, and systemic lupus erythematosus) and oxMIF has been detected in ovarian, colorectal, lung, and pancreatic cancers. In contrast to MIF, oxMIF is specifically detected in plasma and/or tissues of diseased patients, but not in healthy individuals. Therefore, as a druggable isoform of MIF, oxMIF represents a potential new therapeutic target in inflammatory diseases and cancer. Fully human, monoclonal anti-oxMIF antibodies have been shown to selectively bind oxMIF in preclinical and phase I studies; however, additional clinical assessments are necessary to validate their use as either a monotherapy or in combination with standard-of-care regimens (ie, immunomodulatory agents/checkpoint inhibitors, anti-angiogenic drugs, chemotherapeutics, and glucocorticoids).
Collapse
Affiliation(s)
- Michael Thiele
- Biology Research, OncoOne Research & Development GmbH, Vienna, Austria
| | - Seamas C Donnelly
- Department of Medicine, Tallaght University Hospital & Trinity College Dublin, Dublin, Ireland
| | - Robert A Mitchell
- Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, Kentucky, USA
- Department of Surgery, J.G. Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA
- Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky, USA
- Division of Immunotherapy, Department of Surgery, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
5
|
Zhang Z, Zhou X, Guo J, Zhang F, Qian Y, Wang G, Duan M, Wang Y, Zhao H, Yang Z, Liu Z, Jiang X. TA-MSCs, TA-MSCs-EVs, MIF: their crosstalk in immunosuppressive tumor microenvironment. J Transl Med 2022; 20:320. [PMID: 35842634 PMCID: PMC9287873 DOI: 10.1186/s12967-022-03528-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 07/08/2022] [Indexed: 11/24/2022] Open
Abstract
As an important component of the immunosuppressive tumor microenvironment (TME), it has been established that mesenchymal stem cells (MSCs) promote the progression of tumor cells. MSCs can directly promote the proliferation, migration, and invasion of tumor cells via cytokines and chemokines, as well as promote tumor progression by regulating the functions of anti-tumor immune and immunosuppressive cells. MSCs-derived extracellular vesicles (MSCs-EVs) contain part of the plasma membrane and signaling factors from MSCs; therefore, they display similar effects on tumors in the immunosuppressive TME. The tumor-promoting role of macrophage migration inhibitory factor (MIF) in the immunosuppressive TME has also been revealed. Interestingly, MIF exerts similar effects to those of MSCs in the immunosuppressive TME. In this review, we summarized the main effects and related mechanisms of tumor-associated MSCs (TA-MSCs), TA-MSCs-EVs, and MIF on tumors, and described their relationships. On this basis, we hypothesized that TA-MSCs-EVs, the MIF axis, and TA-MSCs form a positive feedback loop with tumor cells, influencing the occurrence and development of tumors. The functions of these three factors in the TME may undergo dynamic changes with tumor growth and continuously affect tumor development. This provides a new idea for the targeted treatment of tumors with EVs carrying MIF inhibitors.
Collapse
Affiliation(s)
- Zhenghou Zhang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiangyu Zhou
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jinshuai Guo
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Fusheng Zhang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yiping Qian
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Guang Wang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Meiqi Duan
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yutian Wang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Haiying Zhao
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Zhi Yang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Zunpeng Liu
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China.
| | - Xiaofeng Jiang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
6
|
Sumaiya K, Langford D, Natarajaseenivasan K, Shanmughapriya S. Macrophage migration inhibitory factor (MIF): A multifaceted cytokine regulated by genetic and physiological strategies. Pharmacol Ther 2021; 233:108024. [PMID: 34673115 DOI: 10.1016/j.pharmthera.2021.108024] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 10/12/2021] [Accepted: 10/14/2021] [Indexed: 02/08/2023]
Abstract
Macrophage migration inhibitory factor (MIF) is a proinflammatory cytokine encoded within a functionally polymorphic genetic locus. MIF was initially recognized as a cytokine generated by activated T cells, but in recent days it has been identified as a multipotent key cytokine secreted by many other cell types involved in immune response and physiological processes. MIF is a highly conserved 12.5 kDa secretory protein that is involved in numerous biological processes. The expression and secretion profile of MIF suggests that MIF to be ubiquitously and constitutively expressed in almost all mammalian cells and is vital for numerous physiological processes. MIF is a critical upstream mediator of host innate and adaptive immunity and survival pathways resulting in the clearance of pathogens thus playing a protective role during infectious diseases. On the other hand, MIF being an immune modulator accelerates detrimental inflammation, promotes cancer metastasis and progression, thus worsening disease conditions. Several reports demonstrated that genetic and physiological factors, including MIF gene polymorphisms, posttranslational regulations, and receptor binding control the functional activities of MIF. Taking into consideration the multi-faceted role of MIF both in physiology and pathology, we thought it is timely to review and summarize the expressional and functional regulation of MIF, its functional mechanisms associated with its beneficial and pathological roles, and MIF-targeting therapies. Thus, our review will provide an overview on how MIF is regulated, its response, and the potency of the therapies that target MIF.
Collapse
Affiliation(s)
- Krishnamoorthi Sumaiya
- Medical Microbiology Laboratory, Department of Microbiology, Centre for Excellence in Life Sciences, Bharathidasan University, Tiruchirappalli 620 024, Tamil Nadu, India
| | - Dianne Langford
- Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Kalimuthusamy Natarajaseenivasan
- Medical Microbiology Laboratory, Department of Microbiology, Centre for Excellence in Life Sciences, Bharathidasan University, Tiruchirappalli 620 024, Tamil Nadu, India; Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA..
| | - Santhanam Shanmughapriya
- Heart and Vascular Institute, Department of Medicine, Department of Cellular and Molecular Physiology, Pennsylvania State University, College of Medicine, Hershey PA-17033, USA.
| |
Collapse
|
7
|
Camargo MC, Song M, Ito H, Oze I, Koyanagi YN, Kasugai Y, Rabkin CS, Matsuo K. Associations of circulating mediators of inflammation, cell regulation and immune response with esophageal squamous cell carcinoma. J Cancer Res Clin Oncol 2021; 147:2885-2892. [PMID: 34128078 DOI: 10.1007/s00432-021-03687-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 06/05/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is the most common histologic subtype of esophageal cancer globally. The development of squamous cell carcinoma has important inflammatory influences and effects. We, therefore, examined circulating levels of inflammation- and immune-related proteins for associations with ESCC. METHODS We used pre-treatment EDTA plasma from 80 ESCC patients (44% clinical stages I and II) and 80 cancer-free control individuals within the Hospital-based Epidemiologic Research Program at Aichi Cancer Center. Levels of 184 biomarkers were measured by high-throughput multiplexed proximity extension assays using Olink's Proseek Cell Regulation and Immuno-Oncology Panels. ESCC odds ratios (OR) per quantile (based on two to four categories) of each biomarker were calculated by unconditional logistic regression models, adjusted for age, sex, cigarette smoking and alcohol consumption. Correlations among continuous biomarker levels were assessed by Spearman's rank correlation. All statistical tests were two-sided with p values < 0.05 considered as significant. Given the exploratory nature of the study, we did not adjust for multiple comparisons. RESULTS Seven proteins were undetectable in nearly all samples. Of the remaining 177 evaluable biomarkers, levels of cluster of differentiation 40 (CD40, per quartile OR 1.64; p trend = 0.018), syntaxin 16 (STX16, per quartile OR 1.63; p trend = 0.008), heme oxygenase 1 (per quartile OR 1.59; p trend = 0.014), and γ-secretase activating protein (GSAP, per quartile OR 1.47; p trend = 0.036) were significantly associated with ESCC. Amongst these significant markers, levels of CD40, STX16, and GSPA were moderately correlated (Rho coefficients 0.46-0.55; p < 0.05). CONCLUSION Our case-control study expands the range of inflammation and immune molecules associated with ESCC. These novel findings warrant replication and functional characterization.
Collapse
Affiliation(s)
- M Constanza Camargo
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, 9609 Medical Center Dr., BG 9609/6E338, Bethesda, MD, 20892, USA.
| | - Minkyo Song
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, 9609 Medical Center Dr., BG 9609/6E338, Bethesda, MD, 20892, USA
| | - Hidemi Ito
- Division of Cancer Information and Control, Department of Preventive Medicine, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Isao Oze
- Division of Cancer Epidemiology and Prevention, Department of Preventive Medicine, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Yuriko N Koyanagi
- Division of Cancer Information and Control, Department of Preventive Medicine, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Yumiko Kasugai
- Division of Cancer Epidemiology and Prevention, Department of Preventive Medicine, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Charles S Rabkin
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, 9609 Medical Center Dr., BG 9609/6E338, Bethesda, MD, 20892, USA
| | - Keitaro Matsuo
- Division of Cancer Epidemiology and Prevention, Department of Preventive Medicine, Aichi Cancer Center Research Institute, Nagoya, Japan.,Department of Cancer Epidemiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
8
|
Gilfillan M, Das P, Shah D, Alam MA, Bhandari V. Inhibition of microRNA-451 is associated with increased expression of Macrophage Migration Inhibitory Factor and mitgation of the cardio-pulmonary phenotype in a murine model of Bronchopulmonary Dysplasia. Respir Res 2020; 21:92. [PMID: 32321512 PMCID: PMC7178994 DOI: 10.1186/s12931-020-01353-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 04/02/2020] [Indexed: 12/13/2022] Open
Abstract
Background Macrophage migration inhibitory factor (MIF) has been implicated as a protective factor in the development of bronchopulmonary dysplasia (BPD) and is known to be regulated by MicroRNA-451 (miR-451). The aim of this study was to evaluate the role of miR-451 and the MIF signaling pathway in in vitro and in vivo models of BPD. Methods Studies were conducted in mouse lung endothelial cells (MLECs) exposed to hyperoxia and in a newborn mouse model of hyperoxia-induced BPD. Lung and cardiac morphometry as well as vascular markers were evaluated. Results Increased expression of miR-451 was noted in MLECs exposed to hyperoxia and in lungs of BPD mice. Administration of a miR-451 inhibitor to MLECs exposed to hyperoxia was associated with increased expression of MIF and decreased expression of angiopoietin (Ang) 2. Treatment with the miR-451 inhibitor was associated with improved lung morphometry indices, significant reduction in right ventricular hypertrophy, decreased mean arterial wall thickness and improvement in vascular density in BPD mice. Western blot analysis demonstrated preservation of MIF expression in BPD animals treated with a miR-451 inhibitor and increased expression of vascular endothelial growth factor-A (VEGF-A), Ang1, Ang2 and the Ang receptor, Tie2. Conclusion We demonstrated that inhibition of miR-451 is associated with mitigation of the cardio-pulmonary phenotype, preservation of MIF expression and increased expression of several vascular growth factors.
Collapse
Affiliation(s)
- Margaret Gilfillan
- Department of Pediatrics, Drexel University College of Medicine, Philadelphia, PA, 19103, USA.,St Christopher's Hospital for Children, Philadelphia, PA, 19134, USA
| | - Pragnya Das
- Department of Pediatrics, Drexel University College of Medicine, Philadelphia, PA, 19103, USA.,Neonatology Research Laboratory, Education and Research Building, Cooper University Hospital, (Room #206), Camden, NJ, 08103, USA
| | - Dilip Shah
- Department of Pediatrics, Drexel University College of Medicine, Philadelphia, PA, 19103, USA.,Neonatology Research Laboratory, Education and Research Building, Cooper University Hospital, (Room #206), Camden, NJ, 08103, USA
| | - Mohammad Afaque Alam
- Department of Pediatrics, Drexel University College of Medicine, Philadelphia, PA, 19103, USA.,Temple University, Philadelphia, PA, 19140, USA
| | - Vineet Bhandari
- Department of Pediatrics, Drexel University College of Medicine, Philadelphia, PA, 19103, USA. .,St Christopher's Hospital for Children, Philadelphia, PA, 19134, USA. .,Neonatology Research Laboratory, Education and Research Building, Cooper University Hospital, (Room #206), Camden, NJ, 08103, USA. .,Temple University, Philadelphia, PA, 19140, USA. .,Pediatrics, Obstetrics and Gynecology and Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, 08103, USA. .,Neonatology, The Children's Regional Hospital at Cooper, One Cooper Plaza, Camden, NJ, 08103, USA.
| |
Collapse
|
9
|
Chu LY, Peng YH, Weng XF, Xie JJ, Xu YW. Blood-based biomarkers for early detection of esophageal squamous cell carcinoma. World J Gastroenterol 2020; 26:1708-1725. [PMID: 32351288 PMCID: PMC7183865 DOI: 10.3748/wjg.v26.i15.1708] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 03/13/2020] [Accepted: 03/19/2020] [Indexed: 02/06/2023] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is a common malignant tumor of the digestive system worldwide, especially in China. Due to the lack of effective early detection methods, ESCC patients often present at an advanced stage at the time of diagnosis, which seriously affects the prognosis of patients. At present, early detection of ESCC mainly depends on invasive and expensive endoscopy and histopathological biopsy. Therefore, there is an unmet need for a non-invasive method to detect ESCC in the early stages. With the emergence of a large class of non-invasive diagnostic tools, serum tumor markers have attracted much attention because of their potential for detection of early tumors. Therefore, the identification of serum tumor markers for early detection of ESCC is undoubtedly one of the most effective ways to achieve early diagnosis and treatment of ESCC. This article reviews the recent advances in the discovery of blood-based ESCC biomarkers, and discusses the origins, clinical applications, and technical challenges of clinical validation of various types of biomarkers.
Collapse
Affiliation(s)
- Ling-Yu Chu
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong Province, China
| | - Yu-Hui Peng
- Department of Clinical Laboratory Medicine, the Cancer Hospital of Shantou University Medical College, Shantou 515041, Guangdong Province, China
- Precision Medicine Research Center, Shantou University Medical College, Shantou 515041, Guangdong Province, China
| | - Xue-Fen Weng
- Department of Clinical Laboratory Medicine, the Cancer Hospital of Shantou University Medical College, Shantou 515041, Guangdong Province, China
- Precision Medicine Research Center, Shantou University Medical College, Shantou 515041, Guangdong Province, China
| | - Jian-Jun Xie
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, Guangdong Province, China
- Precision Medicine Research Center, Shantou University Medical College, Shantou 515041, Guangdong Province, China
| | - Yi-Wei Xu
- Department of Clinical Laboratory Medicine, the Cancer Hospital of Shantou University Medical College, Shantou 515041, Guangdong Province, China
- Precision Medicine Research Center, Shantou University Medical College, Shantou 515041, Guangdong Province, China
| |
Collapse
|
10
|
Mahalingam D, Patel MR, Sachdev JC, Hart LL, Halama N, Ramanathan RK, Sarantopoulos J, Völkel D, Youssef A, de Jong FA, Tsimberidou AM. Phase I study of imalumab (BAX69), a fully human recombinant antioxidized macrophage migration inhibitory factor antibody in advanced solid tumours. Br J Clin Pharmacol 2020; 86:1836-1848. [PMID: 32207164 PMCID: PMC7444762 DOI: 10.1111/bcp.14289] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 02/28/2020] [Accepted: 03/06/2020] [Indexed: 12/15/2022] Open
Abstract
Aim Preclinical evidence suggests that oxidized macrophage migration inhibitory factor (oxMIF) may be involved in carcinogenesis. This phase 1 study (NCT01765790) assessed the safety, tolerability, pharmacokinetics and antitumour activity of imalumab, an oxMIF inhibitor, in patients with advanced cancer using ‘3 + 3’ dose escalation. Methods In Schedule 1, patients with solid tumours received doses from 1 to 50 mg/kg IV every 2 weeks. In Schedule 2, patients with metastatic colorectal adenocarcinoma, non‐small‐cell lung, or ovarian cancer received weekly doses of 10 or 25 mg/kg IV (1 cycle = 28 days). Treatment continued until disease progression, unacceptable toxicity, dose‐limiting toxicity, or withdrawal of consent. Results Fifty of 68 enrolled patients received imalumab. The most common treatment‐related adverse events (TRAEs) included fatigue (10%) and vomiting (6%); four grade 3 serious TRAEs (two patients) occurred. The dose‐limiting toxicity was allergic alveolitis (one patient, 50 mg/kg every 2 weeks). The maximum tolerated and biologically active doses were 37.5 mg/kg every 2 weeks and 10 mg/kg weekly, respectively. Of 39 assessed patients, 13 had stable disease (≥4 months in 8 patients). Conclusions Imalumab had a maximum tolerated dose of 37.5 mg/kg every 2 weeks in patients with advanced solid tumours, with a biologically active dose of 10 mg/kg weekly. Further investigation will help define the role of oxMIF as a cancer treatment target.
Collapse
Affiliation(s)
- Devalingam Mahalingam
- University of Texas Health Science Center, San Antonio, TX, USA.,Robert H Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA
| | - Manish R Patel
- Sarah Cannon Research Institute/Florida Cancer Specialists, Sarasota, FL, USA
| | - Jasgit C Sachdev
- HonorHealth Research Institute/Translational Genomics Research Institute (TGen), Scottsdale, AZ, USA
| | | | - Niels Halama
- National Center for Tumor Diseases, University Medical Center Heidelberg, Heidelberg, Germany
| | | | - John Sarantopoulos
- Institute for Drug Development, Mays Cancer Center at University of Texas Health San Antonio MD Anderson Cancer Center, San Antonio, TX, USA
| | - Dirk Völkel
- Baxalta Innovations GmbH a member of the Takeda group of companies, Vienna, Austria
| | - Ashraf Youssef
- Baxalta US Inc. a member of the Takeda group of companies, Cambridge, MA, USA
| | | | - Apostolia Maria Tsimberidou
- Department of Investigational Cancer Therapeutics (Phase 1 Program), The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
11
|
Huang Z, Zhou B, Li Z, Liu C, Zheng C, Zeng R, Hong C, Xu L, Li E, Peng Y, Xu Y. Serum interleukin-8 as a potential diagnostic biomarker in esophageal squamous cell carcinoma. Cancer Biomark 2020; 29:139-149. [PMID: 32623391 DOI: 10.3233/cbm-201687] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) has poor prognosis mainly due to the difficulty of making early diagnosis. Therefore, novel biomarkers are critically needed. OBJECTIVE We aimed to investigate the diagnostic value of serum interleukin-8 (IL-8) in ESCC. METHODS Data mining of TCGA was used to analyze expression level of IL-8 mRNA in esophageal carcinoma. Serum levels of IL-8 were measured in 103 ESCC patients and 86 normal controls by ELISA. Receiver operating characteristic (ROC) curve was used to evaluate its diagnostic accuracy. RESULTS IL-8 mRNA expression level and serum IL-8 concentration were both statistically higher in patients than normal controls (P< 0.001). ROC curve demonstrated that the optimum diagnostic cut-off for serum IL-8 was 80.082 pg/mL, providing an area under the curve (AUC) of 0.694 (95% CI: 0.620-0.768), with specificity of 86.0% and sensitivity of 42.7%. The AUC for early-stage ESCC was 0.618 (95% CI: 0.499-0.737), with sensitivity of 35.3% and specificity of 86.0%. Kaplan-Meier analysis and the log-rank test indicated that IL-8 may not be a prognostic predictor for ESCC. CONCLUSIONS Serum IL-8 was highly expressed in ESCC patients and may be a potential marker for early diagnosis of ESCC.
Collapse
Affiliation(s)
- Zeting Huang
- Shantou University Medical College, Shantou, Guangdong, China
- Shantou University Medical College, Shantou, Guangdong, China
| | - Bin Zhou
- Shantou University Medical College, Shantou, Guangdong, China
- Shantou University Medical College, Shantou, Guangdong, China
| | - Zheng Li
- Shantou University Medical College, Shantou, Guangdong, China
- Shantou University Medical College, Shantou, Guangdong, China
| | - Cantong Liu
- Department of Clinical Laboratory Medicine, The Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
- Precision Medicine Research Centre, Shantou University Medical College, Shantou, Guangdong, China
| | - Chunwen Zheng
- Shantou University Medical College, Shantou, Guangdong, China
| | - Ruijie Zeng
- Shantou University Medical College, Shantou, Guangdong, China
| | - Chaoqun Hong
- Department of Oncological Laboratory Research, The Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Liyan Xu
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou, Guangdong, China
| | - Enmin Li
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, Guangdong, China
| | - Yuhui Peng
- Department of Clinical Laboratory Medicine, The Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
- Precision Medicine Research Centre, Shantou University Medical College, Shantou, Guangdong, China
| | - Yiwei Xu
- Department of Clinical Laboratory Medicine, The Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, China
- Precision Medicine Research Centre, Shantou University Medical College, Shantou, Guangdong, China
| |
Collapse
|
12
|
Macrophage Migration Inhibitory Factor Promotes the Interaction between the Tumor, Macrophages, and T Cells to Regulate the Progression of Chemically Induced Colitis-Associated Colorectal Cancer. Mediators Inflamm 2019; 2019:2056085. [PMID: 31360118 PMCID: PMC6652048 DOI: 10.1155/2019/2056085] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 06/11/2019] [Indexed: 12/16/2022] Open
Abstract
Colitis-associated colorectal cancer (CRC) development has been shown to be related to chronically enhanced inflammation. Macrophage migration inhibitory factor (MIF) is an inflammatory mediator that favors inflammatory cytokine production and has chemotactic properties for the recruitment of macrophages (Møs) and T cells. Here, we investigated the role of MIF in the inflammatory response and recruitment of immune cells in a murine model of chemical carcinogenesis to establish the impact of MIF on CRC genesis and malignancy. We used BALB/c MIF-knockout (MIF-/-) and wild-type (WT) mice to develop CRC by administering intraperitoneal (i.p.) azoxymethane and dextran sodium sulfate in drinking water. Greater tumor burdens were observed in MIF-/- mice than in WT mice. Tumors from MIF-/- mice were histologically identified to be more aggressive than tumors from WT mice. The localization of MIF suggests that it is also involved in cell differentiation. The relative gene expression of il-17, measured by real-time PCR, was higher in MIF-/- CRC mice, compared to the WT CRC and healthy MIF-/- mice. Importantly, compared to the WT intestinal epithelium, lower percentages of tumor-associated Møs were found in the MIF-/- intestinal epithelium. These results suggest that MIF plays a role in controlling the initial development of CRC by attracting Møs to the tumor, which is a condition that favors the initial antitumor responses.
Collapse
|
13
|
Role of MIF and D-DT in immune-inflammatory, autoimmune, and chronic respiratory diseases: from pathogenic factors to therapeutic targets. Drug Discov Today 2018; 24:428-439. [PMID: 30439447 DOI: 10.1016/j.drudis.2018.11.003] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 10/04/2018] [Accepted: 11/06/2018] [Indexed: 01/03/2023]
Abstract
Macrophage migration inhibitory factor (MIF) is a protein that acts as a cytokine-, enzyme-, endocrine- and chaperon-like molecule. It binds to the cell-surface receptor CD74 in association with CD44, which activates the downstream signal transduction pathway. In addition, MIF acts also as a noncognate ligand for C-X-C chemokine receptor type 2 (CXCR2), type 4 (CXCR4), and type 7 (CXCR7). Recently, D-dopachrome tautomerase (D-DT), a second member of the MIF superfamily, was identified. From a pharmacological and clinical point of view, the nonredundant biological properties of MIF and D-DT anticipate potential synergisms from their simultaneous inhibition. Here, we focus on the role of MIF and D-DT in human immune-inflammatory, autoimmune, and chronic respiratory diseases, providing an update on the progress made in the identification of specific small-molecule inhibitors of these proteins.
Collapse
|
14
|
Song Y, Zhao N, Jiang K, Zheng Z, Wang B, Kong D, Li S. Occurrence of metachronous multiple primary cancers occurred in different parts of the stomach with 2 pathologic features: A case report. Medicine (Baltimore) 2018; 97:e10803. [PMID: 29768377 PMCID: PMC5976309 DOI: 10.1097/md.0000000000010803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
RATIONALE With the increasing survival rate of gastric cancer, more multiple primary cancers (MPC) have been reported. However, few cases involve metachronous multiple primary cancers which both occurred in the stomach. PATIENT CONCERNS An 83-year-old Chinese male had been diagnosed with gastric cardia cancer and underwent proximal gastrectomy. The pathological result was gastric adenocarcinoma. 13 years later the patient's gastroscope result deteriorated. The biopsy of the antrum revealed dysplasia with doubtful focal cancerization. DIAGNOSES Metachronous multiple primary cancers in the stomach. INTERVENTIONS Endoscopic submucosal dissection (ESD) was performed. The pathological result showed an intra-mucosal signet-ring carcinoma. OUTCOMES After treatment, the patient is alive with good condition until now. LESSONS This is an unusual case of MPC with different pathological features in different parts of the same organ in an interval of more than ten years and undergoing different operations.
Collapse
Affiliation(s)
- Yan Song
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin
| | - Ningning Zhao
- Department of Gastroenterology and Hepatology, Hebei Province Cangzhou Hospital of Integrated Traditional and Western Medicine, Cangzhou, Hebei Province
| | - Kui Jiang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin
| | - Zhongqing Zheng
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin
| | - Bangmao Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin
| | - Dalu Kong
- Department of Hepatobiliary Surgery, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Shu Li
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin
| |
Collapse
|
15
|
Abstract
The aim of this study was to investigate the roles of serum interleukin-6 (IL-6), IL-8, IL-10, squamous cell cancer antigen (SCC-Ag), and cytokeratin 21-1 fragment (CYFRA 21-1) in the metastasis and prognosis of breast cancer.A total of 534 breast cancer patients admitted to our department between January 2011 and December 2014 were enrolled in this study. Besides, 452 matched healthy individuals received physical examination at the same period served as the normal control. Serum IL-6, IL-8, IL-10, and tumor necrosis factor-α (TNF-α) were determined using an immunoradiometric assay. SCC-Ag level was evaluated using chemiluminescent microparticle immunoassay. CYFRA 21-1 was determined using the chemiluminescence assay.Compared with the control group, a significant increase was noticed in the serum IL-6, IL-8, and IL-10 in breast cancer patients, especially those with severe conditions (P < .01). Serum IL-6, IL-8, and IL-10 showed a significant increase in the patients with severe breast cancer compared with those with mild conditions (P < .05). For the patients with response after radiotherapy, the serum IL-6, IL-8, and IL-10 were significantly decreased compared with the baseline levels (P < .05). The median survival duration for the patients of SCC-Ag negative patients was 25 months, while that for the SCC-Ag positive group was 16 months. Significant difference was noticed in the survival of SCC-Ag negative group compared with that of SCC-Ag positive group (P < .05).Serum IL-6, IL-8, IL-10, SCC-Ag, and CYFRA 21-1 were considered as potential markers in the metastasis and prognosis of breast cancer.
Collapse
Affiliation(s)
- Haiyan Wang
- Department of Pathology, Weihai Maternal and Child Health Hospital
| | - Xianlu Yang
- Department of Dermatology, Weihai Municipal Hospital, Weihai, China
| |
Collapse
|
16
|
Macrophage migration inhibitory factor promotes tumor aggressiveness of esophageal squamous cell carcinoma via activation of Akt and inactivation of GSK3β. Cancer Lett 2017; 412:289-296. [PMID: 29079416 DOI: 10.1016/j.canlet.2017.10.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 10/11/2017] [Accepted: 10/13/2017] [Indexed: 12/31/2022]
Abstract
The pleiotropic pro-inflammatory cytokine, macrophage migration inhibitory factor (MIF), represents an important link between chronic inflammation and tumorigenesis. Although accumulating evidence demonstrates that MIF overexpression is implicated in the development and progression of multiple cancers, including esophageal squamous cell carcinoma (ESCC), the molecular mechanisms underlying its tumor-promoting roles in ESCC remain unclear. In the present study, we observed that MIF is overexpressed in ESCC and correlated significantly with lymph node metastasis, advanced clinical stage, and poor survival of ESCC. MIF knockdown attenuated the proliferation, migration, and invasion of ESCC cells in vitro and in vivo. Moreover, blockage of MIF expression decreased the activation of the Akt, MEK/ERK, and NF-κB pathways and enhanced sensitivity to apoptosis. Meanwhile, repression of MIF expression resulted in activation of glycogen synthase kinase 3 beta (GSK3β) and subsequent decrease of active β-catenin, as well as its downstream targets including cyclin D1, matrix metalloproteinase (MMP)-7, c-myc, and c-Jun. Collectively, our results provided mechanistic insights into the tumor-promoting role of MIF in ESCC, and suggested that MIF represents a potential therapeutic target for treatment of ESCC.
Collapse
|
17
|
Lechien JR, Nassri A, Kindt N, Brown DN, Journe F, Saussez S. Role of macrophage migration inhibitory factor in head and neck cancer and novel therapeutic targets: A systematic review. Head Neck 2017; 39:2573-2584. [PMID: 28963807 DOI: 10.1002/hed.24939] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Revised: 06/22/2017] [Accepted: 07/27/2017] [Indexed: 12/19/2022] Open
Abstract
Macrophage migration inhibitory factor (MIF) is a proinflammatory cytokine involved in systemic, autoimmune, and inflammatory diseases, such as obesity, rheumatoid arthritis, and systemic lupus erythematosus. For the 2 past decades, MIF has been reported to participate in carcinogenesis, disease prognosis, tumor cell proliferation, invasion, and tumor-induced angiogenesis in many cancers. The purpose of this article is to review published experimental and clinical data for MIF and its involvement in upper aerodigestive tract cancers. Based on the current literature, we propose a biomolecular model describing the mechanisms underlying the involvement of MIF in the initiation, progression, apoptosis, and proliferation of head and neck tumor cells. In reference to this model, potential therapeutic approaches based on the use of MIF antagonists and neutralizing antibodies are described. It is concluded that MIF is a promising target for future therapeutic strategies, both with and without chemoradiation strategies.
Collapse
Affiliation(s)
- Jérôme R Lechien
- Department of Otolaryngology and Head and Neck Surgery, RHMS Baudour, EpiCURA Hospital, Baudour, Belgium.,Laboratory of Phonetics, Faculty of Psychology, Research Institute for Language sciences and Technology, University of Mons (UMONS), Mons, Belgium.,Laboratory of Human Anatomy and Experimental Oncology, Faculty of Medicine, Research Institute for Health Sciences and Technology, University of Mons (UMONS), Mons, Belgium
| | - Amir Nassri
- Laboratory of Human Anatomy and Experimental Oncology, Faculty of Medicine, Research Institute for Health Sciences and Technology, University of Mons (UMONS), Mons, Belgium
| | - Nadege Kindt
- Laboratory of Human Anatomy and Experimental Oncology, Faculty of Medicine, Research Institute for Health Sciences and Technology, University of Mons (UMONS), Mons, Belgium
| | - David N Brown
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Fabrice Journe
- Laboratory of Human Anatomy and Experimental Oncology, Faculty of Medicine, Research Institute for Health Sciences and Technology, University of Mons (UMONS), Mons, Belgium.,Laboratory of Oncology and Experimental Surgery, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Sven Saussez
- Department of Otolaryngology and Head and Neck Surgery, RHMS Baudour, EpiCURA Hospital, Baudour, Belgium.,Laboratory of Human Anatomy and Experimental Oncology, Faculty of Medicine, Research Institute for Health Sciences and Technology, University of Mons (UMONS), Mons, Belgium
| |
Collapse
|
18
|
Ismail MM, Morsi HK, Abdulateef NAB, Noaman MK, Abou El-Ella GA. Evaluation of prothrombin induced by vitamin K absence, macrophage migration inhibitory factor and Golgi protein-73 versus alpha fetoprotein for hepatocellular carcinoma diagnosis and surveillance. Scandinavian Journal of Clinical and Laboratory Investigation 2017; 77:175-183. [PMID: 28276727 DOI: 10.1080/00365513.2017.1286684] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Hepatocellular carcinoma (HCC) represents a challenging malignancy of worldwide importance. It is the third most common cause of cancer-related death globally as most patients present with unresectable disease. Alpha-fetoprotein (AFP) is the widely and solely used biomarker for HCC diagnosis; yet, its usefulness is hampered by low sensitivity and specificity. We aimed to identify more sensitive biomarkers for HCC diagnosis and a surveillance algorithm that may facilitate early detection of HCC. A total of 305 Egyptian and Saudi participants grouped as healthy controls, cancer controls, benign hepatic lesions, chronic viral hepatitis and HCC were included. Serum AFP, prothrombin induced by vitamin K absence-II (PIVKA-II), macrophage migration inhibitory factor (MIF) and Golgi protein-73 (GP-73) levels were quantitated by enzyme immunoassay. Significantly higher levels of GP-73 and PIVKA-II were detected in the HCC group than in all other groups, while MIF showed a highly significant increase in HCC from all groups except the cancer control group. The HCC group showed no significant difference between the studied biomarkers and the type of chronic viral hepatitis. On the basis of multiple ROC curve analyses, GP-73 and PIVKA-II showed the highest sensitivity and specificity for surveillance and diagnosis. In conclusion, PIVKA-II and GP-73 offer an effective approach for early HCC diagnosis and surveillance of high-risk groups with a higher accuracy than AFP. MIF may serve as a promising screening tumor marker for the detection of gastrointestinal tract (GIT) malignancy.
Collapse
Affiliation(s)
- Manar M Ismail
- a Laboratory Medicine Department Faculty of Applied Medical Science , Umm Al Qura University , Kingdom of Saudi Arabia.,b Clinical Pathology Department , National Cancer Institute, Cairo University , Egypt
| | - Heba K Morsi
- a Laboratory Medicine Department Faculty of Applied Medical Science , Umm Al Qura University , Kingdom of Saudi Arabia.,c Medical Biochemistry Department, Faculty of Medicine , Mansoura University , Egypt
| | - Nahla A B Abdulateef
- b Clinical Pathology Department , National Cancer Institute, Cairo University , Egypt.,d Laboratory and Blood Bank Department , KAMC , Makkah , Kingdom of Saudi Arabia
| | - Maissa K Noaman
- e Biostatistics and Cancer Epidemiology , National Cancer Institute, Cairo University , Egypt
| | - Ghada A Abou El-Ella
- a Laboratory Medicine Department Faculty of Applied Medical Science , Umm Al Qura University , Kingdom of Saudi Arabia.,f Department of Animal Medicine, Faculty of Veterinary Medicine , Assiut University , Egypt
| |
Collapse
|
19
|
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]
|
20
|
Wang L, Zheng JN, Pei DS. The emerging roles of Jab1/CSN5 in cancer. Med Oncol 2016; 33:90. [DOI: 10.1007/s12032-016-0805-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 07/04/2016] [Indexed: 12/13/2022]
|
21
|
Chesney JA, Mitchell RA. 25 Years On: A Retrospective on Migration Inhibitory Factor in Tumor Angiogenesis. Mol Med 2015; 21 Suppl 1:S19-24. [PMID: 26605643 DOI: 10.2119/molmed.2015.00055] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 03/16/2015] [Indexed: 01/26/2023] Open
Abstract
Twenty-five years ago marked the publication of the first report describing a functional contribution by the cytokine, macrophage migration inhibitory factor (MIF), to tumor-associated angiogenesis and growth. Since first appearing, this report has been cited 304 times (as of this writing), underscoring not only the importance of this landmark study but also the importance of MIF in tumor neovascularization. Perhaps more importantly, this first link between MIF and stromal cell-dependent tumor angiogenesis presaged the subsequent identification of MIF in mediating protumorigenic contributions to several solid tumor stromal cell types, including monocytes, macrophages, T lymphocytes, NK cells, fibroblasts, endothelial progenitors and mesenchymal stem cells. This retrospective review will broadly evaluate both past and present literature stemming from this initial publication, with an emphasis on cellular sources, cellular effectors, signal transduction mechanisms and the clinical importance of MIF-dependent tumor vascularization.
Collapse
Affiliation(s)
- Jason A Chesney
- Molecular Targets Program, JG Brown Cancer Center, and the Department of Medicine, University of Louisville, Louisville, Kentucky, United States of America
| | - Robert A Mitchell
- Molecular Targets Program, JG Brown Cancer Center, and the Department of Medicine, University of Louisville, Louisville, Kentucky, United States of America
| |
Collapse
|
22
|
No YR, Lee SJ, Kumar A, Yun CC. HIF1α-Induced by Lysophosphatidic Acid Is Stabilized via Interaction with MIF and CSN5. PLoS One 2015; 10:e0137513. [PMID: 26352431 PMCID: PMC4564097 DOI: 10.1371/journal.pone.0137513] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 08/18/2015] [Indexed: 12/29/2022] Open
Abstract
Macrophage migration inhibitory factor (MIF) is a cytokine that has broad effects on immune system and inflammatory response. A growing body of evidence implicates the role of MIF in tumor growth and metastasis. Lysophosphatidic acid (LPA), a bioactive lipid mediator, regulates colon cancer cell proliferation, invasion, and survival through LPA2 receptor. Loss of LPA2 results in decreased expression of MIF in a rodent model of colon cancer, but the mechanism of MIF regulation by LPA is yet to be determined. In this study, we show that LPA transcriptionally regulates MIF expression in colon cancer cells. MIF knockdown decreased LPA-mediated proliferation of HCT116 human adenocarcinoma cells without altering the basal proliferation rates. Conversely, extracellular recombinant MIF stimulated cell proliferation, suggesting that the effect of MIF may in part be mediated through activation of surface receptor. We have shown recently that LPA increases hypoxia-inducible factor 1α (HIF1α) expression. We found that MIF regulation by LPA was ablated by knockdown of HIF1α, indicating that MIF is a transcriptional target of HIF1α. Conversely, knockdown of MIF ablated an increase in HIF1α expression in LPA-treated cells, suggesting a reciprocal relationship between HIF1α and MIF. LPA stimulated co-immunoprecipitation of HIF1α and MIF, indicating that their association is necessary for stabilization of HIF1α. It has been shown previously that CSN9 signalosome subunit 5 (CSN5) interacts with HIF1α to stabilize HIF1α under aerobic conditions. We found that LPA did not alter expression of CSN5, but stimulated its interaction with HIF1α and MIF. Depletion of CSN5 mitigated the association between HIF1α and MIF, indicating that CSN5 acts as a physical link. We suggest that HIF1α, MIF, and CSN5 form a ternary complex whose formation is necessary to prevent degradation of HIF1α under aerobic conditions.
Collapse
Affiliation(s)
- Yi Ran No
- Division of Digestive Diseases, Department of Medicine, Emory University, Atlanta, Georgia, United States of America
| | - Sei-Jung Lee
- Division of Digestive Diseases, Department of Medicine, Emory University, Atlanta, Georgia, United States of America
| | - Ajay Kumar
- Division of Digestive Diseases, Department of Medicine, Emory University, Atlanta, Georgia, United States of America
| | - C Chris Yun
- Division of Digestive Diseases, Department of Medicine, Emory University, Atlanta, Georgia, United States of America; Winship Cancer Institute, Emory University, Atlanta, Georgia, United States of America
| |
Collapse
|
23
|
Cheng SP, Liu CL, Chen MJ, Chien MN, Leung CH, Lin CH, Hsu YC, Lee JJ. CD74 expression and its therapeutic potential in thyroid carcinoma. Endocr Relat Cancer 2015; 22:179-90. [PMID: 25600560 DOI: 10.1530/erc-14-0269] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
CD74, the invariant chain of major histocompatibility complex class II, is also a receptor for macrophage migration inhibitory factor (MIF). CD74 and MIF have been associated with tumor progression and metastasis in hematologic and solid tumors. In this study, we found that 60 and 65% of papillary thyroid cancers were positive for CD74 and MIF immunohistochemical staining respectively. Anaplastic thyroid cancer was negative for MIF, but mostly positive for CD74 expression. Normal thyroid tissue and follicular adenomas were negative for CD74 expression. CD74 expression in papillary thyroid cancer was associated with larger tumor size (P=0.043), extrathyroidal invasion (P=0.021), advanced TNM stage (P=0.006), and higher MACIS score (P=0.026). No clinicopathological parameter was associated with MIF expression. Treatment with anti-CD74 antibody in thyroid cancer cells inhibited cell growth, colony formation, cell migration and invasion, and vascular endothelial growth factor secretion. In contrast, treatment with recombinant MIF induced an increase in cell invasion. Anti-CD74 treatment reduced AKT phosphorylation and stimulated AMPK activation. Our findings suggest that CD74 overexpression in thyroid cancer is associated with advanced tumor stage and may serve as a therapeutic target.
Collapse
MESH Headings
- Antibodies/pharmacology
- Antigens, Differentiation, B-Lymphocyte/genetics
- Antigens, Differentiation, B-Lymphocyte/immunology
- Antigens, Differentiation, B-Lymphocyte/metabolism
- Carcinoma/metabolism
- Carcinoma/pathology
- Carcinoma, Papillary
- Cell Line, Tumor
- Cell Movement
- Gene Expression Regulation, Neoplastic
- Histocompatibility Antigens Class II/genetics
- Histocompatibility Antigens Class II/immunology
- Histocompatibility Antigens Class II/metabolism
- Humans
- Intramolecular Oxidoreductases/genetics
- Intramolecular Oxidoreductases/metabolism
- Macrophage Migration-Inhibitory Factors/genetics
- Macrophage Migration-Inhibitory Factors/metabolism
- Thyroid Cancer, Papillary
- Thyroid Gland/metabolism
- Thyroid Neoplasms/metabolism
- Thyroid Neoplasms/pathology
- Transcriptome
- Tumor Burden
- Wound Healing
Collapse
Affiliation(s)
- Shih-Ping Cheng
- Department of SurgeryDivision of Endocrinology and MetabolismDepartment of Medicine, MacKay Memorial Hospital and Mackay Medical College, Taipei, TaiwanMackay Junior College of MedicineNursing, and Management, No. 92, Sheng-Ching Road, Peitou, Taipei 11260, TaiwanDepartment of PharmacologyGraduate Institute of Medical Sciences, Taipei Medical University, Taipei, TaiwanDepartment of Medical ResearchMacKay Memorial Hospital, Taipei, TaiwanInstitute of Statistical ScienceAcademia Sinica, Taipei, Taiwan Department of SurgeryDivision of Endocrinology and MetabolismDepartment of Medicine, MacKay Memorial Hospital and Mackay Medical College, Taipei, TaiwanMackay Junior College of MedicineNursing, and Management, No. 92, Sheng-Ching Road, Peitou, Taipei 11260, TaiwanDepartment of PharmacologyGraduate Institute of Medical Sciences, Taipei Medical University, Taipei, TaiwanDepartment of Medical ResearchMacKay Memorial Hospital, Taipei, TaiwanInstitute of Statistical ScienceAcademia Sinica, Taipei, Taiwan Department of SurgeryDivision of Endocrinology and MetabolismDepartment of Medicine, MacKay Memorial Hospital and Mackay Medical College, Taipei, TaiwanMackay Junior College of MedicineNursing, and Management, No. 92, Sheng-Ching Road, Peitou, Taipei 11260, TaiwanDepartment of PharmacologyGraduate Institute of Medical Sciences, Taipei Medical University, Taipei, TaiwanDepartment of Medical ResearchMacKay Memorial Hospital, Taipei, TaiwanInstitute of Statistical ScienceAcademia Sinica, Taipei, Taiwan
| | - Chien-Liang Liu
- Department of SurgeryDivision of Endocrinology and MetabolismDepartment of Medicine, MacKay Memorial Hospital and Mackay Medical College, Taipei, TaiwanMackay Junior College of MedicineNursing, and Management, No. 92, Sheng-Ching Road, Peitou, Taipei 11260, TaiwanDepartment of PharmacologyGraduate Institute of Medical Sciences, Taipei Medical University, Taipei, TaiwanDepartment of Medical ResearchMacKay Memorial Hospital, Taipei, TaiwanInstitute of Statistical ScienceAcademia Sinica, Taipei, Taiwan Department of SurgeryDivision of Endocrinology and MetabolismDepartment of Medicine, MacKay Memorial Hospital and Mackay Medical College, Taipei, TaiwanMackay Junior College of MedicineNursing, and Management, No. 92, Sheng-Ching Road, Peitou, Taipei 11260, TaiwanDepartment of PharmacologyGraduate Institute of Medical Sciences, Taipei Medical University, Taipei, TaiwanDepartment of Medical ResearchMacKay Memorial Hospital, Taipei, TaiwanInstitute of Statistical ScienceAcademia Sinica, Taipei, Taiwan
| | - Ming-Jen Chen
- Department of SurgeryDivision of Endocrinology and MetabolismDepartment of Medicine, MacKay Memorial Hospital and Mackay Medical College, Taipei, TaiwanMackay Junior College of MedicineNursing, and Management, No. 92, Sheng-Ching Road, Peitou, Taipei 11260, TaiwanDepartment of PharmacologyGraduate Institute of Medical Sciences, Taipei Medical University, Taipei, TaiwanDepartment of Medical ResearchMacKay Memorial Hospital, Taipei, TaiwanInstitute of Statistical ScienceAcademia Sinica, Taipei, Taiwan Department of SurgeryDivision of Endocrinology and MetabolismDepartment of Medicine, MacKay Memorial Hospital and Mackay Medical College, Taipei, TaiwanMackay Junior College of MedicineNursing, and Management, No. 92, Sheng-Ching Road, Peitou, Taipei 11260, TaiwanDepartment of PharmacologyGraduate Institute of Medical Sciences, Taipei Medical University, Taipei, TaiwanDepartment of Medical ResearchMacKay Memorial Hospital, Taipei, TaiwanInstitute of Statistical ScienceAcademia Sinica, Taipei, Taiwan Department of SurgeryDivision of Endocrinology and MetabolismDepartment of Medicine, MacKay Memorial Hospital and Mackay Medical College, Taipei, TaiwanMackay Junior College of MedicineNursing, and Management, No. 92, Sheng-Ching Road, Peitou, Taipei 11260, TaiwanDepartment of PharmacologyGraduate Institute of Medical Sciences, Taipei Medical University, Taipei, TaiwanDepartment of Medical ResearchMacKay Memorial Hospital, Taipei, TaiwanInstitute of Statistical ScienceAcademia Sinica, Taipei, Taiwan
| | - Ming-Nan Chien
- Department of SurgeryDivision of Endocrinology and MetabolismDepartment of Medicine, MacKay Memorial Hospital and Mackay Medical College, Taipei, TaiwanMackay Junior College of MedicineNursing, and Management, No. 92, Sheng-Ching Road, Peitou, Taipei 11260, TaiwanDepartment of PharmacologyGraduate Institute of Medical Sciences, Taipei Medical University, Taipei, TaiwanDepartment of Medical ResearchMacKay Memorial Hospital, Taipei, TaiwanInstitute of Statistical ScienceAcademia Sinica, Taipei, Taiwan Department of SurgeryDivision of Endocrinology and MetabolismDepartment of Medicine, MacKay Memorial Hospital and Mackay Medical College, Taipei, TaiwanMackay Junior College of MedicineNursing, and Management, No. 92, Sheng-Ching Road, Peitou, Taipei 11260, TaiwanDepartment of PharmacologyGraduate Institute of Medical Sciences, Taipei Medical University, Taipei, TaiwanDepartment of Medical ResearchMacKay Memorial Hospital, Taipei, TaiwanInstitute of Statistical ScienceAcademia Sinica, Taipei, Taiwan
| | - Ching-Hsiang Leung
- Department of SurgeryDivision of Endocrinology and MetabolismDepartment of Medicine, MacKay Memorial Hospital and Mackay Medical College, Taipei, TaiwanMackay Junior College of MedicineNursing, and Management, No. 92, Sheng-Ching Road, Peitou, Taipei 11260, TaiwanDepartment of PharmacologyGraduate Institute of Medical Sciences, Taipei Medical University, Taipei, TaiwanDepartment of Medical ResearchMacKay Memorial Hospital, Taipei, TaiwanInstitute of Statistical ScienceAcademia Sinica, Taipei, Taiwan Department of SurgeryDivision of Endocrinology and MetabolismDepartment of Medicine, MacKay Memorial Hospital and Mackay Medical College, Taipei, TaiwanMackay Junior College of MedicineNursing, and Management, No. 92, Sheng-Ching Road, Peitou, Taipei 11260, TaiwanDepartment of PharmacologyGraduate Institute of Medical Sciences, Taipei Medical University, Taipei, TaiwanDepartment of Medical ResearchMacKay Memorial Hospital, Taipei, TaiwanInstitute of Statistical ScienceAcademia Sinica, Taipei, Taiwan
| | - Chi-Hsin Lin
- Department of SurgeryDivision of Endocrinology and MetabolismDepartment of Medicine, MacKay Memorial Hospital and Mackay Medical College, Taipei, TaiwanMackay Junior College of MedicineNursing, and Management, No. 92, Sheng-Ching Road, Peitou, Taipei 11260, TaiwanDepartment of PharmacologyGraduate Institute of Medical Sciences, Taipei Medical University, Taipei, TaiwanDepartment of Medical ResearchMacKay Memorial Hospital, Taipei, TaiwanInstitute of Statistical ScienceAcademia Sinica, Taipei, Taiwan
| | - Yi-Chiung Hsu
- Department of SurgeryDivision of Endocrinology and MetabolismDepartment of Medicine, MacKay Memorial Hospital and Mackay Medical College, Taipei, TaiwanMackay Junior College of MedicineNursing, and Management, No. 92, Sheng-Ching Road, Peitou, Taipei 11260, TaiwanDepartment of PharmacologyGraduate Institute of Medical Sciences, Taipei Medical University, Taipei, TaiwanDepartment of Medical ResearchMacKay Memorial Hospital, Taipei, TaiwanInstitute of Statistical ScienceAcademia Sinica, Taipei, Taiwan
| | - Jie-Jen Lee
- Department of SurgeryDivision of Endocrinology and MetabolismDepartment of Medicine, MacKay Memorial Hospital and Mackay Medical College, Taipei, TaiwanMackay Junior College of MedicineNursing, and Management, No. 92, Sheng-Ching Road, Peitou, Taipei 11260, TaiwanDepartment of PharmacologyGraduate Institute of Medical Sciences, Taipei Medical University, Taipei, TaiwanDepartment of Medical ResearchMacKay Memorial Hospital, Taipei, TaiwanInstitute of Statistical ScienceAcademia Sinica, Taipei, Taiwan Department of SurgeryDivision of Endocrinology and MetabolismDepartment of Medicine, MacKay Memorial Hospital and Mackay Medical College, Taipei, TaiwanMackay Junior College of MedicineNursing, and Management, No. 92, Sheng-Ching Road, Peitou, Taipei 11260, TaiwanDepartment of PharmacologyGraduate Institute of Medical Sciences, Taipei Medical University, Taipei, TaiwanDepartment of Medical ResearchMacKay Memorial Hospital, Taipei, TaiwanInstitute of Statistical ScienceAcademia Sinica, Taipei, Taiwan Department of SurgeryDivision of Endocrinology and MetabolismDepartment of Medicine, MacKay Memorial Hospital and Mackay Medical College, Taipei, TaiwanMackay Junior College of MedicineNursing, and Management, No. 92, Sheng-Ching Road, Peitou, Taipei 11260, TaiwanDepartment of PharmacologyGraduate Institute of Medical Sciences, Taipei Medical University, Taipei, TaiwanDepartment of Medical ResearchMacKay Memorial Hospital, Taipei, TaiwanInstitute of Statistical ScienceAcademia Sinica, Taipei, Taiwan
| |
Collapse
|
24
|
DE Souza MB, Curioni OA, Kanda JL, DE Carvalho MB. Serum and salivary macrophage migration inhibitory factor in patients with oral squamous cell carcinoma. Oncol Lett 2014; 8:2267-2275. [PMID: 25289107 PMCID: PMC4186499 DOI: 10.3892/ol.2014.2513] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 07/01/2014] [Indexed: 12/14/2022] Open
Abstract
The overexpression of macrophage migration inhibitory factor (MIF) has been identified in a variety of tumors and the investigation of its molecular mechanisms in tumor progression is a key topic of research. The present study aimed to investigate MIF as a potential marker for disease control or recurrence, and to assess the association between serum and salivary MIF and the clinicopathological characteristics of patients with oral squamous cell carcinoma (OSCC). Serum and salivary samples were collected prior to and following the surgical treatment of 50 patients with OSCC. MIF concentrations were assessed by enzyme-linked immunosorbent assay and the adopted level of statistical significance was P<0.05. The results revealed that serum MIF concentrations were significantly reduced following tumor resection in OSCC patients. Furthermore, higher preoperative salivary MIF concentrations were observed in patients with larger tumors and in those who succumbed to the disease. In conclusion, high salivary and serological MIF concentrations were identified in patients with OSCC. Nevertheless, only serological MIF concentrations may be considered as a potential marker for the early detection of OSCC recurrence once the salivary levels, prior and following treatment, do not show any significant differences.
Collapse
Affiliation(s)
- Mariana Barbosa DE Souza
- Department of Radiology and Oncology, Medical School, University of São Paulo, São Paulo 01246903, Brazil ; Laboratory of Molecular Biology, Heliópolis Hospital, São Paulo 04231030, Brazil
| | - Otávio Alberto Curioni
- Laboratory of Molecular Biology, Heliópolis Hospital, São Paulo 04231030, Brazil ; Department of Head and Neck Surgery and Otorhinolaryngology, Heliópolis Hospital, São Paulo 04231030, Brazil
| | - Jossi Ledo Kanda
- Department of Head and Neck Surgery, Padre Anchieta Teaching Hospital, ABC Medical School, São Bernardo do Campo 09715090, Brazil
| | - Marcos Brasilino DE Carvalho
- Laboratory of Molecular Biology, Heliópolis Hospital, São Paulo 04231030, Brazil ; Department of Head and Neck Surgery and Otorhinolaryngology, Heliópolis Hospital, São Paulo 04231030, Brazil
| |
Collapse
|
25
|
Cytokines association with clinical and pathological changes in esophageal squamous cell carcinoma. DISEASE MARKERS 2014; 35:883-93. [PMID: 24427776 PMCID: PMC3877595 DOI: 10.1155/2013/302862] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Carcinogenic transformation of cells in esophageal squamous cell carcinoma (ESCC) is characterized on molecular level by, among other things, changes in protein expression. Among all proteins related to inflammation, cytokines may be implicated as possible biological markers of esophageal cancer. These biomarkers, near imaging techniques, may be helpful in diagnosis and monitoring therapy in ESCC patients. This review demonstrates findings of researches on dysregulation of cytokines in ESCC and their clinical and pathological implications. Articles on cytokines were selected according to the following criteria: (i) the study was performed at protein level, (ii) the differences in cytokines expression or concentration were detected in tissues or serum from ESCC patients, (iii) the alterations of cytokines levels were detected by: immunohistochemistry (IHC), western blot (WB) and enzyme-linked immunosorbent assay (ELISA). Members of VEGF family seem to play an essential role as potential markers in ESCC. The results of all cytokines researches are promising but further studies are necessary to establish the biological significance of these peptydes in ESCC, their potential usefulness for early diagnosis, pre- and postoperative prognosis and monitoring of the respond to chemo- and radiotherapy of cancer patients.
Collapse
|
26
|
The Multifaceted Roles Neutrophils Play in the Tumor Microenvironment. CANCER MICROENVIRONMENT 2014; 8:125-58. [PMID: 24895166 DOI: 10.1007/s12307-014-0147-5] [Citation(s) in RCA: 301] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/01/2014] [Accepted: 05/19/2014] [Indexed: 02/06/2023]
Abstract
Neutrophils are myeloid cells that constitute 50-70 % of all white blood cells in the human circulation. Traditionally, neutrophils are viewed as the first line of defense against infections and as a major component of the inflammatory process. In addition, accumulating evidence suggest that neutrophils may also play a key role in multiple aspects of cancer biology. The possible involvement of neutrophils in cancer prevention and promotion was already suggested more than half a century ago, however, despite being the major component of the immune system, their contribution has often been overshadowed by other immune components such as lymphocytes and macrophages. Neutrophils seem to have conflicting functions in cancer and can be classified into anti-tumor (N1) and pro-tumor (N2) sub-populations. The aim of this review is to discuss the varying nature of neutrophil function in the cancer microenvironment with a specific emphasis on the mechanisms that regulate neutrophil mobilization, recruitment and activation.
Collapse
|
27
|
Pei XJ, Wu TT, Li B, Tian XY, Li Z, Yang QX. Increased expression of macrophage migration inhibitory factor and DJ-1 contribute to cell invasion and metastasis of nasopharyngeal carcinoma. Int J Med Sci 2014; 11:106-15. [PMID: 24396292 PMCID: PMC3880997 DOI: 10.7150/ijms.7264] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 12/09/2013] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND AND AIM Both macrophage migration inhibitory factor (MIF) and DJ-1 protein have been shown to relate with cell invasion and metastasis in tumors. However, the role of DJ-1 in invasion and metastasis of nasopharyngeal carcinoma (NPC) and its relation to MIF expression in NPC are not fully understood. The aim of present study is to determine whether or not MIF and DJ-1 are correlated with tumor invasion and influence a worse outcome in NPC, as well as its related mechanism. METHODS 125 cases of NPC and 45 normal tissues of nasopharynx were collected. The expression of MIF and DJ-1 in tissue microarray was evaluated by immunohistochemical staining. Correlation between immunostainings and clinicopathological parameters, as well as the follow-up data of patients, was analyzed statistically. The association of MIF and DJ-1 with cell invasion and migration in NPC cell line were evaluated by small interfering RNA (siRNA) transfection, invasion assay and Western blotting. RESULTS MIF and DJ-1 staining was diffused and strong in tumor cells, whereas they were generally weaker and less common in normal lining epithelia of nasopharynx. High MIF expression in tumor cells (71.2%, 89/125 cases) were significantly associated with advanced clinical stage, lymph node metastasis, and worse prognosis of NPC patients. High expression of DJ-1 (75.2%, 94/125 cases) were closely correlated to lymph node metastasis and MIF high-expression. Only MIF high expression (P = 0.010) and lymph node metastasis (P = 0.004) emerged as strong independent prognostic factors for overall survival of NPC patients. In vitro, down-regulated expression of DJ-1 in NPC cell lines by siRNA was observed to reduce cell migration and invasion potential, however, exogenous MIF promoted cells invasion. CONCLUSIONS The data provided evidence that increased expression of MIF and DJ-1 induced cell invasion and metastasis of NPC, supporting the idea that MIF and DJ-1 may play important roles as regulators in the progression of NPC.
Collapse
Affiliation(s)
- Xiao-Juan Pei
- 1. Department of Pathology, Huizhou Municipal Central Hospital, 41 Eling Road North, Huizhou 516001, China
| | - Tong-Tong Wu
- 1. Department of Pathology, Huizhou Municipal Central Hospital, 41 Eling Road North, Huizhou 516001, China
| | - Bin Li
- 2. Department of pathology, the First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou 510080, China
| | - Xiao-Ying Tian
- 3. School of Chinese Medicine, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Hong Kong, China
| | - Zhi Li
- 2. Department of pathology, the First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou 510080, China
| | - Qing-Xu Yang
- 1. Department of Pathology, Huizhou Municipal Central Hospital, 41 Eling Road North, Huizhou 516001, China
| |
Collapse
|
28
|
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
|
29
|
Liu N, Jiang N, Guo R, Jiang W, He QM, Xu YF, Li YQ, Tang LL, Mao YP, Sun Y, Ma J. MiR-451 inhibits cell growth and invasion by targeting MIF and is associated with survival in nasopharyngeal carcinoma. Mol Cancer 2013; 12:123. [PMID: 24138931 PMCID: PMC3853142 DOI: 10.1186/1476-4598-12-123] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 10/10/2013] [Indexed: 12/19/2022] Open
Abstract
Background MiRNAs play important roles in diverse biological processes including tumorigenesis. However, little is known about the function and mechanism of miR-451 in nasopharyngeal carcinoma (NPC). Methods Quantitative RT-PCR was used to quantify miR-451 expression in NPC cell lines and clinical tissues. Kaplan-Meier curves were used to estimate the association between miR-451 expression and survival. The MTT, colony formation, Transwell migration and invasion assays, and a xenograft model were performed. A miR-451 target was confirmed using luciferase reporter assays, quantitative RT-PCR, and Western blotting. Results MiR-451 was significantly downregulated in NPC cell lines and clinical tissues (P < 0.01). Patients with low expression of miR-451 had poorer overall survival (HR, 1.98; 95% CI, 1.16-3.34; P = 0.01) and disease-free survival (HR, 1.68; 95% CI, 1.07-2.62; P = 0.02) than patients with high expression. MiR-451 was an independent prognostic factor in NPC in multivariate Cox regression analysis. Ectopic expression of miR-451 suppressed cell viability, colony formation, and cell migration and invasion in vitro, and inhibited xenograft tumor growth in vivo. MIF was verified as a direct target of miR-451, and MIF regulated NPC cell growth and invasion. Conclusions The newly identified miR-451/MIF pathway provides insight into NPC initiation and progression, and may represent a novel therapeutic target.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Jun Ma
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou People's Republic of China.
| |
Collapse
|
30
|
Sun H, Choo-Wing R, Sureshbabu A, Fan J, Leng L, Yu S, Jiang D, Noble P, Homer RJ, Bucala R, Bhandari V. A critical regulatory role for macrophage migration inhibitory factor in hyperoxia-induced injury in the developing murine lung. PLoS One 2013; 8:e60560. [PMID: 23637753 PMCID: PMC3639272 DOI: 10.1371/journal.pone.0060560] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Accepted: 02/27/2013] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND The role and mechanism of action of MIF in hyperoxia-induced acute lung injury (HALI) in the newborn lung are not known. We hypothesized that MIF is a critical regulatory molecule in HALI in the developing lung. METHODOLOGY We studied newborn wild type (WT), MIF knockout (MIFKO), and MIF lung transgenic (MIFTG) mice in room air and hyperoxia exposure for 7 postnatal (PN) days. Lung morphometry was performed and mRNA and protein expression of vascular mediators were analyzed. RESULTS MIF mRNA and protein expression were significantly increased in WT lungs at PN7 of hyperoxia exposure. The pattern of expression of Angiopoietin 2 protein (in MIFKO>WT>MIFTG) was similar to the mortality pattern (MIFKO>WT>MIFTG) in hyperoxia at PN7. In room air, MIFKO and MIFTG had modest but significant increases in chord length, compared to WT. This was associated with decreased expression of Angiopoietin 1 and Tie 2 proteins in the MIFKO and MIFTG, as compared to the WT control lungs in room air. However, on hyperoxia exposure, while the chord length was increased from their respective room air controls, there were no differences between the 3 genotypes. CONCLUSION These data point to the potential roles of Angiopoietins 1, 2 and their receptor Tie2 in the MIF-regulated response in room air and upon hyperoxia exposure in the neonatal lung.
Collapse
Affiliation(s)
- Huanxing Sun
- Department of Pediatrics, Yale University, New Haven, Connecticut, United States of America
| | - Rayman Choo-Wing
- Department of Pediatrics, Yale University, New Haven, Connecticut, United States of America
| | - Angara Sureshbabu
- Department of Pediatrics, Yale University, New Haven, Connecticut, United States of America
| | - Juan Fan
- Department of Medicine, Yale University, New Haven, Connecticut, United States of America
| | - Lin Leng
- Department of Medicine, Yale University, New Haven, Connecticut, United States of America
| | - Shuang Yu
- Department of Medicine, Yale University, New Haven, Connecticut, United States of America
| | - Dianhua Jiang
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Paul Noble
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Robert J. Homer
- Department of Pathology, Yale University, New Haven, Connecticut, United States of America
| | - Richard Bucala
- Department of Medicine, Yale University, New Haven, Connecticut, United States of America
| | - Vineet Bhandari
- Department of Pediatrics, Yale University, New Haven, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
31
|
Kindt N, Preillon J, Kaltner H, Gabius HJ, Chevalier D, Rodriguez A, Johnson BD, Megalizzi V, Decaestecker C, Laurent G, Saussez S. Macrophage migration inhibitory factor in head and neck squamous cell carcinoma: clinical and experimental studies. J Cancer Res Clin Oncol 2013; 139:727-37. [PMID: 23354841 DOI: 10.1007/s00432-013-1375-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Accepted: 10/22/2012] [Indexed: 01/14/2023]
Abstract
PURPOSE The present in vivo/in vitro study was undertaken in order to evaluate the importance of macrophage migration inhibitory factor (MIF) in the progression of head and neck squamous cell carcinoma (HNSCC). METHODS Tumor tissue expression (MIF immunostaining) and plasma levels (ELISA) of MIF were determined in HNSCC patients and correlated with tumor recurrence and metastasis, and overall survival. Furthermore, the impact of MIF expression on cell proliferation and anticancer drug sensitivity was examined in murine squamous carcinoma cell line SCCVII after MIF knockdown (MIF-KD). RESULTS As revealed by quantitative analysis of MIF immunostaining, tumor progression was accompanied by an increase in mean optical density (MOD) and labeling index (LI). Likewise, an elevation of MIF serum levels was noted in HNSCC patients (n = 66) versus healthy individuals (n = 16). Interestingly, comparison of laryngeal carcinoma patients on the basis of MIF tissue expression (high expression, LI ≥ 47, versus low expression, LI < 47) disclosed a significant difference between disease-free survival curves for local and nodal recurrence, and overall survival curve. In vitro, MIF knockdown in murine SCCVII cells resulted in reduced cell proliferation and a decrease in cell motility. In mice inoculated with SCCVII cells, MIF-KD tumors grew more slowly and also appeared more sensitive to chemotherapy. CONCLUSIONS Both clinical observations and experimental data suggest that MIF plays a pivotal role in the progression of HNSCC.
Collapse
Affiliation(s)
- Nadège Kindt
- Laboratory of Anatomy and Cellular Biology, Faculty of Medicine and Pharmacy, University of Mons, Pentagone 2A, 6 Ave du Champ de Mars, 7000 Mons, Belgium
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
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
|
33
|
Zhao Y, Schetter AJ, Yang GB, Nguyen G, Mathé EA, Li P, Cai H, Yu L, Liu F, Hang D, Yang H, Wang XW, Ke Y, Harris CC. microRNA and inflammatory gene expression as prognostic marker for overall survival in esophageal squamous cell carcinoma. Int J Cancer 2012; 132:2901-9. [PMID: 23175214 DOI: 10.1002/ijc.27954] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Accepted: 10/05/2012] [Indexed: 12/11/2022]
Abstract
MicroRNAs (miRNAs) and inflammatory genes have a role in the initiation and development of esophageal squamous cell carcinoma (ESCC). In our study, we examined the potential of using miRNA and inflammatory gene expression patterns as prognostic classifiers for ESCC. Five miRNAs and 25 inflammatory-related genes were measured by quantitative reverse transcriptase PCR in tumor tissues and adjacent noncancerous tissues from 178 Chinese patients with ESCC. The expression levels of miR-21 (p = 0.027), miR-181b (p = 0.002) and miR-146b (p = 0.021) in tumor tissue and miR-21 (p = 0.003) in noncancerous tissue were associated with overall survival of patients. These data were combined to generate a miRNA risk score that was significantly associated with worse prognosis (p = 0.0001), suggesting that these miRNAs may be useful prognostic classifiers for ESCC. To construct an inflammatory gene prognostic classifier, we divided the population into training (n = 124) and test cohorts (n = 54). The expression levels of CRY61, CTGF and IL-18 in tumor tissue and VEGF in adjacent noncancerous tissue were modestly associated with prognosis in the training cohort |Z-score| > 1.5 and were subsequently used to construct a Cox regression-based inflammatory risk score (IRS). IRS was significantly associated with survival in both the training cohort (p = 0.002) and the test cohort (p = 0.005). Furthermore, Cox regression models combining both miRNA risk score and IRS performed significantly better than models with either alone (p < 0.001 likelihood ratio test). Therefore, miRNA and inflammatory gene expression patterns, alone or in combination, have potential as prognostic classifiers for ESCC and may help to guide therapeutic decisions.
Collapse
Affiliation(s)
- Yiqiang Zhao
- Laboratory of Human Carcinogenesis, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Merk M, Mitchell RA, Endres S, Bucala R. D-dopachrome tautomerase (D-DT or MIF-2): doubling the MIF cytokine family. Cytokine 2012; 59:10-7. [PMID: 22507380 PMCID: PMC3367028 DOI: 10.1016/j.cyto.2012.03.014] [Citation(s) in RCA: 126] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Revised: 03/09/2012] [Accepted: 03/16/2012] [Indexed: 12/12/2022]
Abstract
D-dopachrome tautomerase (D-DT) is a newly described cytokine and a member of the macrophage migration inhibitory factor (MIF) protein superfamily. MIF is a broadly expressed pro-inflammatory cytokine that regulates both the innate and the adaptive immune response. MIF activates the MAP kinase cascade, modulates cell migration, and counter-acts the immunosuppressive effects of glucocorticoids. For many cell types, MIF also acts as an important survival or anti-apoptotic factor. Circulating MIF levels are elevated in the serum in different infectious and autoimmune diseases, and neutralization of the MIF protein via antibodies or small molecule antagonists improves the outcome in numerous animal models of human disease. Recently, a detailed investigation of the biological role of the closely homologous protein D-DT, which is encoded by a gene adjacent to MIF, revealed an overlapping functional spectrum with MIF. The D-DT protein also is present in most tissues and circulates in serum at similar concentrations as MIF. D-DT binds the MIF cell surface receptor complex, CD74/CD44, with high affinity and induces similar cell signaling and effector functions. Furthermore, an analysis of the signaling properties of the two proteins showed that they work cooperatively, and that neutralization of D-DT in vivo significantly decreases inflammation. In this review, we highlight the similarities and differences between MIF and D-DT, which we propose to designate "MIF-2", and discuss the implication of D-DT/MIF-2 expression for MIF-based therapies.
Collapse
Affiliation(s)
- Melanie Merk
- Center of Integrated Protein Science Munich, Division of Clinical Pharmacology, LMU Munich, Germany
| | | | - Stefan Endres
- Center of Integrated Protein Science Munich, Division of Clinical Pharmacology, LMU Munich, Germany
| | - Richard Bucala
- Internal Medicine Yale University School of Medicine, New Haven, CT, 06520
| |
Collapse
|
35
|
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
|
36
|
Expression of macrophage migration inhibitory factor and CD74 in cervical squamous cell carcinoma. Int J Gynecol Cancer 2012; 21:1004-12. [PMID: 21792010 DOI: 10.1097/igc.0b013e31821c45b7] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
OBJECTIVE Macrophage migration inhibitory factor (MIF) and CD74 emerge as important players in pathogenesis and angiogenesis of several types of malignant tumors. The purpose of this study was to evaluate the expression of MIF and CD74 in cervical squamous cell carcinoma and explore the potential roles they play in cervical tumor angiogenesis. METHODS Macrophage migration inhibitory factor and CD74 expression was assessed by immunohistochemistry in 209 cases with various degrees of cervical epithelial lesions, including 40 normal cervical epithelia, 43 mild cervical intraepithelial neoplasia 1 (CIN 1), 41 moderate-severe cervical intraepithelial neoplasia (CIN 2 to 3), and 85 cervical squamous cell carcinomas (SCCs). CD34 staining was used for counting microvessel density. Semiquantitative reverse transcription polymerase chain reaction and Western blot were used to detect messenger RNA and protein levels of MIF and CD74 in normal and malignant cervical tissues and cervical cancer cell lines SiHa and C-33A. The concentration of vascular endothelial growth factor (VEGF) in the conditioned media of cervical cancer cells was analyzed by enzyme-linked immunosorbent assay. RESULTS Immunohistochemical analysis showed that MIF and CD74 expression was significantly higher in CIN than in the normal samples and higher in SCC than in CIN. The overexpression of MIF was correlated with deep stromal infiltration but not with the other clinicopathologic features of SCC. Correlation analyses revealed that MIF was positively related to CD74, and both protein levels were associated with microvessel density. Exogenous MIF induced VEGF secretion in SiHa and C-33A cells in a dose-dependent manner, which can be inhibited by MIF-specific inhibitor (ISO-1) or anti-CD74 antibody. CONCLUSION Overexpression of MIF and CD74 in SCC and its precancerous lesions and the up-regulation of VEGF secretion in cervical cancer cells indicate that MIF and CD74 may play critical roles in the pathogenesis and angiogenesis of cervical cancer.
Collapse
|
37
|
Wang XB, Tian XY, Li Y, Li B, Li Z. Elevated expression of macrophage migration inhibitory factor correlates with tumor recurrence and poor prognosis of patients with gliomas. J Neurooncol 2011; 106:43-51. [PMID: 21725855 DOI: 10.1007/s11060-011-0640-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Accepted: 06/17/2011] [Indexed: 10/18/2022]
Abstract
Macrophage migration inhibitory factor (MIF) plays a critical role in tumorigenesis. We aim to examine the association of MIF with tumor recurrence and survival of gliomas, and to determine whether MIF is a valuable prognostic predictor for glioma patients. The expression of MIF and interleukin 8 (IL-8) was evaluated in 36 high-grade gliomas (20 glioblastoma multiforme, 13 anaplastic astrocytoma, and 3 anaplastic oligoastrocytoma) and 32 low-grade gliomas (18 fibrillary astrocytoma, 5 pilocytic astrocytoma, 5 oligodendroglioma, 3 ependymoma and 1 pleomorphic xanthoastrocytoma) by immunostaining. Intratumoral microvessel density (IMD) of tumors in relation to immunostainings and clinicopathological factors were analyzed statistically as well as the follow-up data of patients. High expression of both MIF (58.8%) and IL-8 (52.9%) was significantly associated with high-grade gliomas and increased microvessels in tumors, but only high expression of MIF was closely related to tumor recurrence (P = 0.001). High expression of IL-8 exhibited a close correlation with high expression of MIF in tumors (P = 0.001). Histological grading, high expression of MIF and IL-8 correlated with patients' overall survival in univariate analysis. However, only histological grading and MIF expression exhibited a relationship with survival of patients as independent prognostic factors of glioma by multivariate analysis; the hazard ratios were 28.012 (P = 0.001) and 11.782 (P = 0.001), respectively. Elevated production of MIF in glioma tumor cells may contribute to tumor recurrence and a worse prognosis. MIF may serve as an independent predictive factor for prognosis of glioma patients.
Collapse
Affiliation(s)
- Xiao-Bing Wang
- Department of Pathology, Guangdong General Hospital, 106, Zhongshan Road II, Guangzhou, China
| | | | | | | | | |
Collapse
|
38
|
Brooks HD, Glisson BS, Bekele BN, Ginsberg LE, El-Naggar A, Culotta KS, Takebe N, Wright J, Tran HT, Papadimitrakopoulou VA. Phase 2 study of dasatinib in the treatment of head and neck squamous cell carcinoma. Cancer 2011; 117:2112-9. [PMID: 21523723 PMCID: PMC3117018 DOI: 10.1002/cncr.25769] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2010] [Accepted: 09/13/2010] [Indexed: 11/10/2022]
Abstract
BACKGROUND Treatment options for patients with advanced head and neck squamous cell carcinoma (HNSCC) are scarce. This phase 2 study was conducted to evaluate the safety, tolerability, pharmacokinetics, and efficacy of dasatinib in this setting. METHODS Patients with recurrent and/or metastatic HNSCC after platinum-based therapy were treated with dasatinib either orally or via percutaneous feeding gastrostomy (PFG). Primary endpoints were 12-week progression-free survival (PFS) and objective response rate with a 2-stage design and early withdrawal if the 12-week PFS rate was ≤20% and no patients had an objective response (OR). Forty-nine serum cytokines and angiogenic factors (CAFs) were analyzed from treated patients. RESULTS Of the 15 patients enrolled, 12 were evaluable for response, and all patients were evaluable for toxicity. No OR was observed and 2 patients (16.7%) had stable disease (SD) at 8 weeks. The median treatment duration was 59 days, the median time to disease progression was 3.9 weeks, and the median survival was 26 weeks. One patient required a dose reduction, 3 patients required dose interruptions, and 4 patients were hospitalized for toxicity. Dasatinib inhibited c-Src both when administered orally and via PFG. Greater mean drug exposure, decreased half-life, and greater maximum concentration were observed in patients receiving dasatinib via PFG. Eleven baseline CAFs were associated with treatment outcome and 1 CAF, macrophage migration inhibitory factor, was found to be differentially modulated in correlation with SD versus disease progression. CONCLUSIONS Single-agent dasatinib failed to demonstrate significant activity in patients with advanced HNSCC, despite c-Src inhibition. The toxicity profile was consistent with that reported in other solid tumors, and the drug can be given via PFG tube.
Collapse
Affiliation(s)
- Heather D. Brooks
- Division of Cancer Medicine. Pharmacology Research at the University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA
| | - Bonnie S. Glisson
- Department of Thoracic and Head and Neck Medical Oncology, Pharmacology Research at the University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA
| | - B. Nebiyou Bekele
- Department of Biostatistics, Pharmacology Research at the University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA
| | - Lawrence E. Ginsberg
- Department of Diagnostic Radiology, Pharmacology Research at the University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA
| | - Adel El-Naggar
- Department of Pathology, Pharmacology Research at the University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA
| | - Kirk S. Culotta
- Department of Pharmacy - Pharmacology Research at the University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA
| | - Naoko Takebe
- Cancer Therapy Evaluation Program, DCTD, NCI, NIH
| | - John Wright
- Cancer Therapy Evaluation Program, DCTD, NCI, NIH
| | - Hai T. Tran
- Department of Thoracic and Head and Neck Medical Oncology, Pharmacology Research at the University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA
| | - Vassiliki A. Papadimitrakopoulou
- Department of Thoracic and Head and Neck Medical Oncology, Pharmacology Research at the University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
39
|
Zhao YM, Wang L, Dai Z, Wang DD, Hei ZY, Zhang N, Fu XT, Wang XL, Zhang SC, Qin LX, Tang ZY, Zhou J, Fan J. Validity of plasma macrophage migration inhibitory factor for diagnosis and prognosis of hepatocellular carcinoma. Int J Cancer 2011; 129:2463-72. [PMID: 21213214 DOI: 10.1002/ijc.25918] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2010] [Accepted: 12/22/2010] [Indexed: 11/05/2022]
Abstract
We performed our study to determine whether plasma macrophage migration inhibitory factor (MIF) levels have diagnostic and prognostic value in hepatocellular carcinoma (HCC) patients. Enzyme-linked immunosorbent assay (ELISA) and immunohistochemistry were used to measure the expression of MIF in plasma and tissues, respectively. Plasma MIF levels were compared to HCC occurrence, clinicopathological features and outcomes. Cutpoints of plasma MIF levels for diagnosis and prognosis were, respectively, determined by receiver operating characteristic analysis and X-tile in corresponding training cohort, and then were confirmed in the validation cohort. The postoperative plasma MIF levels of HCC patients were detected in an independent cohort (80 HCC patients). As a result, MIF expression in situ was mainly observed in the cytoplasm of HCC cells. Intratumoral MIF expression was positively correlated with plasma MIF levels (r = 0.759, p < 0.001). Compared to serum α-fetoprotein (AFP), plasma MIF had a higher diagnostic value for discrimination of HCC from controls at 35.3 ng/ml. With determined cutpoints, plasma MIF levels demonstrated a significant association with overall survival (OS) and disease-free survival (DFS) of HCC patients even in patients with normal serum AFP levels and Tumor Node Metastasis (TNM) stage I. In addition, the plasma MIF levels were identified as an independent factor for OS [hazard ratio (HR) = 1.754; p = 0.012] and DFS (HR = 2.121; p < 0.001). Plasma MIF levels decreased markedly within 30 days after tumor resection (p < 0.001). Therefore, plasma MIF levels have potential as a diagnostic and prognostic factor for HCC.
Collapse
Affiliation(s)
- Yi-Ming Zhao
- Liver Cancer Institute, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Liao B, Zhong BL, Li Z, Tian XY, Li Y, Li B. Macrophage migration inhibitory factor contributes angiogenesis by up-regulating IL-8 and correlates with poor prognosis of patients with primary nasopharyngeal carcinoma. J Surg Oncol 2011; 102:844-51. [PMID: 20872800 DOI: 10.1002/jso.21728] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
BACKGROUND AND OBJECTIVES We aim at the association of macrophage migration inhibitory factor (MIF) with neovascularization and survival of nasopharyngeal carcinoma (NPC), and determine whether MIF is a valuable prognostic predictor for NPC patients. METHODS One hundred and forty one cases of NPC and 25 normal tissues of nasopharynx were collected. The expression of MIF and interleukin 8 (IL-8) was evaluated in tissues microarray by immunostaining. Intratumoral microvessel density (IMD) in relation to immunostainings and clinicopathological factors were analyzed statistically as well as the follow-up data of patients. RESULTS High-expression of both MIF (69.5%) and IL-8 (56.0%) were significantly associated with increased microvessels and lymph node metastasis. High-expression of MIF, IL-8 and higher level of IMD were correlated with either patients' overall survival or disease-specific survival in univariate analysis, but only angiogenesis and lymph node status exhibited in relation to survival of patients as independent prognostic factor of NPC by multivariate analysis. In addition, high-expression of MIF and higher level of IMD were closely associated with locoregional failure of NPC patients. CONCLUSIONS MIF may contribute to lymph node metastasis in NPC by inducing angiogenesis through the way of upregulation of IL-8 expression in an autocrine EBV-independent pathway.
Collapse
Affiliation(s)
- Bing Liao
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | | | | | | | | | | |
Collapse
|
41
|
The role of macrophage migration inhibitory factor in ocular surface disease pathogenesis after chemical burn in the murine eye. Mol Vis 2010; 16:2402-11. [PMID: 21152395 PMCID: PMC2994759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2010] [Accepted: 11/11/2010] [Indexed: 11/14/2022] Open
Abstract
PURPOSE To evaluate the role of macrophage migration inhibitory factor (MIF) in the wound healing process following severe chemical burns to the ocular surface. METHODS Chemical burning of the ocular surface was induced in mice (C57BL/6) via the application of 0.1 M NaOH. Macrophage migration inhibitory factor (MIF), tumor necrosis factor-α (TNF-α), and interleukin-1β (IL-1β) mRNA expression in the ocular surface and lacrimal gland was evaluated via real-time reverse transcription PCR on days 2, 7, and 30 after induction of the chemical burn. The expression of MIF protein in the ocular surface and lacrimal gland was evaluated via western blot analysis. Immunohistochemical staining was conducted to detect MIF and vasculoendothelial growth factor in the cornea during the wound healing process. The angiogenic role of MIF was further evaluated using an 8-0 polyglactin suture technique to induce corneal neovascularization. RESULTS MIF, TNF-α, and IL-1β mRNA expression were elevated significantly in the ocular surface up to day 30 after chemical burn induction. TNF-α alone was elevated in the lacrimal gland. MIF protein elevation was confirmed via western blot analysis, and the spatial similarity of MIF and VEGF expression in the cornea was noted during the wound healing process. 8-0 polyglactin sutures soaked in MIF induced significantly higher numbers of new vessels on the mouse cornea after 7 days (p=0.003, Mann-Whitney test). CONCLUSIONS These findings indicate that MIF performs a crucial role in wound healing on the ocular surface after the induction of chemical burns.
Collapse
|
42
|
Bruyère C, Lonez C, Duray A, Cludts S, Ruysschaert JM, Saussez S, Yeaton P, Kiss R, Mijatovic T. Considering temozolomide as a novel potential treatment for esophageal cancer. Cancer 2010; 117:2004-16. [DOI: 10.1002/cncr.25687] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2010] [Revised: 07/13/2010] [Accepted: 09/07/2010] [Indexed: 12/30/2022]
|
43
|
Grieb G, Merk M, Bernhagen J, Bucala R. Macrophage migration inhibitory factor (MIF): a promising biomarker. ACTA ACUST UNITED AC 2010; 23:257-64. [PMID: 20520854 DOI: 10.1358/dnp.2010.23.4.1453629] [Citation(s) in RCA: 164] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Macrophage migration inhibitory factor (MIF) is an immunoregulatory cytokine, the effect of which on arresting random immune cell movement was recognized several decades ago. Despite its historic name, MIF also has a direct chemokine-like function and promotes cell recruitment. Multiple clinical studies have indicated the utility of MIF as a biomarker for different diseases that have an inflammatory component; these include systemic infections and sepsis, autoimmune diseases, cancer, and metabolic disorders such as type 2 diabetes and obesity. The identification of functional promoter polymorphisms in the MIF gene (MIF) and their association with the susceptibility or severity of different diseases has not only served to validate MIF's role in disease development but also opened the possibility of using MIF genotype information to better predict risk and outcome. In this article, we review the clinical data of MIF and discuss its potential as a biomarker for different disease applications.
Collapse
Affiliation(s)
- Gerrit Grieb
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.
| | | | | | | |
Collapse
|
44
|
Dessein AF, Stechly L, Jonckheere N, Dumont P, Monté D, Leteurtre E, Truant S, Pruvot FR, Figeac M, Hebbar M, Lecellier CH, Lesuffleur T, Dessein R, Grard G, Dejonghe MJ, de Launoit Y, Furuichi Y, Prévost G, Porchet N, Gespach C, Huet G. Autocrine induction of invasive and metastatic phenotypes by the MIF-CXCR4 axis in drug-resistant human colon cancer cells. Cancer Res 2010; 70:4644-54. [PMID: 20460542 DOI: 10.1158/0008-5472.can-09-3828] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Metastasis and drug resistance are major problems in cancer chemotherapy. The purpose of this work was to analyze the molecular mechanisms underlying the invasive potential of drug-resistant colon carcinoma cells. Cellular models included the parental HT-29 cell line and its drug-resistant derivatives selected after chronic treatment with either 5-fluorouracil, methotrexate, doxorubicin, or oxaliplatin. Drug-resistant invasive cells were compared with noninvasive cells using cDNA microarray, quantitative reverse transcription-PCR, flow cytometry, immunoblots, and ELISA. Functional and cellular signaling analyses were undertaken using pharmacologic inhibitors, function-blocking antibodies, and silencing by retrovirus-mediated RNA interference. 5-Fluorouracil- and methotrexate-resistant HT-29 cells expressing an invasive phenotype in collagen type I and a metastatic behavior in immunodeficient mice exhibited high expression of the chemokine receptor CXCR4. Macrophage migration-inhibitory factor (MIF) was identified as the critical autocrine CXCR4 ligand promoting invasion in drug-resistant colon carcinoma HT-29 cells. Silencing of CXCR4 and impairing the MIF-CXCR4 signaling pathways by ISO-1, pAb FL-115, AMD-3100, monoclonal antibody 12G5, and BIM-46187 abolished this aggressive phenotype. Induction of CXCR4 was associated with the upregulation of two genes encoding transcription factors previously shown to control CXCR4 expression (HIF-2alpha and ASCL2) and maintenance of intestinal stem cells (ASCL2). Enhanced CXCR4 expression was detected in liver metastases resected from patients with colon cancer treated by the standard FOLFOX regimen. Combination therapies targeting the CXCR4-MIF axis could potentially counteract the emergence of the invasive metastatic behavior in clonal derivatives of drug-resistant colon cancer cells.
Collapse
|
45
|
Abstract
One of the difficulties in using gene expression profiles to predict cancer is how to effectively select a few informative genes to construct accurate prediction models from thousands or ten thousands of genes. We screen highly discriminative genes and gene pairs to create simple prediction models involved in single genes or gene pairs on the basis of soft computing approach and rough set theory. Accurate cancerous prediction is obtained when we apply the simple prediction models for four cancerous gene expression datasets: CNS tumor, colon tumor, lung cancer and DLBCL. Some genes closely correlated with the pathogenesis of specific or general cancers are identified. In contrast with other models, our models are simple, effective and robust. Meanwhile, our models are interpretable for they are based on decision rules. Our results demonstrate that very simple models may perform well on cancerous molecular prediction and important gene markers of cancer can be detected if the gene selection approach is chosen reasonably.
Collapse
Affiliation(s)
- Xiaosheng Wang
- Department of Intelligence Science and Technology, Graduate School of Informatics, Kyoto University, Kyoto, Japan.
| | | |
Collapse
|
46
|
Bandres E, Bitarte N, Arias F, Agorreta J, Fortes P, Agirre X, Zarate R, Diaz-Gonzalez JA, Ramirez N, Sola JJ, Jimenez P, Rodriguez J, Garcia-Foncillas J. microRNA-451 regulates macrophage migration inhibitory factor production and proliferation of gastrointestinal cancer cells. Clin Cancer Res 2009; 15:2281-90. [PMID: 19318487 DOI: 10.1158/1078-0432.ccr-08-1818] [Citation(s) in RCA: 284] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE microRNAs (miRNA) are small RNAs that function as post-transcriptional regulators of gene expression. Recent evidence has shown that some miRNAs can act as oncogenes or tumor suppressors. This study was conducted to evaluate the potential association of miRNA expression with clinical outcome in patients with gastric cancer. EXPERIMENTAL DESIGN Expression of 250 human mature miRNAs was measured by real-time PCR on paraffin-embedded tumor samples of 21 patients with gastric cancer stage III uniformly treated with surgical resection followed by chemoradiation. We identified the miRNAs correlated with disease-free and overall survival times, and the results were evaluated including 24 other patients. In vitro cell proliferation and radiosensitivity studies were done to support clinical data. RESULTS The results revealed that down-regulation of miR-451 was associated with worse prognosis. miR-451 was detected by in situ hybridization in epithelial cells and showed decreased expression in gastric and colorectal cancer versus nontumoral tissues. Overexpression of miR-451 in gastric and colorectal cancer cells reduced cell proliferation and increased sensitivity to radiotherapy. Microarray and bioinformatic analysis identified the novel oncogene macrophage migration inhibitory factor (MIF) as a potential target of miR-451. In fact, overexpression of miR-451 down-regulated mRNA and protein levels of MIF and decreased expression of reporter genes with MIF target sequences. Moreover, we found a significant inverse correlation between miR-451 and MIF expression in tumoral gastric biopsies. CONCLUSIONS These findings support the role of miR-451 as a regulator of cancer proliferation and open new perspectives for the development of effective therapies for chemoradioresistant cancers.
Collapse
Affiliation(s)
- Eva Bandres
- Division of Oncology and Hepatology, Center for Applied Medical Research, Clinica Universitaria, University of Navarra, Pamplona, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Rendon BE, Willer SS, Zundel W, Mitchell RA. Mechanisms of macrophage migration inhibitory factor (MIF)-dependent tumor microenvironmental adaptation. Exp Mol Pathol 2009; 86:180-5. [PMID: 19186177 DOI: 10.1016/j.yexmp.2009.01.001] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2008] [Indexed: 01/08/2023]
Abstract
Since its activity was first reported in the mid-1960s, macrophage migration inhibitory factor (MIF) has gone from a cytokine activity modulating monocyte motility to a pleiotropic regulator of a vast array of cellular and biological processes. Studies in recent years suggest that MIF contributes to malignant disease progression on several different levels. Both circulating and intracellular MIF protein levels are elevated in cancer patients and MIF expression reportedly correlates with stage, metastatic spread and disease-free survival. Additionally, MIF expression positively correlates with angiogenic growth factor expression, microvessel density and tumor-associated neovascularization. Not coincidentally, MIF has recently been shown to contribute to tumoral hypoxic adaptation by promoting hypoxia-induced HIF-1alpha stabilization. Intriguingly, hypoxia is a strong regulator of MIF expression and secretion, suggesting that hypoxia-induced MIF acts as an amplifying factor for both hypoxia and normoxia-associated angiogenic growth factor expression in human malignancies. Combined, these findings suggest that MIF overexpression contributes to tumoral hypoxic adaptation and, by extension, therapeutic responsiveness and disease prognosis. This review summarizes recent literature on the contributions of MIF to tumor-associated angiogenic growth factor expression, neovascularization and hypoxic adaptation. We also will review recent efforts aimed at identifying and employing small-molecule antagonists of MIF as a novel approach to cancer therapeutics.
Collapse
Affiliation(s)
- Beatriz E Rendon
- JG Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| | | | | | | |
Collapse
|
48
|
He XX, Chen K, Yang J, Li XY, Gan HY, Liu CY, Coleman TR, Al-Abed Y. Macrophage migration inhibitory factor promotes colorectal cancer. Mol Med 2009; 15:1-10. [PMID: 19009023 PMCID: PMC2581606 DOI: 10.2119/molmed.2008.00107] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2008] [Accepted: 10/09/2008] [Indexed: 01/07/2023] Open
Abstract
A growing body of evidence implicates macrophage migration inhibitory factor (MIF) in tumorigenesis and metastasis. In this study, we investigated whether MIF expression was associated with clinicopathologic features of colorectal carcinoma (CRC), especially in tumors with hepatic metastasis, and whether neutralization of endogenous MIF using anti-MIF therapeutics would inhibit tumor growth and/or decrease the frequency of colorectal hepatic metastases in a mouse colon carcinoma model. The concentration of serum MIF was positively correlated with an increased risk of hepatic metastasis in human patients with CRC (R = 1.25, 95% confidence internal = 1.02-1.52, P = 0.03). MIF was also dramatically upregulated in human colorectal tissue, with 20-40 times as many MIF-positive cells found in the mucosa of patients with CRC than in normal tissue (P < 0.001 ANOVA). Moreover, in those patients with metastatic colorectal cancer in the liver, MIF-positive cells were similarly increased in the diseased hepatic tissue. This increased MIF expression was restricted to diseased tissue and not found in areas of the liver with normal morphology. In subsequent in vitro experiments, we found that addition of recombinant MIF to colonic cell lines significantly increased their invasive properties and the expression of several genes (for example, matrix metalloproteinase 9 and vascular endothelial growth factor) known to be upregulated in cancerous tissue. Finally, we treated mice that had been given CT26 colon carcinoma cell transplants with anti-MIF therapeutics--either the MIF-specific inhibitor ISO-1 or neutralizing anti-MIF antibodies--and observed a significant reduction in tumor burden relative to vehicle-treated animals. Taken together, these data demonstrate that MIF expression was not only correlated with the presence of colorectal cancer but also may play a direct role in cancer development.
Collapse
Affiliation(s)
- Xing-Xiang He
- Department of Gastroenterology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Ken Chen
- Department of Gastroenterology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Jun Yang
- Department of Epidemiology, Roswell Park Cancer Institute, Buffalo, New York, United States of America
| | - Xiao-Yu Li
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou Medical College, Guangzhou, Guangdong, China
| | - Huo-Ye Gan
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou Medical College, Guangzhou, Guangdong, China
| | - Cheng-Yong Liu
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou Medical College, Guangzhou, Guangdong, China
| | - Thomas R Coleman
- Laboratory of Medicinal Chemistry, The Feinstein Institute for Medical Research, Manhasset, New York, United States of America
| | - Yousef Al-Abed
- Laboratory of Medicinal Chemistry, The Feinstein Institute for Medical Research, Manhasset, New York, United States of America
- New York University School of Medicine, New York, New York, United States of America
- Address correspondence and reprint requests to Yousef Al-Abed, Laboratory of Medicinal Chemistry, The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY 11030. Phone: 516-562-3406; Fax: 516-562-1022; E-mail:
| |
Collapse
|
49
|
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
|
50
|
Han I, Lee MR, Nam KW, Oh JH, Moon KC, Kim HS. Expression of macrophage migration inhibitory factor relates to survival in high-grade osteosarcoma. Clin Orthop Relat Res 2008; 466:2107-13. [PMID: 18563508 PMCID: PMC2492999 DOI: 10.1007/s11999-008-0333-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2007] [Accepted: 05/19/2008] [Indexed: 01/31/2023]
Abstract
UNLABELLED Macrophage migration inhibitory factor (MIF), a proinflammatory cytokine, is implicated in many aspects of tumor progression, including cell proliferation, invasion, and angiogenesis. We asked if MIF expression predicts survival and if it is associated with angiogenesis and cell invasion in osteosarcoma. We performed immunohistochemistry for MIF expression in prechemotherapy biopsy specimens of 58 patients with osteosarcoma. To investigate the role of MIF in angiogenesis, microvessel density was measured and compared with MIF expression. We also treated osteosarcoma cell lines (U2-OS and MG63) with MIF and measured vascular endothelial growth factor, a potent proangiogenic factor, by enzyme-linked immunosorbent assay. To study the role of MIF in cell invasion, Boyden chamber assay was performed after knockdown of MIF by short interfering RNA. MIF independently predicted overall survival and metastasis-free survival. MIF expression correlated with microvessel density and induced a dose-dependent increase in vascular endothelial growth factor. Knockdown of MIF by short interfering RNA resulted in decreased cell invasion. These results suggest MIF could serve as a prognostic marker and a potential therapeutic target for osteosarcoma. LEVEL OF EVIDENCE Level II, prognostic study. See the Guidelines for Authors for a complete description of levels of evidence.
Collapse
Affiliation(s)
- Ilkyu Han
- Department of Orthopaedic Surgery, Seoul National University College of Medicine, 101 Daehangno, Jongno-gu, 110-744 Seoul, Korea
| | - Mi Ra Lee
- Department of Orthopaedic Surgery, Seoul National University College of Medicine, 101 Daehangno, Jongno-gu, 110-744 Seoul, Korea ,Clinical Research Institute of Seoul National University Hospital, Seoul, Korea
| | - Kwang Woo Nam
- Department of Orthopedic Surgery, Cheju National University, Jeju-si, Jeju-do Korea
| | - Joo Han Oh
- Department of Orthopaedic Surgery, Seoul National University College of Medicine, 101 Daehangno, Jongno-gu, 110-744 Seoul, Korea ,Seoul National University Bundang Hospital, Seongnam-si, Gyeonggi-do Korea
| | - Kyung Chul Moon
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
| | - Han-Soo Kim
- Department of Orthopaedic Surgery, Seoul National University College of Medicine, 101 Daehangno, Jongno-gu, 110-744 Seoul, Korea
| |
Collapse
|