501
|
Li L, Wang S, Zheng F, Wu W, Hann SS. Chinese herbal medicine Fuzheng Kang-Ai decoction sensitized the effect of gefitinib on inhibition of human lung cancer cells through inactivating PI3-K/Akt -mediated suppressing MUC1 expression. JOURNAL OF ETHNOPHARMACOLOGY 2016; 194:918-929. [PMID: 27989877 DOI: 10.1016/j.jep.2016.10.077] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 10/17/2016] [Accepted: 10/24/2016] [Indexed: 06/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Chinese herbal medicine (CHM) Fuzheng Kang-Ai (FZKA for short) decoction has been used as adjuvant treatment strategies in lung cancer patients for decades. However, the molecular mechanism underlying the therapeutic potential especially in sensitizing the effect of EGFR-TKI gefitinib has not been well elucidated. MATERIALS AND METHODS Cell viability was detected by MTT assay. Cell cycle distribution was detected by flow cytometry. Western blot were used to examine phosphorylation and protein levels of Akt, p65, p50 and MUC1. The mRNA level of MUC1 was measured by qRT-PCR. Transient transfection experiments were used to overexpression of Akt, p65 and MUC1. Tumor xenograft and bioluminescent imaging experiments were carried out to confirm the in vitro findings. RESULTS Cell viability was inhibited by FZKA treatment and showed more significant when treated with FZKA and gefitinib in combine in lung cancer cells. FZKA induced the cell arrest at G0/G1 phase. Mechanistically, we showed that the phosphorylation of Akt, protein expressions of p65 and MUC1 were suppressed by FZKA and even more responses were observed in the FZKA and gefitinib combining. Overexpressed Akt overcame the effect of FZKA on p65 protein, and exogenously expressed p65 resisted the inhibitory effect of MUC1 protein expression by FZKA. On the contrary, while overexpressed MUC1 had no effect on p65 expression, it feedback increased phosphorylation of Akt, and more importantly, reversed the cell growth inhibition affected by FZKA. In line with the above, our results confirmed the synergistic effects of FZKA and gefitinib combination on tumor growth, the phosphorylation of Akt, and protein expression of p65 and MUC1 in vivo. CONCLUSION This study shows that FZKA decoction inhibits the growth of NSCLC cells through Akt-mediated inhibition of p65, followed by reducing the expression of MUC1. More importantly, there is a synergistic effect of FZKA decoction and gefitinib combination with greater suppression. The positive feedback regulatory loop of MUC1 to Akt signaling pathway further added the important role of MUC1 in mediating the overall responses of FZKA decoction in this process. The in vitro and in vivo study provides an additional and a novel mechanism by which the FZKA decoction enhances the growth inhibition of gefitinib in gefitinib-resistant NSCLC cells.
Collapse
Affiliation(s)
- Longmei Li
- Laboratory of Tumor Molecular Biology and Targeted Therapies of TCM, China; Department of Medical Oncology, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province 510120, China
| | - SuMei Wang
- Department of Medical Oncology, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province 510120, China
| | - Fang Zheng
- Laboratory of Tumor Molecular Biology and Targeted Therapies of TCM, China
| | - WanYin Wu
- Department of Medical Oncology, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province 510120, China.
| | - Swei Sunny Hann
- Laboratory of Tumor Molecular Biology and Targeted Therapies of TCM, China; Department of Medical Oncology, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province 510120, China.
| |
Collapse
|
502
|
Detection of EpCAM-positive microparticles in pleural fluid: A new approach to mini-invasively identify patients with malignant pleural effusions. Oncotarget 2016; 7:3357-66. [PMID: 26689993 PMCID: PMC4823111 DOI: 10.18632/oncotarget.6581] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 11/16/2015] [Indexed: 11/25/2022] Open
Abstract
Pleural biomarkers allowing to mini-invasively discriminate benign from malignant pleural effusions are needed. Among potential candidates, microparticles (MPs) are extracellular vesicles that vectorize antigen derived from the parent cell. We hypothesized that tumor-derived MPs could be present in the pleural liquid and help to identify patients with malignant pleural effusions. Using highly sensitive flow cytometry and cryo-electron microscopy, we showed that large amounts of MPs from hematopoïetic and vascular origin could be detectable in pleural fluids. Their level did not differ between benign (n = 14) and malignant (n = 71) pleural effusions. Analysis of selected tumoral associated antigens (podoplanin, mucin 1 and EpCAM, epithelial-cell-adhesion-molecule) evidenced for the first time the presence of tumor-derived MPs expressing EpCAM in malignant pleural fluids only (Specificity = 93%, Sensitivity = 49% and 45% for flow cytometry and ELISA, respectively). The detection of EpCAM-positive-MPs (EpCAM + MPs) by flow cytometry showed a better specificity and sensitivity than ELISA to distinguish between pleural carcinoma and the others malignant pleural effusions (MPE; Sp: 96% vs 89%; Se: 79% vs 66%). Combining EpCAM+ MPs and cytology improved the diagnosis of MPE compared to cytology alone. This study establishes the basis for using EpCAM+ MPs as a promising new biomarker that could be added to the armamentarium to mini-invasively identify patients with malignant pleural effusions.
Collapse
|
503
|
Wong N, Major P, Kapoor A, Wei F, Yan J, Aziz T, Zheng M, Jayasekera D, Cutz JC, Chow MJ, Tang D. Amplification of MUC1 in prostate cancer metastasis and CRPC development. Oncotarget 2016; 7:83115-83133. [PMID: 27825118 PMCID: PMC5347757 DOI: 10.18632/oncotarget.13073] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Accepted: 10/16/2016] [Indexed: 01/06/2023] Open
Abstract
Evidence supports the upregulation of MUC1 in prostate cancer (PC). However, this has not been thoroughly investigated. We report here an association of MUC1 upregulation with PC metastasis and the development of castration resistant PC (CRPC). MUC1 expression was specifically increased in DU145 cell-derived PC stem-like cells (PCSLCs) in comparison to their non-PCSLCs counterparts. While immunohistochemistry staining of 34 primary PCs revealed variability in MUC1 expression, Nanostring technology demonstrated elevated MUC1 mRNA levels in 4 of 7 PCs compared to their normal matched tissues. By analyzing MUC1 mRNA levels and gene copy number (GCN) using the OncomineTM database, elevations in MUC1 mRNA in 82 metastases versus 280 primary PCs and in MUC1 GCN in 37 metastases over 181 primary tumors were demonstrated. Analysis of genomic datasets within cBioPortal revealed increases in MUC1 GCN in 2% (6/333) of primary PCs, 6% (9/150) of metastatic PCs, and 33% (27/82) of CRPCs; in comparison, the respective increase in androgen receptor (AR) GCN was 1%, 63%, and 56%, revealing a specific increase in MUC1 GCN for CRPC. Furthermore, a 25-gene MUC1 network was amplified in 52% of CRPCs compared to 69% of CRPCs displaying increases in an AR co-regulator group. While genomic alterations in the MUC1 network largely overlap with those in the AR group, 18 CRPCs (66.7% being neuroendocrine PC) showed genomic alterations only in the MUC1 network. Moreover, genomic alterations in the MUC1 network correlated with PC relapse. Collectively, our observations suggest a combination therapy involving MUC1-based immunotherapy and androgen deprivation.
Collapse
MESH Headings
- Adenocarcinoma/genetics
- Adenocarcinoma/metabolism
- Adenocarcinoma/secondary
- Adenocarcinoma/therapy
- Aged
- Aged, 80 and over
- Animals
- Antineoplastic Agents, Phytogenic/pharmacology
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Cell Line, Tumor
- Cell Movement/drug effects
- Computational Biology
- Databases, Genetic
- Disease Progression
- Disease-Free Survival
- Docetaxel
- Gene Amplification
- Gene Dosage
- Gene Expression Regulation, Neoplastic
- Gene Regulatory Networks
- Humans
- Male
- Mice, Inbred NOD
- Mice, SCID
- Middle Aged
- Mucin-1/genetics
- Mucin-1/metabolism
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Neuroendocrine Tumors/genetics
- Neuroendocrine Tumors/metabolism
- Neuroendocrine Tumors/secondary
- Neuroendocrine Tumors/therapy
- Prostatic Neoplasms/genetics
- Prostatic Neoplasms/metabolism
- Prostatic Neoplasms/pathology
- Prostatic Neoplasms/therapy
- Prostatic Neoplasms, Castration-Resistant/genetics
- Prostatic Neoplasms, Castration-Resistant/metabolism
- Prostatic Neoplasms, Castration-Resistant/pathology
- Prostatic Neoplasms, Castration-Resistant/therapy
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Androgen/metabolism
- Signal Transduction
- Survival Analysis
- Taxoids/pharmacology
- Time Factors
- Up-Regulation
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Nicholas Wong
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- Father Sean O'sullivan Research Institute, Hamilton, Ontario, Canada
- The Hamilton Center for Kidney Research, St. Joseph's Hospital, Hamilton, Ontario, Canada
| | - Pierre Major
- Division of Medical Oncology, Department of Oncology, McMaster University, Hamilton, Ontario, Canada
| | - Anil Kapoor
- Father Sean O'sullivan Research Institute, Hamilton, Ontario, Canada
- Department of Surgery, McMaster University, Hamilton, Ontario, Canada
| | - Fengxiang Wei
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- Father Sean O'sullivan Research Institute, Hamilton, Ontario, Canada
- The Hamilton Center for Kidney Research, St. Joseph's Hospital, Hamilton, Ontario, Canada
- The Genetics Laboratory, Longgang District Maternity and Child Healthcare Hospital, Longgang District, Shenzhen, Guangdong, P.R. China
| | - Judy Yan
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- Father Sean O'sullivan Research Institute, Hamilton, Ontario, Canada
- The Hamilton Center for Kidney Research, St. Joseph's Hospital, Hamilton, Ontario, Canada
| | - Tariq Aziz
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Mingxing Zheng
- Department of Respiratory Medicine, Shenzhen 2nd People's Hospital, Shenzhen, Guangdong, China
- Department of Respiratory Disease, The First Affiliated Hospital of Shenzhen University Medical School, Shenzhen, Guangdong, China
| | - Dulitha Jayasekera
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- Father Sean O'sullivan Research Institute, Hamilton, Ontario, Canada
- The Hamilton Center for Kidney Research, St. Joseph's Hospital, Hamilton, Ontario, Canada
| | - Jean-Claude Cutz
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Mathilda Jing Chow
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- Father Sean O'sullivan Research Institute, Hamilton, Ontario, Canada
- The Hamilton Center for Kidney Research, St. Joseph's Hospital, Hamilton, Ontario, Canada
| | - Damu Tang
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- Father Sean O'sullivan Research Institute, Hamilton, Ontario, Canada
- The Hamilton Center for Kidney Research, St. Joseph's Hospital, Hamilton, Ontario, Canada
| |
Collapse
|
504
|
Zheng D, Guo P, Xiong J, Wang S. Streptavidin Modified ZnO Film Bulk Acoustic Resonator for Detection of Tumor Marker Mucin 1. NANOSCALE RESEARCH LETTERS 2016; 11:396. [PMID: 27624339 PMCID: PMC5021656 DOI: 10.1186/s11671-016-1612-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 09/03/2016] [Indexed: 05/11/2023]
Abstract
A ZnO-based film bulk acoustic resonator has been fabricated using a magnetron sputtering technology, which was employed as a biosensor for detection of mucin 1. The resonant frequency of the thin-film bulk acoustic resonator was located near at 1503.3 MHz. The average electromechanical coupling factor [Formula: see text] and quality factor Q were 2.39 % and 224, respectively. Using the specific binding system of avidin-biotin, the streptavidin was self-assembled on the top gold electrode as the sensitive layer to indirectly test the MUC1 molecules. The resonant frequency of the biosensor decreases in response to the mass loading in range of 20-500 nM. The sensor modified with the streptavidin exhibits a high sensitivity of 4642.6 Hz/nM and a good selectivity.
Collapse
Affiliation(s)
- Dan Zheng
- Hubei Collaborative Innovation Center for Advanced Organic Chemical Materials, Faculty of Physics and Electronic Science, Hubei University, Wuhan, 430062 China
- Faculty of Electronic and Engineering, Vocational College of WuHan Software Engineering, WuHan, 430205 China
| | - Peng Guo
- Hubei Collaborative Innovation Center for Advanced Organic Chemical Materials, Faculty of Physics and Electronic Science, Hubei University, Wuhan, 430062 China
| | - Juan Xiong
- Hubei Collaborative Innovation Center for Advanced Organic Chemical Materials, Faculty of Physics and Electronic Science, Hubei University, Wuhan, 430062 China
| | - Shengfu Wang
- Hubei Collaborative Innovation Center for Advanced Organic Chemical Materials, Faculty of Chemistry and Chemical Engineering, Hubei University, Wuhan, 430062 China
| |
Collapse
|
505
|
Dréau D, Moore LJ, Alvarez-Berrios MP, Tarannum M, Mukherjee P, Vivero-Escoto JL. Mucin-1-Antibody-Conjugated Mesoporous Silica Nanoparticles for Selective Breast Cancer Detection in a Mucin-1 Transgenic Murine Mouse Model. J Biomed Nanotechnol 2016; 12:2172-2184. [PMID: 28522938 PMCID: PMC5431076 DOI: 10.1166/jbn.2016.2318] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mucin-1 (MUC1), a transmembrane glycoprotein is aberrantly expressed on ~90% of breast cancer and is an excellent target for nanoparticulate targeted imaging. In this study, the development of a dye-doped NIR emitting mesoporous silica nanoparticles platform conjugated to tumor-specific MUC1 antibody (ab-tMUC1-NIR-MSN) for in vivo optical detection of breast adenocarcinoma tissue is reported. The structural properties, the in vitro and in vivo performance of this nanoparticle-based probe were evaluated. In vitro studies showed that the MSN-based optical imaging nanoprobe is non-cytotoxic and targets efficiently mammary cancer cells overexpressing human tMUC1 protein. In vivo experiments with female C57BL/6 mice indicated that this platform accumulates mainly in the liver and did not induce short-term toxicity. In addition, we demonstrated that the ab-tMUC1-NIR-MSN nanoprobe specifically detects mammary gland tumors overexpressing human tMUC1 in a human MUC1 transgenic mouse model.
Collapse
Affiliation(s)
- Didier Dréau
- Department of Biological Sciences, The University of North Carolina at Charlotte, 9201 University City Blvd, Charlotte NC 28223, USA
- The Center for Biomedical Engineering and Science, The University of North Carolina at Charlotte, Charlotte NC 28223, USA
| | - Laura Jeffords Moore
- Department of Biological Sciences, The University of North Carolina at Charlotte, 9201 University City Blvd, Charlotte NC 28223, USA
| | - Merlis P. Alvarez-Berrios
- Department of Chemistry, The University of North Carolina at Charlotte, Charlotte NC 28223, USA; 9201 University City Blvd, Charlotte NC 28223, USA
| | - Mubin Tarannum
- Department of Chemistry, The University of North Carolina at Charlotte, Charlotte NC 28223, USA; 9201 University City Blvd, Charlotte NC 28223, USA
- The Center for Biomedical Engineering and Science, The University of North Carolina at Charlotte, Charlotte NC 28223, USA
| | - Pinku Mukherjee
- Department of Biological Sciences, The University of North Carolina at Charlotte, 9201 University City Blvd, Charlotte NC 28223, USA
- The Center for Biomedical Engineering and Science, The University of North Carolina at Charlotte, Charlotte NC 28223, USA
| | - Juan L. Vivero-Escoto
- Department of Chemistry, The University of North Carolina at Charlotte, Charlotte NC 28223, USA; 9201 University City Blvd, Charlotte NC 28223, USA
- The Center for Biomedical Engineering and Science, The University of North Carolina at Charlotte, Charlotte NC 28223, USA
| |
Collapse
|
506
|
Cascio S, Finn OJ. Intra- and Extra-Cellular Events Related to Altered Glycosylation of MUC1 Promote Chronic Inflammation, Tumor Progression, Invasion, and Metastasis. Biomolecules 2016; 6:biom6040039. [PMID: 27754373 PMCID: PMC5197949 DOI: 10.3390/biom6040039] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 08/29/2016] [Accepted: 09/27/2016] [Indexed: 12/12/2022] Open
Abstract
Altered glycosylation of mucin 1 (MUC1) on tumor cells compared to normal epithelial cells was previously identified as an important antigenic modification recognized by the immune system in the process of tumor immunosurveillance. This tumor form of MUC1 is considered a viable target for cancer immunotherapy. The importance of altered MUC1 glycosylation extends also to its role as a promoter of chronic inflammatory conditions that lead to malignant transformation and cancer progression. We review here what is known about the role of specific cancer-associated glycans on MUC1 in protein-protein interactions and intracellular signaling in cancer cells and in their adhesion to each other and the tumor stroma. The tumor form of MUC1 also creates a different landscape of inflammatory cells in the tumor microenvironment by controlling the recruitment of inflammatory cells, establishing specific interactions with dendritic cells (DCs) and macrophages, and facilitating tumor escape from the immune system. Through multiple types of short glycans simultaneously present in tumors, MUC1 acquires multiple oncogenic properties that control tumor development, progression, and metastasis at different steps of the process of carcinogenesis.
Collapse
Affiliation(s)
- Sandra Cascio
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA.
- Fondazione Ri.Med, via Bandiera 11, Palermo 90133, Italy.
| | - Olivera J Finn
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| |
Collapse
|
507
|
Szabo R, Skropeta D. Advancement of Sialyltransferase Inhibitors: Therapeutic Challenges and Opportunities. Med Res Rev 2016; 37:219-270. [DOI: 10.1002/med.21407] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 07/14/2016] [Accepted: 08/03/2016] [Indexed: 01/06/2023]
Affiliation(s)
- Rémi Szabo
- School of Chemistry; University of Wollongong; Wollongong NSW 2522 Australia
| | - Danielle Skropeta
- School of Chemistry; University of Wollongong; Wollongong NSW 2522 Australia
- Centre for Medical & Molecular Bioscience; University of Wollongong; Wollongong NSW 2522 Australia
| |
Collapse
|
508
|
Novel Immunotherapeutic Approaches for Head and Neck Squamous Cell Carcinoma. Cancers (Basel) 2016; 8:cancers8100087. [PMID: 27669306 PMCID: PMC5082377 DOI: 10.3390/cancers8100087] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 08/16/2016] [Accepted: 09/08/2016] [Indexed: 12/11/2022] Open
Abstract
The immune system plays a key role in preventing tumor formation by recognizing and destroying malignant cells. For over a century, researchers have attempted to harness the immune response as a cancer treatment, although this approach has only recently achieved clinical success. Head and neck squamous cell carcinoma (HNSCC) is the sixth most common cancer worldwide and is associated with cigarette smoking, alcohol consumption, betel nut use, and human papillomavirus infection. Unfortunately, worldwide mortality from HNSCC remains high, partially due to limits on therapy secondary to the significant morbidity associated with current treatments. Therefore, immunotherapeutic approaches to HNSCC treatment are attractive for their potential to reduce morbidity while improving survival. However, the application of immunotherapies to this disease has been challenging because HNSCC is profoundly immunosuppressive, resulting in decreased absolute lymphocyte counts, impaired natural killer cell function, reduced antigen-presenting cell function, and a tumor-permissive cytokine profile. Despite these challenges, numerous clinical trials testing the safety and efficacy of immunotherapeutic approaches to HNSCC treatment are currently underway, many of which have produced promising results. This review will summarize immunotherapeutic approaches to HNSCC that are currently undergoing clinical trials.
Collapse
|
509
|
Asadi MH, Khalifeh K, Mowla SJ. OCT4 spliced variants are highly expressed in brain cancer tissues and inhibition of OCT4B1 causes G2/M arrest in brain cancer cells. J Neurooncol 2016; 130:455-463. [PMID: 27585657 DOI: 10.1007/s11060-016-2255-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 08/21/2016] [Indexed: 12/14/2022]
Abstract
The new claim about the origin of cancer known as Cancer Stem Cell theory states that a somatic differentiated cell can dedifferentiated or reprogrammed for regaining the cancer cell features. It has been recently shown that expression of stemness factors such as Oct4, Sox2, Nanog and Klf4, in a variety of somatic cancers can leads to development of tumorogenesis. Here, the expression of Oct4 variants were evaluated in brain tumor tissues by quantitative RT-PCR and immunohistochemical (IHC) analysis. In next phase of our study, the expression of Oct4B1 was knock-down in brain cancer cell lines and its effect on cell cycle was assessed. Finally, in order to get insights into sequence-structure-function relationships of Oct4 isofroms, their sequences were analysed using bioinformatic tools. Our data revealed that all three variants of Oct4 are expressed in different types of brain cancer. The expression level of Oct4B1, in contast to Oct4B, was much higher in high-grade brain tumors compared with low-grade ones. In line with qPCR, the expression of Oct4A and B isofroms was confirmed with IHC in different types of brain tumors. Moreover, as a result of the suppression of Oct4B1 expression, the brain cancer cells were arrested in G2/M phase of cell cycle. Bioinfromatics data indicated that the predicted Oct4B1 protein have DNA binding properties. All together, our findings suggest that Oct4B1 has a potential role in tumorigenesis of brain cancer and can be considered as a new tumor marker with potential value in diagnosis and treatment of brain cancer.
Collapse
Affiliation(s)
- Malek Hossein Asadi
- Department of Biotechnology, Institute of Science and High Technology and Environmental Sciences, Graduate University of Advanced Technology, Kerman, Iran.
| | - Khosrow Khalifeh
- Department of Biology, Faculty of Sciences, University of Zanjan, Zanjan, Iran
| | - Seyed Javad Mowla
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
510
|
Fehringer G, Kraft P, Pharoah PD, Eeles RA, Chatterjee N, Schumacher FR, Schildkraut JM, Lindström S, Brennan P, Bickeböller H, Houlston RS, Landi MT, Caporaso N, Risch A, Amin Al Olama A, Berndt SI, Giovannucci EL, Grönberg H, Kote-Jarai Z, Ma J, Muir K, Stampfer MJ, Stevens VL, Wiklund F, Willett WC, Goode EL, Permuth JB, Risch HA, Reid BM, Bezieau S, Brenner H, Chan AT, Chang-Claude J, Hudson TJ, Kocarnik JK, Newcomb PA, Schoen RE, Slattery ML, White E, Adank MA, Ahsan H, Aittomäki K, Baglietto L, Blomquist C, Canzian F, Czene K, Dos-Santos-Silva I, Eliassen AH, Figueroa JD, Flesch-Janys D, Fletcher O, Garcia-Closas M, Gaudet MM, Johnson N, Hall P, Hazra A, Hein R, Hofman A, Hopper JL, Irwanto A, Johansson M, Kaaks R, Kibriya MG, Lichtner P, Liu J, Lund E, Makalic E, Meindl A, Müller-Myhsok B, Muranen TA, Nevanlinna H, Peeters PH, Peto J, Prentice RL, Rahman N, Sanchez MJ, Schmidt DF, Schmutzler RK, Southey MC, Tamimi R, Travis RC, Turnbull C, Uitterlinden AG, Wang Z, Whittemore AS, Yang XR, Zheng W, Buchanan DD, Casey G, Conti DV, Edlund CK, Gallinger S, Haile RW, Jenkins M, Le Marchand L, Li L, Lindor NM, Schmit SL, Thibodeau SN, Woods MO, Rafnar T, Gudmundsson J, Stacey SN, Stefansson K, Sulem P, Chen YA, Tyrer JP, Christiani DC, Wei Y, Shen H, Hu Z, Shu XO, Shiraishi K, Takahashi A, Bossé Y, Obeidat M, Nickle D, Timens W, Freedman ML, Li Q, Seminara D, Chanock SJ, Gong J, Peters U, Gruber SB, Amos CI, Sellers TA, Easton DF, Hunter DJ, Haiman CA, Henderson BE, Hung RJ. Cross-Cancer Genome-Wide Analysis of Lung, Ovary, Breast, Prostate, and Colorectal Cancer Reveals Novel Pleiotropic Associations. Cancer Res 2016; 76:5103-14. [PMID: 27197191 PMCID: PMC5010493 DOI: 10.1158/0008-5472.can-15-2980] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 04/05/2016] [Indexed: 01/26/2023]
Abstract
Identifying genetic variants with pleiotropic associations can uncover common pathways influencing multiple cancers. We took a two-stage approach to conduct genome-wide association studies for lung, ovary, breast, prostate, and colorectal cancer from the GAME-ON/GECCO Network (61,851 cases, 61,820 controls) to identify pleiotropic loci. Findings were replicated in independent association studies (55,789 cases, 330,490 controls). We identified a novel pleiotropic association at 1q22 involving breast and lung squamous cell carcinoma, with eQTL analysis showing an association with ADAM15/THBS3 gene expression in lung. We also identified a known breast cancer locus CASP8/ALS2CR12 associated with prostate cancer, a known cancer locus at CDKN2B-AS1 with different variants associated with lung adenocarcinoma and prostate cancer, and confirmed the associations of a breast BRCA2 locus with lung and serous ovarian cancer. This is the largest study to date examining pleiotropy across multiple cancer-associated loci, identifying common mechanisms of cancer development and progression. Cancer Res; 76(17); 5103-14. ©2016 AACR.
Collapse
Affiliation(s)
- Gordon Fehringer
- Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, Toronto, Canada
| | - Peter Kraft
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | | | | | | | | | | | - Sara Lindström
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Paul Brennan
- International Agency for Research on Cancer, Lyon, France
| | | | | | | | | | - Angela Risch
- Division of Cancer Genetics/Epigenetics, Department of Molecular Biology, University of Salzburg, Salzburg, Austria. Division of Epigenomics and Cancer Risk Factors, DKFZ - German Cancer Research Center, Heidelberg, Germany. Translational Lung Research Center Heidelberg (TLRC-H), German Center for Lung Research (DZL), Heidelberg, Germany
| | | | | | | | | | | | - Jing Ma
- Harvard Medical School, Boston Massachusetts. Brigham and Women's Hospital, Boston, Massachusetts
| | - Kenneth Muir
- University of Manchester, Manchester, United Kingdom. The University of Warwick, Coventry, United Kingdom
| | | | - Victoria L Stevens
- Epidemiology Research Program, American Cancer Society, Atlanta, Georgia
| | | | - Walter C Willett
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | | | | | | | | | | | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany. Division of Preventive Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany. German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Andrew T Chan
- Massachusetts General Hospital, Boston, Massachusetts
| | - Jenny Chang-Claude
- National Center for Tumor Diseases and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | | | | | - Robert E Schoen
- University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | | | - Emily White
- Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Muriel A Adank
- VU University Medical Center, Amsterdam, the Netherlands
| | | | - Kristiina Aittomäki
- University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | | | - Carl Blomquist
- University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Federico Canzian
- Genomic Epidemiology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Isabel Dos-Santos-Silva
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - A Heather Eliassen
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts. Harvard Medical School, Boston Massachusetts
| | | | | | - Olivia Fletcher
- Breakthrough Research Centre, The Institute of Cancer Research, London, United Kingdom
| | | | - Mia M Gaudet
- Epidemiology Research Program, American Cancer Society, Atlanta, Georgia
| | - Nichola Johnson
- Breakthrough Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Per Hall
- Karolinska Institutet, Stockholm, Sweden
| | - Aditi Hazra
- Harvard Medical School, Boston Massachusetts
| | - Rebecca Hein
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ) Heidelberg, Germany. Institute of Medical Statistics, Informatics and Epidemiology, University of Cologne, Cologne, Germany
| | - Albert Hofman
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - John L Hopper
- Melbourne School of Population Health, University of Melbourne, Melbourne, Victoria, Australia
| | | | - Mattias Johansson
- International Agency for Research on Cancer, Lyon, France. Department of Biobank Research, Umea University, Umea, Sweden
| | - Rudolf Kaaks
- National Center for Tumor Diseases and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Peter Lichtner
- German Research Center for Environmental Health, Neuherberg, Germany
| | | | - Eiliv Lund
- Institute of Community Medicine, UiT The Arctic University of Norway, Tromso, Norway
| | - Enes Makalic
- Melbourne School of Population Health, University of Melbourne, Melbourne, Victoria, Australia
| | | | | | - Taru A Muranen
- University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Heli Nevanlinna
- University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland
| | - Petra H Peeters
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, the Netherlands
| | - Julian Peto
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | | | | | - Maria Jose Sanchez
- Escuela Andaluza de Salud Publica, Instituto de Investigacion Biosanitaria ibs. GRANADA, Granada, Spain. Hospitales Universitarios de Granada/Universidad de Granada, Granada, Spain. CIBER de Epidemiología y Salud Pública CIBERESP, Madrid, Spain
| | - Daniel F Schmidt
- Melbourne School of Population Health, University of Melbourne, Melbourne, Victoria, Australia
| | | | | | | | - Ruth C Travis
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | | | - Andre G Uitterlinden
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands. Department of Internal Medicine, Erasmus MC, Rotterdam, the Netherlands
| | | | | | | | - Wei Zheng
- Vanderbilt University, Nashville, Tennessee
| | | | - Graham Casey
- University of Southern California, Los Angeles, California
| | - David V Conti
- University of Southern California, Los Angeles, California
| | | | - Steven Gallinger
- Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, Toronto, Canada
| | | | - Mark Jenkins
- The University of Melbourne, Melbourne, Victoria, Australia
| | - Loïc Le Marchand
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - Li Li
- Case Comprehensive Cancer Center and Mary Ann Swetland Center for Environmental Health, Case Western Reserve University, Cleveland, Ohio
| | | | | | | | - Michael O Woods
- Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| | | | | | | | | | | | | | | | | | - Yongyue Wei
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Hongbing Shen
- Nanjing Medical University School of Public Health, Nanjing, China
| | - Zhibin Hu
- Nanjing Medical University School of Public Health, Nanjing, China
| | | | - Kouya Shiraishi
- Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan
| | - Atsushi Takahashi
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Yohan Bossé
- Department of Molecular Medicine, Institut universitaire de cardiologie et de pneumologie de Québec, Laval University, Québec, Canada
| | - Ma'en Obeidat
- University of British Columbia Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, Canada
| | - David Nickle
- Merck & Co, Merck Research Laboratories, Seattle, Washington
| | - Wim Timens
- University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, the Netherlands
| | | | | | | | | | - Jian Gong
- Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Ulrike Peters
- Fred Hutchinson Cancer Research Center, Seattle, Washington
| | | | | | | | | | - David J Hunter
- Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | | | | | - Rayjean J Hung
- Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, Toronto, Canada.
| |
Collapse
|
511
|
Amini A, Masoumi-Moghaddam S, Ehteda A, Liauw W, Morris DL. Depletion of mucin in mucin-producing human gastrointestinal carcinoma: Results from in vitro and in vivo studies with bromelain and N-acetylcysteine. Oncotarget 2016; 6:33329-44. [PMID: 26436698 PMCID: PMC4741769 DOI: 10.18632/oncotarget.5259] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 09/18/2015] [Indexed: 01/02/2023] Open
Abstract
Aberrant expression of membrane-associated and secreted mucins, as evident in epithelial tumors, is known to facilitate tumor growth, progression and metastasis, and to provide protection against adverse growth conditions, chemotherapy and immune surveillance. Emerging evidence provides support for the oncogenic role of MUC1 in gastrointestinal carcinomas and relates its expression to an invasive phenotype. Similarly, mucinous differentiation of gastrointestinal tumors, in particular increased or de novo expression of MUC2 and/or MUC5AC, is widely believed to imply an adverse clinicopathological feature. Through formation of viscous gels, too, MUC2 and MUC5AC significantly contribute to the biology and pathogenesis of mucin-secreting gastrointestinal tumors. Here, we investigated the mucin-depleting effects of bromelain (BR) and N-acetylcysteine (NAC), in nine different regimens as single or combination therapy, in in vitro (MKN45, KATOIII and LS174T cell lines) and in vivo (female nude mice bearing intraperitoneal MKN45 and LS174T) settings. The inhibitory effects of the treatment on cancer cell growth and proliferation were also evaluated in vivo. Our results suggest that a combination of BR and NAC with dual effects on growth and mucin products of mucin-expressing tumor cells is a promising candidate towards the development of novel approaches to gastrointestinal malignancies with the involvement of mucin pathology. This capability supports the use of this combination formulation in locoregional approaches for reducing the adverse effects of the aberrantly secreted gel-forming mucins, as in pseudomyxoma peritonei and similar pathologies with ectopic production of mucin.
Collapse
Affiliation(s)
- Afshin Amini
- Department of Surgery, St George Hospital, The University of New South Wales, Kogarah, Sydney NSW 2217, Australia
| | - Samar Masoumi-Moghaddam
- Department of Surgery, St George Hospital, The University of New South Wales, Kogarah, Sydney NSW 2217, Australia
| | - Anahid Ehteda
- Department of Surgery, St George Hospital, The University of New South Wales, Kogarah, Sydney NSW 2217, Australia
| | - Winston Liauw
- Cancer Care Center, St George Hospital, The University of New South Wales, Kogarah, Sydney NSW 2217, Australia
| | - David L Morris
- Department of Surgery, St George Hospital, The University of New South Wales, Kogarah, Sydney NSW 2217, Australia
| |
Collapse
|
512
|
Mittal S, Kaur H, Gautam N, Mantha AK. Biosensors for breast cancer diagnosis: A review of bioreceptors, biotransducers and signal amplification strategies. Biosens Bioelectron 2016; 88:217-231. [PMID: 27567264 DOI: 10.1016/j.bios.2016.08.028] [Citation(s) in RCA: 136] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 08/10/2016] [Accepted: 08/11/2016] [Indexed: 11/19/2022]
Abstract
Breast cancer is highly prevalent in females and accounts for second highest number of deaths, worldwide. Cumbersome, expensive and time consuming detection techniques presently available for detection of breast cancer potentiates the need for development of novel, specific and ultrasensitive devices. Biosensors are the promising and selective detection devices which hold immense potential as point of care (POC) tools. Present review comprehensively scrutinizes various breast cancer biosensors developed so far and their technical evaluation with respect to efficiency and potency of selected bioreceptors and biotransducers. Use of glycoproteins, DNA biomarkers, micro-RNA, circulatory tumor cells (CTC) and some potential biomarkers are introduced briefly. The review also discusses various strategies used in signal amplification such as nanomaterials, redox mediators, p19 protein, duplex specific nucleases (DSN) and redox cycling.
Collapse
Affiliation(s)
- Sunil Mittal
- Centre for Environmental Science and Technology, Central University of Punjab, Bathinda, 151001 India.
| | - Hardeep Kaur
- Centre for Environmental Science and Technology, Central University of Punjab, Bathinda, 151001 India.
| | - Nandini Gautam
- Centre for Environmental Science and Technology, Central University of Punjab, Bathinda, 151001 India.
| | - Anil K Mantha
- Centre for Animal Sciences, Central University of Punjab, Bathinda, 151001 India.
| |
Collapse
|
513
|
Sheng YH, He Y, Hasnain SZ, Wang R, Tong H, Clarke DT, Lourie R, Oancea I, Wong KY, Lumley JW, Florin TH, Sutton P, Hooper JD, McMillan NA, McGuckin MA. MUC13 protects colorectal cancer cells from death by activating the NF-κB pathway and is a potential therapeutic target. Oncogene 2016; 36:700-713. [PMID: 27399336 PMCID: PMC5541270 DOI: 10.1038/onc.2016.241] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Revised: 05/24/2016] [Accepted: 06/01/2016] [Indexed: 02/07/2023]
Abstract
MUC13 is a transmembrane mucin glycoprotein that is over produced by many cancers, although its functions are not fully understood. Nuclear factor-κB (NF-κB) is a key transcription factor promoting cancer cell survival, but therapeutically targeting this pathway has proved difficult because NF-κB has pleiotropic functions. Here, we report that MUC13 prevents colorectal cancer cell death by promoting two distinct pathways of NF-kB activation, consequently upregulating BCL-XL. MUC13 promoted tumor necrosis factor (TNF)-induced NF-κB activation by interacting with TNFR1 and the E3 ligase, cIAP1, to increase ubiquitination of RIPK1. MUC13 also promoted genotoxin-induced NF-κB activation by increasing phosphorylation of ATM and SUMOylation of NF-κB essential modulator. Moreover, elevated expression of cytoplasmic MUC13 and NF-κB correlated with colorectal cancer progression and metastases. Our demonstration that MUC13 enhances NF-κB signaling in response to both TNF and DNA-damaging agents provides a new molecular target for specific inhibition of NF-κB activation. As proof of principle, silencing MUC13 sensitized colorectal cancer cells to killing by cytotoxic drugs and inflammatory signals and abolished chemotherapy-induced enrichment of CD133+ CD44+ cancer stem cells, slowed xenograft growth in mice, and synergized with 5-fluourouracil to induce tumor regression. Therefore, these data indicate that combining chemotherapy and MUC13 antagonism could improve the treatment of metastatic cancers.
Collapse
Affiliation(s)
- Y H Sheng
- Inflammatory Disease Biology and Therapeutics Group-Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Y He
- Cancer Biology Group, Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - S Z Hasnain
- Inflammatory Disease Biology and Therapeutics Group-Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - R Wang
- Inflammatory Disease Biology and Therapeutics Group-Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - H Tong
- Inflammatory Disease Biology and Therapeutics Group-Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - D T Clarke
- Molecular Basis of Disease Program, School of Medical Sciences, Griffith University, Gold Coast Campus, Southport, Queensland, Australia
| | - R Lourie
- Inflammatory Disease Biology and Therapeutics Group-Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia.,Inflammatory Bowel Diseases Group, Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - I Oancea
- Inflammatory Disease Biology and Therapeutics Group-Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia.,Inflammatory Bowel Diseases Group, Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - K Y Wong
- Inflammatory Disease Biology and Therapeutics Group-Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - J W Lumley
- Wesley Hospital, Auchenflower, Australia
| | - T H Florin
- Inflammatory Bowel Diseases Group, Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - P Sutton
- Mucosal Immunology, Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, Melbourne, Victoria, Australia.,Centre for Animal Biotechnology, School of Veterinary and Agricultural Science, University of Melbourne, Melbourne, Victoria, Australia.,Department of Paediatrics, University of Melbourne, Victoria, Australia
| | - J D Hooper
- Cancer Biology Group, Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - N A McMillan
- Molecular Basis of Disease Program, School of Medical Sciences, Griffith University, Gold Coast Campus, Southport, Queensland, Australia
| | - M A McGuckin
- Inflammatory Disease Biology and Therapeutics Group-Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| |
Collapse
|
514
|
Is mucin a determinant of peritoneal dissemination of gastrointestinal cancer? Analysis of mucin depletion in two preclinical models. Clin Transl Oncol 2016; 19:261-264. [PMID: 27193208 DOI: 10.1007/s12094-016-1519-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 05/10/2016] [Indexed: 01/28/2023]
Abstract
BACKGROUND Mucinous gastrointestinal cancers may indicate a higher propensity for widespread peritoneal seeding than their non-mucinous counterparts. We hypothesized that mucin content of gastrointestinal cancer cells and tumors is an indicator of cell viability and a determinant of the peritoneal tumor burden and tested our hypothesis in relevant experimental models. METHODS MKN45 and LS174T models of human gastrointestinal cancer were treated with known mucin-depleting agents in vitro and in vivo, their mucin production was evaluated with Western blot immunohistochemistry, PAS staining and ELISA, and its correlation with cell viability and peritoneal tumor burden was analyzed. RESULTS A relationship was found between the viability of cancer cells and their mucin levels in vitro. In agreement, when treated animal models were categorized into low- and high-burden groups (based on the weight and number of the peritoneal nodules), tumoral mucin levels were found to be significantly higher in the latter group. CONCLUSIONS Tumoral mucin is apparently among the factors that dictate the pattern and extent of the peritoneal spread of gastrointestinal cancer, where it allows for enhanced dissemination and redistribution. If further tested and validated, our hypothesis could lay the basis for the development of novel mucin-targeted strategies.
Collapse
|
515
|
Zhang H, Liu C, Zhang F, Geng F, Xia Q, Lu Z, Xu P, Xie Y, Wu H, Yu B, Wu J, Yu X, Kong W. MUC1 and survivin combination tumor gene vaccine generates specific immune responses and anti-tumor effects in a murine melanoma model. Vaccine 2016; 34:2648-55. [DOI: 10.1016/j.vaccine.2016.04.045] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 03/17/2016] [Accepted: 04/17/2016] [Indexed: 12/13/2022]
|
516
|
Wu N, Bao L, Ding L, Ju H. A Single Excitation-Duplexed Imaging Strategy for Profiling Cell Surface Protein-Specific Glycoforms. Angew Chem Int Ed Engl 2016. [DOI: 10.1002/ange.201601233] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Na Wu
- State Key Laboratory of Analytical Chemistry for Life Science; School of Chemistry and Chemical Engineering; Nanjing University; Nanjing 210023 P.R. China
| | - Lei Bao
- State Key Laboratory of Analytical Chemistry for Life Science; School of Chemistry and Chemical Engineering; Nanjing University; Nanjing 210023 P.R. China
| | - Lin Ding
- State Key Laboratory of Analytical Chemistry for Life Science; School of Chemistry and Chemical Engineering; Nanjing University; Nanjing 210023 P.R. China
| | - Huangxian Ju
- State Key Laboratory of Analytical Chemistry for Life Science; School of Chemistry and Chemical Engineering; Nanjing University; Nanjing 210023 P.R. China
| |
Collapse
|
517
|
Wu N, Bao L, Ding L, Ju H. A Single Excitation-Duplexed Imaging Strategy for Profiling Cell Surface Protein-Specific Glycoforms. Angew Chem Int Ed Engl 2016; 55:5220-4. [PMID: 27001418 DOI: 10.1002/anie.201601233] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Indexed: 11/10/2022]
Abstract
This work develops a site-specific duplexed luminescence resonance energy transfer system on cell surface for simultaneous imaging of two kinds of monosaccharides on a specific protein by single near-infrared excitation. The single excitation-duplexed imaging system utilizes aptamer modified upconversion luminescent nanoparticles as an energy donor to target the protein, and two fluorescent dye acceptors to tag two kinds of cell surface monosaccharides by a dual metabolic labeling technique. Upon excitation at 980 nm, only the dyes linked to protein-specific glycans can be lit up by the donor by two parallel energy transfer processes, for in situ duplexed imaging of glycoforms on specific protein. Using MUC1 as the model, this strategy can visualize distinct glycoforms of MUC1 on various cell types and quantitatively track terminal monosaccharide pattern. This approach provides a versatile platform for profiling protein-specific glycoforms, thus contributing to the study of the regulation mechanisms of protein functions by glycosylation.
Collapse
Affiliation(s)
- Na Wu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P.R. China
| | - Lei Bao
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P.R. China
| | - Lin Ding
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P.R. China.
| | - Huangxian Ju
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P.R. China.
| |
Collapse
|
518
|
Das S, Rachagani S, Torres-Gonzalez MP, Lakshmanan I, Majhi PD, Smith LM, Wagner KU, Batra SK. Carboxyl-terminal domain of MUC16 imparts tumorigenic and metastatic functions through nuclear translocation of JAK2 to pancreatic cancer cells. Oncotarget 2016; 6:5772-87. [PMID: 25691062 PMCID: PMC4467401 DOI: 10.18632/oncotarget.3308] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Accepted: 01/02/2015] [Indexed: 02/07/2023] Open
Abstract
MUC16 (CA125) is a type-I transmembrane glycoprotein that is up-regulated in multiple cancers including pancreatic cancer (PC). However, the existence and role of carboxyl-terminal MUC16 generated following its cleavage in PC is unknown. Our previous study using a systematic dual-epitope tagged domain deletion approach of carboxyl-terminal MUC16 has demonstrated the generation of a 17-kDa cleaved MUC16 (MUC16-Cter). Here, we demonstrate the functional significance of MUC16-Cter in PC using the dual-epitope tagged version (N-terminal FLAG- and C-terminal HA-tag) of 114 carboxyl-terminal residues of MUC16 (F114HA). In vitro analyses using F114HA transfected MiaPaCa-2 and T3M4 cells showed enhanced proliferation, motility and increased accumulation of cells in the G2/M phase with apoptosis resistance, a feature associated with cancer stem cells (CSCs). This was supported by enrichment of ALDH+ CSCs along with enhanced drug-resistance. Mechanistically, we demonstrate a novel function of MUC16-Cter that promotes nuclear translocation of JAK2 resulting in phosphorylation of Histone-3 up-regulating stemness-specific genes LMO2 and NANOG. Jak2 dependence was demonstrated using Jak2+/+ and Jak2−/− cells. Using eGFP-Luciferase labeled cells, we demonstrate enhanced tumorigenic and metastatic potential of MUC16-Cter in vivo. Taken together, we demonstrate that MUC16-Cter mediated enrichment of CSCs is partly responsible for tumorigenic, metastatic and drug-resistant properties of PC cells.
Collapse
Affiliation(s)
- Srustidhar Das
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Satyanarayana Rachagani
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Maria P Torres-Gonzalez
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Imayavaramban Lakshmanan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Prabin D Majhi
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Lynette M Smith
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kay-Uwe Wagner
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.,Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.,Department of Pathology, University of Nebraska Medical Center, Omaha, NE, USA.,Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
519
|
Chou CH, Huang MJ, Chen CH, Shyu MK, Huang J, Hung JS, Huang CS, Huang MC. Up-regulation of C1GALT1 promotes breast cancer cell growth through MUC1-C signaling pathway. Oncotarget 2016; 6:6123-35. [PMID: 25762620 PMCID: PMC4467426 DOI: 10.18632/oncotarget.3045] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 01/06/2015] [Indexed: 11/25/2022] Open
Abstract
Aberrant glycosylation is frequently observed in cancers. Core 1 β1,3-galactosyltransferase (C1GALT1) is an exclusive enzyme in humans that catalyzes the biosynthesis of core 1 O-glycan structure, Gal-GalNAc-O-Ser/Thr, whose expression is commonly up-regulated during tumorigenesis. Little is known about the function of C1GALT1 in breast cancer. This study aims to determine the correlation between C1GALT1 expression and breast cancer clinicopathological features and roles of C1GALT1 in breast cancer malignant phenotypes. Public databases and our data showed that C1GALT1 mRNA and C1GALT1 protein are frequently up-regulated in breast cancer; and increased C1GALT1 expression correlates with higher histological grade and advanced tumor stage. Overexpression of C1GALT1 enhanced breast cancer cell growth, migration, and invasion in vitro as well as tumor growth in vivo. Conversely, C1GALT1 knockdown suppressed these malignant phenotypes. Furthermore, C1GALT1 modulates O-glycan structures on Mucin (MUC) 1 and promotes MUC1-C/β-catenin signaling in breast cancer cells. These findings suggest that C1GALT1 enhances breast cancer malignant progression through promoting MUC1-C/β-catenin signaling pathway. Unveiling the function of C1GALT1 in breast cancer opens new insights to the roles of C1GALT1 and O-glycosylation in tumorigenesis and renders the potential of C1GALT1 as a target of novel therapeutic agent development.
Collapse
Affiliation(s)
- Chih-Hsing Chou
- Graduate Institute of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Miao-Juei Huang
- Graduate Institute of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chi-Hau Chen
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei, Taiwan
| | - Ming-Kwang Shyu
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei, Taiwan
| | - John Huang
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Ji-Shiang Hung
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Chiun-Sheng Huang
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Min-Chuan Huang
- Graduate Institute of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei, Taiwan.,Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
520
|
Pre-clinical toxicity and immunogenicity evaluation of a MUC1-MBP/BCG anti-tumor vaccine. Int Immunopharmacol 2016; 33:108-18. [PMID: 26896668 DOI: 10.1016/j.intimp.2016.02.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 01/28/2016] [Accepted: 02/05/2016] [Indexed: 12/23/2022]
Abstract
Mucin 1 (MUC1), as an oncogene, plays a key role in the progression and tumorigenesis of many human adenocarcinomas and is an attractive target in tumor immunotherapy. Our previous study showed that the MUC1-MBP/BCG anti-tumor vaccine induced a MUC1-specific Th1-dominant immune response, simulated MUC1-specific cytotoxic T lymphocyte killing activity, and could significantly inhibit MUC1-expression B16 cells' growth in mice. To help move the vaccine into a Phase I clinical trial, in the current study, a pre-clinical toxicity and immunogenicity evaluation of the vaccine was conducted. The evaluation was comprised of a single-dose acute toxicity study in mice, repeat-dose chronic toxicity and immunogenicity studies in rats, and pilot toxicity and immunogenicity studies in cynomolgus monkeys. The results showed that treatment with the MUC1-MBP/BCG anti-tumor vaccine did not cause any organ toxicity, except for arthritis or local nodules induced by BCG in several rats. Furthermore, the vaccine significantly increased the levels of IFN-γ in rats, indicating that Th1 cells were activated. In addition, the results showed that the MUC1-MBP/BCG anti-tumor vaccine induced a MUC1-specific IgG antibody response both in rats and cynomolgus monkeys. Collectively, these data are beneficial to move the MUC1-MBP/BCG anti-tumor vaccine into a Phase I clinical trial.
Collapse
|
521
|
Electrochemiluminescence Aptasensor for the MUC1 Protein Based on Multi-functionalized Graphene Oxide Nanocomposite. ELECTROANAL 2016. [DOI: 10.1002/elan.201501068] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
522
|
Ham SY, Kwon T, Bak Y, Yu JH, Hong J, Lee SK, Yu DY, Yoon DY. Mucin 1-mediated chemo-resistance in lung cancer cells. Oncogenesis 2016; 5:e185. [PMID: 26779808 PMCID: PMC4728677 DOI: 10.1038/oncsis.2015.47] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 11/11/2015] [Accepted: 11/20/2015] [Indexed: 02/07/2023] Open
Abstract
Paclitaxel (PTX) is a commonly used drug to treat diverse cancer types. However, its treatment can generate resistance and the mechanisms of PTX-resistance in lung cancers are still unclear. We demonstrated that non-small cell lung cancers (NSCLCs) survive PTX treatment. Compared with the progenitor NSCLC A549 cells, the PTX-resistant A549 cells (A549/PTX) displayed enhanced sphere-formation ability. The proportion of the cancer stem cell marker, aldehyde dehydrogenase-positive cells, and epithelial-mesenchymal transition signaling protein levels were also elevated in A549/PTX. Importantly, the levels of oncoproteins phosphoinositide-3 kinase/Akt, mucin 1 cytoplasmic domain (MUC1-C) and β-catenin were also significantly elevated in A549/PTX. Furthermore, nuclear translocation of MUC1-C and β-catenin increased in A549/PTX. The c-SRC protein, an activator of MUC1-C, was also overexpressed in A549/PTX. These observations led to the hypothesis that enhanced expression of MUC1-C is associated with stemness and PTX resistance in NSCLCs. To test this, we knocked down or overexpressed MUC1-C in A549/PTX and found that inhibition of MUC1-C expression coupled with PTX treatment was sufficient to reduce the sphere-forming ability and survival of A549/PTX. In summary, our in vitro and in vivo studies have revealed a potential mechanism of MUC1-C-mediated PTX resistance and provided insights into a novel therapeutic measure for lung cancers.
Collapse
Affiliation(s)
- S Y Ham
- Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University, Seoul, Republic of Korea
| | - T Kwon
- Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University, Seoul, Republic of Korea
| | - Y Bak
- Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University, Seoul, Republic of Korea
| | - J-H Yu
- Department of Bacteriology and Genetic, Food Research Institute (FRI), Molecular and Environmental Toxicology Center (METC), University of Wisconsin, Madison, WI, USA
| | - J Hong
- College of Pharmacy, Medical Research Center, Chungbuk National University, Cheongju, Republic of Korea
| | - S K Lee
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - D-Y Yu
- Aging Intervention Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - D-Y Yoon
- Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
523
|
Ferreira JA, Peixoto A, Neves M, Gaiteiro C, Reis CA, Assaraf YG, Santos LL. Mechanisms of cisplatin resistance and targeting of cancer stem cells: Adding glycosylation to the equation. Drug Resist Updat 2016; 24:34-54. [DOI: 10.1016/j.drup.2015.11.003] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 11/09/2015] [Accepted: 11/18/2015] [Indexed: 02/06/2023]
|
524
|
Karaulov AV, Gurina NN, Novikov DV, Fomina SG, Novikov VV. [Role of MUC1 Expression in Tumor Progression]. ACTA ACUST UNITED AC 2016; 71:392-6. [PMID: 29297994 DOI: 10.15690/vramn736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Mucin 1 (MUC1) is a multistructural and multifunctional protein that is involved in regulating diverse cellular activities. This strongly glycosylated transmembrane protein forms a mucous gel on the surface of epithelial cells that protects the cells from injury. MUC1 acts as a signaling molecule and transcription factor modulating metabolism and resistance to bacterial-induced inflammation. This article presents a review of the relationship between structural and functional changes of the MUC1 and the characteristics of cancer cells. The alteration in MUC1 expression level, a number of structural forms, protein glycosylation and localization occurs in cancer cells. These alterations lead to metabolic reprogramming associated with proliferation, resistance to hypoxia and angiogenesis which affects the survival of cancer cells. Furthermore, cancer cells can take advantage of MUC1 interaction with adhesion molecules for invasion and metastasis. Thus, MUC1 plays a key role both in the homeostasis of epithelial cells and in cancer progression. Understanding the role of MUC1 expression in tumor cells survival is important for the development of new monitoring and therapeutic approaches for the treatment MUC1 positive maligancies.
Collapse
|
525
|
Mucin1 shifts Smad3 signaling from the tumor-suppressive pSmad3C/p21(WAF1) pathway to the oncogenic pSmad3L/c-Myc pathway by activating JNK in human hepatocellular carcinoma cells. Oncotarget 2015; 6:4253-65. [PMID: 25714018 PMCID: PMC4414187 DOI: 10.18632/oncotarget.2973] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 12/16/2014] [Indexed: 12/31/2022] Open
Abstract
Mucin1 (MUC1) is a transmembrane glycoprotein that acts as an oncogene in human hepatic tumorigenesis. Hepatocellular carcinoma (HCC) cells often gain advantage by reducing the tumor-suppressive activity of transforming growth factor beta (TGF-β) together with stimulation of its oncogenic activity as in MUC1 expressing HCC cells; however, molecular mechanisms remain largely unknown. Type I TGF-β receptor (TβRI) and c-Jun NH2-terminal kinase (JNK) differentially phosphorylate Smad3 mediator to create 2 phosphorylated forms: COOH-terminally phosphorylated Smad3 (pSmad3C) and linker-phosphorylated Smad3 (pSmad3L). Here, we report that MUC1 overexpression in HCC cell lines suppresses TβRI-mediated pSmad3C signaling which involves growth inhibition by up-regulating p21WAF1. Instead, MUC1 directly activates JNK to stimulate oncogenic pSmad3L signaling, which fosters cell proliferation by up-regulating c-Myc. Conversely, MUC1 gene silencing in MUC1 expressing HCC cells results in preserved tumor-suppressive function via pSmad3C, while eliminating pSmad3L-mediated oncogenic activity both in vitro and in vivo. In addition, high correlation between MUC1 and pSmad3L/c-Myc but not pSmad3C/p21WAF1 expression was observed in HCC tissues from patients. Collectively, these results indicate that MUC1 shifts Smad3 signaling from a tumor-suppressive pSmad3C/p21WAF1 to an oncogenic pSmad3L/c-Myc pathway by directly activating JNK in HCC cells, suggesting that MUC1 is an important target for HCC therapy.
Collapse
|
526
|
Apostolopoulos V, Stojanovska L, Gargosky SE. MUC1 (CD227): a multi-tasked molecule. Cell Mol Life Sci 2015; 72:4475-500. [PMID: 26294353 PMCID: PMC11113675 DOI: 10.1007/s00018-015-2014-z] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 07/23/2015] [Accepted: 08/06/2015] [Indexed: 12/16/2022]
Abstract
Mucin 1 (MUC1 [CD227]) is a high-molecular weight (>400 kDa), type I membrane-tethered glycoprotein that is expressed on epithelial cells and extends far above the glycocalyx. MUC1 is overexpressed and aberrantly glycosylated in adenocarcinomas and in hematological malignancies. As a result, MUC1 has been a target for tumor immunotherapeutic studies in mice and in humans. MUC1 has been shown to have anti-adhesive and immunosuppressive properties, protects against infections, and is involved in the oncogenic process as well as in cell signaling. In addition, MUC1 plays a key role in the reproductive tract, in the immune system (affecting dendritic cells, monocytes, T cells, and B cells), and in chronic inflammatory diseases. Evidence for all of these roles for MUC1 is discussed herein and demonstrates that MUC1 is truly a multitasked molecule.
Collapse
Affiliation(s)
- Vasso Apostolopoulos
- Centre for Chronic Disease, College of Health and Biomedicine, Victoria University, Melbourne, VIC, Australia.
| | - Lily Stojanovska
- Centre for Chronic Disease, College of Health and Biomedicine, Victoria University, Melbourne, VIC, Australia
| | | |
Collapse
|
527
|
Wen R, Gao F, Zhou CJ, Jia YB. Polymorphisms in mucin genes in the development of gastric cancer. World J Gastrointest Oncol 2015; 7:328-337. [PMID: 26600932 PMCID: PMC4644855 DOI: 10.4251/wjgo.v7.i11.328] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2015] [Revised: 07/01/2015] [Accepted: 09/07/2015] [Indexed: 02/05/2023] Open
Abstract
Gastric cancer (GC) is the third leading cause of cancer-related death worldwide. In areas of high prevalence, such as Japan, South Korea and China, most cases of GC are related to Helicobacter pylori (H. pylori), which involves well-characterized sequential stages, including infection, atrophic gastritis, intestinal metaplasia, dysplasia, and GC. Mucins are the most abundant high-molecular-weight glycoproteins in mucus, which is the first line of defense and plays a major role in blocking pathogenic factors. Normal gastric mucosa shows expression of MUC1, MUC5AC and MUC6 that is specific to cell type. However, the specific pattern of MUC1, MUC5AC and MUC6 expression is changed in gastric carcinogenesis, accompanied by de novo expression of secreted MUC2. Recent studies have provided evidence that variations in these mucin genes affect many steps of GC development, such as H. pylori infection, and gastric precancerous lesions. In this review, we focus on studies of the association between polymorphisms in mucin genes and development of GC. This information should be helpful for the early detection, surveillance, and treatment of GC.
Collapse
|
528
|
A Cell ELISA for the quantification of MUC1 mucin (CD227) expressed by cancer cells of epithelial and neuroectodermal origin. Cell Immunol 2015; 298:96-103. [DOI: 10.1016/j.cellimm.2015.09.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Revised: 09/18/2015] [Accepted: 09/25/2015] [Indexed: 12/13/2022]
|
529
|
Lakshmanan I, Ponnusamy MP, Macha MA, Haridas D, Majhi PD, Kaur S, Jain M, Batra SK, Ganti AK. Mucins in lung cancer: diagnostic, prognostic, and therapeutic implications. J Thorac Oncol 2015; 10:19-27. [PMID: 25319180 DOI: 10.1097/jto.0000000000000404] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Aberrant expression of mucins is associated with cancer development and metastasis. An overexpression of few mucins contributes to oncogenesis by enhancing cancer cell growth and providing constitutive survival signals. This review focuses on the importance of mucins both in the normal bronchial epithelial cells and the malignant tumors of the lung and their contribution in the diagnosis and prognosis of lung cancer patients. During lung cancer progression, mucins either alone or through their interaction with many receptor tyrosine kinases mediate cell signals for growth and survival of cancer cells. Also, stage-specific expression of certain mucins, like MUC1, is associated with poor prognosis from lung cancer. Thus, mucins are emerging as attractive targets for developing novel therapeutic approaches for lung cancer. Several strategies targeting mucin expression and function are currently being investigated to control lung cancer progression.
Collapse
Affiliation(s)
- Imayavaramban Lakshmanan
- *Department of Biochemistry and Molecular Biology, †Department of Pathology and Microbiology, ‡Eppley Institute for Research in Cancer and Allied Diseases, §Department of Internal Medicine, VA Nebraska-Western Iowa Health Care System, Omaha, Nebraska, and ‖Division of Oncology-Hematology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | | | | | | | | | | | | | | | | |
Collapse
|
530
|
Kannan K, Kordestani GK, Galagoda A, Coarfa C, Yen L. Aberrant MUC1-TRIM46-KRTCAP2 Chimeric RNAs in High-Grade Serous Ovarian Carcinoma. Cancers (Basel) 2015; 7:2083-93. [PMID: 26492273 PMCID: PMC4695878 DOI: 10.3390/cancers7040878] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 09/28/2015] [Accepted: 10/08/2015] [Indexed: 01/08/2023] Open
Abstract
High-grade serous ovarian cancer (HGSC) is among the most lethal forms of cancer in women. By analyzing the mRNA-seq reads from The Cancer Genome Atlas (TCGA), we uncovered a novel cancer-enriched chimeric RNA as the result of splicing between MUC1, a highly glycosylated transmembrane mucin, TRIM46, a tripartite motif containing protein, and KRTCAP2, a keratinocyte associated protein. Experimental analyses by RT-PCR (reverse transcription PCR) and Sanger sequencing using an in-house cohort of 59 HGSC patient tumors revealed a total of six MUC1-TRIM46-KRTCAP2 isoforms joined by different annotated splice sites between these genes. These chimeric isoforms are not detected in non-cancerous ovaries, yet are present in three out of every four HGSC patient tumors, a significant frequency given the exceedingly heterogeneous nature of this disease. Transfection of the cDNA of MUC1-TRIM46-KRTCAP2 isoforms in mammalian cells led to the translation of mutant MUC1 fusion proteins that are unglycosylated and cytoplasmically localized as opposed to the cell membrane, a feature resembling the tumor-associated MUC1. Because the parental MUC1 is overexpressed in 90% of HGSC tumors and has been proposed as a clinical biomarker and therapeutic target, the chimeric MUC1-TRIM46-KRTCAP2 isoforms identified in this report could represent significantly better MUC1 variants for the same clinical utilities.
Collapse
Affiliation(s)
- Kalpana Kannan
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Gona Karimi Kordestani
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Anika Galagoda
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Cristian Coarfa
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA.
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Laising Yen
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, USA.
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA.
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA.
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
531
|
Glycosylation-Based Serum Biomarkers for Cancer Diagnostics and Prognostics. BIOMED RESEARCH INTERNATIONAL 2015; 2015:490531. [PMID: 26509158 PMCID: PMC4609776 DOI: 10.1155/2015/490531] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 05/28/2015] [Accepted: 05/31/2015] [Indexed: 12/13/2022]
Abstract
Cancer is the second most common cause of death in developed countries with approximately 14 million newly diagnosed individuals and over 6 million cancer-related deaths in 2012. Many cancers are discovered at a more advanced stage but better survival rates are correlated with earlier detection. Current clinically approved cancer biomarkers are most effective when applied to patients with widespread cancer. Single biomarkers with satisfactory sensitivity and specificity have not been identified for the most common cancers and some biomarkers are ineffective for the detection of early stage cancers. Thus, novel biomarkers with better diagnostic and prognostic performance are required. Aberrant protein glycosylation is well known hallmark of cancer and represents a promising source of potential biomarkers. Glycoproteins enter circulation from tissues or blood cells through active secretion or leakage and patient serum is an attractive option as a source for biomarkers from a clinical and diagnostic perspective. A plethora of technical approaches have been developed to address the challenges of glycosylation structure detection and determination. This review summarises currently utilised glycoprotein biomarkers and novel glycosylation-based biomarkers from the serum glycoproteome under investigation as cancer diagnostics and for monitoring and prognostics and includes details of recent high throughput and other emerging glycoanalytical techniques.
Collapse
|
532
|
Yuan H, Wang J, Wang F, Zhang N, Li Q, Xie F, Chen T, Zhai R, Wang F, Guo Y, Ni W, Tai G. Mucin 1 gene silencing inhibits the growth of SMMC-7721 human hepatoma cells through Bax-mediated mitochondrial and caspase-8-mediated death receptor apoptotic pathways. Mol Med Rep 2015; 12:6782-8. [PMID: 26398332 PMCID: PMC4626135 DOI: 10.3892/mmr.2015.4323] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 08/25/2015] [Indexed: 01/04/2023] Open
Abstract
Mucin 1 (MUC1) is an oncogene that has a crucial role in the pathogenesis and progression of the majority of epithelial malignant tumors. Our previous study demonstrated that MUC1 gene silencing inhibited the growth of SMMC-7721 cells in vitro and in vivo, however, whether this growth inhibition is associated with apoptotic cell death remains to be elucidated. In the present study, it was found that MUC1 gene silencing not only resulted in the inhibition of SMMC-7721 cell growth, determined using a clone formation assay in vitro and a tumor xenograft mouse model with an in vivo imaging system, but also induced apoptotic alterations in SMMC-7721 cells, determined using Hoechst 33342 staining, flow cytometry with an Annexin V-PE staining and a DNA ladder assay. Further investigation using western blotting revealed that cytochrome c was released from the mitochondria into the cytoplasm, and caspase-8 and caspase-9 were activated in MUC1 gene-silenced SMMC-7721 cells. The pro-apoptotic protein Bcl-2-associated X protein (Bax) and the tumor suppressor p53 were increased, while the anti-apoptotic protein B-cell lymphoma 2 was decreased in MUC1 gene-silenced cells. In addition, results from the co-immunoprecipitation experiments demonstrated that the MUC1 cytoplasmic tail can bind directly to Bax or caspase-8 and these interactions were reduced upon MUC1 gene silencing in SMMC-7721 cells. The above results indicate that MUC1 gene silencing induces growth inhibition in SMMC-7721 cells through Bax-mediated mitochondrial and caspase-8-mediated death receptor apoptotic pathways.
Collapse
Affiliation(s)
- Hongyan Yuan
- Department of Immunology, College of Basic Medical Science, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Juan Wang
- Department of Immunology, College of Basic Medical Science, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Fengli Wang
- Department of Clinical Laboratory, Dalian Municipal Central Hospital, Dalian, Liaoning 116033, P.R. China
| | - Nannan Zhang
- Department of Immunology, College of Basic Medical Science, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Qiongshu Li
- Department of Immunology, College of Basic Medical Science, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Fei Xie
- Department of Immunology, College of Basic Medical Science, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Tanxiu Chen
- Department of Immunology, College of Basic Medical Science, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Ruiping Zhai
- Department of Immunology, College of Basic Medical Science, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Fang Wang
- Department of Immunology, College of Basic Medical Science, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yingying Guo
- Department of Immunology, College of Basic Medical Science, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Weihua Ni
- Department of Immunology, College of Basic Medical Science, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Guixiang Tai
- Department of Immunology, College of Basic Medical Science, Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
533
|
Chinen AB, Guan CM, Ferrer JR, Barnaby SN, Merkel TJ, Mirkin CA. Nanoparticle Probes for the Detection of Cancer Biomarkers, Cells, and Tissues by Fluorescence. Chem Rev 2015; 115:10530-74. [PMID: 26313138 DOI: 10.1021/acs.chemrev.5b00321] [Citation(s) in RCA: 629] [Impact Index Per Article: 69.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Alyssa B Chinen
- Department of Chemistry, ‡Department of Chemical Engineering, §Department of Interdepartmental Biological Sciences, and ∥International Institute for Nanotechnology, Northwestern University , 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Chenxia M Guan
- Department of Chemistry, ‡Department of Chemical Engineering, §Department of Interdepartmental Biological Sciences, and ∥International Institute for Nanotechnology, Northwestern University , 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Jennifer R Ferrer
- Department of Chemistry, ‡Department of Chemical Engineering, §Department of Interdepartmental Biological Sciences, and ∥International Institute for Nanotechnology, Northwestern University , 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Stacey N Barnaby
- Department of Chemistry, ‡Department of Chemical Engineering, §Department of Interdepartmental Biological Sciences, and ∥International Institute for Nanotechnology, Northwestern University , 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Timothy J Merkel
- Department of Chemistry, ‡Department of Chemical Engineering, §Department of Interdepartmental Biological Sciences, and ∥International Institute for Nanotechnology, Northwestern University , 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Chad A Mirkin
- Department of Chemistry, ‡Department of Chemical Engineering, §Department of Interdepartmental Biological Sciences, and ∥International Institute for Nanotechnology, Northwestern University , 2145 Sheridan Road, Evanston, Illinois 60208, United States
| |
Collapse
|
534
|
Luo H, Guo W, Wang F, You Y, Wang J, Chen X, Wang J, Wang Y, Du Y, Chen X, Xue C, Song G, Wang F. miR-1291 targets mucin 1 inhibiting cell proliferation and invasion to promote cell apoptosis in esophageal squamous cell carcinoma. Oncol Rep 2015; 34:2665-73. [PMID: 26324125 DOI: 10.3892/or.2015.4206] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 07/10/2015] [Indexed: 11/05/2022] Open
Abstract
MicroRNAs (miRNAs) are well known as important regulators in various cancer development. In the present study, we focused on the expression and biological function of miR-1291 in esophageal squamous cell carcinoma (ESCC). Compared with adjacent non-tumorous tissue samples, qRT-PCR data showed significant downregulation of miR-1291 in 54 ESCC tissue samples (P<0.05), which was also significantly associated with lymph node metastases and clinical stage (P<0.05). Cell Counting Kit-8 (CCK-8), colony formation, Transwell and flow cytometric apoptosis assays were performed to detect the effect of miR-1291 upregulation, and the results showed inhibition of the proliferation, invasion and promotion of apoptosis in EC9706 and EC-1 cells. Using bioinformatic analyses, we found that mucin 1 (MUC1) was a potential target for miR-1291. Luciferase assays were performed to reveal that miR-1291 inhibited MUC1 expression by targeting the seed region of MUC1 3'-untranslated region (3'UTR). We also found that the expression of MUC1 lacking in 3'UTR abrogated the anti-invasion and pro-apoptosis function of miR-1291. Our results demonstrated the importance of miR-1291 in targeting MUC1 for the regulation of esophagus cancer growth, invasion and apoptosis, and may be helpful for developing new targets for early diagnosis or new therapeutic targets for ESCC.
Collapse
Affiliation(s)
- Hailan Luo
- Department of Basic Medical Sciences, Luohe Medical College, Luohe, Henan 462002, P.R. China
| | - Wentao Guo
- Department of Basic Medical Sciences, Luohe Medical College, Luohe, Henan 462002, P.R. China
| | - Fei Wang
- Department of Basic Medical Sciences, Luohe Medical College, Luohe, Henan 462002, P.R. China
| | - Yanjie You
- Department of Basic Medical Sciences, Luohe Medical College, Luohe, Henan 462002, P.R. China
| | - Jianguo Wang
- Department of Basic Medical Sciences, Luohe Medical College, Luohe, Henan 462002, P.R. China
| | - Xudong Chen
- Department of Basic Medical Sciences, Luohe Medical College, Luohe, Henan 462002, P.R. China
| | - Jihong Wang
- Department of Basic Medical Sciences, Luohe Medical College, Luohe, Henan 462002, P.R. China
| | - Yuanyuan Wang
- College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Yuwen Du
- College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Xiaonan Chen
- College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Changgui Xue
- College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Guohua Song
- Department of Basic Medical Sciences, Luohe Medical College, Luohe, Henan 462002, P.R. China
| | - Fuqing Wang
- Department of Basic Medical Sciences, Luohe Medical College, Luohe, Henan 462002, P.R. China
| |
Collapse
|
535
|
Yang CW, Chang CYY, Lai MT, Chang HW, Lu CC, Chen Y, Chen CM, Lee SC, Tsai PW, Yang SH, Lin CH, Sheu JJC, Tsai FJ. Genetic variations of MUC17 are associated with endometriosis development and related infertility. BMC MEDICAL GENETICS 2015; 16:60. [PMID: 26285705 PMCID: PMC4593232 DOI: 10.1186/s12881-015-0209-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 08/04/2015] [Indexed: 12/13/2022]
Abstract
BACKGROUND Genetic alterations of mucin genes, such as MUC2 and MUC4, were previously identified to be associated with endometriosis and related infertility. Additionally, gene expression profiling has confirmed MUC17 to be overexpressed in mucinous ovarian carcinoma; however, its associated risk for endometriosis remains unclear. This study was focused on the potential impact of genetic variations in MUC17 on endometriosis development and associated clinical features. METHODS The study subjects included 189 female Taiwanese patients with pathology-proven endometriosis and 191 healthy Taiwanese women as controls. Five single-nucleotide polymorphisms (rs4729645, rs10953316, rs74974199, rs4729655, and rs4729656) within the MUC17 gene were selected and genotyped using the Taqman genotyping assay to examine the allele frequency and genotype distributions of MUC17 polymorphisms. RESULTS Genotyping revealed that the A allele at rs10953316 in MUC17 was a protective genetic factor in endometriosis development (p = 0.008; OR = 0.53; 95% CI: 0.36-0.79). Genetic variation of rs4729655 protected patients against endometriosis-induced infertility, but was associated with a higher cancer antigen 125 (CA125) level. Base-pairing analysis, called MaxExpect, predicted an additional loop in the mRNA structure caused by rs10953316 polymorphism, possibly influencing ribosome sliding and translation efficiency. Such predictions were confirmed by immunohistochemistry that patients with AA genotype at rs10953316 showed low MUC17 levels in their endometrium, patients with GA genotype showed moderate levels, and strong staining could be found in patients with GG genotype. CONCLUSIONS MUC17 polymorphisms are involved in endometriosis development and the associated infertility in the Taiwanese population.
Collapse
Affiliation(s)
- Ching-Wen Yang
- The Institute of Basic Medical Sciences, National Cheng Kung University, Tainan, Taiwan. .,Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan.
| | - Cherry Yin-Yi Chang
- Department of Obstetrics and Gynecology, China Medical University, Taichung, Taiwan. .,Institute of Environmental Health, China Medical University, Taichung, Taiwan.
| | - Ming-Tsung Lai
- Department of Pathology, Taichung Hospital, Taichung, Taiwan.
| | - Hui-Wen Chang
- School of Medicine, China Medical University, Taichung, Taiwan.
| | - Cheng-Chan Lu
- The Institute of Basic Medical Sciences, National Cheng Kung University, Tainan, Taiwan. .,Department of Pathology, Medical College, National Cheng Kung University, Tainan, Taiwan.
| | - Yi Chen
- Human Genetic Center, China Medical University Hospital, Taichung, Taiwan.
| | - Chih-Mei Chen
- Human Genetic Center, China Medical University Hospital, Taichung, Taiwan.
| | - Shan-Chih Lee
- Collage of Medical Science and Technology, Chung Shan Medical University, Taichung, Taiwan.
| | - Pei-Wen Tsai
- Human Genetic Center, China Medical University Hospital, Taichung, Taiwan.
| | - Su-Han Yang
- Human Genetic Center, China Medical University Hospital, Taichung, Taiwan.
| | - Chih-Hung Lin
- Department of Pathology, Kaohsiung University Hospital, Kaohsiung, Taiwan.
| | - Jim Jinn-Chyuan Sheu
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan. .,Human Genetic Center, China Medical University Hospital, Taichung, Taiwan. .,School of Chinese Medicine, China Medical University, Taichung, Taiwan. .,Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan.
| | - Fuu-Jen Tsai
- Human Genetic Center, China Medical University Hospital, Taichung, Taiwan. .,School of Post-Baccalaureate Chinese Medicine, China Medical University, Taichung, Taiwan.
| |
Collapse
|
536
|
Wang J, Liu G, Li Q, Wang F, Xie F, Zhai R, Guo Y, Chen T, Zhang N, Ni W, Yuan H, Tai G. Mucin1 promotes the migration and invasion of hepatocellular carcinoma cells via JNK-mediated phosphorylation of Smad2 at the C-terminal and linker regions. Oncotarget 2015; 6:19264-78. [PMID: 26057631 PMCID: PMC4662489 DOI: 10.18632/oncotarget.4267] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 05/13/2015] [Indexed: 02/07/2023] Open
Abstract
Mucin1 (MUC1), as an oncogene, plays a key role in the progression and tumorigenesis of many human adenocarcinomas. In this study, wound-healing, transwell migration and matrigel invasion assays showed that MUC1 promotes human hepatocellular carcinoma (HCC) cell migration and invasion by MUC1 gene silencing and overexpressing. Treatment with exogenous transforming growth factor beta (TGF-β)1, TGF-β type I receptor (TβRI) inhibitor, TGF-β1 siRNAs, or activator protein 1 (AP-1) inhibitor to MUC1-overexpressing HCC cells revealed that MUC1-induced autocrine TGF-β via JNK/AP-1 pathway promotes the cell migration and invasion. In addition, the migration and invasion of HCC cells were more significantly inhibited by JNK inhibitor compared with that by TβRI inhibitor or TGF-β1 siRNAs. Further studies demonstrated that MUC1-mediated JNK activation not only enhances the phosphorylation of Smad2 C-terminal at Ser-465/467 site (Smad2C) through TGF-β/TβRI, but also directly enhances the phosphorylation of Smad2 linker region at Ser-245/250/255 site (Smad2L), and then both of them collaborate to upregulate matrix metalloproteinase (MMP)-9-mediated cell migration and invasion of HCC. These results indicate that MUC1 is an attractive target in liver cancer therapy.
Collapse
Affiliation(s)
- Juan Wang
- Department of Immunology, College of Basic Medical Science, Jilin University, Changchun, China
| | - Guomu Liu
- Department of Immunology, College of Basic Medical Science, Jilin University, Changchun, China
| | - Qiongshu Li
- Department of Immunology, College of Basic Medical Science, Jilin University, Changchun, China
| | - Fang Wang
- Department of Immunology, College of Basic Medical Science, Jilin University, Changchun, China
| | - Fei Xie
- Department of Immunology, College of Basic Medical Science, Jilin University, Changchun, China
| | - Ruiping Zhai
- Department of Immunology, College of Basic Medical Science, Jilin University, Changchun, China
| | - Yingying Guo
- Department of Immunology, College of Basic Medical Science, Jilin University, Changchun, China
| | - Tanxiu Chen
- Department of Immunology, College of Basic Medical Science, Jilin University, Changchun, China
| | - Nannan Zhang
- Department of Immunology, College of Basic Medical Science, Jilin University, Changchun, China
| | - Weihua Ni
- Department of Immunology, College of Basic Medical Science, Jilin University, Changchun, China
| | - Hongyan Yuan
- Department of Immunology, College of Basic Medical Science, Jilin University, Changchun, China
| | - Guixiang Tai
- Department of Immunology, College of Basic Medical Science, Jilin University, Changchun, China
| |
Collapse
|
537
|
Mocellin S, Verdi D, Pooley KA, Nitti D. Genetic variation and gastric cancer risk: a field synopsis and meta-analysis. Gut 2015; 64:1209-19. [PMID: 25731870 DOI: 10.1136/gutjnl-2015-309168] [Citation(s) in RCA: 135] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2015] [Accepted: 02/06/2015] [Indexed: 12/19/2022]
Abstract
BACKGROUND Data on genetic susceptibility to sporadic gastric carcinoma have been published at a growing pace, but to date no comprehensive overview and quantitative summary has been available. METHODS We conducted a systematic review and meta-analysis of the evidence on the association between DNA variation and risk of developing stomach cancer. To assess result credibility, summary evidence was graded according to the Venice criteria and false positive report probability (FPRP) was calculated to further validate result noteworthiness. Meta-analysis was also conducted for subgroups, which were defined by ethnicity (Asian vs Caucasian), tumour histology (intestinal vs diffuse), tumour site (cardia vs non-cardia) and Helicobacter pylori infection status (positive vs negative). RESULTS Literature search identified 824 eligible studies comprising 2 530 706 subjects (cases: 261 386 (10.3%)) and investigating 2841 polymorphisms involving 952 distinct genes. Overall, we performed 456 primary and subgroup meta-analyses on 156 variants involving 101 genes. We identified 11 variants significantly associated with disease risk and assessed to have a high level of summary evidence: MUC1 rs2070803 at 1q22 (diffuse carcinoma subgroup), MTX1 rs2075570 at 1q22 (diffuse), PSCA rs2294008 at 8q24.2 (non-cardia), PRKAA1 rs13361707 5p13 (non-cardia), PLCE1 rs2274223 10q23 (cardia), TGFBR2 rs3087465 3p22 (Asian), PKLR rs3762272 1q22 (diffuse), PSCA rs2976392 (intestinal), GSTP1 rs1695 11q13 (Asian), CASP8 rs3834129 2q33 (mixed) and TNF rs1799724 6p21.3 (mixed), with the first nine variants characterised by a low FPRP. We also identified polymorphisms with lower quality significant associations (n=110). CONCLUSIONS We have identified several high-quality biomarkers of gastric cancer susceptibility. These data will form the backbone of an annually updated online resource that will be integral to the study of gastric carcinoma genetics and may inform future screening programmes.
Collapse
Affiliation(s)
- Simone Mocellin
- Department of Surgery Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Daunia Verdi
- Department of Surgery Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Karen A Pooley
- Department of Public Health and Primary Care, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, UK
| | - Donato Nitti
- Department of Surgery Oncology and Gastroenterology, University of Padova, Padova, Italy
| |
Collapse
|
538
|
Abstract
OBJECTIVE Eighty percent of pancreatic ductal adenocarcinomas (PDAs) overexpress mucin 1 (MUC1), a transmembrane mucin glycoprotein. MUC1(high) PDA patients also express high levels of cyclooxygenase 2 (COX-2) and show poor prognosis. The cytoplasmic tail of MUC1 (MUC1-CT) partakes in oncogenic signaling, resulting in accelerated cancer progression. Our aim was to understand the regulation of Cox-2 expression by MUC1. METHODS Levels of COX-2 and MUC1 were determined in MUC1(-/-), MUC1(low), and MUC1(high) PDA cells and tumors using reverse transcriptase-polymerase chain reaction, Western blot, and immunohistochemistry. Proliferative and invasive potential was assessed using MTT and Boyden chamber assays. Chromatin immunoprecipitation was performed to evaluate binding of MUC1-CT to the promoter of COX-2 gene. RESULTS Significantly higher levels of COX-2 mRNA and protein were detected in MUC1(high) versus MUC1(low/null) cells, which were recapitulated in vivo. In addition, deletion of MUC1 gene and transient knockdown of MUC1 led to decreased COX-2 level. Also, MUC1-CT associated with the COX-2 promoter at ∼1000 base pairs upstream of the transcription start site, the same gene locus where nuclear factor κB p65 associates with the COX-2 promoter. CONCLUSIONS Data supports a novel regulation of COX-2 gene by MUC1 in PDA, the intervention of which may lead to a better therapeutic targeting in PDA patients.
Collapse
|
539
|
Establishment of a heterotypic 3D culture system to evaluate the interaction of TREG lymphocytes and NK cells with breast cancer. J Immunol Methods 2015. [PMID: 26215372 DOI: 10.1016/j.jim.2015.07.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Three-dimensional (3D) culture approaches to investigate breast tumour progression are yielding information more reminiscent of the in vivo microenvironment. We have established a 3D Matrigel system to determine the interactions of luminal phenotype MCF-7 cells and basal phenotype MDA-MB-231 cells with regulatory T lymphocytes and Natural Killer cells. Immune cells were isolated from peripheral blood using magnetic cell sorting and their phenotype validated using flow cytometry both before and after activation with IL-2 and phytohaemagglutinin. Following the establishment of the heterotypic culture system, tumour cells displayed morphologies and cell-cell associations distinct to that observed in 2D monolayer cultures, and associated with tissue remodelling and invasion processes. We found that the level of CCL4 secretion was influenced by breast cancer phenotype and immune stimulation. We further established that for RNA extraction, the use of proteinase K in conjunction with the Qiagen RNeasy Mini Kit and only off-column DNA digestion gave the best RNA yield, purity and integrity. We also investigated the efficacy of the culture system for immunolocalisation of the biomarkers oestrogen receptor-α and the glycoprotein mucin 1 in luminal phenotype breast cancer cells; and epidermal growth factor receptor in basal phenotype breast cancer cells, in formalin-fixed, paraffin-wax embedded cultures. The expression of these markers was shown to vary under immune mediation. We thus demonstrate the feasibility of using this co-culture system for downstream applications including cytokine analysis, immunolocalisation of tumour biomarkers on serial sections and RNA extraction in accordance with MIQE guidelines.
Collapse
|
540
|
Lu Y, Lu F, Zeng S, Sun S, Lu L, Liu L. Genetics and gastric cancer susceptibility. Int J Clin Exp Med 2015; 8:8377-8383. [PMID: 26309491 PMCID: PMC4538020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 04/23/2015] [Indexed: 06/04/2023]
Abstract
Gastric cancer has high morbidity and mortality in China. It is ranked first in malignant tumors of the digestive system. Its etiology and pathogenesis are still unclear, but they may be associated with a variety of factors. Genetic susceptibility genes have become a research hotspot in China. Elucidating the genetic mechanisms of gastric cancer can facilitate achieving individualized prevention and developing more effective methods to reduce clinical adverse consequences, which has important clinical significance. Genetic susceptibility results from the influence of genetic factors or specific genetic defects that endow an individual's offspring with certain physiological and metabolic features that are prone to certain diseases. Currently, studies on the genetic susceptibility genes of gastric cancer have become a hotspot. The purpose is to screen for the etiology of gastric cancer, search for gene therapy methods, and ultimately provide a scientific basis for the prevention and control of gastric cancer. This article reviews the current progress of studies on genetic susceptibility genes for gastric cancer.
Collapse
Affiliation(s)
- Yan Lu
- Department of Gastroenterology, Liaocheng People’s HospitalLiaocheng 252000, China
| | - Fang Lu
- Department of Traditional Chinese Medicine, Liaocheng People’s HospitalLiaocheng 252000, China
| | - Sha Zeng
- Central Laboratory, Liaocheng People’s HospitalLiaocheng 252000, China
| | - Suqing Sun
- Foreign Affairs Office, Liaocheng People’s HospitalLiaocheng 252000, China
| | - Li Lu
- Department of Pharmacy, Liaocheng International Peace HospitalLiaocheng 252000, China
| | - Lifeng Liu
- Department of Gastroenterology, Liaocheng People’s HospitalLiaocheng 252000, China
| |
Collapse
|
541
|
Madi A, Bransburg-Zabary S, Maayan-Metzger A, Dar G, Ben-Jacob E, Cohen IR. Tumor-associated and disease-associated autoantibody repertoires in healthy colostrum and maternal and newborn cord sera. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2015; 194:5272-81. [PMID: 25917091 PMCID: PMC4432729 DOI: 10.4049/jimmunol.1402771] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 03/31/2015] [Indexed: 02/07/2023]
Abstract
In this work, we studied autoantibody repertoires and Ig isotypes in 71 mothers and their 104 healthy newborns (including twins and triplets delivered term or premature). Newborns receive maternal IgG Abs via the placenta before birth, but developing infants must produce their own IgM and IgA Abs. We used an Ag microarray analysis to detect binding to a selection of 295 self-Ags, compared with 27 standard foreign Ags. The magnitude of binding to specific self-Ags was found to be not less than that to the foreign Ags. As expected, each newborn shared with its mother a similar IgG repertoire-manifest as early as the 24th week of gestation. IgM and IgA autoantibody repertoires in cord sera were highly correlated among the newborns and differed from their mothers' repertoires; the latter differed in sera and milk. The autoantibodies bound to self-Ags known to be associated with tumors and to autoimmune diseases. Thus, autoantibody repertoires in healthy humans--the immunological homunculus--arise congenitally, differ in maternal milk and sera, and mark the potential of the immune system to attack tumors, beneficially, or healthy tissues, harmfully; regulation of the tissue site, the dynamics, and the response phenotype of homuncular autoimmunity very likely affects health.
Collapse
Affiliation(s)
- Asaf Madi
- Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel; School of Physics and Astronomy, Tel Aviv University, 69978 Tel Aviv, Israel; Department of Neonatology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, 5262100 Ramat Gan, Israel
| | - Sharron Bransburg-Zabary
- Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel; School of Physics and Astronomy, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Ayala Maayan-Metzger
- Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel; Department of Neonatology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, 5262100 Ramat Gan, Israel
| | - Gittit Dar
- School of Physics and Astronomy, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Eshel Ben-Jacob
- School of Physics and Astronomy, Tel Aviv University, 69978 Tel Aviv, Israel; Center for Theoretical Biological Physics, Rice University, Houston, TX 77005; and
| | - Irun R Cohen
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
542
|
Multivalent aptamer/gold nanoparticle-modified graphene oxide for mass spectrometry-based tumor tissue imaging. Sci Rep 2015; 5:10292. [PMID: 25973571 PMCID: PMC4431351 DOI: 10.1038/srep10292] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 04/08/2015] [Indexed: 12/14/2022] Open
Abstract
The protein mucin1 (MUC1) is an attractive target for cancer biomarkers because it is overexpressed in most adenocarcinomas. In this study, we exploited a MUC1-binding aptamer (AptMUC1) as a targeting agent for nanoparticle-based imaging systems coupled with laser desorption/ionization mass spectrometry (LDI-MS). We found that AptMUC1-conjugated gold nanoparticles immobilized, through hydrophobic and π–π interactions, on graphene oxide (AptMUC1–Au NPs/GO) bound effectively to MUC1 units on tumor cell membranes. The ultrahigh density and high flexibility of AptMUC1 on the GO surface enhanced the platform’s cooperative and multivalent binding affinity for MUC1 on cell membranes. After we had labeled MUC1-overexpressing MCF-7 cells (human breast adenocarcinoma cell line) with AptMUC1–Au NPs/GO, we used LDI-MS to monitor Au cluster ions ([Aun]+; n = 1–3), resulting in the detection of as few as 100 MCF-7 cells. We also employed this AptMUC1–Au NPs/GO–LDI-MS system to analyze four different MUC1 expression cell lines. In addition, the AptMUC1–Au NPs/GO platform could be used further as a labeling agent for tumor tissue imaging when coupled with LDI-MS. Thus, Apt–Au NPs/GO can function as a highly amplified signal transducer through the formation of large Au clusters ions during LDI-MS analysis.
Collapse
|
543
|
Chiu WJ, Ling TK, Chiang HP, Lin HJ, Huang CC. Monitoring Cluster Ions Derived from Aptamer-Modified Gold Nanofilms under Laser Desorption/Ionization for the Detection of Circulating Tumor Cells. ACS APPLIED MATERIALS & INTERFACES 2015; 7:8622-8630. [PMID: 25855859 DOI: 10.1021/acsami.5b00731] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
In this paper, we describe the use of pulsed laser desorption/ionization mass spectrometry (LDI-MS) for the detection of tumor cells through the analysis of gold cluster ions [Aun](+) from aptamer-modified gold nanofilms (Au NFs). We observed not only the transformation of the Au NFs into gold nanoparticles (Au NPs) but also the formation of gaseous gold cluster ions ([Au(n)](+); n = 1-5) under irradiation with a nanosecond pulsed laser. The size and density of the formed Au NPs and the abundance of [Au(n)](+) ions were both highly dependent on the thickness of the Au NFs (10-100 nm). Thin Au NFs tended to form highly dense Au NPs on the substrate and favored the desorption and ionization of gold cluster ions. The signal intensities of the [Au(n)](+) species, monitoring using mass spectrometry, decreased upon increasing the thickness of the Au NF from 10 to 100 nm and after modification with thiolated DNA. Furthermore, we found that Au NFs modified with mucin1-binding aptamer (AptMUC1-Au NFs) could selectively enrich MCF-7 cells (human breast adenocarcinoma cell line) in blood samples; coupled with LDI-MS analysis of the [Au(n)](+) ions, we could detect MCF-7 cells selectively in blood samples at abundances as low as 10 cells. This approach offers the advantages of high sensitivity, selectivity, and throughput for the detection of circulating tumor cells, and has great potential for use as a powerful analytical platform for clinical diagnoses of tumor metastasis.
Collapse
Affiliation(s)
| | | | | | | | - Chih-Ching Huang
- ⊥School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| |
Collapse
|
544
|
Complex of MUC1, CIN85 and Cbl in Colon Cancer Progression and Metastasis. Cancers (Basel) 2015; 7:342-52. [PMID: 25675408 PMCID: PMC4381262 DOI: 10.3390/cancers7010342] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 02/03/2015] [Indexed: 12/30/2022] Open
Abstract
We previously reported that CIN85, an 85 KDa protein known to be involved in tumor cell migration and metastasis through its interaction with Cbl, associates with MUC1 in tumor cells. MUC1/CIN85 complex also regulates migration and invasion of tumor cells in vitro. Here, we examined specifically human colon carcinoma tissue microarrays (TMA) by immunohistochemistry for the expression of MUC1 and CIN85 and their potential role in cancer progression and metastasis. We detected a significant increase in expression of both MUC1 and CIN85 associated with advanced tumor stage and lymph node metastasis. We further investigated if Cbl could also be present in the MUC1/CIN85 complex. Co-immunoprecipitation assay showed that Cbl co-localized both with CIN85 and with MUC1 in a human colon cancer cell line. To begin to investigate the in vivo relevance of MUC1 overexpression and association with CIN85 and Cbl in cancer development and progression, we used human MUC1 transgenic mice that express MUC1 on the colonic epithelial cells, treated with azoxymethane to initiate and dextran sulfate sodium (AOM/DSS) to promote colorectal carcinogenesis. MUC1.Tg mice showed higher tumor incidence and decreased survival when compared with wild-type mice. Consistent with the in vitro data, the association of MUC1, CIN85 and Cbl was detected in colon tissues of AOM/DSS-treated MUC1 transgenic mice. MUC1/CIN85/Cbl complex appears to contribute to promotion and progression of colon cancer and thus increased expression of MUC1, CIN85 and Cbl in early stage colon cancer might be predictive of poor prognosis.
Collapse
|
545
|
Drake RR, Jones EE, Powers TW, Nyalwidhe JO. Altered glycosylation in prostate cancer. Adv Cancer Res 2015; 126:345-82. [PMID: 25727153 DOI: 10.1016/bs.acr.2014.12.001] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Prostate cancer is annually the most common newly diagnosed cancer in men. The prostate functions as a major secretory gland for the production of glycoproteins critical to sperm activation and reproduction. Prostate-specific antigen (PSA), produced by the prostate, is one of the most commonly assayed glycoproteins in blood, serving as a biomarker for early detection and progression of prostate cancer. The single site of N-glycosylation on PSA has been the target of multiple glycan characterization studies. In this review, the extensive number of studies that have characterized the changes in O-linked and N-linked glycosylations associated with prostate cancer development and progression will be summarized. This includes analysis of the glycosylation of PSA, and other prostate glycoproteins, in tissues, clinical biofluids, and cell line models. Other studies are summarized in the context of understanding the complexities of these glycan changes in order to address the many confounding questions associated with prostate cancer, as well as efforts to improve prostate cancer biomarker assays using targeted glycomic-based strategies.
Collapse
Affiliation(s)
- Richard R Drake
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, South Carolina, USA.
| | - E Ellen Jones
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Thomas W Powers
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Julius O Nyalwidhe
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, Virginia, USA
| |
Collapse
|
546
|
Viticchiè G, Muller PAJ. c-Met and Other Cell Surface Molecules: Interaction, Activation and Functional Consequences. Biomedicines 2015; 3:46-70. [PMID: 28536399 PMCID: PMC5344229 DOI: 10.3390/biomedicines3010046] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 01/08/2015] [Indexed: 12/18/2022] Open
Abstract
The c-Met receptor, also known as the HGF receptor, is one of the most studied tyrosine kinase receptors, yet its biological functions and activation mechanisms are still not fully understood. c-Met has been implicated in embryonic development and organogenesis, in tissue remodelling homeostasis and repair and in cancer metastasis. These functions are indicative of the many cellular processes in which the receptor plays a role, including cell motility, scattering, survival and proliferation. In the context of malignancy, sustained activation of c-Met leads to a signalling cascade involving a multitude of kinases that initiate an invasive and metastatic program. Many proteins can affect the activation of c-Met, including a variety of other cell surface and membrane-spanning molecules or receptors. Some cell surface molecules share structural homology with the c-Met extracellular domain and can activate c-Met via clustering through this domain (e.g., plexins), whereas other receptor tyrosine kinases can enhance c-Met activation and signalling through intracellular signalling cascades (e.g., EGFR). In this review, we provide an overview of c-Met interactions and crosstalk with partner molecules and the functional consequences of these interactions on c-Met activation and downstream signalling, c-Met intracellular localization/recycling and c-Met degradation.
Collapse
Affiliation(s)
- Giuditta Viticchiè
- MRC (Medical Research Council) Toxicology Unit, Lancaster Road, Leicester LE1 9HN, UK.
| | - Patricia A J Muller
- MRC (Medical Research Council) Toxicology Unit, Lancaster Road, Leicester LE1 9HN, UK.
| |
Collapse
|
547
|
Abstract
MicroRNAs (miRNAs) are an important class of small non-coding RNAs that direct post-transcriptional gene regulation by different mechanisms. Mounting evidences from numerous studies in the last decade have unraveled that deregulated miRNAs, and their cognate target expressions are strongly implicated in the carcinogenesis. Recent advances have highlighted miRNA-mediated regulation of mucins that have critical role in inflammation and cancer biology. The aberrant expression and differential glycosylation of mucins cause tumorigenesis, metastasis, chemoresistance and poor outcome of cancer patients, thus recognizing them as attractive therapeutic targets. Though current mucin-based therapies (antibodies, vaccines, immunotherapy, peptide inhibitors) have shown preclinical utility but only a handful promise for clinical transferability. In this context, understanding miRNA-mediated modulation of multiple mucin(s) expression and function gives a new hope for future anti-tumor therapeutics. Herein, we reviewed miRNA biogenesis, mechanism of action, and their role in tumor development. Further, we provide an overview of miRNA-mediated mucin regulation and their application as anticancer therapeutics.
Collapse
Affiliation(s)
- Shiv Ram Krishn
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA. Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Sukhwinder Kaur
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
548
|
Harms PW, Fullen DR, Patel RM, Chang D, Shalin SC, Ma L, Wood B, Beer TW, Siddiqui J, Carskadon S, Wang M, Palanisamy N, Fisher GJ, Andea A. Cutaneous basal cell carcinosarcomas: evidence of clonality and recurrent chromosomal losses. Hum Pathol 2015; 46:690-7. [PMID: 25704628 DOI: 10.1016/j.humpath.2015.01.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 01/03/2015] [Indexed: 10/24/2022]
Abstract
Cutaneous carcinosarcomas are heterogeneous group of tumors composed of malignant epithelial and mesenchymal components. Although mutation analyses have identified clonal changes between these morphologically disparate components in some subtypes of cutaneous carcinosarcoma, few cases have been analyzed thus far. To our knowledge, copy number variations (CNVs) and copy-neutral loss of heterozygosity (CN-LOH) have not been investigated in cutaneous carcinosarcomas. We analyzed 4 carcinosarcomas with basal cell carcinoma and osteosarcomatous components for CNVs/CN-LOH by comparative genomic hybridization/single-nucleotide polymorphism array, TP53 hot spot mutations by polymerase chain reaction and Sanger sequencing, and TP53 genomic rearrangements by fluorescence in situ hybridization. All tumors displayed multiple CNV/CN-LOH events (median, 7.5 per tumor). Three of 4 tumors displayed similar CNV/CN-LOH patterns between the epithelial and mesenchymal components within each tumor, supporting a common clonal origin. Recurrent changes included allelic loss at 9p21 (CDKN2A), 9q (PTCH1), and 17p (TP53). Allelic losses of chromosome 16 including CDH1 (E-cadherin) were present in 2 tumors and were restricted to the sarcomatous component. TP53 mutation analysis revealed an R248L mutation in both epithelial and mesenchymal components of 1 tumor. No TP53 rearrangements were identified. Our findings indicate that basal cell carcinosarcomas harbor CNV/CN-LOH changes similar to conventional basal cell carcinoma, with additional changes including recurrent 9p21 losses and a relatively high burden of copy number changes. In addition, most cutaneous carcinosarcomas show evidence of clonality between epithelial and mesenchymal components.
Collapse
Affiliation(s)
- Paul W Harms
- Department of Pathology, University of Michigan Health System, Ann Arbor MI 48109; Department of Dermatology, University of Michigan Health System, Ann Arbor MI 48109; Michigan Center for Translational Pathology, University of Michigan Health System, Ann Arbor MI 48109.
| | - Douglas R Fullen
- Department of Pathology, University of Michigan Health System, Ann Arbor MI 48109; Department of Dermatology, University of Michigan Health System, Ann Arbor MI 48109
| | - Rajiv M Patel
- Department of Pathology, University of Michigan Health System, Ann Arbor MI 48109; Department of Dermatology, University of Michigan Health System, Ann Arbor MI 48109
| | - Dannie Chang
- Department of Dermatology, University of Michigan Health System, Ann Arbor MI 48109
| | - Sara C Shalin
- Departments of Pathology and Dermatology, University of Arkansas for Medical Sciences, Little Rock, AR 72205
| | - Linglei Ma
- Miraca Life Sciences, Glen Burnie, MD 21061
| | - Benjamin Wood
- School of Pathology and Laboratory Medicine, University of Western Australia, Perth, WA 6009, Australia
| | - Trevor W Beer
- CliniPath Pathology, Osborne Park, WA 6017, Australia
| | - Javed Siddiqui
- Department of Pathology, University of Michigan Health System, Ann Arbor MI 48109; Michigan Center for Translational Pathology, University of Michigan Health System, Ann Arbor MI 48109
| | | | - Min Wang
- Department of Pathology, University of Michigan Health System, Ann Arbor MI 48109
| | - Nallasivam Palanisamy
- Department of Pathology, University of Michigan Health System, Ann Arbor MI 48109; Michigan Center for Translational Pathology, University of Michigan Health System, Ann Arbor MI 48109; Department of Urology, Henry Ford Health System, Detroit, MI 48202; King Saud University, Riyadh, Saudi Arabia 11362
| | - Gary J Fisher
- Department of Dermatology, University of Michigan Health System, Ann Arbor MI 48109
| | - Aleodor Andea
- Department of Pathology, University of Michigan Health System, Ann Arbor MI 48109; Department of Dermatology, University of Michigan Health System, Ann Arbor MI 48109
| |
Collapse
|
549
|
Macha MA, Krishn SR, Jahan R, Banerjee K, Batra SK, Jain M. Emerging potential of natural products for targeting mucins for therapy against inflammation and cancer. Cancer Treat Rev 2015; 41:277-88. [PMID: 25624117 DOI: 10.1016/j.ctrv.2015.01.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 12/31/2014] [Accepted: 01/07/2015] [Indexed: 02/07/2023]
Abstract
Deregulated mucin expression is a hallmark of several inflammatory and malignant pathologies. Emerging evidence suggests that, apart from biomarkers, these deregulated mucins are functional contributors to the pathogenesis in inflammation and cancer. Both overexpression and downregulation of mucins in various organ systems is associated with pathobiology of inflammation and cancer. Restoration of mucin homeostasis has become an important goal for therapy and management of such disorders has fueled the quest for selective mucomodulators. With improved understanding of mucin regulation and mechanistic insights into their pathobiological roles, there is optimism to find selective non-toxic agents capable of modulating mucin expression and function. Recently, natural compounds derived from dietary sources have drawn attention due to their anti-inflammatory and anti-oxidant properties and low toxicity. Considerable efforts have been directed towards evaluating dietary natural products as chemopreventive and therapeutic agents; identification, characterization and synthesis of their active compounds; and improving their delivery and bioavailability. We describe the current understanding of mucin regulation, rationale for targeting mucins with natural products and discuss some natural products that modulate mucin expression and functions. We further discuss the approaches and parameters that should guide future research to identify and evaluate selective natural mucomodulators for therapy.
Collapse
Affiliation(s)
- Muzafar A Macha
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | - Shiv Ram Krishn
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Rahat Jahan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Kasturi Banerjee
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA; Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Maneesh Jain
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA; Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
550
|
Acres B, Lacoste G, Limacher JM. Targeted Immunotherapy Designed to Treat MUC1-Expressing Solid Tumour. Curr Top Microbiol Immunol 2015; 405:79-97. [PMID: 25702159 DOI: 10.1007/82_2015_429] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Several approaches to antigen-specific immunotherapy of cancer antigen-specific immunotherapy of cancer have been tested clinically. In this chapter, we will describe studies done with the antigen MUC1. Tested MUC1 therapeutic vaccines include the following: monoclonal antibodies (MAbs) specific for MUC1; synthetic and recombinant polypeptides from the protein sequence of MUC1; dendritic cells carrying MUC1; RNA and DNA vaccinations; and recombinant viruses carrying the MUC1 DNA sequence. Chemotherapy of cancer aims to be toxic to the cancer cells with manageable side effects to the patient. In contrast, antigen-specific immunotherapy of cancer aims to treat the patient, such that the patient is then able to control and eventually eliminate their cancer cells. It is therefore important to know the immune status of each cancer patient prior to therapy.
Collapse
Affiliation(s)
| | - Gisele Lacoste
- Department of Medical Affairs, Transgene SA, 400 Blvd Gonthier d'Andernach, Parc d'Innovation CS80166, 67405, Illkirch-Graffenstaden Cedex, France.
| | - Jean-Marc Limacher
- Department of Medical Affairs, Transgene SA, 400 Blvd Gonthier d'Andernach, Parc d'Innovation CS80166, 67405, Illkirch-Graffenstaden Cedex, France
| |
Collapse
|