1
|
Pregnancy-Specific Glycoprotein 9 Enhances Store-Operated Calcium Entry and Nitric Oxide Release in Human Umbilical Vein Endothelial Cells. Diagnostics (Basel) 2023; 13:diagnostics13061134. [PMID: 36980442 PMCID: PMC10047280 DOI: 10.3390/diagnostics13061134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 03/10/2023] [Accepted: 03/11/2023] [Indexed: 03/19/2023] Open
Abstract
We explored changes in pregnancy-specific glycoprotein 9 (PSG9) levels in the serum of patients with preeclampsia and the effects and underlying mechanisms of PSG9 effects on calcium (Ca2+) homeostasis and nitric oxide (NO) release in human umbilical vein endothelial cells (HUVECs). Western blotting was used to detect protein expression levels, and an NO fluorescence probe was used to examine NO production. Intracellular Ca2+ concentrations were measured using a Ca2+-sensitive fluorescent dye under a fluorescence microscope. Compared with those in healthy pregnant women, serum PSG9 levels were significantly decreased in patients with preeclampsia. PSG9 (0.1 μg/mL) treatment of HUVECs significantly enhanced the expression levels of store-operated calcium entry (SOCE) channel proteins Orai1 and Orai2, but not Orai3, and of endothelial nitric oxide synthase (eNOS) and NO production. Pretreatment with an inhibitor of SOCE (BTP2) abolished PSG9-enhanced Orai1, Orai2, and eNOS expression levels and NO production in HUVECs. The mechanisms underlying SOCE that were PSG9 enhanced in HUVECs appear to involve the Ca2+/eNOS/NO signaling pathway. These findings suggest that serum PSG9 levels may be a potential biomarker for monitoring the occurrence or development of preeclampsia in pregnancy and that PSG9 may be a potential therapeutic target for the treatment of preeclampsia.
Collapse
|
2
|
Kandel M, MacDonald TM, Walker SP, Cluver C, Bergman L, Myers J, Hastie R, Keenan E, Hannan NJ, Cannon P, Nguyen TV, Pritchard N, Tong S, Kaitu'u-Lino TJ. PSG7 and 9 (Pregnancy-Specific β-1 Glycoproteins 7 and 9): Novel Biomarkers for Preeclampsia. J Am Heart Assoc 2022; 11:e024536. [PMID: 35322669 PMCID: PMC9075453 DOI: 10.1161/jaha.121.024536] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Preeclampsia is pregnancy specific, involving significant maternal endothelial dysfunction. Predictive biomarkers are lacking. We evaluated the biomarker potential, expression, and function of PSG7 (pregnancy‐specific β‐1 glycoprotein 7) and PSG9 (pregnancy‐specific β‐1 glycoprotein 9) in preeclampsia. Methods and Results At 36 weeks gestation preceding term preeclampsia diagnosis, PSG7 and PSG9 (in Australian cohorts of n=918 and n=979, respectively) were significantly increased before the onset of term preeclampsia (PSG7, P=0.013; PSG9, P=0.0011). In samples collected at 28 to 32 weeks from those with preexisting cardiovascular disease and at high risk of preeclampsia (Manchester Antenatal Vascular Service, UK cohort, n=235), both PSG7 and PSG9 were also significantly increased preceding preeclampsia onset (PSG7, P<0.0001; PSG9, P=0.0003) relative to controls. These changes were validated in the plasma and placentas of patients with established preeclampsia who delivered at <34 weeks gestation (PSG7, P=0.0008; PSG9, P<0.0001). To examine whether PSG7 and PSG9 are associated with increasing disease severity, we measured them in a cohort from South Africa stratified for this outcome, the PROVE (Preeclampsia Obstetric Adverse Events) cohort (n=72). PSG7 (P=0.0027) and PSG9 (P=0.0028) were elevated among patients who were preeclamptic with severe features (PROVE cohort), but not significantly changed in those without severe features or with eclampsia. In syncytialized first trimester cytotrophoblast stem cells, exposure to TNFα (tumor necrosis factor α) or IL‐6 (interleukin 6) significantly increased the expression and secretion of PSG7 and PSG9. In contrast, when we treated primary endothelial cells with recombinant PSG7 and PSG9, we only observed modest changes in Flt‐1 (FMS‐like tyrosine kinase‐1) expression and Plgf (placental growth factor) expression, and no other effects on proangiogenic/antiangiogenic or endothelial dysfunction markers were observed. Conclusions Circulating PSG7 and PSG9 are increased before preeclampsia onset and among those with established disease with their production and release potentially driven by placental inflammation.
Collapse
Affiliation(s)
- Manju Kandel
- Translational Obstetrics Group Mercy Hospital for Women Heidelberg Victoria Australia.,Department of Obstetrics and Gynaecology Mercy Hospital for Women University of Melbourne Heidelberg Victoria Australia
| | - Teresa M MacDonald
- Department of Obstetrics and Gynaecology Mercy Hospital for Women University of Melbourne Heidelberg Victoria Australia.,Mercy PerinatalMercy Hospital for Women Victoria Australia
| | - Susan P Walker
- Department of Obstetrics and Gynaecology Mercy Hospital for Women University of Melbourne Heidelberg Victoria Australia.,Mercy PerinatalMercy Hospital for Women Victoria Australia
| | - Catherine Cluver
- Translational Obstetrics Group Mercy Hospital for Women Heidelberg Victoria Australia.,Mercy PerinatalMercy Hospital for Women Victoria Australia.,Department of Obstetrics and Gynecology Stellenbosch University Cape Town South Africa
| | - Lina Bergman
- Department of Obstetrics and Gynecology Stellenbosch University Cape Town South Africa.,Department of Women's and Children's Health Uppsala University Uppsala Sweden.,Department of Obstetrics and Gynecology Institute of clinical sciencesSahlgrenska AcademyUniversity of Gothenburg Gothenburg Sweden
| | - Jenny Myers
- Division of Developmental Biology and Medicine University of ManchesterManchester Academic Health Science CentreSt Mary's Hospital Manchester United Kingdom
| | - Roxanne Hastie
- Translational Obstetrics Group Mercy Hospital for Women Heidelberg Victoria Australia.,Department of Obstetrics and Gynaecology Mercy Hospital for Women University of Melbourne Heidelberg Victoria Australia
| | - Emerson Keenan
- Department of Obstetrics and Gynaecology Mercy Hospital for Women University of Melbourne Heidelberg Victoria Australia
| | - Natalie J Hannan
- Translational Obstetrics Group Mercy Hospital for Women Heidelberg Victoria Australia.,Department of Obstetrics and Gynaecology Mercy Hospital for Women University of Melbourne Heidelberg Victoria Australia.,Mercy PerinatalMercy Hospital for Women Victoria Australia
| | - Ping Cannon
- Translational Obstetrics Group Mercy Hospital for Women Heidelberg Victoria Australia.,Department of Obstetrics and Gynaecology Mercy Hospital for Women University of Melbourne Heidelberg Victoria Australia
| | - Tuong-Vi Nguyen
- Translational Obstetrics Group Mercy Hospital for Women Heidelberg Victoria Australia.,Department of Obstetrics and Gynaecology Mercy Hospital for Women University of Melbourne Heidelberg Victoria Australia
| | - Natasha Pritchard
- Translational Obstetrics Group Mercy Hospital for Women Heidelberg Victoria Australia.,Department of Obstetrics and Gynaecology Mercy Hospital for Women University of Melbourne Heidelberg Victoria Australia.,Mercy PerinatalMercy Hospital for Women Victoria Australia
| | - Stephen Tong
- Translational Obstetrics Group Mercy Hospital for Women Heidelberg Victoria Australia.,Department of Obstetrics and Gynaecology Mercy Hospital for Women University of Melbourne Heidelberg Victoria Australia.,Mercy PerinatalMercy Hospital for Women Victoria Australia
| | - Tu'uhevaha J Kaitu'u-Lino
- Translational Obstetrics Group Mercy Hospital for Women Heidelberg Victoria Australia.,Department of Obstetrics and Gynaecology Mercy Hospital for Women University of Melbourne Heidelberg Victoria Australia.,Mercy PerinatalMercy Hospital for Women Victoria Australia
| |
Collapse
|
3
|
McNulty SN, Schwetye KE, Ferguson C, Storer CE, Ansstas G, Kim AH, Gutmann DH, Rubin JB, Head RD, Dahiya S. BRAF mutations may identify a clinically distinct subset of glioblastoma. Sci Rep 2021; 11:19999. [PMID: 34625582 PMCID: PMC8501013 DOI: 10.1038/s41598-021-99278-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 09/09/2021] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma (GBM) is the most common primary malignant brain tumor in adults. Prior studies examining the mutational landscape of GBM revealed recurrent alterations in genes that regulate the same growth control pathways. To this regard, ~ 40% of GBM harbor EGFR alterations, whereas BRAF variants are rare. Existing data suggests that gain-of-function mutations in these genes are mutually exclusive. This study was designed to explore the clinical, pathological, and molecular differences between EGFR- and BRAF-mutated GBM. We reviewed retrospective clinical data from 89 GBM patients referred for molecular testing between November 2012 and December 2015. Differences in tumor mutational profile, location, histology, and survival outcomes were compared in patients with EGFR- versus BRAF-mutated tumors, and microarray data from The Cancer Genome Atlas was used to assess differential gene expression between the groups. Individuals with BRAF-mutant tumors were typically younger and survived longer relative to those with EGFR-mutant tumors, even in the absence of targeted treatments. BRAF-mutant tumors lacked distinct histomorphology but exhibited unique localization in the brain, typically arising adjacent to the lateral ventricles. Compared to EGFR- and IDH1-mutant tumors, BRAF-mutant tumors showed increased expression of genes related to a trophoblast-like phenotype, specifically HLA-G and pregnancy specific glycoproteins, that have been implicated in invasion and immune evasion. Taken together, these observations suggest a distinct clinical presentation, brain location, and gene expression profile for BRAF-mutant tumors. Pending further study, this may prove useful in the stratification and management of GBM.
Collapse
Affiliation(s)
- Samantha N McNulty
- Department of Pathology and Immunology, Washington University School of Medicine, 660 South Euclid Ave, St. Louis, MO, 63110, USA
| | - Katherine E Schwetye
- Department of Pathology and Immunology, Washington University School of Medicine, 660 South Euclid Ave, St. Louis, MO, 63110, USA
| | - Cole Ferguson
- Department of Pathology and Immunology, Washington University School of Medicine, 660 South Euclid Ave, St. Louis, MO, 63110, USA
| | - Chad E Storer
- Department of Genetics, Washington University School of Medicine, 660 South Euclid Ave, St. Louis, MO, 63110, USA
| | - George Ansstas
- Division of Medical Oncology, Washington University School of Medicine, St. Louis, MO, USA.,Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Albert H Kim
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA.,Department of Neurosurgery, Washington University School of Medicine, St. Louis, MO, USA
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Joshua B Rubin
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA.,Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA.,Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, USA
| | - Richard D Head
- Department of Genetics, Washington University School of Medicine, 660 South Euclid Ave, St. Louis, MO, 63110, USA.
| | - Sonika Dahiya
- Department of Pathology and Immunology, Washington University School of Medicine, 660 South Euclid Ave, St. Louis, MO, 63110, USA. .,Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
4
|
Hao J, Dai X, Gao J, Li Y, Hou Z, Chang Z, Wang Y. Curcumin suppresses colorectal tumorigenesis via the Wnt/β-catenin signaling pathway by downregulating Axin2. Oncol Lett 2021; 21:186. [PMID: 33574925 PMCID: PMC7816292 DOI: 10.3892/ol.2021.12447] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 11/20/2020] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer worldwide, with high incidence and mortality rates. Conventional therapies, including surgery, chemotherapy and radiation, are extensively used for the treatment of CRC. However, patients present with adverse effects, such as toxicity, hepatic injury and drug resistance. Thus, there is an urgent requirement to identify effective and safe therapy for CRC. Curcumin (CUR), a polyphenol substrate extracted from the rhizome of Curcuma longa, has been extensively studied for the treatment of CRC due to its high efficacy and fewer side effects. Previous studies have reported that several signaling pathways, such as NF-κB, Wnt/β-catenin, are involved in the antitumor effects of CUR in vitro. However, the effect and mechanisms in vivo are not yet fully understood. The present study aimed to determine the molecular mechanism of colorectal cancer in vivo. Reverse transcription-quantitative PCR, western blot and immunohistochemistry analyses were performed to determine the underlying molecular mechanism of curcumin's anti-cancer effect in azoxymethane-dextran sodium sulfate induced colorectal cancer. The results of the present study demonstrated that CUR suppressed tumorigenesis in AOM-DSS induced CRC in mice, and anticancer effects were exerted by suppressing the expression of pro-inflammatory cytokines, and downregulating Axin2 in the Wnt/β-catenin signaling pathway. Taken together, these results exhibit the potential in vivo mechanisms of the anticancer effects of CUR, and highlight Axin2 as a potential therapeutic target for CRC.
Collapse
Affiliation(s)
- Jiaxue Hao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education/College of Life Science, Northwest University, Xi'an, Shaanxi 710069, P.R. China
| | - Xufen Dai
- Food and Drug Technology Research Center, Shaanxi Province Food and Drug Supervision and Inspection Research, Shaanxi Institute for Food and Drug Control, Xi'an, Shaanxi 710065, P.R. China
| | - Juan Gao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education/College of Life Science, Northwest University, Xi'an, Shaanxi 710069, P.R. China
| | - Yuexuan Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education/College of Life Science, Northwest University, Xi'an, Shaanxi 710069, P.R. China
| | - Zhaoling Hou
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education/College of Life Science, Northwest University, Xi'an, Shaanxi 710069, P.R. China
| | - Zhongman Chang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education/College of Life Science, Northwest University, Xi'an, Shaanxi 710069, P.R. China
| | - Yuxin Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education/College of Life Science, Northwest University, Xi'an, Shaanxi 710069, P.R. China
| |
Collapse
|
5
|
Liu Y, Zhang S, Yu T, Zhang F, Yang F, Huang Y, Ma D, Liu G, Shao Z, Li D. Pregnancy-specific glycoprotein 9 acts as both a transcriptional target and a regulator of the canonical TGF-β/Smad signaling to drive breast cancer progression. Clin Transl Med 2020; 10:e245. [PMID: 33377651 PMCID: PMC7733318 DOI: 10.1002/ctm2.245] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 11/16/2020] [Accepted: 11/27/2020] [Indexed: 12/24/2022] Open
Abstract
Pregnancy-specific glycoprotein 9 (PSG9) is a placental glycoprotein essential for the maintenance of normal gestation in mammals. Bioinformatics analysis of multiple publicly available datasets revealed aberrant PSG9 expression in breast tumors, but its functional and mechanistic role in breast cancer remains unexplored. Here, we report that PSG9 expression levels were elevated in tumor tissues and plasma specimens from breast cancer patients, and were associated with poor prognosis. Gain- or loss-of-function studies demonstrated that PSG9 promoted breast cancer cell proliferation, migration, and invasionin vitro, and enhanced tumor growth and lung colonization in vivo. Mechanistically, transforming growth factor-β1 (TGF-β1) transcriptionally activated PSG9 expression through enhancing the enrichment of Smad3 and Smad4 onto PSG9 promoter regions containing two putative Smad-binding elements (SBEs). Mutation of both SBEs in the PSG9 promoter, or knockdown of TGF-β receptor 1 (TGFBR1), TGFBR2, Smad3, or Smad4 impaired the ability of TGF-β1 to induce PSG9 expression. Consequently, PSG9 contributed to TGF-β1-induced epithelial-mesenchymal transition (EMT) and breast cancer cell migration and invasion. Moreover, PSG9 enhanced the stability of Smad2, Smad3, and Smad4 proteins by blocking their proteasomal degradation, and regulated the expression of TGF-β1 target genes involved in EMT and breast cancer progression, thus further amplifying the canonical TGF-β/Smad signaling in breast cancer cells. Collectively, these findings establish PSG9 as a novel player in breast cancer progressionvia hijacking the canonical TGF-β/Smad signaling, and identify PSG9 as a potential plasma biomarker for the early detection of breast cancer.
Collapse
Affiliation(s)
- Ying‐Ying Liu
- Fudan University Shanghai Cancer Center and Shanghai Key Laboratory of Medical EpigeneticsInternational Co‐laboratory of Medical Epigenetics and MetabolismMinistry of Science and TechnologyInstitutes of Biomedical SciencesFudan UniversityShanghaiChina
- Cancer InstituteShanghai Medical College, Fudan UniversityShanghaiChina
- Department of OncologyShanghai Medical College, Fudan UniversityShanghaiChina
- Department of Breast SurgeryShanghai Medical College, Fudan UniversityShanghaiChina
| | - Sa Zhang
- Fudan University Shanghai Cancer Center and Shanghai Key Laboratory of Medical EpigeneticsInternational Co‐laboratory of Medical Epigenetics and MetabolismMinistry of Science and TechnologyInstitutes of Biomedical SciencesFudan UniversityShanghaiChina
| | - Tian‐Jian Yu
- Fudan University Shanghai Cancer Center and Shanghai Key Laboratory of Medical EpigeneticsInternational Co‐laboratory of Medical Epigenetics and MetabolismMinistry of Science and TechnologyInstitutes of Biomedical SciencesFudan UniversityShanghaiChina
- Cancer InstituteShanghai Medical College, Fudan UniversityShanghaiChina
- Department of OncologyShanghai Medical College, Fudan UniversityShanghaiChina
- Department of Breast SurgeryShanghai Medical College, Fudan UniversityShanghaiChina
| | - Fang‐Lin Zhang
- Fudan University Shanghai Cancer Center and Shanghai Key Laboratory of Medical EpigeneticsInternational Co‐laboratory of Medical Epigenetics and MetabolismMinistry of Science and TechnologyInstitutes of Biomedical SciencesFudan UniversityShanghaiChina
- Cancer InstituteShanghai Medical College, Fudan UniversityShanghaiChina
- Department of OncologyShanghai Medical College, Fudan UniversityShanghaiChina
| | - Fan Yang
- Fudan University Shanghai Cancer Center and Shanghai Key Laboratory of Medical EpigeneticsInternational Co‐laboratory of Medical Epigenetics and MetabolismMinistry of Science and TechnologyInstitutes of Biomedical SciencesFudan UniversityShanghaiChina
- Cancer InstituteShanghai Medical College, Fudan UniversityShanghaiChina
- Department of OncologyShanghai Medical College, Fudan UniversityShanghaiChina
- Department of Breast SurgeryShanghai Medical College, Fudan UniversityShanghaiChina
| | - Yan‐Ni Huang
- Department of Breast SurgeryShanghai Medical College, Fudan UniversityShanghaiChina
| | - Ding Ma
- Department of Breast SurgeryShanghai Medical College, Fudan UniversityShanghaiChina
| | - Guang‐Yu Liu
- Department of OncologyShanghai Medical College, Fudan UniversityShanghaiChina
- Department of Breast SurgeryShanghai Medical College, Fudan UniversityShanghaiChina
| | - Zhi‐Ming Shao
- Fudan University Shanghai Cancer Center and Shanghai Key Laboratory of Medical EpigeneticsInternational Co‐laboratory of Medical Epigenetics and MetabolismMinistry of Science and TechnologyInstitutes of Biomedical SciencesFudan UniversityShanghaiChina
- Cancer InstituteShanghai Medical College, Fudan UniversityShanghaiChina
- Department of OncologyShanghai Medical College, Fudan UniversityShanghaiChina
- Department of Breast SurgeryShanghai Medical College, Fudan UniversityShanghaiChina
- Shanghai Key Laboratory of Breast CancerShanghai Medical College, Fudan UniversityShanghaiChina
| | - Da‐Qiang Li
- Fudan University Shanghai Cancer Center and Shanghai Key Laboratory of Medical EpigeneticsInternational Co‐laboratory of Medical Epigenetics and MetabolismMinistry of Science and TechnologyInstitutes of Biomedical SciencesFudan UniversityShanghaiChina
- Cancer InstituteShanghai Medical College, Fudan UniversityShanghaiChina
- Department of OncologyShanghai Medical College, Fudan UniversityShanghaiChina
- Department of Breast SurgeryShanghai Medical College, Fudan UniversityShanghaiChina
- Shanghai Key Laboratory of Breast CancerShanghai Medical College, Fudan UniversityShanghaiChina
| |
Collapse
|
6
|
A Support Vector Machine Model Predicting the Risk of Duodenal Cancer in Patients with Familial Adenomatous Polyposis at the Transcript Levels. BIOMED RESEARCH INTERNATIONAL 2020; 2020:5807295. [PMID: 32626748 PMCID: PMC7315318 DOI: 10.1155/2020/5807295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 05/14/2020] [Accepted: 05/19/2020] [Indexed: 11/18/2022]
Abstract
Objective Familial adenomatous polyposis (FAP) is one major type of inherited duodenal cancer. The estimate of duodenal cancer risk in patients with FAP is critical for selecting the optimal treatment strategy. Methods Microarray datasets related with FAP were retrieved from the Gene Expression Omnibus (GEO) database. Differentially expressed genes were identified by FAP vs. normal samples and FAP and duodenal cancer vs. normal samples. Furthermore, functional enrichment analyses of these differentially expressed genes were performed. A support vector machine (SVM) was performed to train and validate cancer risk prediction model. Results A total of 196 differentially expressed genes were identified between FAP compared with normal samples. 177 similarly expressed genes were identified both in FAP and duodenal cancer, which were mainly enriched in pathways in cancer and metabolic-related pathway, indicating that these genes in patients with FAP could contribute to duodenal cancer. Among them, Cyclin D1, SDF-1, AXIN, and TCF were significantly upregulated in FAP tissues using qRT-PCR. Based on the 177 genes, an SVM model was constructed for prediction of the risk of cancer in patients with FAP. After validation, the model can accurately distinguish FAP patients with high risk from those with low risk for duodenal cancer. Conclusion This study proposed a cancer risk prediction model based on an SVM at the transcript levels.
Collapse
|
7
|
Expression of Pregnancy Specific β-1 Glycoprotein 1 in Cervical Cancer Cells. Arch Med Res 2020; 51:504-514. [PMID: 32546445 DOI: 10.1016/j.arcmed.2020.05.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 04/25/2020] [Accepted: 05/07/2020] [Indexed: 11/24/2022]
Abstract
BACKGROUND Cervical Cancer (CC) is a worldwide public health concern associated with genetic alterations, among these the gain of the 19q chromosome harboring the Pregnancy Specific Glycoproteins (PSG) gene family. These proteins play a critical role in pregnancy, with participation in immunotolerance, angiogenesis, and invasion processes, which are also observed in carcinogenesis. The aim of this study was to determine the molecular alterations of PSG1 and its relationship with CC. METHODS PSG1 Copy Number Variation (CNV) was evaluated in 31 CC and eight normal cervical tissues by qPCR. PSG1 expression was correlated with HPV detection and IL-10 and TGF-β expression in CC samples. Finally, PSG1 protein expression was evaluated by immunofluorescence in CC cell lines, by immunohistochemistry in a tissue microarray, and by immunoblotting in the sera of women with normal cervix, pre-invasive lesions, and CC. RESULTS PSG1 showed a gain of 25.6% in CNV and gene expression in CC. There was a lack of PSG1 expression in normal cervical epithelium and positive immunostaining in 57% of CC tissues, while all CC cell lines expressed PSG1. Finally, PSG1 was immunodetected in 90% of pre-invasive lesions and in all CC serum samples, but not in healthy women. PSG1 expression correlates with the expression of IL-10 and TGF-β in CC tissues, but not with the presence of HPV. CONCLUSION These data show evidence of the differential expression of PSG1 in CC that could explain its participation in tumor-biology and immunotolerance mechanisms. Further, its immunodetection could provide early detection of this cancer.
Collapse
|
8
|
Interaction of Pregnancy-Specific Glycoprotein 1 With Integrin Α5β1 Is a Modulator of Extravillous Trophoblast Functions. Cells 2019; 8:cells8111369. [PMID: 31683744 PMCID: PMC6912793 DOI: 10.3390/cells8111369] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 10/23/2019] [Accepted: 10/25/2019] [Indexed: 01/23/2023] Open
Abstract
Human pregnancy-specific glycoproteins (PSGs) serve immunomodulatory and pro-angiogenic functions during pregnancy and are mainly expressed by syncytiotrophoblast cells. While PSG mRNA expression in extravillous trophoblasts (EVTs) was reported, the proteins were not previously detected. By immunohistochemistry and immunoblotting, we show that PSGs are expressed by invasive EVTs and co-localize with integrin 5. In addition, we determined that native and recombinant PSG1, the most highly expressed member of the family, binds to 51 and induces the formation of focal adhesion structures resulting in adhesion of primary EVTs and EVT-like cell lines under 21% oxygen and 1% oxygen conditions. Furthermore, we found that PSG1 can simultaneously bind to heparan sulfate in the extracellular matrix and to 51 on the cell membrane. Wound healing assays and single-cell movement tracking showed that immobilized PSG1 enhances EVT migration. Although PSG1 did not affect EVT invasion in the in vitro assays employed, we found that the serum PSG1 concentration is lower in African-American women diagnosed with early-onset and late-onset preeclampsia, a pregnancy pathology characterized by shallow trophoblast invasion, than in their respective healthy controls only when the fetus was a male; therefore, the reduced expression of this molecule should be considered in the context of preeclampsia as a potential therapy.
Collapse
|
9
|
Vanichtantikul A, Hodge KG, Somparn P, Saethang T, Triratanachat S, Pisitkun T, Lertkhachonsuk R. Proteomic identification of predictive biomarkers for malignant transformation in complete hydatidiform moles. Placenta 2019; 77:58-64. [PMID: 30827356 DOI: 10.1016/j.placenta.2019.02.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Revised: 12/11/2018] [Accepted: 02/04/2019] [Indexed: 10/27/2022]
Abstract
INTRODUCTION Protein expression in cells are associated with oncogenesis. This study aims to explore proteomic profiles and discover potential biomarkers that can predict malignant transformation of hydatidiform mole. METHODS Retrospective analysis was done in 14 cases of remission hydatidiform mole and 14 cases of hydatidiform mole who later developed malignancy (GTN group). Molar tissues were retrieved from -70 °C frozen tissue. Subsequently, a large-scale proteomic analysis was performed to identify proteins and compare their abundance levels in the preserved molar tissues from these two groups using a dimethyl-labeling technique coupled with liquid chromatography-tandem mass spectrometry (LC-MS/MS). RESULTS A total of 2,153 proteins were identified from all samples. 22 and 10 proteins were significantly up-regulated and down-regulated, respectively, in the GTN group compared with the mole group. These altered proteins were found in several biological groups such as cell-cell adhesion, secreted proteins, and ribonucleoproteins. Several hormone-related proteins were among the most up-regulated proteins in the GTN group including choriogonadotropin subunit beta (β-hCG) and alpha (α-hCG), growth/differentiation factor 15, as well as both pregnancy-specific beta-1-glycoproteins 2 and 3. In contrast, protein S100-A11 and l-lactate dehydrogenase A chain, were down-regulated in molar tissue from most patients in the GTN group. DISCUSSION This study identified a set of differentially expressed proteins in molar tissues that could potentially be further examined as predictive biomarkers for the malignant transformation of CHMs. A molar proteome database was constructed and can be accessible online at http://sysbio.chula.ac.th/Database/GTD_DB/Supplementary_Data.xlsx.
Collapse
Affiliation(s)
- Asama Vanichtantikul
- Placental Related Disease Research Unit, Department of Obstetrics and Gynecology, Faculty of Medicine, Chulalongkorn University, Thailand
| | - Kenneth G Hodge
- Systems Biology Center, Faculty of Medicine, Chulalongkorn University, Thailand
| | - Poorichaya Somparn
- Systems Biology Center, Faculty of Medicine, Chulalongkorn University, Thailand
| | - Thammakorn Saethang
- Systems Biology Center, Faculty of Medicine, Chulalongkorn University, Thailand
| | - Surang Triratanachat
- Division of Gynecologic Cytopathology, Department of Obstetrics and Gynecology, Faculty of Medicine, Chulalongkorn University, Thailand
| | - Trairak Pisitkun
- Systems Biology Center, Faculty of Medicine, Chulalongkorn University, Thailand
| | - Ruangsak Lertkhachonsuk
- Placental Related Disease Research Unit, Department of Obstetrics and Gynecology, Faculty of Medicine, Chulalongkorn University, Thailand.
| |
Collapse
|
10
|
hnRNPDL extensively regulates transcription and alternative splicing. Gene 2018; 687:125-134. [PMID: 30447347 DOI: 10.1016/j.gene.2018.11.026] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 08/30/2018] [Accepted: 11/08/2018] [Indexed: 01/31/2023]
Abstract
RNA binding proteins (RBPs) are key players of genome regulation. Here we report the transcriptome study of HnRNP D-Like protein, which belongs to the hnRNP family. We used RNA-seq to analyze the global transcript level and alternative splicing on hnRNPDL shRNA-treated cells and control. Sh-hnRNPDL extensively increased in the expression of genes involved in female pregnancy, cell apoptosis, cell proliferation and cell migration. HnRNPDL regulated alternative splicing of hundreds of genes enriched in transcription regulation and signaling pathways including NOD-like receptor signaling, Notch signaling, and TNF signaling. This study provides the first transcriptome-wide analysis of hnRNPDL regulation of gene expression, which adds to the understanding of critical hnRNPDL functions.
Collapse
|
11
|
Post-surgical resection prognostic value of combined OPN, MMP7, and PSG9 plasma biomarkers in hepatocellular carcinoma. Front Med 2018; 13:250-258. [PMID: 29770948 DOI: 10.1007/s11684-018-0632-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 12/18/2017] [Indexed: 12/11/2022]
Abstract
Biomarkers for hepatocellular carcinoma (HCC) following curative resection are not currently sufficient for prognostic indication of overall survival (OS) and disease-free survival (DFS). The aim of this study was to investigate the prognostic performance of osteopontin (OPN), matrix metalloproteinase 7 (MMP7), and pregnancy specific glycoprotein 9 (PSG9) in patients with HCC. A total of 179 prospective patients with HCC provided plasma before hepatectomy. Plasma OPN, MMP7, and PSG9 levels were determined by enzyme-linked immunosorbent assay. Correlations between plasma levels, clinical parameters, and outcomes (OS and DFS) were overall analyzed. High OPN ( ⩾ 149.97 ng/mL), MMP7 ( ⩾ 2.28 ng/mL), and PSG9 ( ⩾ 45.59 ng/mL) were prognostic indicators of reduced OS (P < 0.001, P < 0.001, and P = 0.007, respectively). Plasma PSG9 protein level was an independent factor in predicting OS (P = 0.008) and DFS (P = 0.038). Plasma OPN + MMP7 + PSG9 elevation in combination was a prognostic factor for OS (P < 0.001). OPN was demonstrated to be a risk factorassociated OS in stage I patients with HCC and patients with low α-fetoprotein levels ( < 20 ng/mL). These findings suggested that OPN, MMP7, PSG9 and their combined panels may be useful for aiding in tumor recurrence and mortality risk prediction of patients with HCC, particularly in the early stage of HCC carcinogenesis.
Collapse
|
12
|
Yang L, Hu S, Tan J, Zhang X, Yuan W, Wang Q, Xu L, Liu J, Liu Z, Jia Y, Huang X. Pregnancy-specific glycoprotein 9 (PSG9), a driver for colorectal cancer, enhances angiogenesis via activation of SMAD4. Oncotarget 2018; 7:61562-61574. [PMID: 27528036 PMCID: PMC5308672 DOI: 10.18632/oncotarget.11146] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 07/27/2016] [Indexed: 12/12/2022] Open
Abstract
PSG9 is a member of the pregnancy-specific glycoprotein (PSG) family and has been shown to contribute to the progression of colorectal cancer (CRC) and cancer-related angiogenesis. Here, we aim to investigate abnormal PSG9 levels in patients with CRC and to emphasize the role of PSG9 in driving tumorigenesis. Serum from 140 patients with CRC and 125 healthy controls as well as 74 paired tumors and adjacent normal tissue were used to determine PSG9 levels. We discovered that PSG9 was significantly increased in serum (P<0.001) and in tumor tissues (P<0.001) from patients with CRC. Interestingly, the increased PSG9 levels correlated with poor survival (P=0.009) and microvessel density (MVD) (P=0.034). The overexpression of PSG9 strongly promoted the proliferation and migration of HCT-116 and HT-29 cells. However, PSG9 depletion inhibited the proliferation of SW-480 cells. Using a human umbilical vein endothelial cell tube-forming assay, we found that PSG9 promoted angiogenesis. The overexpression of PSG9 also increased the growth of tumor xenografts in nude mice. Co-immunoprecipitation experiments revealed that PSG9 was bound to SMAD4. The PSG9/SMAD4 complex recruited cytoplasmic SMAD2/3 to form a complex, which enhanced SMAD4 nuclear retention. The PSG9 and SMAD4 complex activated the expression of multiple angiogenesis-related genes (included IGFBP-3, PDGF-AA, GM-CSF, and VEGFA). Together, our findings illustrate the innovative mechanism by which PSG9 drives the progression of CRC and tumor angiogenesis. This occurs via nuclear translocation of PSG9/SMAD4, which activates angiogenic cytokines. Therefore, our study may provide evidence for novel treatment strategies by targeting PSG9 in antiangiogenic cancer therapy.
Collapse
Affiliation(s)
- Lei Yang
- Medical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing, P.R. China
| | - Shusheng Hu
- Clinical Laboratory Department, Tianjin Medical University Cancer Institute and Hospital, Tianjin, P.R. China
| | - Jinjing Tan
- Department of Cellular and Molecular Biology, Beijing Chest Hospital, Capital Medical University, Beijing, P.R. China
| | - Xiaojing Zhang
- Oncology Department, Beijing Chaoyang Hospital, Capital Medical University, Beijing, P.R. China
| | - Wen Yuan
- Medical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing, P.R. China
| | - Qian Wang
- Oncology Department, Beijing Chaoyang Hospital, Capital Medical University, Beijing, P.R. China
| | - Lingling Xu
- Oncology Department, Beijing Chaoyang Hospital, Capital Medical University, Beijing, P.R. China
| | - Jian Liu
- Medical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing, P.R. China
| | - Zheng Liu
- Medical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing, P.R. China
| | - Yanjun Jia
- Medical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing, P.R. China
| | - Xiaoxi Huang
- Medical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing, P.R. China
| |
Collapse
|
13
|
PSG9 promotes angiogenesis by stimulating VEGFA production and is associated with poor prognosis in hepatocellular carcinoma. SCIENCE CHINA-LIFE SCIENCES 2017; 60:528-535. [PMID: 28078509 DOI: 10.1007/s11427-016-0226-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 11/04/2016] [Indexed: 12/11/2022]
Abstract
Hepatocellular carcinoma (HCC) is a common malignant solid tumor characterized by rich vascularization. Pregnancy-specific glycoprotein 9 (PSG9) is a member of the carcinoembryonic antigen (CEA)/PSG family and is produced at high levels during pregnancy. We previously identified PSG9 as an HCC-related protein. However, the expression of PSG9 and its regulation during HCC carcinogenesis remain poorly explored. In the present study, we first found that the levels of PSG9 protein were significantly increased in the plasma of HCC patients. PSG9 overexpression also increased the proliferation ability of an HCC cell line. High expression of PSG9 was associated with angiogenesis by accelerating VEGFA expression. In addition, Cox's proportional hazards model analysis revealed that the plasma level of PSG9 was an independent prognostic factor for overall survival. We propose that PSG9 is a novel indicator of prognosis in patients with HCC and could serve as a novel therapeutic target for HCC. Furthermore, our results indicate that PSG9 protein may facilitate the development of HCC by fostering angiogenesis via promoting VEGFA production in cancer cells.
Collapse
|
14
|
Shahinian JH, Fuellgraf H, Tholen S, Mastroianni J, Knopf JD, Kuehs M, Mayer B, Schlimpert M, Kulemann B, Kuesters S, Hoeppner J, Wellner UF, Werner M, Hopt UT, Zeiser R, Bronsert P, Schilling O. Pregnancy Specific β-1 Glycoprotein 1 is Expressed in Pancreatic Ductal Adenocarcinoma and its Subcellular Localization Correlates with Overall Survival. J Cancer 2016; 7:2018-2027. [PMID: 27877217 PMCID: PMC5118665 DOI: 10.7150/jca.15864] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Accepted: 09/18/2016] [Indexed: 12/14/2022] Open
Abstract
Proteins of the pregnancy specific β-1 glycoprotein (PSG) family are renowned for their elevated expression during pregnancy. Only few reports have investigated their expression in adenocarcinomas. We studied the expression of PSG1 in pancreatic adenocarcinoma (PDAC). In a cohort of 104 patient samples, immunohistochemical analysis determined PSG1 expression in every specimen. PSG1 was found at apical and cytoplasmic localization or solely at cytoplasmic localization, with the latter case being correlated to shortened median survival (25 vs 11 months, logrank p-value < 0.001). At the same time, enzyme linked immunosorbent assay (ELISA) did not detect elevated PSG1 levels in the plasma of PDAC patients as opposed to the plasma of healthy, non-pregnant control individuals. We also probed the impact of PSG1 expression in a murine tumor model system, using subcutaneous injection of Colo-26 cells into immunocompetent BALB/c mice. Here, tumor growth was not affected by the expression of human PSG1. Our study reaffirms interest into the tumor-contextual biology of PSG proteins.
Collapse
Affiliation(s)
- Jasmin H Shahinian
- Division of Cardiac Surgery, University Hospital Basel, Basel, Switzerland
| | - Hannah Fuellgraf
- Institute of Surgical Pathology, University Medical Center Freiburg, Germany
| | - Stefan Tholen
- Institute of Molecular Medicine and Cell Research, University of Freiburg, Germany
| | - Justin Mastroianni
- Department of Hematology and Oncology, University Medical Center Freiburg, Germany
| | - Julia Daniela Knopf
- Institute of Molecular Medicine and Cell Research, University of Freiburg, Germany;; present address: Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), DKFZ-ZMBH Allianz, Im Neuenheimer Feld 282, 69120 Heidelberg, Germany
| | - Markus Kuehs
- Institute of Surgical Pathology, University Medical Center Freiburg, Germany;; Comprehensive Cancer Center Freiburg, Germany
| | - Bettina Mayer
- Institute of Molecular Medicine and Cell Research, University of Freiburg, Germany
| | - Manuel Schlimpert
- Institute of Molecular Medicine and Cell Research, University of Freiburg, Germany
| | - Birte Kulemann
- Clinic for General and Visceral Surgery, University Medical Center Freiburg, Germany
| | - Simon Kuesters
- Clinic for General and Visceral Surgery, University Medical Center Freiburg, Germany
| | - Jens Hoeppner
- Clinic for General and Visceral Surgery, University Medical Center Freiburg, Germany;; Comprehensive Cancer Center Freiburg, Germany
| | - Ulrich F Wellner
- Clinic for Surgery, University Clinic Schleswig-Holstein Campus Lübeck, Germany
| | - Martin Werner
- Institute of Surgical Pathology, University Medical Center Freiburg, Germany;; Comprehensive Cancer Center Freiburg, Germany;; German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, German
| | - Ulrich T Hopt
- Clinic for General and Visceral Surgery, University Medical Center Freiburg, Germany
| | - Robert Zeiser
- Department of Hematology and Oncology, University Medical Center Freiburg, Germany;; BIOSS Centre for Biological Signaling Studies, University of Freiburg, D-79104 Freiburg, Germany
| | - Peter Bronsert
- Institute of Surgical Pathology, University Medical Center Freiburg, Germany;; Comprehensive Cancer Center Freiburg, Germany;; German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, German
| | - Oliver Schilling
- Institute of Molecular Medicine and Cell Research, University of Freiburg, Germany;; BIOSS Centre for Biological Signaling Studies, University of Freiburg, D-79104 Freiburg, Germany;; German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, German
| |
Collapse
|
15
|
Houston A, Williams JM, Rovis TL, Shanley DK, O'Riordan RT, Kiely PA, Ball M, Barry OP, Kelly J, Fanning A, MacSharry J, Mandelboim O, Singer BB, Jonjic S, Moore T. Pregnancy-specific glycoprotein expression in normal gastrointestinal tract and in tumors detected with novel monoclonal antibodies. MAbs 2016; 8:491-500. [PMID: 26926266 DOI: 10.1080/19420862.2015.1134410] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Pregnancy-specific glycoproteins (PSGs) are immunoglobulin superfamily members related to the carcinoembryonic antigen-related cell adhesion molecule (CEACAM) family and are encoded by 10 genes in the human. They are secreted at high levels by placental syncytiotrophoblast into maternal blood during pregnancy, and are implicated in immunoregulation, thromboregulation, and angiogenesis. To determine whether PSGs are expressed in tumors, we characterized 16 novel monoclonal antibodies to human PSG1 and used 2 that do not cross-react with CEACAMs to study PSG expression in tumors and in the gastrointestinal (GI) tract using tissue arrays and immunohistochemistry. Staining was frequently observed in primary squamous cell carcinomas and colonic adenocarcinomas and was correlated with the degree of tumor differentiation, being largely absent from metastatic samples. Staining was also observed in normal oesophageal and colonic epithelium. PSG expression in the human and mouse GI tract was confirmed using quantitative RT-PCR. However, mRNA expression was several orders of magnitude lower in the GI tract compared to placenta. Our results identify a non-placental site of PSG expression in the gut and associated tumors, with implications for determining whether PSGs have a role in tumor progression, and utility as tumor biomarkers.
Collapse
Affiliation(s)
- Aileen Houston
- a School of Medicine , University College Cork , Ireland
| | - John M Williams
- b School of Biochemistry and Cell Biology, University College Cork , Ireland
| | - Tihana Lenac Rovis
- c Department of Histology and Embryology/Center for Proteomics , Faculty of Medicine, University of Rijeka , Croatia
| | - Daniel K Shanley
- b School of Biochemistry and Cell Biology, University College Cork , Ireland
| | - Ronan T O'Riordan
- b School of Biochemistry and Cell Biology, University College Cork , Ireland
| | - Patrick A Kiely
- d Department of Life Sciences , Materials and Surface Science Institute and Stokes Institute, University of Limerick
| | - Melanie Ball
- b School of Biochemistry and Cell Biology, University College Cork , Ireland
| | - Orla P Barry
- e Department of Pharmacology , Alimentary Pharmabiotic Center, University College Cork , Ireland
| | - Jacquie Kelly
- a School of Medicine , University College Cork , Ireland
| | - Aine Fanning
- f Alimentary Pharmabiotic Center, University College Cork , Ireland
| | - John MacSharry
- f Alimentary Pharmabiotic Center, University College Cork , Ireland
| | - Ofer Mandelboim
- g Lautenberg Center for General and Tumor Immunology, Institute for Medical Research Israel-Canada, Hebrew University-Hadassah Medical School , Jerusalem , Israel
| | - Bernhard B Singer
- h Institute of Anatomy, University Hospital, University Duisburg-Essen , Essen , Germany
| | - Stipan Jonjic
- c Department of Histology and Embryology/Center for Proteomics , Faculty of Medicine, University of Rijeka , Croatia
| | - Tom Moore
- b School of Biochemistry and Cell Biology, University College Cork , Ireland
| |
Collapse
|
16
|
Januar V, Desoye G, Novakovic B, Cvitic S, Saffery R. Epigenetic regulation of human placental function and pregnancy outcome: considerations for causal inference. Am J Obstet Gynecol 2015; 213:S182-96. [PMID: 26428498 DOI: 10.1016/j.ajog.2015.07.011] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 07/03/2015] [Accepted: 07/13/2015] [Indexed: 12/14/2022]
Abstract
Epigenetic mechanisms, often defined as regulating gene activity independently of underlying DNA sequence, are crucial for healthy development. The sum total of epigenetic marks within a cell or tissue (the epigenome) is sensitive to environmental influence, and disruption of the epigenome in utero has been associated with adverse pregnancy outcomes. Not surprisingly, given its multifaceted functions and important role in regulating pregnancy outcome, the placenta shows unique epigenetic features. Interestingly however, many of these are only otherwise seen in human malignancy (the pseudomalignant placental epigenome). Epigenetic variation in the placenta is now emerging as a candidate mediator of environmental influence on placental functioning and a key regulator of pregnancy outcome. However, replication of findings is generally lacking, most likely due to small sample sizes and a lack of standardization of analytical approaches. Defining DNA methylation "signatures" in the placenta associated with maternal and fetal outcomes offers tremendous potential to improve pregnancy outcomes, but care must be taken in interpretation of findings. Future placental epigenetic research would do well to address the issues present in epigenetic epidemiology more generally, including careful consideration of sample size, potentially confounding factors, issues of tissue heterogeneity, reverse causation, and the role of genetics in modulating epigenetic profile. The importance of animal or in vitro models in establishing a functional role of epigenetic variation identified in human beings, which is key to establishing causation, should not be underestimated.
Collapse
Affiliation(s)
- Vania Januar
- Cancer and Disease Epigenetics, Murdoch Childrens Research Institute, Royal Children's Hospital, Parkville, Australia
| | - Gernot Desoye
- Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria
| | - Boris Novakovic
- Cancer and Disease Epigenetics, Murdoch Childrens Research Institute, Royal Children's Hospital, Parkville, Australia
| | - Silvija Cvitic
- Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria
| | - Richard Saffery
- Cancer and Disease Epigenetics, Murdoch Childrens Research Institute, Royal Children's Hospital, Parkville, Australia; Department of Pediatrics, University of Melbourne, Parkville, Australia.
| |
Collapse
|
17
|
Yang L, Rong W, Xiao T, Zhang Y, Xu B, Liu Y, Wang L, Wu F, Qi J, Zhao X, Wang H, Han N, Guo S, Wu J, Gao Y, Cheng S. Secretory/releasing proteome-based identification of plasma biomarkers in HBV-associated hepatocellular carcinoma. SCIENCE CHINA-LIFE SCIENCES 2013; 56:638-46. [PMID: 23749381 DOI: 10.1007/s11427-013-4497-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Accepted: 05/17/2013] [Indexed: 12/11/2022]
Abstract
For successful therapy, hepatocellular carcinoma (HCC) must be detected at an early stage. Herein, we used a proteomic approach to analyze the secretory/releasing proteome of HCC tissues to identify plasma biomarkers. Serum-free conditioned media (CM) were collected from primary cultures of cancerous tissues and surrounding noncancerous tissues. Proteomic analysis of the CM proteins permitted the identification of 1365 proteins. The enriched molecular functions and biological processes of the CM proteins, such as hydrolase activity and catabolic processes, were consistent with the liver being the most important metabolic organ. Moreover, 19% of the proteins were characterized as extracellular or membrane-bound. For validation, secretory proteins involved in transforming growth factor-β signaling pathways were validated in plasma samples. Alphafetoprotein (AFP), metalloproteinase (MMP)1, osteopontin (OPN), and pregnancy-specific beta-1-glycoprotein (PSG)9 were significantly increased in HCC patients. The overall performance of MMP1 and OPN in the diagnosis of HCC remained greater than that of AFP. In addition, this study represents the first report of MMP1 as a biomarker with a higher sensitivity and specificity than AFP. Thus, this study provides a valuable resource of the HCC secretome with the potential to investigate serological biomarkers. MMP1 and OPN could be used as novel biomarkers for the early detection of HCC and to improve the sensitivity of biomarkers compared with AFP.
Collapse
Affiliation(s)
- Lei Yang
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory for Carcinogenesis and Cancer Prevention, Cancer Institute (Hospital), Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100021, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Morishita Y, Watanabe M, Nakazawa E, Ishibashi K, Kusano E. The interaction of LFA-1 on mononuclear cells and ICAM-1 on tubular epithelial cells accelerates TGF-β1-induced renal epithelial-mesenchymal transition. PLoS One 2011; 6:e23267. [PMID: 21850266 PMCID: PMC3151298 DOI: 10.1371/journal.pone.0023267] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2010] [Accepted: 07/14/2011] [Indexed: 12/24/2022] Open
Abstract
The epithelial-mesenchymal transition (EMT) of renal epithelial cells (RTECs) has pivotal roles in the development of renal fibrosis. Although the interaction of lymphocyte function-associated antigen 1 (LFA-1) on leukocytes and its ligand, intracellular adhesion molecule 1 (ICAM-1), plays essential roles in most inflammatory reactions, its pathogenetic role in the EMT of RTECs remains to be clarified. In the present study, we investigated the effect of the interaction of LFA-1 on peripheral blood mononuclear cells (PBMCs) and ICAM-1 on HK-2 cells after stimulation with TGF-β(1) on the EMT of RTECs. ICAM-1 was highly expressed in HK-2 cells. After TGF-β(1) stimulation, the chemokines CCL3 and CXCL12 increased on HK-2 cells. After co-culture of PBMCs and HK-2 cells pre-stimulated with TGF-β(1) (0.1 ng/ml) (HK-2-TGF-β(1) (0.1)), the expression of the active form of LFA-1 increased on PBMCs; however, total LFA-1 expression did not change. The expression of the active form of LFA-1 on PBMCs did not increase after co-culture with not CCL3 but CXCL12 knockdown HK-2-TGF-β(1) (0.1). The expression of epithelial cell junction markers (E-cadherin and occludin) further decreased and that of mesenchymal markers (vimentin and fibronectin) further increased in HK-2-TGF-β(1) (0.1) after co-culture with PBMCs for 24 hrs (HK-2-TGF-β(1) (0.1)-PBMCs). The phosphorylation of ERK 1/2 but not smad2 and smad3 increased in HK-2-TGF-β(1) (0.1)-PBMCs. The snail and slug signaling did not increase HK-2-TGF-β(1) (0.1)-PBMCs. Although the migration and invasion of HK-2 cells induced full EMT by a high dose (10.0 ng/ml) and long-term (72-96 hrs) TGF-β(1) stimulation increased, that of HK-2-TGF-β(1) (0.1)-PBMCs did not increase. These results suggested that HK-2 cells stimulated with TGF-β(1) induced conformational activation of LFA-1 on PBMCs by increased CXCL12. Then, the direct interaction of LFA-1 on PBMCs and ICAM-1 on HK-2 cells activated ERK1/2 signaling to accelerate the part of EMT of HK-2 cells induced by TGF-β(1).
Collapse
Affiliation(s)
- Yoshiyuki Morishita
- Division of Nephrology, Department of Medicine, Jichi Medical University, Tochigi, Japan.
| | | | | | | | | |
Collapse
|
19
|
Morishita Y, Hanawa S, Miki T, Sugase T, Sugaya Y, Chinda J, Iimura O, Tsunematsu S, Ishibashi K, Kusano E. The association of plasma prorenin level with an oxidative stress marker, 8-OHdG, in nondiabetic hemodialysis patients. Clin Exp Nephrol 2011; 15:398-404. [PMID: 21234784 DOI: 10.1007/s10157-010-0398-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2010] [Accepted: 12/20/2010] [Indexed: 11/25/2022]
Abstract
BACKGROUND Circulating prorenin contributes to the pathogenesis of tissue damage leading to cardiovascular disease (CVD) in hypertension and diabetic mellitus (DM) by activating the tissue renin-angiotensin-aldosterone (RAS) system; however, little is known about its roles in hemodialysis (HD) patients. METHODS We evaluated plasma prorenin level and prorenin receptor [(P)RR] expression in peripheral blood mononuclear cells (PBMCs) in 49 nondiabetic HD (non-DM-HD) patients. Then we investigated the association between plasma prorenin level or (P)RR expression level in PBMCs and CVD-predictive biomarkers. RESULTS The plasma prorenin level increased in non-DM-HD patients [147.1 ± 118.9 pg/ml (standard value <100 pg/ml)]. The (P)RR mRNA expression level in PBMCs also increased 1.41 ± 0.39-fold in non-DM-HD patients compared with that in healthy control subjects (p < 0.001). Although plasma prorenin level did not correlate with plasma BNP level and plasma high-sensitivity C-reactive protein level, it significantly correlated with plasma 8-hydroxydeoxyguanosine (8-OHdG) level (r = 0.535, p < 0.001). The plasma prorenin level did not correlate with plasma renin activity (PRA), plasma angiotensin I (AT I) level, plasma angiotensin II (AT II) level and plasma aldosterone (Ald) level. PRA, plasma AT I level, plasma AT II level and plasma Ald level did not correlate with the level of any CVD predictive biomarker. (P)RR expression level in PBMCs did not correlate with the level of any CVD predictive biomarker. CONCLUSIONS The plasma prorenin level and (P)RR expression level in PBMCs increased, and the plasma prorenin level was associated with plasma 8-OHdG level independent of circulating RAS in non-DM-HD patients.
Collapse
Affiliation(s)
- Yoshiyuki Morishita
- Division of Nephrology, Department of Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan.
| | - Shiho Hanawa
- Division of Nephrology, Department of Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Takuya Miki
- Division of Nephrology, Department of Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Taro Sugase
- Division of Nephrology, Department of Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | | | - Junko Chinda
- Division of Nephrology, Department of Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | | | | | - Kenichi Ishibashi
- Department of Medical Physiology, Meiji Pharmaceutical University, Tokyo, Japan
| | - Eiji Kusano
- Division of Nephrology, Department of Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| |
Collapse
|
20
|
Miura K, Miura S, Yamasaki K, Shimada T, Kinoshita A, Niikawa N, Yoshiura KI, Masuzaki H. The possibility of microarray-based analysis using cell-free placental mRNA in maternal plasma. Prenat Diagn 2010; 30:849-61. [DOI: 10.1002/pd.2570] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
21
|
Expression and transcriptional regulation of individual pregnancy-specific glycoprotein genes in differentiating trophoblast cells. Placenta 2010; 31:312-9. [PMID: 20116096 DOI: 10.1016/j.placenta.2010.01.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2009] [Revised: 12/31/2009] [Accepted: 01/05/2010] [Indexed: 11/23/2022]
Abstract
Human pregnancy-specific glycoproteins (PSGs), encoded by eleven highly conserved genes, are the major placental polypeptides. Low PSG levels in maternal circulation have been associated with complicated pregnancies. However, expression of each PSG gene and their regulation during cytotrophoblast cell differentiation remain poorly explored. Herein, we analyze the expression of five PSG genes and demonstrate that they are almost undetectable in undifferentiated trophoblast, but are all transcribed in differentiated cells. Among them, PSG1, PSG3 and PSG5 genes achieve high mRNA levels while PSG7 and PSG9 are poorly expressed. In addition, total PSG proteins and transcripts markedly increase during trophoblast differentiation, preceding morphological syncytialization and betahCG expression. The 5' regulatory region contributes to the transcriptional control of PSG gene induction in trophoblast cells undergoing differentiation. This responsive region in PSG3 maps within a 130 bp promoter sequence, which overlaps the transcription start site and requires a functional Retinoic Acid Responsive Element (RARE) and a GA-binding protein (GABP) consensus site for basal and differentiation-dependent promoter activity, respectively. Present findings provide novel data for understanding the control of PSG gene expression and demonstrate that their proteins and transcripts represent early markers of trophoblast differentiation.
Collapse
|
22
|
Holtan SG, Creedon DJ, Haluska P, Markovic SN. Cancer and Pregnancy: Parallels in Growth, Invasion, and Immune Modulation and Implications for Cancer Therapeutic Agents. Mayo Clin Proc 2009. [DOI: 10.4065/84.11.985] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
23
|
Holtan SG, Creedon DJ, Haluska P, Markovic SN. Cancer and pregnancy: parallels in growth, invasion, and immune modulation and implications for cancer therapeutic agents. Mayo Clin Proc 2009; 84:985-1000. [PMID: 19880689 PMCID: PMC2770910 DOI: 10.1016/s0025-6196(11)60669-1] [Citation(s) in RCA: 214] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Many proliferative, invasive, and immune tolerance mechanisms that support normal human pregnancy are also exploited by malignancies to establish a nutrient supply and evade or edit the host immune response. In addition to the shared capacity for invading through normal tissues, both cancer cells and cells of the developing placenta create a microenvironment supportive of both immunologic privilege and angiogenesis. Systemic alterations in immunity are also detectable, particularly with respect to a helper T cell type 2 polarization evident in advanced cancers and midtrimester pregnancy. This review summarizes the similarities between growth and immune privilege in cancer and pregnancy and identifies areas for further investigation. Our PubMed search strategy included combinations of terms such as immune tolerance, pregnancy, cancer, cytokines, angiogenesis, and invasion. We did not place any restrictions on publication dates. The knowledge gained from analyzing similarities and differences between the physiologic state of pregnancy and the pathologic state of cancer could lead to identification of new potential targets for cancer therapeutic agents.
Collapse
Affiliation(s)
| | | | | | - Svetomir N. Markovic
- From the Division of Hematology (S.G.H., S.N.M.), Department of Oncology (S.G.H., P.H., S.N.M.), and Department of Obstetrics and Gynecology (D.J.C.), Mayo Clinic, Rochester, MN
| |
Collapse
|
24
|
Stockwin LH, Vistica DT, Kenney S, Schrump DS, Butcher DO, Raffeld M, Shoemaker RH. Gene expression profiling of alveolar soft-part sarcoma (ASPS). BMC Cancer 2009; 9:22. [PMID: 19146682 PMCID: PMC2635365 DOI: 10.1186/1471-2407-9-22] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2008] [Accepted: 01/15/2009] [Indexed: 11/25/2022] Open
Abstract
Background Alveolar soft-part sarcoma (ASPS) is an extremely rare, highly vascular soft tissue sarcoma affecting predominantly adolescents and young adults. In an attempt to gain insight into the pathobiology of this enigmatic tumor, we performed the first genome-wide gene expression profiling study. Methods For seven patients with confirmed primary or metastatic ASPS, RNA samples were isolated immediately following surgery, reverse transcribed to cDNA and each sample hybridized to duplicate high-density human U133 plus 2.0 microarrays. Array data was then analyzed relative to arrays hybridized to universal RNA to generate an unbiased transcriptome. Subsequent gene ontology analysis was used to identify transcripts with therapeutic or diagnostic potential. A subset of the most interesting genes was then validated using quantitative RT-PCR and immunohistochemistry. Results Analysis of patient array data versus universal RNA identified elevated expression of transcripts related to angiogenesis (ANGPTL2, HIF-1 alpha, MDK, c-MET, VEGF, TIMP-2), cell proliferation (PRL, IGFBP1, NTSR2, PCSK1), metastasis (ADAM9, ECM1, POSTN) and steroid biosynthesis (CYP17A1 and STS). A number of muscle-restricted transcripts (ITGB1BP3/MIBP, MYF5, MYF6 and TRIM63) were also identified, strengthening the case for a muscle cell progenitor as the origin of disease. Transcript differentials were validated using real-time PCR and subsequent immunohistochemical analysis confirmed protein expression for several of the most interesting changes (MDK, c-MET, VEGF, POSTN, CYP17A1, ITGB1BP3/MIBP and TRIM63). Conclusion Results from this first comprehensive study of ASPS gene expression identifies several targets involved in angiogenesis, metastasis and myogenic differentiation. These efforts represent the first step towards defining the cellular origin, pathogenesis and effective treatment strategies for this atypical malignancy.
Collapse
Affiliation(s)
- Luke H Stockwin
- Screening Technologies Branch, Developmental Therapeutics Program, National Cancer Institute at Frederick, Frederick, MD 21702, USA.
| | | | | | | | | | | | | |
Collapse
|
25
|
Wong N, Novakovic B, Weinrich B, Dewi C, Andronikos R, Sibson M, Macrae F, Morley R, Pertile M, Craig J, Saffery R. Methylation of the adenomatous polyposis coli (APC) gene in human placenta and hypermethylation in choriocarcinoma cells. Cancer Lett 2008; 268:56-62. [DOI: 10.1016/j.canlet.2008.03.033] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2007] [Revised: 03/13/2008] [Accepted: 03/19/2008] [Indexed: 11/29/2022]
|
26
|
Novakovic B, Rakyan V, Ng HK, Manuelpillai U, Dewi C, Wong NC, Morley R, Down T, Beck S, Craig JM, Saffery R. Specific tumour-associated methylation in normal human term placenta and first-trimester cytotrophoblasts. Mol Hum Reprod 2008; 14:547-54. [PMID: 18708652 DOI: 10.1093/molehr/gan046] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Human placentation displays many similarities with tumourigenesis, including rapid cell division, migration and invasion, overlapping gene expression profiles and escape from immune detection. Recent data have identified promoter methylation in the Ras association factor and adenomatous polyposis coli tumour suppressor genes as part of this process. However, the extent of tumour-associated methylation in the placenta remains unclear. Using whole genome methylation data as a starting point, we have examined this phenomenon in placental tissue. We found no evidence for methylation of the majority of common tumour suppressor genes in term placentas, but identified methylation in several genes previously described in some human tumours. Notably, promoter methylation of four independent negative regulators of Wnt signalling has now been identified in human placental tissue and purified trophoblasts. Methylation is present in baboon, but not in mouse placentas. This supports a role for elevated Wnt signalling in primate trophoblast invasiveness and placentation. Examination of invasive choriocarcinoma cell lines revealed altered methylation patterns consistent with a role of methylation change in gestational trophoblastic disease. This distinct pattern of tumour-associated methylation implicates a coordinated series of epigenetic silencing events, similar to those associated with some tumours, in the distinct features of normal human placental invasion and function.
Collapse
Affiliation(s)
- B Novakovic
- Developmental Epigenetics Research, Murdoch Children's Research Institute, Royal Children's Hospital, VIC 3052, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Mols J, van den Berg A, Otsuka M, Zheng M, Chen J, Han J. TNF-alpha stimulation inhibits siRNA-mediated RNA interference through a mechanism involving poly-(A) tail stabilization. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2008; 1779:712-9. [PMID: 18423387 DOI: 10.1016/j.bbagrm.2008.03.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2007] [Revised: 03/21/2008] [Accepted: 03/22/2008] [Indexed: 12/27/2022]
Abstract
The control of mRNA stability is a complex biological process that involves numerous factors, including microRNA (miRNA) and short interfering RNA (siRNA). Here, we show that short interfering RNA (siRNA) and microRNA share some similarities in their response to cellular stress. miR16 expedites the degradation of mRNAs containing AU-rich elements (ARE) in their 3' untranslated region (UTR). si20 is an siRNA designed to target a non-ARE sequence in the TNF 3'UTR. We found that both si20 and miR16/ARE-mediated degradation of mRNAs can be inhibited by stimulating cells with different stresses. By analyzing TNF-alpha stimulation-mediated stabilization of si20- and miR16-targeted mRNA, we show that this stabilization is not caused by modifying si20 and miR16 loading into Ago2 complexes, or mRNA targeting to Ago2, but by inhibiting mRNA deadenylation. This is the first report showing that a specific siRNA-mediated mRNA degradation can be regulated by inflammatory stimuli, and that deadenylation is involved in this siRNA-mediated mRNA decay.
Collapse
Affiliation(s)
- Johann Mols
- Department of Immunology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | | | | | | | |
Collapse
|
28
|
Bowden NA, Croft A, Scott RJ. Gene expression profiling in familial adenomatous polyposis adenomas and desmoid disease. Hered Cancer Clin Pract 2007; 5:79-96. [PMID: 19725988 PMCID: PMC2736996 DOI: 10.1186/1897-4287-5-2-79] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2007] [Accepted: 04/25/2007] [Indexed: 02/26/2023] Open
Abstract
Gene expression profiling is a powerful method by which alterations in gene expression can be interrogated in a single experiment. The disease familial adenomatous polyposis (FAP) is associated with germline mutations in the APC gene, which result in aberrant β-catenin control. The molecular mechanisms underlying colorectal cancer development in FAP are being characterised but limited information is available about other symptoms that occur in this disorder. Although extremely rare in the general population, desmoid tumours in approximately 10% of FAP patients. The aim of this study was to determine the similarities and differences in gene expression profiles in adenomas and compare them to those observed in desmoid tumours. Illumina whole genome gene expression BeadChips were used to measure gene expression in FAP adenomas and desmoid tumours. Similarities between gene expression profiles and mechanisms important in regulating formation of FAP adenomas and desmoid tumours were identified. This study furthers our understanding of the mechanisms underlying FAP and desmoid tumour formation.
Collapse
Affiliation(s)
- Nikola A Bowden
- School of Biomedical Sciences, University of Newcastle, Callaghan, NSW, Australia
| | | | | |
Collapse
|
29
|
Del Rio M, Molina F, Bascoul-Mollevi C, Copois V, Bibeau F, Chalbos P, Bareil C, Kramar A, Salvetat N, Fraslon C, Conseiller E, Granci V, Leblanc B, Pau B, Martineau P, Ychou M. Gene expression signature in advanced colorectal cancer patients select drugs and response for the use of leucovorin, fluorouracil, and irinotecan. J Clin Oncol 2007; 25:773-80. [PMID: 17327601 PMCID: PMC2257989 DOI: 10.1200/jco.2006.07.4187] [Citation(s) in RCA: 145] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
PURPOSE In patients with advanced colorectal cancer, leucovorin, fluorouracil, and irinotecan (FOLFIRI) is considered as one of the reference first-line treatments. However, only about half of treated patients respond to this regimen, and there is no clinically useful marker that predicts response. A major clinical challenge is to identify the subset of patients who could benefit from this chemotherapy. We aimed to identify a gene expression profile in primary colon cancer tissue that could predict chemotherapy response. PATIENTS AND METHODS Tumor colon samples from 21 patients with advanced colorectal cancer were analyzed for gene expression profiling using Human Genome GeneChip arrays U133. At the end of the first-line treatment, the best observed response, according to WHO criteria, was used to define the responders and nonresponders. Discriminatory genes were first selected by the significance analysis of microarrays algorithm and the area under the receiver operating characteristic curve. A predictor classifier was then constructed using support vector machines. Finally, leave-one-out cross validation was used to estimate the performance and the accuracy of the output class prediction rule. RESULTS We determined a set of 14 predictor genes of response to FOLFIRI. Nine of nine responders (100% specificity) and 11 of 12 nonresponders (92% sensitivity) were classified correctly, for an overall accuracy of 95%. CONCLUSION After validation in an independent cohort of patients, our gene signature could be used as a decision tool to assist oncologists in selecting colorectal cancer patients who could benefit from FOLFIRI chemotherapy, both in the adjuvant and the first-line metastatic setting.
Collapse
Affiliation(s)
- Maguy Del Rio
- Centre National de la Recherche Scientifique Unité Mixte de Recherche 5160, Service d'Anatomie pathologique, Service d'Oncologie Digestive, Montpellier, France.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|