1
|
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
|
2
|
Eletr LF, Ibnouf SH, Salih TA, Ibrahim HI, Mustafa MI, Alhashmi NA, Alfaki M. Comprehensive Analysis Reveals Deoxyribonuclease 1 as a Potential Prognostic and Diagnostic Biomarker in Human Cancers. Cureus 2024; 16:e56171. [PMID: 38618458 PMCID: PMC11015913 DOI: 10.7759/cureus.56171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2024] [Indexed: 04/16/2024] Open
Abstract
BACKGROUND Deoxyribonuclease 1 (DNASE1) is an important gene associated with several cancers, including liver, bladder, and gastric cancer. It has been linked to autoimmune illnesses, including systemic lupus erythematosus, which may lead to cancer formation. However, the role of DNASE1 in cancer has not been studied. MATERIALS AND METHODS We performed a pan-cancer analysis using bioinformatics tools, including Tumor Immune Estimation Resource (TIMER), Gene Expression Profiling Interactive Analysis (GEPIA), and University of Alabama at Birmingham Cancer Data Analysis Portal (UALCAN) databases, Kaplan-Meier plotter, and cBioPortal, to investigate the expression of DNASE1 across various cancers as well as its association with immune infiltration and genetic alterations. Public datasets were used to validate DNASE1 expression in kidney renal clear cell carcinoma (KIRC) and kidney papillary renal cell carcinoma (KIRP) samples. RESULTS DNASE1 was found to be highly expressed in many cancers, such as bladder urothelial carcinoma (BLCA), breast invasive carcinoma (BRCA), head and neck squamous cell carcinoma (HNSC), and was lowly expressed in other cancers, including KIRC, KIRP, and thyroid carcinoma (THCA). Additionally, TIMER results showed an association of DNASE1 with immune cell infiltration in KIRC and KIRP. Survival analysis indicated that high DNASE1 expression was associated with poor prognosis in KIRC. We also discovered that altered DNASE1 expression was related to poor prognosis in The Cancer Genome Atlas (TCGA) tumors. CONCLUSION DNASE1 could potentially be used as a prognostic and diagnostic biomarker for KIRC and as a diagnostic biomarker for KIRP.
Collapse
Affiliation(s)
- Loai F Eletr
- Computing and Bioinformatics, Faculty of Science, Port Said University, Port Said, EGY
| | | | | | - Hadba I Ibrahim
- Zoology, Faculty of Science, University of Khartoum, Khartoum, SDN
| | - Mustafa I Mustafa
- Internal Medicine, Sudan Medical Specialization Board, Khartoum, SDN
- Clinical Immunology, Sudan Medical Specialization Board, Khartoum, SDN
- Neurology, King Abdulaziz Medical City Jeddah, Jeddah, SAU
| | | | | |
Collapse
|
3
|
Zhou S, Cao C, Hu J. Long Non-Coding RNA Small Nucleolar RNA Host Gene 4 Induced by Transcription Factor SP1 Promoted the Progression of Nasopharyngeal Carcinoma Through Modulating microRNA-510-5p/Centromere Protein F Axis. Biochem Genet 2023; 61:1967-1986. [PMID: 36899270 DOI: 10.1007/s10528-023-10351-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 02/15/2023] [Indexed: 03/12/2023]
Abstract
Long non-coding RNAs (LncRNAs) are implicated with tumorigenesis and the development of nasopharyngeal carcinoma (NPC). Previous studies suggested that long non-coding RNA small nucleolar RNA host gene 4 (SNHG4) exerted oncogenic roles in various cancers. However, the function and molecular mechanism of SNHG4 in NPC have not been investigated. In our study, it was confirmed that the SNHG4 level was enriched in NPC tissues and cells. Functional assays indicated that SNHG4 depletion inhibited the proliferation and metastasis but promoted apoptosis of NPC cells. Furthermore, we identified miR-510-5p as a downstream gene of SNHG4 in NPC cells and SNHG4 upregulated CENPF expression by binding to miR-510-5p. Moreover, there was a positive (or negative) association between CENPF and SNHG4 (or miR-510-5p) expression in NPC. In addition, rescue experiments verified that CENPF overexpression or miR-510-5p silencing abrogated inhibitory effects on NPC tumorigenesis caused by SNHG4 deficiency. The study demonstrated that SNHG4 promoted NPC progression via miR-510-5p/CENPF axis, providing a novel potential therapeutic target for NPC treatments.
Collapse
Affiliation(s)
- Shao Zhou
- Department of Otorhinolaryngology, The Affiliated People's Hospital of Ningbo University, No. 251 East Baizhang Road, Ningbo, 315000, Zhejiang, China.
| | - Cheng Cao
- Department of Otorhinolaryngology, The Affiliated People's Hospital of Ningbo University, No. 251 East Baizhang Road, Ningbo, 315000, Zhejiang, China
| | - Jiandao Hu
- Department of Otorhinolaryngology, The Affiliated People's Hospital of Ningbo University, No. 251 East Baizhang Road, Ningbo, 315000, Zhejiang, China
| |
Collapse
|
4
|
Liang T, Xiao D, Lu S, Ye X, Xiao Z. Prognostic Value of a Serum Panel of Inflammatory Factors in Non-Metastatic Nasopharyngeal Carcinoma Patients Undergoing Radical Radiotherapy with Adjuvant Chemotherapy. Cancer Manag Res 2022; 14:2763-2772. [PMID: 36148318 PMCID: PMC9488185 DOI: 10.2147/cmar.s371922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 08/17/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose To evaluate the prognostic value of interleukin (IL)-6, IL-8, granulocyte-macrophage colony-stimulating factor (GM-CSF), leukemia inhibitory factor (LIF), and macrophage migration inhibitory factor (MIF) in non-metastatic nasopharyngeal carcinoma (NPC) patients undergoing radical radiotherapy. Patients and Methods A serum panel compromising the inflammatory factors was analyzed in 372 NPC patients before and after radiotherapy. Independent prognostic factors were screened out using multivariate Cox regression analysis. A prediction model was built based on the training set data and validated using the test set data. The prognostic value of these factors was evaluated using the time-dependent receiver operating characteristic (ROC) curve and an integrated time-averaged area under the curve (AUC). Results The baseline levels of IL-6, GM-CSF, and MIF were independent factors associated with poor OS and DMFS. A predictive model base established combining the baseline levels of these factors. The AUC values for the test set were 0.9828, 0.9968, and 0.9571 at 1, 3, and 5 years, respectively, compared to 0.9978, 0.9981, and 0.9222 for the training set, respectively. The AUC values for DMFS at 1, 3, and 5-years for the training set were 0.8744, 0.8951, and 0.9358, respectively, compared to 0.9525, 0.9663, and 0.9625 for the test set, respectively. The combination of post-treatment levels of IL-6, GM-CSF, and LIF also had good predictive value for OS with an AUC value > 0.85 during follow-up. Conclusion IL-6, GM-CSF, and MIF baseline levels are powerful prognostic factors for non-metastatic NPC patients. The combination of these factors effectively predicts OS and DMFS in non-metastatic NPC patients.
Collapse
Affiliation(s)
- Ting Liang
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, People’s Republic of China
- Research Center of Carcinogenesis and Targeted Therapy, Xiangya Hospital, Central South University, Changsha, 410008, People’s Republic of China
- Higher Educational Key Laboratory for Cancer Proteomics and Translational Medicine of Hunan Province, Xiangya Hospital, Central South University, Changsha, 410008, People’s Republic of China
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, 410008, People’s Republic of China
- Hunan Hematology Oncology Clinical Medical Research Center, Changsha, 410008, People’s Republic of China
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, 215006, People’s Republic of China
| | - Ding Xiao
- Research Center of Carcinogenesis and Targeted Therapy, Xiangya Hospital, Central South University, Changsha, 410008, People’s Republic of China
- Higher Educational Key Laboratory for Cancer Proteomics and Translational Medicine of Hunan Province, Xiangya Hospital, Central South University, Changsha, 410008, People’s Republic of China
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410008, People’s Republic of China
| | - Shanshan Lu
- Research Center of Carcinogenesis and Targeted Therapy, Xiangya Hospital, Central South University, Changsha, 410008, People’s Republic of China
- Higher Educational Key Laboratory for Cancer Proteomics and Translational Medicine of Hunan Province, Xiangya Hospital, Central South University, Changsha, 410008, People’s Republic of China
| | - Xu Ye
- Department of Radiation Oncology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410008, People’s Republic of China
| | - Zhiqiang Xiao
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, People’s Republic of China
- Research Center of Carcinogenesis and Targeted Therapy, Xiangya Hospital, Central South University, Changsha, 410008, People’s Republic of China
- Higher Educational Key Laboratory for Cancer Proteomics and Translational Medicine of Hunan Province, Xiangya Hospital, Central South University, Changsha, 410008, People’s Republic of China
| |
Collapse
|
5
|
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
|
6
|
Feng G, Xu Y, Ma N, Midorikawa K, Oikawa S, Kobayashi H, Nakamura S, Ishinaga H, Zhang Z, Huang G, Takeuchi K, Murata M. Influence of Epstein-Barr virus and human papillomavirus infection on macrophage migration inhibitory factor and macrophage polarization in nasopharyngeal carcinoma. BMC Cancer 2021; 21:929. [PMID: 34407796 PMCID: PMC8371777 DOI: 10.1186/s12885-021-08675-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 07/30/2021] [Indexed: 12/18/2022] Open
Abstract
Background To assess the effects of Epstein–Barr virus (EBV) and human papillomavirus (HPV) infection on the tumor microenvironment, we examined the relationship between viral infection status, macrophage migration inhibitory factor (MIF), and tumor-associated macrophages in nasopharyngeal carcinoma (NPC). Methods A tissue microarray containing 150 cores from 90 patients with NPC and six with chronic inflammation was used. EBV and HPV status were detected using in situ hybridization with commercial EBER1 and HPV16/18 probes. Immunofluorescence double staining of MIF, pan-macrophage marker CD68, M1 macrophage marker CD11c, and M2 macrophage marker CD163 were analyzed using the same tissue microarray. The levels of these markers between NPC and inflammation cases and between tumor nests and stroma were compared. Correlations among these markers were analyzed. Results We found EBER1(+) cases in 90% of NPC patients, including 10% EBV/HPV co-infection. M1 macrophages mainly infiltrated the tumor nest, while M2 macrophages infiltrated the tumor stroma. We found a significant positive correlation between EBER1 levels and MIF levels in tumor nests and a significant positive correlation between HPV16/18 and CD11c(+) cell levels in NPC tissues. Conclusions It is suggested that MIF is associated with EBV, and M1 macrophage infiltration is affected by HPV status in NPC. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-08675-x.
Collapse
Affiliation(s)
- Guofei Feng
- Department of Environmental and Molecular Medicine, Mie University Graduate School of Medicine, Tsu, 514-8507, Japan.,Department of Otorhinolaryngology-Head and Neck Surgery, Mie University Graduate School of Medicine, Tsu, 514-8507, Japan
| | - Yifei Xu
- Department of Environmental and Molecular Medicine, Mie University Graduate School of Medicine, Tsu, 514-8507, Japan.,Department of Otorhinolaryngology-Head and Neck Surgery, Mie University Graduate School of Medicine, Tsu, 514-8507, Japan.,Department of Otorhinolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Ning Ma
- Graduate School of Health Science, Suzuka University of Medical Science, Suzuka, 510-0226, Japan
| | - Kaoru Midorikawa
- Department of Environmental and Molecular Medicine, Mie University Graduate School of Medicine, Tsu, 514-8507, Japan
| | - Shinji Oikawa
- Department of Environmental and Molecular Medicine, Mie University Graduate School of Medicine, Tsu, 514-8507, Japan
| | - Hatasu Kobayashi
- Department of Environmental and Molecular Medicine, Mie University Graduate School of Medicine, Tsu, 514-8507, Japan
| | - Satoshi Nakamura
- Department of Otorhinolaryngology-Head and Neck Surgery, Mie University Graduate School of Medicine, Tsu, 514-8507, Japan
| | - Hajime Ishinaga
- Department of Otorhinolaryngology-Head and Neck Surgery, Mie University Graduate School of Medicine, Tsu, 514-8507, Japan
| | - Zhe Zhang
- Department of Otorhinolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Guangwu Huang
- Key Laboratory of High-Incidence-Tumor Prevention & Treatment, Guangxi Medical University, Nanning, 530021, China
| | - Kazuhiko Takeuchi
- Department of Otorhinolaryngology-Head and Neck Surgery, Mie University Graduate School of Medicine, Tsu, 514-8507, Japan.
| | - Mariko Murata
- Department of Environmental and Molecular Medicine, Mie University Graduate School of Medicine, Tsu, 514-8507, Japan.
| |
Collapse
|
7
|
Chen Q, Wang Y, Li F, Cheng X, Xiao Y, Chen S, Xiao B, Tao Z. (S,R)3-(4-Hydroxyphenyl)-4,5-Dihydro-5-Isoxazole Acetic Acid Methyl Ester Inhibits Epithelial-to-Mesenchymal Transition through TGF-β/Smad4 Axis in Nasopharyngeal Carcinoma. Anticancer Agents Med Chem 2021; 22:1080-1090. [PMID: 34229595 DOI: 10.2174/1871520621666210706101442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 03/22/2021] [Accepted: 04/19/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Macrophage migration inhibitory factor (MIF), originally reported as an inflammation regulating molecule, is elevated in various cancer cells, which may promote carcinogenesis. Meanwhile, ISO-1 is a potent small molecular inhibitor of MIF, which has not been investigated in nasopharyngeal carcinoma (NPC); hence the impact of ISO-1 on NPC cells remains to be illustrated. OBJECTIVE This study intended to explore the biological function of ISO-1 in NPC cells in vitro and prove a possibility of ISO-1 being a novel agent in NPC treatments. METHODS Gene expression of MIF in Head and Neck squamous cell carcinoma were obtained from The Cancer Genome Atlas (TCGA) database. Nasal pharyngeal tissues were collected from adult patients undergoing nasopharyngeal biopsy for MIF level detection. Proliferation of NPC cell lines 5-8B and 6-10B was studied using Cell Counting Kit-8 (CCK-8) assay and plate-colony-formation assay, apoptosis was determined by flow cytometry and TUNEL staining, migration and invasion capacities were measured by wound-healing assay and transwell assay, all to explore the function of ISO-1 in NPC cells in vitro. Epithelial-to-mesenchymal transition (EMT) level of NPC cells was determined by Western blot analysis and immunofluorescence assay. RESULTS Transcript level of MIF was significantly higher in head and neck squamous cell carcinoma. Protein MIF was overexpressed in human NPC tissues compared to non-cancerous ones, and its expression could be compromised by ISO-1 in vitro. 100μM ISO-1 significantly hindered NPC cells migration and invasion capacities in vitro but acted relatively poorly on proliferation and apoptosis. Immunofluorescence assay and Western blotting implied a down-regulated EMT level through TGF-β/Smad4 axis in ISO-1 treated NPC cells compared to the vehicle. CONCLUSION This study indicated that MIF antagonist ISO-1 holds impact on NPC progression by influencing the migration and invasion of NPC cells ISO-1 inhibits the EMT process of NPC cells through TGF-β/Smad4 axis, supporting that prudent application of ISO-1 may be a potential adjuvant treatment for NPC.
Collapse
Affiliation(s)
- Qibing Chen
- Department of Otolaryngology-Head and Neck Surgery, Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yan Wang
- Department of Otolaryngology-Head and Neck Surgery, Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Fen Li
- Institute of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiang Cheng
- Department of Otolaryngology-Head and Neck Surgery, Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yu Xiao
- Department of Otolaryngology-Head and Neck Surgery, Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shiming Chen
- Department of Otolaryngology-Head and Neck Surgery, Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Bokui Xiao
- Department of Otolaryngology-Head and Neck Surgery, Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zezhang Tao
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, 238 Jie-Fang Road, Wuhan, Hubei 430060, China
| |
Collapse
|
8
|
Parol-Kulczyk M, Gzil A, Maciejewska J, Bodnar M, Grzanka D. Clinicopathological significance of the EMT-related proteins and their interrelationships in prostate cancer. An immunohistochemical study. PLoS One 2021; 16:e0253112. [PMID: 34157052 PMCID: PMC8219170 DOI: 10.1371/journal.pone.0253112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 05/29/2021] [Indexed: 11/19/2022] Open
Abstract
The chronic inflammation influences a microenvironment, where as a result of losing control over tissue homeostatic mechanisms, the carcinogenesis process may be induced. Inflammatory response cells can secrete a number of factors that support both initiation and progression of cancer and also they may consequently induct an epithelial-mesenchymal transition (EMT), the process responsible for development of distant metastasis. Macrophage migration inhibitory factor (MIF) acts as a pro-inflammatory cytokine that is considered as a link between chronic inflammation and tumor development. MIF can function as a modulator of important cancer-related genes expression, as well as an activator of signaling pathways that promotes the development of prostate cancer. The study was performed on FFPE tissues resected from patients who underwent radical prostatectomy. To investigate the relationship of studied proteins with involvement in tumor progression and initiation of epithelial-to-mesenchymal transition (EMT) process, we selected clinicopathological parameters related to tumor progression. Immunohistochemical analyses of MIF, SOX-4, β-catenin and E-cadherin were performed on TMA slides. We found a statistically significant correlation of overall β-catenin expression with the both lymph node metastasis (p<0.001) and presence of angioinvasion (p = 0.012). Membrane β-catenin expression was associated with distant metastasis (p = 0.021). In turn, nuclear MIF was correlated with lymph node metastasis (p = 0.003). The positive protein-protein correlations have been shown between the total β-catenin protein expression level with level of nuclear SOX-4 protein expression (r = 0.27; p<0.05) as well as negative correlation of β-catenin expression with level of nuclear MIF protein expression (r = -0.23; p<0.05). Our results seem promising and strongly highlight the potential role of MIF in development of nodal metastases as well as may confirm an involvement of β-catenin in disease spread in case of prostate cancer.
Collapse
Affiliation(s)
- Martyna Parol-Kulczyk
- Department of Clinical Pathomorphology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Toruń, Poland
| | - Arkadiusz Gzil
- Department of Clinical Pathomorphology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Toruń, Poland
| | - Joanna Maciejewska
- Department of Clinical Pathomorphology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Toruń, Poland
| | - Magdalena Bodnar
- Department of Clinical Pathomorphology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Toruń, Poland
| | - Dariusz Grzanka
- Department of Clinical Pathomorphology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Toruń, Poland
| |
Collapse
|
9
|
Qin L, Qin J, Lv X, Yin C, Zhang Q, Zhang J. MIF promoter polymorphism increases peripheral blood expression levels, contributing to increased susceptibility and poor prognosis in hepatocellular carcinoma. Oncol Lett 2021; 22:549. [PMID: 34093770 PMCID: PMC8170199 DOI: 10.3892/ol.2021.12810] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 04/28/2021] [Indexed: 12/13/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignancies in the world. The etiology and pathogenesis of HCC remain unclear. Macrophage migration inhibitory factor (MIF) plays a critical role in the pathogenesis of hepatocellular carcinoma. The association between MIF polymorphisms and its expression level in HCC has rarely been demonstrated. In the present study, the peripheral blood of 202 patients with HCC (HCC group), 242 patients with chronic hepatitis B (CHB group), 215 patients with liver cirrhosis (LC group) and 227 healthy volunteers (normal group) were collected, DNA was extracted and the target fragment of MIF gene was amplified using PCR. The products were then sequenced, and the expression levels of MIF protein were tested using ELISA. The results showed that the MIF rs755622 polymorphism was associated with an increased susceptibility and metastasis of HCC, and that the genotypes GC and CC were associated with poor prognosis of HCC. Compared with the normal, CHB and LC groups, the expression of MIF in the peripheral blood of the HCC group was significantly increased, and the high expression was associated with to poor prognosis. In the HCC group, MIF protein levels for genotypes GC and CC were increased compared with those of genotype GG. The current study indicated that the MIF rs755622 polymorphism is associated with susceptibility and metastasis of HCC, and that the GC and CC genotypes may be indicators of poor prognosis, which may be ascribed to the MIF rs755622 polymorphism leading to elevated MIF protein expression in peripheral blood.
Collapse
Affiliation(s)
- Lifeng Qin
- Department of Gastroenterology, Minda Hospital of Hubei Minzu University, Enshi, Hubei 445000, P.R. China
| | - Jinmei Qin
- Department of Infectious Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Xiaoping Lv
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Caiqiao Yin
- Department of Gastroenterology, Minda Hospital of Hubei Minzu University, Enshi, Hubei 445000, P.R. China
| | - Qian'e Zhang
- Department of Gastroenterology, Minda Hospital of Hubei Minzu University, Enshi, Hubei 445000, P.R. China
| | - Jiqiao Zhang
- Department of Gastroenterology, Minda Hospital of Hubei Minzu University, Enshi, Hubei 445000, P.R. China
| |
Collapse
|
10
|
Ejma M, Madetko N, Brzecka A, Guranski K, Alster P, Misiuk-Hojło M, Somasundaram SG, Kirkland CE, Aliev G. The Links between Parkinson's Disease and Cancer. Biomedicines 2020; 8:biomedicines8100416. [PMID: 33066407 PMCID: PMC7602272 DOI: 10.3390/biomedicines8100416] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/08/2020] [Accepted: 10/12/2020] [Indexed: 12/12/2022] Open
Abstract
Epidemiologic studies indicate a decreased incidence of most cancer types in Parkinson’s disease (PD) patients. However, some neoplasms are associated with a higher risk of occurrence in PD patients. Both pathologies share some common biological pathways. Although the etiologies of PD and cancer are multifactorial, some factors associated with PD, such as α-synuclein aggregation; mutations of PINK1, PARKIN, and DJ-1; mitochondrial dysfunction; and oxidative stress can also be involved in cancer proliferation or cancer suppression. The main protein associated with PD, i.e., α-synuclein, can be involved in some types of neoplastic formations. On the other hand, however, its downregulation has been found in the other cancers. PINK1 can act as oncogenic or a tumor suppressor. PARKIN dysfunction may lead to some cancers’ growth, and its expression may be associated with some tumors’ suppression. DJ-1 mutation is involved in PD pathogenesis, but its increased expression was found in some neoplasms, such as melanoma or breast, lung, colorectal, uterine, hepatocellular, and nasopharyngeal cancers. Both mitochondrial dysfunction and oxidative stress are involved in PD and cancer development. The aim of this review is to summarize the possible associations between PD and carcinogenesis.
Collapse
Affiliation(s)
- Maria Ejma
- Department of Neurology, Wroclaw Medical University, Borowska 213, 50-556 Wrocław, Poland; (M.E.); (N.M.); (K.G.)
| | - Natalia Madetko
- Department of Neurology, Wroclaw Medical University, Borowska 213, 50-556 Wrocław, Poland; (M.E.); (N.M.); (K.G.)
| | - Anna Brzecka
- Department of Pulmonology and Lung Oncology, Wroclaw Medical University, Grabiszyńska 105, 53-439 Wroclaw, Poland;
| | - Konstanty Guranski
- Department of Neurology, Wroclaw Medical University, Borowska 213, 50-556 Wrocław, Poland; (M.E.); (N.M.); (K.G.)
| | - Piotr Alster
- Department of Neurology, Medical University of Warsaw, Kondratowicza 8, 03-242 Warszawa, Poland;
| | - Marta Misiuk-Hojło
- Department of Ophthalmology, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland;
| | - Siva G. Somasundaram
- Department of Biological Sciences, Salem University, Salem, WV 26426, USA; (S.G.S.); (C.E.K.)
| | - Cecil E. Kirkland
- Department of Biological Sciences, Salem University, Salem, WV 26426, USA; (S.G.S.); (C.E.K.)
| | - Gjumrakch Aliev
- Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, bld. 2, 119991 Moscow, Russia
- Research Institute of Human Morphology, Russian Academy of Medical Science, Street Tsyurupa 3, 117418 Moscow, Russia
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, Chernogolovka, 142432 Moscow Region, Russia
- GALLY International Research Institute, 7733 Louis Pasteur Drive, #330, San Antonio, TX 78229, USA
- Correspondence: or ; Tel.: +1-210-442-8625 or +1-440-263-7461
| |
Collapse
|
11
|
Koh HM, Kim DC. Prognostic significance of macrophage migration inhibitory factor expression in cancer patients: A systematic review and meta-analysis. Medicine (Baltimore) 2020; 99:e21575. [PMID: 32769903 PMCID: PMC7592988 DOI: 10.1097/md.0000000000021575] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Recent studies showed that Macrophage migration inhibitory factor (MIF) is overexpressed and closely associated with prognosis in cancer patients. The present study was systematically evaluated the prognostic significance of MIF expression in cancer patients. METHODS PubMed, Cochrane library and Scopus were searched for eligible studies up to January 2020. Pooled hazard ratio with confidence interval (CI) was determined to assess the relationship between MIF expression and survival in cancer patients. RESULTS A total of 8 studies comprising 847 cancer patients were included in this meta-analysis. For overall survival, the pooled hazard ratio was 2.23 (95% CI 1.67-2.99, P < .001). For disease-free survival, the pooled hazard ratio was 2.24 (95% CI 1.69-2.96, P < .001). The results suggested that high expression of MIF was significantly related to poor overall survival and disease-free survival in cancer patients. CONCLUSION MIF expression could be a valuable prognostic factor in cancer patients.
Collapse
Affiliation(s)
- Hyun Min Koh
- Department of Pathology, Gyeongsang National University Changwon Hospital, Changwon
| | - Dong Chul Kim
- Department of Pathology, Gyeongsang National University School of Medicine
- Department of Pathology, Gyeongsang National University Hospital
- Gyeongsang Institute of Health Science, Jinju, Republic of Korea
| |
Collapse
|
12
|
Xue N, Xing S, Ma W, Sheng J, Huang Z, Xu Q. Combination of Plasma MIF and VCA-IgA Improves the Diagnostic Specificity for Patients With Nasopharyngeal Carcinoma. Technol Cancer Res Treat 2020; 19:1533033820935773. [PMID: 32578505 PMCID: PMC7315673 DOI: 10.1177/1533033820935773] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION The purpose of this study is to evaluate the diagnostic value of macrophage migration inhibitory factor in patients with nasopharyngeal carcinoma. MATERIALS AND METHODS The expression levels of macrophage migration inhibitory factor in nasopharyngeal carcinoma cell lines, tumor tissues, and plasma were measured by real-time polymerase chain reaction, Western blotting, enzyme-linked immunosorbent assay, and immunohistochemistry. Plasma Epstein-Barr virus viral capsid antigen was determined by immunoenzymatic techniques. RESULTS Both the messenger RNA and protein expression levels of macrophage migration inhibitory factor were upregulated in nasopharyngeal carcinoma cell lines and nasopharyngeal carcinoma tissues. Macrophage migration inhibitory factor in plasma was significantly elevated in patients with nasopharyngeal carcinoma compared to Epstein-Barr virus viral capsid antigen-negative and Epstein-Barr virus viral capsid antigen-positive healthy donors. The combination of macrophage migration inhibitory factor and Epstein-Barr virus viral capsid antigen was better for diagnosing nasopharyngeal carcinoma (area under receiver operating characteristic curve = 0.925, 95% CI: 0.898-0.951) than macrophage migration inhibitory factor (area under receiver operating characteristic curve = 0.778, 95% CI: 0.732-0.824) and Epstein-Barr virus viral capsid antigen. Combining macrophage migration inhibitory factor and Epstein-Barr virus viral capsid antigen had higher specificity (82.40% vs 69.96%) and higher positive predictive value (79.17% vs 67.44%) without an obvious reduction in sensitivity (95.25%) compared to Epstein-Barr virus viral capsid antigen alone. Macrophage migration inhibitory factor was highly expressed in nasopharyngeal carcinoma cell lines, whereas it was not associated with Epstein-Barr virus infection. The level of macrophage migration inhibitory factor in plasma was not related to the titer of Epstein-Barr virus viral capsid antigen. CONCLUSION The combination of macrophage migration inhibitory factor and Epstein-Barr virus viral capsid antigen increases the specificity and positive predictive value of detecting nasopharyngeal carcinoma and improves the diagnostic accuracy of nasopharyngeal carcinoma in high-risk individuals.
Collapse
Affiliation(s)
- Ning Xue
- Department of Clinical Laboratory, Affiliated Tumor Hospital of Zhengzhou University, Henan Tumor Hospital, Zhengzhou, China
| | - Shan Xing
- Department of Clinical Laboratory, Sun Yat-Sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Weiguo Ma
- Department of Clinical Laboratory, Affiliated Tumor Hospital of Zhengzhou University, Henan Tumor Hospital, Zhengzhou, China
| | - Jiahe Sheng
- Department of Clinical Laboratory, Affiliated Tumor Hospital of Zhengzhou University, Henan Tumor Hospital, Zhengzhou, China
| | - Zhiliang Huang
- Department of Thoracic Surgery, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen, Fujian, China
| | - Qingxia Xu
- Department of Clinical Laboratory, Affiliated Tumor Hospital of Zhengzhou University, Henan Tumor Hospital, Zhengzhou, China
| |
Collapse
|
13
|
Utispan K, Koontongkaew S. Macrophage migration inhibitory factor modulates proliferation, cell cycle, and apoptotic activity in head and neck cancer cell lines. J Dent Sci 2020; 16:342-348. [PMID: 33384818 PMCID: PMC7770260 DOI: 10.1016/j.jds.2020.02.008] [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: 01/14/2020] [Revised: 02/06/2020] [Indexed: 11/20/2022] Open
Abstract
Background/purpose Macrophage migration inhibitory factor (MIF) is a multifunctional cytokine that contributes to the progression of several cancers. MIF overexpression has been reported in head and neck squamous cell carcinoma (HNSCC) patients. However, the exact role of MIF in HNSCC is not fully understood. Our aim was to evaluate the amount of secreted MIF and the role of MIF in the proliferation, cell cycle, and apoptosis in HNSCC cell lines. Materials and methods Genetically matched HNSCC cell lines derived from primary (HN18 and HN30) and metastatic sites (HN17 and HN31) from the same patient were used in this study. The MIF levels in conditioned media from the HNSCC cell lines were evaluated using ELISA. The HNSCC cell lines were treated with recombinant MIF at concentrations 25, 50 and 100 ng/ml, and cell proliferation was evaluated by MTT assay. A proliferative dose of MIF was used to treat the cells then, cell cycle, and apoptotic status were determined by flow cytometry. Results The HNSCC-secreted MIF concentration ranged from 49.33 to 973 pg/ml. Exogenous MIF (25 ng/ml) significantly increased HN18, HN30, and HN31 cell proliferation. Moreover, MIF induced cell cycle progression and inhibited apoptosis in these cells. However, MIF did not affect growth or apoptosis in HN17 cell. Conclusion MIF secreted from the HNSCC cell lines were evaluated. Exogenous MIF promotes various effects on proliferation, cell cycle, and apoptosis in HNSCC cells.
Collapse
Affiliation(s)
- Kusumawadee Utispan
- Oral Biology Research Unit, Faculty of Dentistry, Thammasat University (Rangsit Campus), Pathum Thani, Thailand
| | - Sittichai Koontongkaew
- Oral Biology Research Unit, Faculty of Dentistry, Thammasat University (Rangsit Campus), Pathum Thani, Thailand.,Walailak University International College of Dentistry, Walailak University, Bangkok, Thailand
| |
Collapse
|
14
|
Salazar C, Ruiz-Hincapie P, Ruiz LM. The Interplay among PINK1/PARKIN/Dj-1 Network during Mitochondrial Quality Control in Cancer Biology: Protein Interaction Analysis. Cells 2018; 7:cells7100154. [PMID: 30274236 PMCID: PMC6210981 DOI: 10.3390/cells7100154] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 09/14/2018] [Accepted: 09/25/2018] [Indexed: 12/18/2022] Open
Abstract
PARKIN (E3 ubiquitin ligase PARK2), PINK1 (PTEN induced kinase 1) and DJ-1 (PARK7) are proteins involved in autosomal recessive parkinsonism, and carcinogenic processes. In damaged mitochondria, PINK1’s importing into the inner mitochondrial membrane is prevented, PARKIN presents a partial mitochondrial localization at the outer mitochondrial membrane and DJ-1 relocates to mitochondria when oxidative stress increases. Depletion of these proteins result in abnormal mitochondrial morphology. PINK1, PARKIN, and DJ-1 participate in mitochondrial remodeling and actively regulate mitochondrial quality control. In this review, we highlight that PARKIN, PINK1, and DJ-1 should be regarded as having an important role in Cancer Biology. The STRING database and Gene Ontology (GO) enrichment analysis were performed to consolidate knowledge of well-known protein interactions for PINK1, PARKIN, and DJ-1 and envisage new ones. The enrichment analysis of KEGG pathways showed that the PINK1/PARKIN/DJ-1 network resulted in Parkinson disease as the main feature, while the protein DJ-1 showed enrichment in prostate cancer and p53 signaling pathway. Some predicted transcription factors regulating PINK1, PARK2 (PARKIN) and PARK7 (DJ-1) gene expression are related to cell cycle control. We can therefore suggest that the interplay among PINK1/PARKIN/DJ-1 network during mitochondrial quality control in cancer biology may occur at the transcriptional level. Further analysis, like a systems biology approach, will be helpful in the understanding of PINK1/PARKIN/DJ-1 network.
Collapse
Affiliation(s)
- Celia Salazar
- Instituto de Investigaciones Biomédicas, Universidad Autónoma de Chile, Santiago 8910060, Chile.
| | - Paula Ruiz-Hincapie
- School of Engineering and Technology, University of Hertfordshire, Hatfield AL 10 9AB, UK.
| | - Lina María Ruiz
- Instituto de Investigaciones Biomédicas, Universidad Autónoma de Chile, Santiago 8910060, Chile.
| |
Collapse
|
15
|
Wang Q, Li F, Shi W, Zhang Q, Wang J, Yan X, Chai L, Li M. Overexpression of DJ-1 correlates with aggressive clinicopathological characteristics and poor prognosis in malignant tumors: a meta-analysis. Onco Targets Ther 2018; 11:3931-3942. [PMID: 30022836 PMCID: PMC6042497 DOI: 10.2147/ott.s162045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Purpose A number of studies have investigated the role of DJ-1 in the development and progression of malignant tumors. This meta-analysis aims to systematically estimate the rela-tionship between the expression level of DJ-1 and the malignant biological behaviors of tumors and to assess the clinical significances of DJ-1 in the prognosis and diagnosis of cancer. Materials and methods We searched PubMed, Web of Science, China National Knowledge Infrastructure and Wanfang databases from inception to December 1, 2017. Pooled odds ratio (OR) and hazard ratio (HR) with their 95% confidence interval and the diagnostic value of DJ-1 were calculated. Results Fourteen eligible studies with a total of 1,947 subjects were enrolled in our meta-analysis. The results showed that DJ-1 was overexpressed in cancer patients compared with noncancer patients (OR = 30.72), and elevated expression of DJ-1 was demonstrated to be closely associated with high tumor-node-metastasis stage (OR = 5.52), poor differentiated degree (OR = 2.46), positive lymph node metastasis (OR = 4.12) and worse overall survival (HR = 2.23). In addition, the combined sensitivity and specificity for DJ-1 to discern malignant tumors were 0.73 and 0.93, respectively. The diagnostic OR was 34.87, and the area under the summary receiver operating characteristic curve was 0.88. Conclusion This meta-analysis demonstrated that DJ-1 was an important biomarker in tumor assessment and prognosis prediction.
Collapse
Affiliation(s)
- Qingting Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China,
| | - Fangwei Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China,
| | - Wenhua Shi
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China,
| | - Qianqian Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China,
| | - Jian Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China,
| | - Xin Yan
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China,
| | - Limin Chai
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China,
| | - Manxiang Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China,
| |
Collapse
|
16
|
Kang Y, Zhang Y, Sun Y. Macrophage migration inhibitory factor is a novel prognostic marker for human oral squamous cell carcinoma. Pathol Res Pract 2018; 214:1192-1198. [PMID: 30041958 DOI: 10.1016/j.prp.2018.06.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 06/11/2018] [Accepted: 06/25/2018] [Indexed: 01/30/2023]
Abstract
Macrophage migration inhibitory factor (MIF) is considered a pro-tumour factor. However, its clinical relevance in oral squamous cell carcinoma (OSCC) remains unclear. The objective of this study was to investigate the expression of MIF and its receptor CD74 in OSCC tissues, and to study the function of MIF in OSCC cells. Tissues of 90 patients with OSCC from the School of Stomatology, China Medical University were collected, and immunohistochemical staining and quantitative reverse transcription polymerase chain reaction were performed for MIF and CD74. The possible correlations between MIF and CD74 and clinical characteristics were analysed. The Kaplan-Meier analysis was used to determine the survival rates of patients. In addition, the proliferation and invasion of OSCC cells were evaluated after transfection with siRNA against MIF. MIF and CD74 levels were significantly higher in tissues of patients with OSCC than in control tissues. Moreover, MIF levels in patients with OSCC were significantly associated with cell differentiation and TNM classification. MIF expression was closely related to CD74 expression. Kaplan-Meier analysis indicated that OSCC patients with high MIF levels showed reduced overall survival and recurrenc-free survival. Furthermore, MIF expression promoted proliferation and invasion of OSCC cells. Collectively, our results reveal that MIF expression is a significant independent prognostic factor for patients with OSCC and may be a novel prognostic marker for OSCC.
Collapse
Affiliation(s)
- Yuanyuan Kang
- Department of Emergency and Oral medicine, The School of Stomatology, China Medical University & Liaoning Institute of Dental Research & Liaoning Province Key Laboratory of Oral Diseases & Liaoning Province Translational Medicine Research Center of Oral Diseases, Shenyang, Liaoning, 110001, PR China.
| | - Ying Zhang
- Department of Emergency and Oral medicine, The School of Stomatology, China Medical University & Liaoning Institute of Dental Research & Liaoning Province Key Laboratory of Oral Diseases & Liaoning Province Translational Medicine Research Center of Oral Diseases, Shenyang, Liaoning, 110001, PR China
| | - Yan Sun
- Department of Emergency and Oral medicine, The School of Stomatology, China Medical University & Liaoning Institute of Dental Research & Liaoning Province Key Laboratory of Oral Diseases & Liaoning Province Translational Medicine Research Center of Oral Diseases, Shenyang, Liaoning, 110001, PR China
| |
Collapse
|
17
|
Liu R, Yang YN, Yi L, Qing J, Li QY, Wang WS, Wang J, Tang YX, Tan H. Diallyl disulfide effect on the invasion and migration ability of HL-60 cells with a high expression of DJ-1 in the nucleus through the suppression of the Src signaling pathway. Oncol Lett 2018; 15:6377-6385. [PMID: 29725397 PMCID: PMC5920463 DOI: 10.3892/ol.2018.8139] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 01/04/2018] [Indexed: 12/18/2022] Open
Abstract
The present study examined the effect of diallyl disulfide (DADS) on the invasion and migration ability of HL-60 cells with a high expression of parkinsonism associated deglycase (DJ-1) in the nucleus (HHDN), and its molecular mechanism. A western blot assay was used to measure the effects of DADS and an Src inhibitor on the expression of DJ-1 and the Src signal pathway in HHDN. The effects of DADS and Src inhibitors on the invasion and migration ability of HHDN was detected using Transwell migration and invasion chamber experiments. The experiments were divided into three groups: A control group (HL-60 cells), an empty vector group and a high expression group (HHDN cells). Western blot assays revealed that the expression of DJ-1 in HHDN was inhibited in a time-dependent manner following treatment with DADS for 24, 48 and 72 h. Following DADS treatment, the expression of phosphorylated Src (p-Src) and phosphorylated Fak (p-Fak) were significantly decreased in all groups compared with the untreated groups, however the expression level of Src, Fak and integrin did not change significantly. Western blot analysis results revealed that following treatment with DADS and Src inhibitor, the expression levels of p-Src and p-Fak significantly decreased in all three groups compared with untreated groups, whereas the expression levels of Src, Fak and integrin did not change significantly. The expression of DJ-1 in HHND was inhibited in time-dependent manner following treatment with DADS and Src inhibitor for 24, 48 and 72 h. Transwell migration and invasion assay results revealed that DADS and Src inhibitors may suppress migration and invasion in leukemic cells, and a combination of the two treatments may result in more efficient suppression. DADS may downregulate DJ-1-mediated invasion and migration in leukemic cells through suppressing the Src-Fak-Integrin signaling pathway, and the Src inhibitor may enhance the antitumor effect of DADS.
Collapse
Affiliation(s)
- Ran Liu
- Cancer Research Institute, Key Laboratory of Tumor Cellular and Molecular Pathology, College of Hunan Province, University of South China, Hunan 421001, P.R. China.,Department of Pathology, The First Hospital of Changsha, Changsha, Hunan 410005, P.R. China
| | - Ye-Ning Yang
- Department of Pathology, The First People's Hospital of Youxian, Youxian, Hunan 412300, P.R. China
| | - Lan Yi
- Cancer Research Institute, Key Laboratory of Tumor Cellular and Molecular Pathology, College of Hunan Province, University of South China, Hunan 421001, P.R. China
| | - Jing Qing
- Cancer Research Institute, Key Laboratory of Tumor Cellular and Molecular Pathology, College of Hunan Province, University of South China, Hunan 421001, P.R. China
| | - Qing-Ye Li
- Cancer Research Institute, Key Laboratory of Tumor Cellular and Molecular Pathology, College of Hunan Province, University of South China, Hunan 421001, P.R. China
| | - Wen-Song Wang
- Cancer Research Institute, Key Laboratory of Tumor Cellular and Molecular Pathology, College of Hunan Province, University of South China, Hunan 421001, P.R. China
| | - Juan Wang
- Cancer Research Institute, Key Laboratory of Tumor Cellular and Molecular Pathology, College of Hunan Province, University of South China, Hunan 421001, P.R. China
| | - Yu-Xian Tang
- Cancer Research Institute, Key Laboratory of Tumor Cellular and Molecular Pathology, College of Hunan Province, University of South China, Hunan 421001, P.R. China
| | - Hui Tan
- Cancer Research Institute, Key Laboratory of Tumor Cellular and Molecular Pathology, College of Hunan Province, University of South China, Hunan 421001, P.R. China
| |
Collapse
|
18
|
Wang SS, Cen X, Liang XH, Tang YL. Macrophage migration inhibitory factor: a potential driver and biomarker for head and neck squamous cell carcinoma. Oncotarget 2018; 8:10650-10661. [PMID: 27788497 PMCID: PMC5354689 DOI: 10.18632/oncotarget.12890] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 10/19/2016] [Indexed: 02/05/2023] Open
Abstract
Macrophage migration inhibitory factor (MIF), a pleiotropic proinflammatory cytokine, has been showed to be associated with the immunopathogenesis of many diseases. Recent study demonstrated that MIF promoted tumorigenesis and tumor progression and played a critical role in various kinds of human cancer including head and neck squamous cell carcinoma(HNSCC). Hence, in this paper we retrospected the relationship between MIF and angiogenesis, epithelial-mesenchymal transition (EMT), inflammation, immune response, hypoxia microenvironment, and discussed whether it is a promising biomarker for diagnosis and supervisor of HNSCC.
Collapse
Affiliation(s)
- Sha-Sha Wang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu Sichuan, People's Republic of China.,Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University, Chengdu Sichuan, People's Republic of China
| | - Xiao Cen
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu Sichuan, People's Republic of China.,Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University, Chengdu Sichuan, People's Republic of China
| | - Xin-Hua Liang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu Sichuan, People's Republic of China.,Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University, Chengdu Sichuan, People's Republic of China
| | - Ya-Ling Tang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu Sichuan, People's Republic of China.,Department of Oral Pathology, West China Hospital of Stomatology, Sichuan University, Chengdu Sichuan, People's Republic of China
| |
Collapse
|
19
|
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
|
20
|
Kindt N, Journe F, Laurent G, Saussez S. Involvement of macrophage migration inhibitory factor in cancer and novel therapeutic targets. Oncol Lett 2016; 12:2247-2253. [PMID: 27698786 DOI: 10.3892/ol.2016.4929] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 05/16/2016] [Indexed: 12/18/2022] Open
Abstract
Macrophage migration inhibitory factor (MIF) was originally identified in 1966 by Bloom and Bennett as a pro-inflammatory cytokine involved in the inhibition of macrophage motility. Since then, studies have investigated the functional contribution of this pro-inflammatory cytokine in several immune diseases, including rheumatoid arthritis and lupus erythematous. Recently, MIF has been reported to be involved in a variety of neoplastic diseases. The present review discusses previous cancer research studies that have investigated the involvement of MIF in carcinogenesis, disease prognosis, tumor cell proliferation and invasion, and tumor-induced angiogenesis. Finally, potential therapeutic approaches based on the use of MIF antagonists and neutralizing antibodies are examined. The review concludes that MIF could be a good prognostic biomarker in several types of cancer, but also that the inhibition of MIF could represent a novel therapy against cancer.
Collapse
Affiliation(s)
- Nadège Kindt
- Laboratory of Anatomy and Cellular Biology, Faculty of Medicine and Pharmacy, University of Mons, Mons 7000, Belgium
| | - Fabrice Journe
- Laboratory of Anatomy and Cellular Biology, Faculty of Medicine and Pharmacy, University of Mons, Mons 7000, Belgium; Laboratory of Oncology and Experimental Surgery, Institut Jules Bordet, Free University of Brussels, Brussels 1000, Belgium
| | - Guy Laurent
- Laboratory of Histology, Faculty of Medicine and Pharmacy, University of Mons, Mons 7000, Belgium
| | - Sven Saussez
- Laboratory of Anatomy and Cellular Biology, Faculty of Medicine and Pharmacy, University of Mons, Mons 7000, Belgium; Department of Otorhinolaryngology, Faculty of Medicine, Free University of Brussels, Brussels 1000, Belgium
| |
Collapse
|
21
|
Schumann C, Chan S, Khalimonchuk O, Khal S, Moskal V, Shah V, Alani AWG, Taratula O, Taratula O. Mechanistic Nanotherapeutic Approach Based on siRNA-Mediated DJ-1 Protein Suppression for Platinum-Resistant Ovarian Cancer. Mol Pharm 2016; 13:2070-83. [PMID: 27170529 DOI: 10.1021/acs.molpharmaceut.6b00205] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
We report an efficient therapeutic modality for platinum resistant ovarian cancer based on siRNA-mediated suppression of a multifunctional DJ-1 protein that is responsible for the proliferation, growth, invasion, oxidative stress, and overall survival of various cancers. The developed therapeutic strategy can work alone or in concert with a low dose of the first line chemotherapeutic agent cisplatin, to elicit a maximal therapeutic response. To achieve an efficient DJ-1 knockdown, we constructed the polypropylenimine dendrimer-based nanoplatform targeted to LHRH receptors overexpressed on ovarian cancer cells. The quantitative PCR and Western immunoblotting analysis revealed that the delivered DJ-1 siRNA downregulated the expression of targeted mRNA and corresponding protein by more than 80% in various ovarian cancer cells. It was further demonstrated that siRNA-mediated DJ-1 suppression dramatically impaired proliferation, viability, and migration of the employed ovarian cancer cells. Finally, the combinatorial approach led to the most pronounced therapeutic response in all the studied cell lines, outperforming both siRNA-mediated DJ-1 knockdown and cisplatin treatment alone. It is noteworthy that the platinum-resistant cancer cells (A2780/CDDP) with the highest basal level of DJ-1 protein are most susceptible to the developed therapy and this susceptibility declines with decreasing basal levels of DJ-1. Finally, we interrogate the molecular underpinnings of the DJ-1 knockdown effects in the treatment of the ovarian cancer cells. By using various experimental techniques, it was revealed that DJ-1 depletion (1) decreases the activity of the Akt pathway, thereby reducing cellular proliferation and migration and increasing the antiproliferative effect of cisplatin on ovarian cancer cells; (2) enhances the activity of p53 tumor suppressor protein therefore restoring cell cycle arrest functionality and upregulating the Bax-caspase pathway, triggering cell death; and (3) weakens the cellular defense mechanisms against inherited oxidative stress thereby increasing toxic intracellular radicals and amplifying the reactive oxygen species created by the administration of cisplatin.
Collapse
Affiliation(s)
- Canan Schumann
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University , Portland, Oregon 97201, United States
| | - Stephanie Chan
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University , Portland, Oregon 97201, United States
| | - Oleh Khalimonchuk
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center , Omaha, Nebraska 68198, United States
| | - Shannon Khal
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University , Portland, Oregon 97201, United States
| | - Vitaliya Moskal
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University , Portland, Oregon 97201, United States
| | - Vidhi Shah
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University , Portland, Oregon 97201, United States
| | - Adam W G Alani
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University , Portland, Oregon 97201, United States
| | - Olena Taratula
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University , Portland, Oregon 97201, United States
| | - Oleh Taratula
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University , Portland, Oregon 97201, United States
| |
Collapse
|
22
|
Lv W, Chen N, Lin Y, Ma H, Ruan Y, Li Z, Li X, Pan X, Tian X. Macrophage migration inhibitory factor promotes breast cancer metastasis via activation of HMGB1/TLR4/NF kappa B axis. Cancer Lett 2016; 375:245-255. [PMID: 26952810 DOI: 10.1016/j.canlet.2016.02.005] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 02/01/2016] [Accepted: 02/02/2016] [Indexed: 12/15/2022]
Abstract
Macrophage migration inhibitory factor (MIF) is up-regulated in diverse solid tumors and acts as the critical link between immune response and tumorigenesis. In this study, we demonstrated that MIF overexpression promoted migration of breast cancer cells by elevating TLR4 expression. Further investigation evidenced that MIF induced ROS generation. MIF-induced ROS led to ERK phosphorylation, which facilitated HMGB1 release from the nucleus to the cytoplasm. MIF overexpression also induced caveolin-1 phosphorylation. Caveolin-1 phosphorylation contributed to HMGB1 secretion from the cytoplasm to the extracellular matrix. The extracellular HMGB1 activated TLR4 signaling including NF-κB phosphorylation, which was responsible for the transcription of Snail and Twist as well as MMP2 activation. Furthermore, MIF-induced caveolin-1-dependent HMGB1 secretion might control the recruitment of CD11b+ immune cells. Our data suggested that MIF affected the intrinsic properties of tumors and the host immune response in tumor microenvironment by regulating the TLR4/HMGB1 axis, leading to metastasis of breast cancer.
Collapse
Affiliation(s)
- Wei Lv
- Department of Breast and Thyroid Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, China
| | - Na Chen
- Department of Breast and Thyroid Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, China
| | - Yanliang Lin
- Department of Breast and Thyroid Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, China
| | - Hongyan Ma
- Department of Breast and Thyroid Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, China
| | - Yongwei Ruan
- Department of Breast and Thyroid Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, China
| | - Zhiwei Li
- Department of Breast and Thyroid Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, China
| | - Xungeng Li
- Department of Breast and Thyroid Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, China
| | - Xiaohua Pan
- Department of Breast and Thyroid Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, China
| | - Xingsong Tian
- Department of Breast and Thyroid Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, China.
| |
Collapse
|
23
|
Liu G, Xu Z, Hao D. MicroRNA‑451 inhibits neuroblastoma proliferation, invasion and migration by targeting macrophage migration inhibitory factor. Mol Med Rep 2016; 13:2253-60. [PMID: 26783235 DOI: 10.3892/mmr.2016.4770] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Accepted: 11/19/2015] [Indexed: 11/05/2022] Open
Abstract
Neuroblastoma (NB) is the most prevalent type of extracranial solid tumour in young children. To improve current understanding of the mechanisms, which modulate cancer cell proliferation, invasion and migration, investigations have focused on microRNAs (miRs), a class of small non‑coding RNAs, which post‑transcriptionally regulate gene expression during various crucial cell processes. The present study aimed to investigate the role of miR‑451 in NB. Human NB tissue and adjacent normal tissue were surgically removed, and the expression of miR‑451, and development and pathological characteristics of NB were investigated. The expression of miR‑451 was reduced in the NB tissue, compared with that in the adjacent tissue, and correlations between the reduction in miR‑451 and unfavourable variables included tumour size (P=0.0081), differentiation (P=0.0217), lymph node metastasis (P=0.0489), tumour‑node‑metastasis stage (0.0220) and distant metastases (P=0.0201). Transfection of the SK‑N‑SH and GI‑LA‑N NB cell lines with miR‑451 inhibited cell growth, invasion and migration. Furthermore, the present study demonstrated that macrophage migration inhibitory factor (MIF) was regulated directly by miR‑451 and was a critical mediator of the biological effects of miR‑451 in NB. The re‑expression of MIF markedly reversed the carcinogenic inhibitory property of miR‑451. These data provide a more detailed understanding of the essential role of miR‑451 in NB, which relies on regulation of the expression of MIF.
Collapse
Affiliation(s)
- Geng Liu
- Department of Hand Surgery, Hong‑Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710054, P.R. China
| | - Zhengwei Xu
- Department of Spine Surgery, Hong‑Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710054, P.R. China
| | - Dingjun Hao
- Department of Spine Surgery, Hong‑Hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710054, P.R. China
| |
Collapse
|
24
|
Wang K, Liang Q, Wei L, Zhang W, Zhu P. MicroRNA-608 acts as a prognostic marker and inhibits the cell proliferation in hepatocellular carcinoma by macrophage migration inhibitory factor. Tumour Biol 2015; 37:3823-30. [PMID: 26474589 DOI: 10.1007/s13277-015-4213-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 10/09/2015] [Indexed: 11/28/2022] Open
Abstract
Human hepatocellular carcinoma (HCC) is one of the most prevalent malignancies in the world. Research on HCC has recently focused on microRNAs (miRNAs) that play crucial roles in cancer development and progression of HCC. In this study, we aimed to analyze the expression and function of a metastasis-associated microRNA-608 (miR-608) in HCC. Samples of human HCC and matched adjacent normal tissues were surgically removed, and miR-608 expression and the pathological characteristics of HCC were investigated. In this study, we found that miR-608 expression was significantly reduced in HCC and its expression levels were highly associated with tumor size, differentiation, clinical stage, and overall and disease-free survival of HCC. Overexpression of miR-608 in HCC cell lines HepG2 and SK-Hep-1 inhibited cell proliferation by G1 arrest. Macrophage migration inhibitory factor (MIF), a potential target gene of miR-608, was inversely correlated with miR-608 expression in HCC tissues and cell lines. Furthermore, we demonstrated that MIF was directly regulated by miR-608 and the restoration of MIF expression reversed the inhibitory effects of miR-608 on HCC cell proliferation. Taken together, these findings collectively demonstrate that miR-608 exerts its anti-cancer function by directly targeting MIF in HCC, indicating a potential novel prognostic biomarker and therapeutic target for HCC.
Collapse
Affiliation(s)
- Kejia Wang
- Department of Pathology, No. 401 Hospital of PLA, 22 Minjiang Road, Qingdao, 266071, China
| | - Qing Liang
- Department of Physiology and Pathophysiology, Fudan University Shanghai Medical College, Shanghai, 200032, China
| | - Li Wei
- Department of Pathology, No. 401 Hospital of PLA, 22 Minjiang Road, Qingdao, 266071, China
| | - Wei Zhang
- Department of Pathology, No. 401 Hospital of PLA, 22 Minjiang Road, Qingdao, 266071, China.
| | - Ping Zhu
- Department of Obstetrics and Gynecology, No. 401 Hospital of PLA, 22 Minjiang Road, Qingdao, 266071, China.
| |
Collapse
|
25
|
DJ-1 is activated in medulloblastoma and is associated with cell proliferation and differentiation. World J Surg Oncol 2014; 12:373. [PMID: 25475127 PMCID: PMC4289263 DOI: 10.1186/1477-7819-12-373] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 11/18/2014] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND DJ-1 is a key regulator in human tumorigenesis, including brain malignancies. The mechanisms by which DJ-1 contributes to the pathogenesis of medulloblastoma (MB) remain unclear, and its impact on the prognosis for patients with MB has not been identified. The aim of this study was to determine whether the DJ-1 protein is associated with tumorigenesis of MBs, and whether DJ-1 is a valuable factor for predicting the prognosis of patients with MB. METHODS We collected 66 pairs of MB and adjacent normal cerebellum samples. Expression of DJ-1, Ser 473-phosphorylated-Akt (p-Akt), PTEN, and Ki-67 (MIB-1) was detected by immunohistochemical staining, and the correlation of these immunostaining results with the clinicopathological features of patients with MB was determined. RESULTS High DJ-1 expression (48.5%, 32/66) in tumor cells of MBs was significantly associated with the classic MB variant (P = 0.003), high proliferative activity (P = 0.002) and undifferentiated tumor (P = 0.001), whereas high p-Akt expression (56.1%, 37/66) was associated with tumor metastasis stage (P = 0.007), undifferentiated tumor (P = 0.007), and high-risk tumor (P = 0.002). High DJ-1 expression also correlated with high p-Akt expression and high MIB-1 index. However, only high levels of DJ-1(P = 0.009) and high MIB-1 index (P = 0.001) were strong independent prognostic factors associated with worse overall survival. CONCLUSIONS Although the validity of the preliminary data in this study needs to be confirmed by a larger number of cases, our study indicates that DJ-1, PTEN, and p-Akt might play important roles in cell proliferation and differentiation of MBs. The evaluation of expression of DJ-1 and related proteins might be useful for predicting the prognosis of patients with MB.
Collapse
|
26
|
Wen Q, Li J, Wang W, Xie G, Xu L, Luo J, Chu S, She L, Li D, Huang D, Fan S. Increased expression of flotillin-2 protein as a novel biomarker for lymph node metastasis in nasopharyngeal carcinoma. PLoS One 2014; 9:e101676. [PMID: 25014228 PMCID: PMC4094483 DOI: 10.1371/journal.pone.0101676] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 06/09/2014] [Indexed: 11/18/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a head and neck malignant tumor rare throughout most of the world but common in Southeast Asia, especially in Southern China. Flotillin-2 (Flot-2) is not only an important component of cellular membrane, but also involves in various cellular processes such as membrane trafficking, T cell and B cell activation, regulation of several signaling pathways associated with cell growth and malignant transformation, keeping structure and junction of epidermal cells and formation of filopodia. Although such molecular effects of Flot-2 have been reported, whether the expression of Flot-2 protein is associated with clinicopathologic implication for NPC has not been reported. The purpose of this research is to investigate the expression of Flot-2 protein in NPC and control nasopharyngeal epithelial tissues by immunohistochemistry and elucidate the association between the expression of Flot-2 protein and clinicopathological characteristics of NPC. The results showed that the positive percentage of Flot-2 expression in the NPC, nasopharyngeal epithelia with atypical hyperplasia and in the control nasopharyngeal mucosa epithelia was 88.8% (119/134), 76.9% (10/13) and 5.7% (5/88), respectively. There was significantly higher expression of Flot-2 protein in NPC and nasopharyngeal epithelia with atypical hyperplasia compared to the control nasopharyngeal mucosa epithelia (P<0.001, respectively). The positive percentage of Flot-2 protein expression in NPC patients with lymph node metastasis was significantly higher than those without lymph node metastasis. Increasing of Flot-2 expression was obviously correlated with clinical stages of NPC patients. The expression of Flot-2 was proved to be the independent predicted factor for lymph node metastasis by multivariate analysis. The sensitivity of Flot-2 for predicting lymph node metastasis of NPC patients was 93%. Taken together, our results suggest that the increased expression of Flot-2 protein is a novel higher sensitivity biomarker that can predict lymph node metastases in NPC.
Collapse
Affiliation(s)
- Qiuyuan Wen
- Department of Pathology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jiao Li
- Department of Pathology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Weiyuan Wang
- Department of Pathology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Guiyuan Xie
- Department of Oncology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lina Xu
- Department of Pathology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jiadi Luo
- Department of Pathology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shuzhou Chu
- Department of Pathology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lei She
- Department of Pathology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Duo Li
- Department of Pathology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Donghai Huang
- Department of Otorhinolaryngology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Songqing Fan
- Department of Pathology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- * E-mail:
| |
Collapse
|
27
|
Morris KT, Nofchissey RA, Pinchuk IV, Beswick EJ. Chronic macrophage migration inhibitory factor exposure induces mesenchymal epithelial transition and promotes gastric and colon cancers. PLoS One 2014; 9:e98656. [PMID: 24887129 PMCID: PMC4041794 DOI: 10.1371/journal.pone.0098656] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 05/06/2014] [Indexed: 02/07/2023] Open
Abstract
Macrophage Migration Inhibitory Factor (MIF) is an inflammatory cytokine that is highly produced in gastrointestinal cancers. Since chronic inflammation is a risk factor for tumorigenesis in these cancers, in this study, the role of MIF in pro-tumorigenic events was examined. MIF and its receptor, CD74, were examined in gastric and colon tumors and found to be increased in most tumors with significantly higher expression in tumors from patients with lymph node metastasis. MIF was also found to be highly produced by cancer associated fibroblasts isolated from human tumors compared to fibroblasts from matched normal tissues from uninvolved areas. Fibroblast-produced MIF highly increased GI cancer cell proliferation, which was decreased upon neutralizing MIF or CD74. Chronic MIF treatment led to sustained proliferation and signaling events in non-transformed GI fibroblast cells, which was maintained upon removing MIF treatment for 8 weeks. Additionally, chronic treatment of normal GI cells expressing fibroblast markers for up to 16 weeks with MIF led to a drastic decrease of fibroblast markers with concurrent increase of epithelial markers. Transformation was examined by telomerase and focus forming assays. These results suggest the MIF promotes mesenchymal epithelial transition, cell transformation and tumorigenesis in GI cancers, and thus may be an important link between chronic inflammation and tumorigenesis.
Collapse
Affiliation(s)
- Katherine T. Morris
- Department of Surgery, University of New Mexico, Albuquerque, New Mexico, United States of America
| | - Robert A. Nofchissey
- Department of Molecular Genetics and Microbiology, University of New Mexico, Albuquerque, New Mexico, United States of America
| | - Irina V. Pinchuk
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Ellen J. Beswick
- Department of Molecular Genetics and Microbiology, University of New Mexico, Albuquerque, New Mexico, United States of America
- * E-mail:
| |
Collapse
|