1
|
Huang DH, Li YZ, Xu HL, Liu FH, Li XY, Xiao Q, Chen X, Liu KX, Wang DD, Men YX, Cao YN, Gao S, Zhao YH, Gong TT, Wu QJ. Proteomics for Biomarker Discovery in Gynecological Cancers: A Systematic Review. J Proteome Res 2025; 24:1-12. [PMID: 39698999 DOI: 10.1021/acs.jproteome.4c00675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Abstract
The present study aims to summarize the current biomarker landscape in gynecological cancers (GCs) and incorporate bioinformatics analysis to highlight specific biological processes. The literature was retrieved from PubMed, Web of Science, Embase, Scopus, Ovid Medline, and Cochrane Library. The final search was conducted on December 7, 2022. Prospective registration was completed with the PROSPERO with registration number CRD42023477145. This systematic review covered proteomic research on biomarkers for cervical, endometrial, and ovarian cancers. The PANTHER classification system was used to classify the shortlisted candidate biomarkers (CBs), and the STRING database was utilized to visualize protein-protein interaction networks. A total of 23 articles were included in this systematic review. Consistently regulated CBs in the GCs include collagen alpha-2(I) chain, collagen alpha-1(III) chain, collagen alpha-2(V) chain, calreticulin, protein disulfide-isomerase A3, heat shock protein family A (Hsp70) member 5, prolyl 4-hydroxylase, beta polypeptide, fibrinogen alpha chain, fibrinogen gamma chain, apolipoprotein B-100, apolipoprotein C-IV, and apolipoprotein M. In conclusion, collagens, fibrinogens, chaperones, and apolipoproteins were revealed to be replicated in GCs and to be regulated consistently. These CBs contribute to GC etiology and physiology by participating in collagen fibril organization, blood coagulation, protein folding in endoplasmic reticulum, and lipid transporter activity.
Collapse
Affiliation(s)
- Dong-Hui Huang
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang 110004, China
- Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang 110022, China
- Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Benxi 117004, China
| | - Yi-Zi Li
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang 110004, China
- Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang 110022, China
- Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Benxi 117004, China
| | - He-Li Xu
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang 110004, China
- Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang 110022, China
- Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Benxi 117004, China
| | - Fang-Hua Liu
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang 110004, China
- Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang 110022, China
- Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Benxi 117004, China
| | - Xiao-Ying Li
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang 110004, China
- Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang 110022, China
- Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Benxi 117004, China
| | - Qian Xiao
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang 110004, China
- Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang 110022, China
- Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Benxi 117004, China
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110022, China
| | - Xing Chen
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang 110004, China
- Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang 110022, China
- Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Benxi 117004, China
| | - Ke-Xin Liu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110022, China
| | - Dong-Dong Wang
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang 110004, China
- Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang 110022, China
- Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Benxi 117004, China
| | - Yi-Xuan Men
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110022, China
| | - Yi-Ning Cao
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang 110004, China
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110022, China
| | - Song Gao
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110022, China
| | - Yu-Hong Zhao
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang 110004, China
- Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang 110022, China
- Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Benxi 117004, China
| | - Ting-Ting Gong
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110022, China
- NHC Key Laboratory of Advanced Reproductive Medicine and Fertility (China Medical University), National Health Commission, Shenyang 110022, China
| | - Qi-Jun Wu
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang 110004, China
- Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang 110022, China
- Liaoning Key Laboratory of Precision Medical Research on Major Chronic Disease, Shengjing Hospital of China Medical University, Benxi 117004, China
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110022, China
- NHC Key Laboratory of Advanced Reproductive Medicine and Fertility (China Medical University), National Health Commission, Shenyang 110022, China
| |
Collapse
|
2
|
Tang B, Wu F, Peng L, Leng X, Han Y, Wang Q, Wu J, Orlandini LC. Computed tomography-based radiomics nomogram for prediction of lympho-vascular and perineural invasion in esophageal squamous cell cancer patients: a retrospective cohort study. Cancer Imaging 2024; 24:131. [PMID: 39367492 PMCID: PMC11451056 DOI: 10.1186/s40644-024-00781-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 09/30/2024] [Indexed: 10/06/2024] Open
Abstract
PURPOSE Lympho-vascular invasion (LVI) and perineural invasion (PNI) have been established as prognostic factors in various types of cancers. The preoperative prediction of LVI and PNI has the potential to guide personalized medicine strategies for patients with esophageal squamous cell cancer (ESCC). This study investigates whether radiomics features derived from preoperative contrast-enhanced CT could predict LVI and PNI in ESCC patients. METHODS AND MATERIALS A retrospective cohort of 544 ESCC patients who underwent esophagectomy were included in this study. Preoperative contrast-enhanced CT images, pathological results of PNI and LVI, and clinical characteristics were collected. For each patient, the gross tumor volume (GTV-T) and lymph nodes volume (GTV-N) were delineated and four categories of radiomics features (first-order, shape, textural and wavelet) were extracted from GTV-T and GTV-N. The Mann-Whitney U test was used to select significant features associated with LVI and PNI in turn. Subsequently, radiomics signatures for LVI and PNI were constructed using LASSO regression with ten-fold cross-validation. Significant clinical characteristics were combined with radiomics signature to develop two nomogram models for predicting LVI and PNI, respectively. The area under the curve (AUC) and calibration curve were used to evaluate the predictive performance of the models. RESULTS The radiomics signature for LVI prediction consisted of 28 features, while the PNI radiomics signature comprised 14 features. The AUCs of the LVI radiomics signature were 0.77 and 0.74 in the training and validation groups, respectively, while the AUCs of the PNI radiomics signature were 0.69 and 0.68 in the training and validation groups. The nomograms incorporating radiomics signatures and significant clinical characteristics such as age, gender, thrombin time and D-Dimer showed improved predictive performance for both LVI (AUC: 0.82 and 0.80 in the training and validation group) and PNI (AUC: 0.75 and 0.72 in the training and validation groups) compared to the radiomics signature alone. CONCLUSION The radiomics features extracted from preoperative contrast-enhanced CT of gross tumor and lymph nodes have demonstrated their potential in predicting LVI and PNI in ESCC patients. Furthermore, the incorporation of clinical characteristics has shown additional value, resulting in improved predictive performance.
Collapse
Affiliation(s)
- Bin Tang
- Department of Radiation Oncology, Radiation Onocology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital & Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041, China
| | - Fan Wu
- Department of Radiation Oncology, Radiation Onocology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital & Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041, China
| | - Lin Peng
- Department of Thoracic Surgery, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital & Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041, China
| | - Xuefeng Leng
- Department of Thoracic Surgery, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital & Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041, China
| | - Yongtao Han
- Department of Thoracic Surgery, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital & Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041, China
| | - Qifeng Wang
- Department of Radiation Oncology, Radiation Onocology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital & Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041, China.
| | - Junxiang Wu
- Department of Radiation Oncology, Radiation Onocology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital & Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041, China.
| | - Lucia Clara Orlandini
- Department of Radiation Oncology, Radiation Onocology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital & Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041, China
| |
Collapse
|
3
|
Postl M, Danisch M, Schrott F, Kofler P, Petrov P, Aust S, Concin N, Polterauer S, Bartl T. The Predictive Value of the Fibrinogen-Albumin-Ratio Index on Surgical Outcomes in Patients with Advanced High-Grade Serous Ovarian Cancer. Cancers (Basel) 2024; 16:3295. [PMID: 39409916 PMCID: PMC11476045 DOI: 10.3390/cancers16193295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/14/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND/OBJECTIVES The present study evaluates predictive implications of the pretherapeutic Fibrinogen-Albumin-Ratio Index (FARI) in high-grade serous ovarian cancer (HGSOC) patients undergoing primary cytoreductive surgery. METHODS This retrospective study included 161 patients with HGSOC International Federation of Gynecology and Obstetrics (FIGO) stage ≥ IIb, who underwent primary cytoreductive surgery followed by platinum-based chemotherapy. Associations between the FARI and complete tumor resection status were described by receiver operating characteristics, and binary logistic regression models were fitted. RESULTS Higher preoperative FARI values correlated with higher ascites volumes (r = 0.371, p < 0.001), and higher CA125 levels (r = 0.271, p = 0.001). A high FARI cut at its median (≥11.06) was associated with lower rates of complete tumor resection (OR 3.13, 95% CI [1.63-6.05], p = 0.001), and retrained its predictive value in a multivariable model independent of ascites volumes, CA125 levels, FIGO stage, and Charlson Comorbidity Index (CCI). CONCLUSIONS The FARI appears to act as a surrogate for higher intra-abdominal tumor load. After clinical validation, FARI could serve as a readily available serologic biomarker to complement preoperative patient assessment, helping to identify patients who are likely to achieve complete tumor resection during primary cytoreductive surgery.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Stephan Polterauer
- Department of Obstetrics and Gynecology, Division of General Gynecology and Gynecologic Oncology, Medical University Vienna, 1090 Vienna, Austria; (M.P.); (M.D.); (F.S.); (P.K.); (P.P.); (S.A.); (N.C.); (T.B.)
| | | |
Collapse
|
4
|
Torkildsen CF, Austdal M, Jarmund AH, Kleinmanns K, Lamark EK, Nilsen EB, Stefansson I, Sande RK, Iversen AC, Thomsen LCV, Bjørge L. New immune phenotypes for treatment response in high-grade serous ovarian carcinoma patients. Front Immunol 2024; 15:1394497. [PMID: 38947323 PMCID: PMC11211251 DOI: 10.3389/fimmu.2024.1394497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 06/03/2024] [Indexed: 07/02/2024] Open
Abstract
Despite advances in surgical and therapeutic approaches, high-grade serous ovarian carcinoma (HGSOC) prognosis remains poor. Surgery is an indispensable component of therapeutic protocols, as removal of all visible tumor lesions (cytoreduction) profoundly improves the overall survival. Enhanced predictive tools for assessing cytoreduction are essential to optimize therapeutic precision. Patients' immune status broadly reflects the tumor cell biological behavior and the patient responses to disease and treatment. Serum cytokine profiling is a sensitive measure of immune adaption and deviation, yet its integration into treatment paradigms is underexplored. This study is part of the IMPACT trial (NCT03378297) and aimed to characterize immune responses before and during primary treatment for HGSOC to identify biomarkers for treatment selection and prognosis. Longitudinal serum samples from 22 patients were collected from diagnosis until response evaluation. Patients underwent primary cytoreductive surgery or neoadjuvant chemotherapy (NACT) based on laparoscopy scoring. Twenty-seven serum cytokines analyzed by Bio-Plex 200, revealed two immune phenotypes at diagnosis: Immune High with marked higher serum cytokine levels than Immune Low. The immune phenotypes reflected the laparoscopy scoring and allocation to surgical treatment. The five Immune High patients undergoing primary cytoreductive surgery exhibited immune mobilization and extended progression-free survival, compared to the Immune Low patients undergoing the same treatment. Both laparoscopy and cytoreductive surgery induced substantial and transient changes in serum cytokines, with upregulation of the inflammatory cytokine IL-6 and downregulation of the multifunctional cytokines IP-10, Eotaxin, IL-4, and IL-7. Over the study period, cytokine levels uniformly decreased in all patients, leading to the elimination of the initial immune phenotypes regardless of treatment choice. This study reveals distinct pre-treatment immune phenotypes in HGSOC patients that might be informative for treatment stratification and prognosis. This potential novel biomarker holds promise as a foundation for improved assessment of treatment responses in patients with HGSOC. ClinicalTrials.gov Identifier: NCT03378297.
Collapse
Affiliation(s)
- Cecilie Fredvik Torkildsen
- Department of Obstetrics and Gynecology, Stavanger University Hospital, Stavanger, Norway
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Marie Austdal
- Department of Research, Stavanger University Hospital, Stavanger, Norway
| | - Anders Hagen Jarmund
- Department of Clinical and Molecular Medicine, and Centre of Molecular Inflammation Research (CEMIR), Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Katrin Kleinmanns
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway
| | - Eva Karin Lamark
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway
| | - Elisabeth Berge Nilsen
- Department of Obstetrics and Gynecology, Stavanger University Hospital, Stavanger, Norway
| | - Ingunn Stefansson
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Ragnar Kvie Sande
- Department of Obstetrics and Gynecology, Stavanger University Hospital, Stavanger, Norway
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Ann-Charlotte Iversen
- Department of Clinical and Molecular Medicine, and Centre of Molecular Inflammation Research (CEMIR), Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Liv Cecilie Vestrheim Thomsen
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway
| | - Line Bjørge
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
5
|
Amniouel S, Yalamanchili K, Sankararaman S, Jafri MS. Evaluating Ovarian Cancer Chemotherapy Response Using Gene Expression Data and Machine Learning. BIOMEDINFORMATICS 2024; 4:1396-1424. [PMID: 39149564 PMCID: PMC11326537 DOI: 10.3390/biomedinformatics4020077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Background Ovarian cancer (OC) is the most lethal gynecological cancer in the United States. Among the different types of OC, serous ovarian cancer (SOC) stands out as the most prevalent. Transcriptomics techniques generate extensive gene expression data, yet only a few of these genes are relevant to clinical diagnosis. Methods Methods for feature selection (FS) address the challenges of high dimensionality in extensive datasets. This study proposes a computational framework that applies FS techniques to identify genes highly associated with platinum-based chemotherapy response on SOC patients. Using SOC datasets from the Gene Expression Omnibus (GEO) database, LASSO and varSelRF FS methods were employed. Machine learning classification algorithms such as random forest (RF) and support vector machine (SVM) were also used to evaluate the performance of the models. Results The proposed framework has identified biomarkers panels with 9 and 10 genes that are highly correlated with platinum-paclitaxel and platinum-only response in SOC patients, respectively. The predictive models have been trained using the identified gene signatures and accuracy of above 90% was achieved. Conclusions In this study, we propose that applying multiple feature selection methods not only effectively reduces the number of identified biomarkers, enhancing their biological relevance, but also corroborates the efficacy of drug response prediction models in cancer treatment.
Collapse
Affiliation(s)
- Soukaina Amniouel
- School of System Biology, George Mason University, Fairfax, VA 22030, USA
| | - Keertana Yalamanchili
- School of System Biology, George Mason University, Fairfax, VA 22030, USA
- School of Engineering, Brown University, Providence, RI 02912, USA
| | - Sreenidhi Sankararaman
- School of System Biology, George Mason University, Fairfax, VA 22030, USA
- Department of Biomedical Engineering, The John Hopkins University, Baltimore, MD 21218, USA
| | - Mohsin Saleet Jafri
- School of System Biology, George Mason University, Fairfax, VA 22030, USA
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
6
|
Shettar PS, Hiremath MB, Kumbar VM. Anti-proliferative Activity of Vitex negundo Leaf Extracts on PA1 Human Ovarian Cancer Cell Lines. ARCHIVES OF RAZI INSTITUTE 2024; 79:426-436. [PMID: 39463706 PMCID: PMC11512179 DOI: 10.32592/ari.2024.79.2.426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 09/03/2023] [Indexed: 10/29/2024]
Abstract
In Ayurveda, Vitex negundo (VN) is used as a drug to manage pain, inflammation, and problems related to polycystic ovary disease and the menstrual cycle. The bioactive compounds isolated from this plant exhibit anti-inflammatory, anti-oxidant, anti-cancer, and microbicidal properties. The shrub VN is known for its role in the modulation of cellular events like apoptosis and cell cycle. There is still a scarcity of data in the literature on the cytotoxic activity of VN extracts on ovarian cancer. Therefore, in the present study, the phytochemical composition, anti-oxidant, and anti-cancer activities of leaf extracts were evaluated. The chloroform and methanol fractions exhibited higher phenolic content (161.04 ± 0.02 mg/g GAE and 152.56 ± 0.05 mg/g GAE, respectively) than those of other fractions. The aqueous and petroleum ether fractions exhibited higher flavonoid content (215.27 ± 0.28 mg/g QE and 111.82 ± 0.05 mg/g QE, respectively). The acetone and methanol extracts showed significant anti-oxidant capacities. Both leaf extracts of VN inhibited PA1 cancer cell growth in a dose-dependent manner with IC50 values of 88.01 ± 3.14 and 112.30 ± 1.93 μg/ml, respectively, as compared to the standard drug Doxorubicin with IC50 value 2.91 μg/ml (P<0.05, One-way ANOVA). The gas chromatography-mass spectroscopy (GC-MS) analysis allowed us to identify twenty-five bioactive compounds in acetone extract and twenty-two in methanol extract. Therefore, further studies should focus on the isolation of novel compounds that are more effective and less toxic, and that constitute interesting substitutes for the development of anti-cancer drugs.
Collapse
Affiliation(s)
- P S Shettar
- Department of Biotechnology and Microbiology, Karnatak University, Dharwad- 580003. Karnataka, India
| | - M B Hiremath
- Department of Biotechnology and Microbiology, Karnatak University, Dharwad- 580003. Karnataka, India
| | - V M Kumbar
- Basic Science Research Centre, KLE Academy of Higher Education and Research (KLE University), Belagavi, Karnataka, India
| |
Collapse
|
7
|
Lyu Y, Wu C, Sun W, Li Z. Regional analysis to delineate intrasample heterogeneity with RegionalST. Bioinformatics 2024; 40:btae186. [PMID: 38579257 PMCID: PMC11026142 DOI: 10.1093/bioinformatics/btae186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 03/06/2024] [Accepted: 04/03/2024] [Indexed: 04/07/2024] Open
Abstract
MOTIVATION Spatial transcriptomics has greatly contributed to our understanding of spatial and intra-sample heterogeneity, which could be crucial for deciphering the molecular basis of human diseases. Intra-tumor heterogeneity, e.g. may be associated with cancer treatment responses. However, the lack of computational tools for exploiting cross-regional information and the limited spatial resolution of current technologies present major obstacles to elucidating tissue heterogeneity. RESULTS To address these challenges, we introduce RegionalST, an efficient computational method that enables users to quantify cell type mixture and interactions, identify sub-regions of interest, and perform cross-region cell type-specific differential analysis for the first time. Our simulations and real data applications demonstrate that RegionalST is an efficient tool for visualizing and analyzing diverse spatial transcriptomics data, thereby enabling accurate and flexible exploration of tissue heterogeneity. Overall, RegionalST provides a one-stop destination for researchers seeking to delve deeper into the intricacies of spatial transcriptomics data. AVAILABILITY AND IMPLEMENTATION The implementation of our method is available as an open-source R/Bioconductor package with a user-friendly manual available at https://bioconductor.org/packages/release/bioc/html/RegionalST.html.
Collapse
Affiliation(s)
- Yue Lyu
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
- Department of Biostatistics and Data Science, The University of Texas Health Science Center at Houston, Houston, TX 77030, United States
| | - Chong Wu
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Wei Sun
- Biostatistics Program, Public Health Science Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, United States
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27516, United States
- Department of Biostatistics, University of Washington, Seattle, WA 98195, United States
| | - Ziyi Li
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| |
Collapse
|
8
|
Wang S, Zhang J, Li J, Wang J, Liu W, Zhang Z, Yu H. Label-free quantitative proteomics reveals the potential mechanisms of insoluble dietary fiber from okara in improving hepatic lipid metabolism of high-fat diet-induced mice. J Proteomics 2023; 287:104980. [PMID: 37499746 DOI: 10.1016/j.jprot.2023.104980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 03/21/2023] [Accepted: 07/24/2023] [Indexed: 07/29/2023]
Abstract
The high purity insoluble dietary fiber (IDF) from okara is a natural component with a potentially positive effect on a high-fat diet (HFD)-induced hepatic metabolic disorders, although its regulatory mechanism remains unclear. This study aims to elucidate the potential pathways and key proteins of IDF for the amelioration of hepatic lipid metabolism in mice fed with HFD. Here, we used label-free quantitative proteomics technology to quantity and identify differentially expressed proteins in the liver that are associated with IDF treatment. The differentially expressed proteins were assessed by GO annotation and KEGG pathways. Western blot and qRT-PCR analyses were conducted to validate the potential targets regulated by IDF. In total, 73 differentially expressed proteins were identified, of which 27 were up-regulated (FC > 1.5) and 46 were down-regulated (FC < 0.667). GO analysis suggested that differentially expressed proteins were mainly located in the cell and organelles, regulated biological processes, and were associated with enzyme activity and molecular binding. The KEGG pathway enrichment analysis further demonstrated glycolysis/gluconeogenesis, pyruvate metabolism, TCA cycle, arginine biosynthesis, alanine, aspartate and glutamate metabolism, and retinol metabolism were affected. The combination of proteomics, Western blot, and qRT-PCR suggested that ACS, ACLY, GOT1, GLS2, NAGS, CYP4A10, CYP3A25, and CYP2A5 in these pathways might be key proteins for IDF intervention. Taken together, our findings elucidate new mechanisms involved in how IDF affects hepatic metabolism, provide important information for the functional food industries, and improve the added value of okara. SIGNIFICANCE: Okara is evidenced as a high-quality by-product with several nutritional components, especially dietary fiber (50-60%) labeled as "The Seventh Nutrient". Previous studies have shown that IDF has a positive potential effect on a high-fat diet (HFD)-induced hepatic metabolic disorders, but its molecular mechanism remains unclear. To elucidate the therapeutic mechanism of IDF at the protein level, a label-free quantitative proteomic analysis was used to identify the dynamic changes of the liver proteome between HIDF and HFD groups in this study. These results provide a new perspective for exploring the therapeutic mechanism of IDF at the protein level and enlightenment for promoting the comprehensive utilization of okara.
Collapse
Affiliation(s)
- Sainan Wang
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, Jilin 130118, China; Division of Soybean Processing, Soybean Research & Development Center, Chinese Agricultural Research System, Changchun, Jilin 130118, China
| | - Jiarui Zhang
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, Jilin 130118, China; Division of Soybean Processing, Soybean Research & Development Center, Chinese Agricultural Research System, Changchun, Jilin 130118, China
| | - Jiaxin Li
- Division of Soybean Processing, Soybean Research & Development Center, Chinese Agricultural Research System, Changchun, Jilin 130118, China; Universidade de Vigo, Nutrition and Bromatology Group, Department of Analytical and Food Chemistry, Faculty of Sciences, Ourense, 32004, Spain
| | - Junyao Wang
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, Jilin 130118, China; Division of Soybean Processing, Soybean Research & Development Center, Chinese Agricultural Research System, Changchun, Jilin 130118, China
| | - Wenhao Liu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, Jilin 130118, China; Division of Soybean Processing, Soybean Research & Development Center, Chinese Agricultural Research System, Changchun, Jilin 130118, China
| | - Zhao Zhang
- Shandong Sinoglory Health Food Co., Ltd., Liaocheng, Shandong 252000, China
| | - Hansong Yu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, Jilin 130118, China; Division of Soybean Processing, Soybean Research & Development Center, Chinese Agricultural Research System, Changchun, Jilin 130118, China.
| |
Collapse
|
9
|
Buzza MS, Pawar NR, Strong AA, Antalis TM. Intersection of Coagulation and Fibrinolysis by the Glycosylphosphatidylinositol (GPI)-Anchored Serine Protease Testisin. Int J Mol Sci 2023; 24:9306. [PMID: 37298257 PMCID: PMC10252689 DOI: 10.3390/ijms24119306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/16/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Hemostasis is a delicate balance between coagulation and fibrinolysis that regulates the formation and removal of fibrin, respectively. Positive and negative feedback loops and crosstalk between coagulation and fibrinolytic serine proteases maintain the hemostatic balance to prevent both excessive bleeding and thrombosis. Here, we identify a novel role for the glycosylphosphatidylinositol (GPI)-anchored serine protease testisin in the regulation of pericellular hemostasis. Using in vitro cell-based fibrin generation assays, we found that the expression of catalytically active testisin on the cell surface accelerates thrombin-dependent fibrin polymerization, and intriguingly, that it subsequently promotes accelerated fibrinolysis. We find that the testisin-dependent fibrin formation is inhibited by rivaroxaban, a specific inhibitor of the central prothrombin-activating serine protease factor Xa (FXa), demonstrating that cell-surface testisin acts upstream of factor X (FX) to promote fibrin formation at the cell surface. Unexpectedly, testisin was also found to accelerate fibrinolysis by stimulating the plasmin-dependent degradation of fibrin and enhancing plasmin-dependent cell invasion through polymerized fibrin. Testisin was not a direct activator of plasminogen, but it is able to induce zymogen cleavage and the activation of pro-urokinase plasminogen activator (pro-uPA), which converts plasminogen to plasmin. These data identify a new proteolytic component that can regulate pericellular hemostatic cascades at the cell surface, which has implications for angiogenesis, cancer biology, and male fertility.
Collapse
Affiliation(s)
- Marguerite S. Buzza
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (N.R.P.); (A.A.S.); (T.M.A.)
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Research and Development Service, VA Maryland Health Care System, Baltimore, MD 21201, USA
| | - Nisha R. Pawar
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (N.R.P.); (A.A.S.); (T.M.A.)
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Amando A. Strong
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (N.R.P.); (A.A.S.); (T.M.A.)
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Toni M. Antalis
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (N.R.P.); (A.A.S.); (T.M.A.)
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Research and Development Service, VA Maryland Health Care System, Baltimore, MD 21201, USA
| |
Collapse
|
10
|
Jing H, Wu X, Xiang M, Wang C, Novakovic VA, Shi J. Microparticle Phosphatidylserine Mediates Coagulation: Involvement in Tumor Progression and Metastasis. Cancers (Basel) 2023; 15:cancers15071957. [PMID: 37046617 PMCID: PMC10093313 DOI: 10.3390/cancers15071957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/17/2023] [Accepted: 03/17/2023] [Indexed: 04/14/2023] Open
Abstract
Tumor progression and cancer metastasis has been linked to the release of microparticles (MPs), which are shed upon cell activation or apoptosis and display parental cell antigens, phospholipids such as phosphatidylserine (PS), and nucleic acids on their external surfaces. In this review, we highlight the biogenesis of MPs as well as the pathophysiological processes of PS externalization and its involvement in coagulation activation. We review the available evidence, suggesting that coagulation factors (mainly tissue factor, thrombin, and fibrin) assist in multiple steps of tumor dissemination, including epithelial-mesenchymal transition, extracellular matrix remodeling, immune escape, and tumor angiogenesis to support the formation of the pre-metastatic niche. Platelets are not just bystander cells in circulation but are functional players in primary tumor growth and metastasis. Tumor-induced platelet aggregation protects circulating tumor cells (CTCs) from the blood flow shear forces and immune cell attack while also promoting the binding of CTCs to endothelial cells and extravasation, which activates tumor invasion and sustains metastasis. Finally, in terms of therapy, lactadherin can inhibit coagulation by competing effectively with coagulation factors for PS binding sites and may similarly delay tumor progression. Furthermore, we also investigate the therapeutic potential of coagulation factor inhibitors within the context of cancer treatment. The development of multiple therapies targeting platelet activation and platelet-tumor cell interactions may not only reduce the lethal consequences of thrombosis but also impede tumor growth and spread.
Collapse
Affiliation(s)
- Haijiao Jing
- Department of Hematology, The First Hospital, Harbin Medical University, Harbin 150001, China
| | - Xiaoming Wu
- Department of Hematology, The First Hospital, Harbin Medical University, Harbin 150001, China
| | - Mengqi Xiang
- Department of Hematology, The First Hospital, Harbin Medical University, Harbin 150001, China
| | - Chengyue Wang
- Department of Hematology, The First Hospital, Harbin Medical University, Harbin 150001, China
| | - Valerie A Novakovic
- Department of Research, VA Boston Healthcare System, Harvard Medical School, Boston, MA 02132, USA
| | - Jialan Shi
- Department of Hematology, The First Hospital, Harbin Medical University, Harbin 150001, China
- Department of Research, VA Boston Healthcare System, Harvard Medical School, Boston, MA 02132, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02132, USA
| |
Collapse
|
11
|
Kim JG, Kim SI, Song SH, Gu JY, Lee M, Kim HK. Diagnostic and prognostic role of circulating neutrophil extracellular trap markers and prekallikrein in patients with high-grade serous ovarian cancer. Front Oncol 2022; 12:992056. [PMID: 36620601 PMCID: PMC9813379 DOI: 10.3389/fonc.2022.992056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Abstract
Objective Tumor-promoting inflammation is among the hallmarks of cancer. Prekallikrein is among the acute-phase reactants in the inflammatory response; moreover, neutrophils release nuclear contents into the extracellular space to create neutrophil extracellular traps (NET). We aimed to investigate the diagnostic and prognostic utilities of circulating plasma NET markers and prekallikrein for high-grade serous ovarian cancer (HGSOC). Methods Circulating levels of three NET markers (histone-DNA complex, cell-free DNA, and neutrophil elastase) and prekallikrein were measured in 75 patients with HGSOC and 23 healthy controls. We used an area under the receiver operating characteristic curve (AUC) analysis to investigate their diagnostic and prognostic utilities for HGSOC. Results Compared with healthy controls, patients with HGSOC showed significantly higher levels of the three NET markers and prekallikrein. Patients with advanced-stage HGSOC showed significantly higher levels of the cell-free DNA (87.4 vs. 79.5 ng/ml; P = 0.013), compared with those with early-stage HGSOC. Further, the levels of histone-DNA complex, neutrophil elastase, and prekallikrein did not significantly differ according to the cancer stage. All markers showed significant diagnostic utility. Notably, a logistic regression-based model that comprised all four markers showed the strongest diagnostic power (AUC, 0.966; 95% confidence interval [CI], 0.933-1.000). Specifically, neutrophil elastase was identified as an independent poor prognostic factor for overall survival (adjusted hazard ratio [aHR], 10.17; 95% CI, 1.09-94.97; P = 0.042) and progression-free survival (aHR, 14.47; 95% CI, 1.52-137.35; P = 0.020) in patients with HGSOC. Conclusions The levels of the three NET markers and prekallikrein might be novel diagnostic and prognostic markers for HGSOC.
Collapse
Affiliation(s)
- Jisoo G. Kim
- Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Se Ik Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, South Korea
| | - Sang Hoon Song
- Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul, South Korea,Department of Laboratory Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Ja-Yoon Gu
- Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul, South Korea,Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Maria Lee
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, South Korea,Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul, South Korea,*Correspondence: Maria Lee, ; Hyun Kyung Kim,
| | - Hyun Kyung Kim
- Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul, South Korea,Department of Laboratory Medicine, Seoul National University Hospital, Seoul, South Korea,Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea,*Correspondence: Maria Lee, ; Hyun Kyung Kim,
| |
Collapse
|
12
|
Yokomizo R, Lopes TJS, Takashima N, Hirose S, Kawabata A, Takenaka M, Iida Y, Yanaihara N, Yura K, Sago H, Okamoto A, Umezawa A. O3C Glass-Class: A Machine-Learning Framework for Prognostic Prediction of Ovarian Clear-Cell Carcinoma. Bioinform Biol Insights 2022. [DOI: 10.1177/11779322221134312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Ovarian clear cell carcinoma (OCCC), one of the histopathological types of ovarian cancer, has a poor prognosis when it recurs; however, it is difficult to precisely predict the risk of recurrence. Here, we analyzed pathological images of OCCC to elucidate the relationship between pathological findings and recurrence, and using machine learning, we established a classifier to predict the recurrence and several other prognosis indicators of this disease. In total, 110 patients with OCCC treated with primary surgery at a single institution were enrolled in this study. We used the deep-learning neural networks to process the whole slide images of OCCC obtained by digitally scanning the original hematoxylin and eosin-stained glass slides. The images were preprocessed and used as input to the machine learning pipeline. We fine-tuned its parameters to predict the recurrence, progression-free survival, and the overall survival days of all patients. We predicted the recurrence of OCCC with an overall accuracy of 93%, area under the receiver operating characteristic curve of 0.98, and sensitivity/specificity above 0.92 using Resnet 34. Furthermore, we predicted progression-free survival/overall survival of the patients with ~90% accuracy. In conclusion, our study demonstrates the feasibility of using a machine learning system to predict different features of OCCC samples using histopathological images as input. This novel application provides accurate prognosis information and aids in the development of personalized treatment strategies.
Collapse
Affiliation(s)
- Ryo Yokomizo
- Center for Regenerative Medicine, National Center for Child Health and Development Research Institute, Setagaya-ku, Japan
- Center for Maternal-Fetal, Neonatal and Reproductive Medicine, National Center for Child Health and Development, Setagaya-ku, Japan
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Minato-ku, Japan
| | - Tiago JS Lopes
- Center for Regenerative Medicine, National Center for Child Health and Development Research Institute, Setagaya-ku, Japan
| | - Nagisa Takashima
- Center for Regenerative Medicine, National Center for Child Health and Development Research Institute, Setagaya-ku, Japan
- Divison of Life Sciences, Graduate School of Humanities and Sciences, Ochanomizu University, Bunkyo-ku, Japan
| | - Sou Hirose
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Minato-ku, Japan
| | - Ayako Kawabata
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Minato-ku, Japan
| | - Masataka Takenaka
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Minato-ku, Japan
| | - Yasushi Iida
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Minato-ku, Japan
| | - Nozomu Yanaihara
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Minato-ku, Japan
| | - Kei Yura
- Divison of Life Sciences, Graduate School of Humanities and Sciences, Ochanomizu University, Bunkyo-ku, Japan
- Department of Life Science & Medical Bioscience, School of Advanced Science and Engineering, Waseda University, Shinjuku-ku, Japan
| | - Haruhiko Sago
- Center for Maternal-Fetal, Neonatal and Reproductive Medicine, National Center for Child Health and Development, Setagaya-ku, Japan
| | - Aikou Okamoto
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Minato-ku, Japan
| | - Akihiro Umezawa
- Center for Regenerative Medicine, National Center for Child Health and Development Research Institute, Setagaya-ku, Japan
| |
Collapse
|
13
|
Yazdian M, Groeben H, Ataseven B, Schneider S, Baert T, Bommert M, Traut A, Elfers-Wassenhoven A, Brüß U, Schwameis R, du Bois A, Wagner U, Harter P. The role of factor XIII in surgery for advanced stage of epithelial ovarian cancer. Arch Gynecol Obstet 2021; 305:1311-1318. [PMID: 34724570 DOI: 10.1007/s00404-021-06308-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 10/21/2021] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Hereditary factor (F) XIII-deficiency is a known risk factor for postoperative complications, but data of acquired FXIII-deficiency in malignancies are limited. Therefore, we evaluated the role of acquired FXIII-deficiency in surgery for advanced epithelial ovarian cancer (EOC). MATERIALS AND METHODS We performed a retrospective analysis of patients with known serum FXIII status and treatment between 2011 and 2018 at our center. We defined cohorts according to FXIII with values > 75% as normal (group A), 55-75% as reduced (group B) and < 55% as low (group C). Complications were classified according to the Clavien-Dindo Classification, class III-V complications were defined as severe. RESULTS 347 patients with EOC were identified. 180 patients (51.2%) were in group A, 82 patients (23.6%) in group B, and 85 patients (24.4%) in group C. Lower levels of FXIII were associated with higher amount of ascites, FIGO IV, high grade serous histology, low albumin, and higher CA-125 levels. Regarding intraoperative variables, low FXIII was associated with longer duration of surgery, higher blood loss, higher surgical complexity score/number of bowel anastomosis and a higher probability for macroscopic residual disease. The risk of severe complications in group A was 12.2%, 24.4% in group B, and 31.8% in group C. In a multivariate model, low FXIII (OR 2.8), > 1 bowel anastomosis (OR 2.7), age-adjusted Charlson comorbidity index ≥ 4 (OR 3.6) and a longer duration of surgery (> 285 min.) were significant predictive factors for severe complications. CONCLUSION FXIII is associated with tumor and treatment burden. A low level of FXIII is associated with postoperative complications. The knowledge about the presurgical serum FXIII-level might be helpful to plan the treatment strategy.
Collapse
Affiliation(s)
- Mahtab Yazdian
- Department of Gynecology and Gynecologic Oncology, Ev. Kliniken Essen-Mitte, 45136, Essen, Germany
- Department of Gynecology and Obstetrics, Philipps-University, Marburg, Germany
| | - Harald Groeben
- Department of Anesthesiology, Ev. Kliniken-Essen-Mitte, Essen, Germany
| | - Beyhan Ataseven
- Department of Gynecology and Gynecologic Oncology, Ev. Kliniken Essen-Mitte, 45136, Essen, Germany
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Munich, Germany
| | - Stephanie Schneider
- Department of Gynecology and Gynecologic Oncology, Ev. Kliniken Essen-Mitte, 45136, Essen, Germany
| | - Thais Baert
- Department of Gynecology and Gynecologic Oncology, Ev. Kliniken Essen-Mitte, 45136, Essen, Germany
| | - Mareike Bommert
- Department of Gynecology and Gynecologic Oncology, Ev. Kliniken Essen-Mitte, 45136, Essen, Germany
| | - Alexander Traut
- Department of Gynecology and Gynecologic Oncology, Ev. Kliniken Essen-Mitte, 45136, Essen, Germany
| | | | - Ulrich Brüß
- Department of Anesthesiology, Ev. Kliniken-Essen-Mitte, Essen, Germany
| | - Richard Schwameis
- Department of Gynecology and Gynecologic Oncology, Ev. Kliniken Essen-Mitte, 45136, Essen, Germany
| | - Andreas du Bois
- Department of Gynecology and Gynecologic Oncology, Ev. Kliniken Essen-Mitte, 45136, Essen, Germany
| | - Uwe Wagner
- Department of Gynecology and Obstetrics, Philipps-University, Marburg, Germany
| | - Philipp Harter
- Department of Gynecology and Gynecologic Oncology, Ev. Kliniken Essen-Mitte, 45136, Essen, Germany.
- Department of Gynecology and Obstetrics, Philipps-University, Marburg, Germany.
| |
Collapse
|
14
|
Huang Y, Yang Z, Huang C, Jiang X, Yan Y, Zhuang K, Wen Y, Liu F, Li P. Identification of N6-Methylandenosine-Related lncRNAs for Subtype Identification and Risk Stratification in Gastric Adenocarcinoma. Front Oncol 2021; 11:725181. [PMID: 34646770 PMCID: PMC8504261 DOI: 10.3389/fonc.2021.725181] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 09/02/2021] [Indexed: 01/11/2023] Open
Abstract
Objectives The purpose of this study was to investigate the role of m6A-related lncRNAs in gastric adenocarcinoma (STAD) and to determine their prognostic value. Methods Gene expression and clinicopathological data were obtained from The Cancer Genome Atlas (TCGA) database. Correlation analysis and univariate Cox regression analysis were conducted to identify m6A-related prognostic lncRNAs. Subsequently, different clusters of patients with STAD were identified via consensus clustering analysis, and a prognostic signature was established by least absolute shrinkage and selection operator (LASSO) Cox regression analyses. The clinicopathological characteristics, tumor microenvironment (TME), immune checkpoint genes (ICGs) expression, and the response to immune checkpoint inhibitors (ICIs) in different clusters and subgroups were explored. The prognostic value of the prognostic signature was evaluated using the Kaplan-Meier method, receiver operating characteristic curves, and univariate and multivariate regression analyses. Additionally, Gene Set Enrichment Analysis (GSEA), Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway, and Gene Ontology (GO) analysis were performed for biological functional analysis. Results Two clusters based on 19 m6A-related lncRNAs were identified, and a prognostic signature comprising 14 m6A-related lncRNAs was constructed, which had significant value in predicting the OS of patients with STAD, clinicopathological characteristics, TME, ICGs expression, and the response to ICIs. Biological processes and pathways associated with cancer and immune response were identified. Conclusions We revealed the role and prognostic value of m6A-related lncRNAs in STAD. Together, our finding refreshed the understanding of m6A-related lncRNAs and provided novel insights to identify predictive biomarkers and immunotherapy targets for STAD.
Collapse
Affiliation(s)
- Yuancheng Huang
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zehong Yang
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chaoyuan Huang
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaotao Jiang
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yanhua Yan
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Kunhai Zhuang
- Department of Gastroenterology, Baiyun Branch of the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yi Wen
- Department of Gastroenterology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Fengbin Liu
- Department of Gastroenterology, Baiyun Branch of the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Peiwu Li
- Department of Gastroenterology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
15
|
Zhou K, Arslanturk S, Craig DB, Heath E, Draghici S. Discovery of primary prostate cancer biomarkers using cross cancer learning. Sci Rep 2021; 11:10433. [PMID: 34001952 PMCID: PMC8128891 DOI: 10.1038/s41598-021-89789-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 04/30/2021] [Indexed: 02/03/2023] Open
Abstract
Prostate cancer (PCa), the second leading cause of cancer death in American men, is a relatively slow-growing malignancy with multiple early treatment options. Yet, a significant number of low-risk PCa patients are over-diagnosed and over-treated with significant and long-term quality of life effects. Further, there is ever increasing evidence of metastasis and higher mortality when hormone-sensitive or castration-resistant PCa tumors are treated indistinctively. Hence, the critical need is to discover clinically-relevant and actionable PCa biomarkers by better understanding the biology of PCa. In this paper, we have discovered novel biomarkers of PCa tumors through cross-cancer learning by leveraging the pathological and molecular similarities in the DNA repair pathways of ovarian, prostate, and breast cancer tumors. Cross-cancer disease learning enriches the study population and identifies genetic/phenotypic commonalities that are important across diseases with pathological and molecular similarities. Our results show that ADIRF, SLC2A5, C3orf86, HSPA1B are among the most significant PCa biomarkers, while MTRNR2L1, EEPD1, TEPP and VN1R2 are jointly important biomarkers across prostate, breast and ovarian cancers. Our validation results have further shown that the discovered biomarkers can predict the disease state better than any randomly selected subset of differentially expressed prostate cancer genes.
Collapse
Affiliation(s)
- Kaiyue Zhou
- Department of Computer Science, Wayne State University, Detroit, 48201, USA
| | - Suzan Arslanturk
- Department of Computer Science, Wayne State University, Detroit, 48201, USA.
| | - Douglas B Craig
- Department of Oncology, Wayne State University, Detroit, 48201, USA
- Bioinformatics and Biostatistics Core, Barbara Ann Karmanos Cancer Institute, Detroit, 48201, USA
| | - Elisabeth Heath
- Department of Oncology, Wayne State University, Detroit, 48201, USA
- Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Detroit, 48201, USA
| | - Sorin Draghici
- Department of Computer Science, Wayne State University, Detroit, 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, 48201, USA
| |
Collapse
|
16
|
Chaudhari S, Dey Pereira S, Asare-Warehene M, Naha R, Kabekkodu SP, Tsang BK, Satyamoorthy K. Comorbidities and inflammation associated with ovarian cancer and its influence on SARS-CoV-2 infection. J Ovarian Res 2021; 14:39. [PMID: 33632295 PMCID: PMC7906086 DOI: 10.1186/s13048-021-00787-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 02/09/2021] [Indexed: 12/29/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) caused by the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) worldwide is a major public health concern. Cancer patients are considered a vulnerable population to SARS-CoV-2 infection and may develop several COVID-19 symptoms. The heightened immunocompromised state, prolonged chronic pro-inflammatory milieu coupled with comorbid conditions are shared in both disease conditions and may influence patient outcome. Although ovarian cancer (OC) and COVID-19 are diseases of entirely different primary organs, both diseases share similar molecular and cellular characteristics in their microenvironment suggesting a potential cooperativity leading to poor outcome. In COVID-19 related cases, hospitalizations and deaths worldwide are lower in women than in males; however, comorbidities associated with OC may increase the COVID-19 risk in women. The women at the age of 50-60 years are at greater risk of developing OC as well as SARS-CoV-2 infection. Increased levels of gonadotropin and androgen, dysregulated renin-angiotensin-aldosterone system (RAAS), hyper-coagulation and chronic inflammation are common conditions observed among OC and severe cases of COVID-19. The upregulation of common inflammatory cytokines and chemokines such as tumor necrosis factor α (TNF-α), interleukin (IL)-1β, IL-2, IL-6, IL-10, interferon-γ-inducible protein 10 (IP-10), granulocyte colony-stimulating factor (G-CSF), monocyte chemoattractant protein-1 (MCP-1), macrophage colony-stimulating factor (M-CSF), among others in the sera of COVID-19 and OC subjects suggests potentially similar mechanism(s) involved in the hyper-inflammatory condition observed in both disease states. Thus, it is conceivable that the pathogenesis of OC may significantly contribute to the potential infection by SARS-CoV-2. Our understanding of the influence and mechanisms of SARS-CoV-2 infection on OC is at an early stage and in this article, we review the underlying pathogenesis presented by various comorbidities of OC and correlate their influence on SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Sima Chaudhari
- Department of Cell and Molecular Biology, Manipal School of Life Science, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Satyajit Dey Pereira
- Department of Cell and Molecular Biology, Manipal School of Life Science, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Meshach Asare-Warehene
- Chronic Disease Program, Ottawa Hospital Research Institute and Department of Obstetrics & Gynecology and Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, K1N 6N5, Canada
| | - Ritam Naha
- Department of Cell and Molecular Biology, Manipal School of Life Science, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Science, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Benjamin K Tsang
- Chronic Disease Program, Ottawa Hospital Research Institute and Department of Obstetrics & Gynecology and Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, K1N 6N5, Canada
| | - Kapaettu Satyamoorthy
- Department of Cell and Molecular Biology, Manipal School of Life Science, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
17
|
Chen X, Sun J, Wang X, Yuan Y, Cai L, Xie Y, Fan Z, Liu K, Jiao X. A Meta-Analysis of Proteomic Blood Markers of Colorectal Cancer. Curr Med Chem 2021; 28:1176-1196. [PMID: 32338203 DOI: 10.2174/0929867327666200427094054] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 02/23/2020] [Accepted: 03/24/2020] [Indexed: 02/05/2023]
Abstract
BACKGROUND Early diagnosis will significantly improve the survival rate of colorectal cancer (CRC); however, the existing methods for CRC screening were either invasive or inefficient. There is an emergency need for novel markers in CRC's early diagnosis. Serum proteomics has gained great potential in discovering novel markers, providing markers that reflect the early stage of cancer and prognosis prediction of CRC. In this paper, the results of proteomics of CRC studies were summarized through a meta-analysis in order to obtain the diagnostic efficiency of novel markers. METHODS A systematic search on bibliographic databases was performed to collect the studies that explore blood-based markers for CRC applying proteomics. The detection and validation methods, as well as the specificity and sensitivity of the biomarkers in these studies, were evaluated. Newcastle- Ottawa Scale (NOS) case-control studies version was used for quality assessment of included studies. RESULTS Thirty-four studies were selected from 751 studies, in which markers detected by proteomics were summarized. In total, fifty-nine proteins were classified according to their biological function. The sensitivity, specificity, or AUC varied among these markers. Among them, Mammalian STE20-like protein kinase 1/ Serine threonine kinase 4 (MST1/STK4), S100 calcium-binding protein A9 (S100A9), and Tissue inhibitor of metalloproteinases 1 (TIMP1) were suitable for effect sizes merging, and their diagnostic efficiencies were recalculated after merging. MST1/STK4 obtained a sensitivity of 68% and a specificity of 78%. S100A9 achieved a sensitivity of 72%, a specificity of 83%, and an AUC of 0.88. TIMP1 obtained a sensitivity of 42%, a specificity of 88%, and an AUC of 0.71. CONCLUSION MST1/STK4, S100A9, and TIMP1 showed excellent performance for CRC detection. Several other markers also presented optimized diagnostic efficacy for CRC early detection, but further verification is still needed before they are suitable for clinical use. The discovering of more efficient markers will benefit CRC treatment.
Collapse
Affiliation(s)
- Xiang Chen
- The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Jiayu Sun
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Xue Wang
- Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Yumeng Yuan
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Leshan Cai
- The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Yanxuan Xie
- The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Zhiqiang Fan
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Kaixi Liu
- Shantou Central Hospital, Shantou, Guangdong 515041, China
| | - Xiaoyang Jiao
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong 515041, China
| |
Collapse
|
18
|
John A, Günes C, Bolenz C, Vidal-Y-Sy S, Bauer AT, Schneider SW, Gorzelanny C. Bladder cancer-derived interleukin-1 converts the vascular endothelium into a pro-inflammatory and pro-coagulatory surface. BMC Cancer 2020; 20:1178. [PMID: 33267794 PMCID: PMC7709388 DOI: 10.1186/s12885-020-07548-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 10/21/2020] [Indexed: 12/31/2022] Open
Abstract
Background Bladder cancer cells orchestrate tumour progression by pro-inflammatory cytokines. Cytokines modulate the local tumour microenvironment and increase the susceptibility of tumour distant tissues for metastasis. Here, we investigated the impact of human bladder cancer cell derived factors on the ability to modulate and activate human vascular endothelial cells. Methods The pro-inflammatory and pro-coagulatory potential of four different bladder cancer cell lines was accessed by qRT-PCR arrays and ELISA. Modulation and activation of endothelial cells was studied in microfluidic devices. Clinical relevance of our findings was confirmed by immune histology in tissue samples of bladder cancer patients and public transcriptome data. Results The unbalanced ratio between interleukin (IL)-1 and IL-1 receptor antagonist (IL-1ra) in the secretome of bladder cancer cells converted the quiescent vascular endothelium into a pro-adhesive, pro-inflammatory, and pro-coagulatory surface. Microfluidic experiments showed that tumour cell induced endothelial cell activation promoted leukocyte recruitment and platelet adhesion. Human bladder cancer tissue analysis confirmed that loss of IL-1ra and elevated IL-1 expression was associated with enhanced cancer progression. Conclusions Our data indicate that IL-1 and IL-1ra were dysregulated in bladder cancer and could facilitate tumour dissemination through endothelial cell activation. Targeting the IL-1/IL-1ra axis might attenuate tumour-mediated inflammation and metastasis formation. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-020-07548-z.
Collapse
Affiliation(s)
- A John
- Department of Urology, University of Ulm, Ulm, Germany
| | - C Günes
- Department of Urology, University of Ulm, Ulm, Germany
| | - C Bolenz
- Department of Urology, University of Ulm, Ulm, Germany
| | - S Vidal-Y-Sy
- Department of Dermatology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - A T Bauer
- Department of Dermatology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - S W Schneider
- Department of Dermatology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - C Gorzelanny
- Department of Dermatology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.
| |
Collapse
|
19
|
Wang J, Jiang N, Sang X, Yang N, Feng Y, Chen R, Wang X, Chen Q. Protein Modification Characteristics of the Malaria Parasite Plasmodium falciparum and the Infected Erythrocytes. Mol Cell Proteomics 2020; 20:100001. [PMID: 33517144 PMCID: PMC7857547 DOI: 10.1074/mcp.ra120.002375] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 11/04/2020] [Indexed: 12/14/2022] Open
Abstract
Malaria elimination is still pending on the development of novel tools that rely on a deep understanding of parasite biology. Proteins of all living cells undergo myriad posttranslational modifications (PTMs) that are critical to multifarious life processes. An extensive proteome-wide dissection revealed a fine PTM map of most proteins in both Plasmodium falciparum, the causative agent of severe malaria, and the infected red blood cells. More than two-thirds of proteins of the parasite and its host cell underwent extensive and dynamic modification throughout the erythrocytic developmental stage. PTMs critically modulate the virulence factors involved in the host-parasite interaction and pathogenesis. Furthermore, P. falciparum stabilized the supporting proteins of erythrocyte origin by selective demodification. Collectively, our multiple omic analyses, apart from having furthered a deep understanding of the systems biology of P. falciparum and malaria pathogenesis, provide a valuable resource for mining new antimalarial targets.
Collapse
Affiliation(s)
- Jianhua Wang
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Shenyang Agricultural University, Shengyang, China; The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang, China; College of Food Science, Shenyang Agricultural Sciences, Shenyang, China
| | - Ning Jiang
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Shenyang Agricultural University, Shengyang, China; The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang, China
| | - Xiaoyu Sang
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Shenyang Agricultural University, Shengyang, China; The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang, China
| | - Na Yang
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Shenyang Agricultural University, Shengyang, China; The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang, China
| | - Ying Feng
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Shenyang Agricultural University, Shengyang, China; The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang, China
| | - Ran Chen
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Shenyang Agricultural University, Shengyang, China; The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang, China
| | - Xinyi Wang
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Shenyang Agricultural University, Shengyang, China; College of Basic Sciences, Shenyang Agricultural University, Shenyang, China
| | - Qijun Chen
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Shenyang Agricultural University, Shengyang, China; The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang, China.
| |
Collapse
|
20
|
Ahn HS, Yeom J, Yu J, Kwon YI, Kim JH, Kim K. Convergence of Plasma Metabolomics and Proteomics Analysis to Discover Signatures of High-Grade Serous Ovarian Cancer. Cancers (Basel) 2020; 12:cancers12113447. [PMID: 33228226 PMCID: PMC7709037 DOI: 10.3390/cancers12113447] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/10/2020] [Accepted: 11/17/2020] [Indexed: 12/12/2022] Open
Abstract
Simple Summary In-time diagnosing ovarian cancer, intractable cancer that has no symptoms can increase the survival of women. The aim of this study was to discover biomarkers from liquid biopsy samples using multi-omics approach, metabolomics and proteomics for the diagnosis of ovarian cancer. To verify our biomarker candidates, we conducted comparative analysis with other previous published studies. Despite the limitations of non-invasive samples, our findings are able to discover emerging properties through the interplay between metabolites and proteins and mechanism-based biomarkers through integrated protein and metabolite analysis. Abstract The 5-year survival rate in the early and late stages of ovarian cancer differs by 63%. In addition, a liquid biopsy is necessary because there are no symptoms in the early stage and tissue collection is difficult without using invasive methods. Therefore, there is a need for biomarkers to achieve this goal. In this study, we found blood-based metabolite or protein biomarker candidates for the diagnosis of ovarian cancer in the 20 clinical samples (10 ovarian cancer patients and 10 healthy control subjects). Plasma metabolites and proteins were measured and quantified using mass spectrometry in ovarian cancer patients and control groups. We identified the differential abundant biomolecules (34 metabolites and 197 proteins) and statistically integrated molecules of different dimensions to better understand ovarian cancer signal transduction and to identify novel biological mechanisms. In addition, the biomarker reliability was verified through comparison with existing research results. Integrated analysis of metabolome and proteome identified emerging properties difficult to grasp with the single omics approach, more reliably interpreted the cancer signaling pathway, and explored new drug targets. Especially, through this analysis, proteins (PPCS, PMP2, and TUBB) and metabolites (L-carnitine and PC-O (30:0)) related to the carnitine system involved in cancer plasticity were identified.
Collapse
Affiliation(s)
- Hee-Sung Ahn
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Korea; (H.-S.A.); (J.Y.)
| | - Jeonghun Yeom
- Convergence Medicine Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Korea;
| | - Jiyoung Yu
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Korea; (H.-S.A.); (J.Y.)
| | | | - Jae-Hoon Kim
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06237, Korea
- Correspondence: (J.-H.K.); (K.K.); Tel.: +82-2-2019-3436 (J.-H.K.); +82-2-1688-7575 (K.K.)
| | - Kyunggon Kim
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Korea; (H.-S.A.); (J.Y.)
- Convergence Medicine Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Korea;
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul 05505, Korea
- Clinical Proteomics Core Laboratory, Convergence Medicine Research Center, Asan Medical Center, Seoul 05505, Korea
- Bio-Medical Institute of Technology, Asan Medical Center, Seoul 05505, Korea
- Correspondence: (J.-H.K.); (K.K.); Tel.: +82-2-2019-3436 (J.-H.K.); +82-2-1688-7575 (K.K.)
| |
Collapse
|
21
|
Yi S, Zhou W. Tumorigenesis-related key genes in adolescents and young adults with HR(+)/HER2(-) breast cancer. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2020; 13:2701-2709. [PMID: 33165441 PMCID: PMC7642711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 09/14/2020] [Indexed: 06/11/2023]
Abstract
Breast cancer (BC) in adolescents and young adults (AYAs) accounts for 5.6% of BC in all women. BC in this population is often characterized as aggressive. Two-thirds of BC in AYAs belong to the hormone-receptor positive (HR(+))/human epidermal growth factor receptor 2 negative (HER2(-)) subtype. However, the underlying pathogenesis of this subtype has not been fully elucidated. To study HR(+)/HER2(-) BC in AYAs, we downloaded the available RNA-seq data from The Cancer Genome Atlas (TCGA) database and then performed differential expression gene screening and constructed a protein-protein interaction (PPI) network, identified the key genes, and did gene set enrichment analysis (GSEA). Based on the analyses, 32.26% of patients were in stage III. Additionally, we identified 1671 differentially expressed genes (DEGs) and 35 key genes. In addition, GSEA showed that ether lipid metabolism and complement and coagulation cascades were significantly enriched in the GNAI1 high expression phenotype. The key genes CXCL2, CXCL5, CXCL3, GPR37L1, NPY2R, OXGR1, NPW, CCL21, GNAI1, SAA1, GRM4, HCAR2, CX3CL1, GRM8, CCL28, SSTR1, PENK, P2RY12, NMUR1, NMU, ADCY5, TAS1R1, OXER1, GNG13, CCL16, CCR8, NPY5R, CXCL11, CXCL10, CXCL9, CXCL1, CXCL6, CCR4, and ANXA1 may be molecular markers of tumorigenesis of HR(+)/HER2(-) BC in AYAs. In addition, ether lipid metabolism and complement and coagulation cascades may be key pathways for GNAI1 regulation in HR(+)/HER2(-) BC in AYAs.
Collapse
Affiliation(s)
- Shun Yi
- Department of Breast Surgery, The Affiliated Zhuzhou Hospital, Xiangya Medical College CSU Zhuzhou, Hunan, PR China
| | - Wei Zhou
- Department of Breast Surgery, The Affiliated Zhuzhou Hospital, Xiangya Medical College CSU Zhuzhou, Hunan, PR China
| |
Collapse
|
22
|
Zhang J, Chen M, Zhao Y, Xiong H, Sneh T, Fan Y, Wang J, Zhou X, Gong C. Complement and coagulation cascades pathway correlates with chemosensitivity and overall survival in patients with soft tissue sarcoma. Eur J Pharmacol 2020; 879:173121. [PMID: 32339514 DOI: 10.1016/j.ejphar.2020.173121] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/30/2020] [Accepted: 04/15/2020] [Indexed: 10/24/2022]
Abstract
Chemotherapy is an indispensable method in treatment of Soft tissue sarcomas (STS), but variability in sensitivity as a result of tumor heterogeneity is a key factor in determining patient outcome. Several studies have investigated the phenomenon of chemotherapy resistance in STS, while its precise complex mechanism is still unknown. This study aims to identify potential biomarkers for predicting the STS chemosensitivity, with the goal of both aiding patient treatment determination in the clinic and providing insight into key parts of the underlying mechanism. Gene expression profiles of 265 patients were obtained from The Cancer Genome Atlas dataset and differentially expressed genes (DEGs) associated with chemosensitivity were identified in groups of varying chemosensitivity, including 177 upregulated and 21 downregulated genes (P < 0.05). Then, DEGs were found to be enriched in complement and coagulation cascades and the osteoclast differentiation pathway. Protein-protein interaction analysis showed 15 genes (52 edges) enriched in the complement and coagulation cascades while 11 genes (28 edges) enriched in the osteoclast differentiation pathway. Notably, all the genes that significantly correlated to disease-free survival (DFS) and overall survival (OS), such as C1QC, C3AR1, C7, CFI and SERPINE1, are enriched in complement and coagulation cascades pathway. The differential expression of these genes was further verified by using the GSE database. Our findings support that C1QC, C3AR1, C7, CFI and SERPINE1 in the complement and coagulation cascade pathway are potential biomarkers for chemotherapy resistance and survival of STS patients.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Maoshan Chen
- Myeloma Research Group, Australian Centre for Blood Diseases (ACBD), Clinical Central School, Monash University, Melbourne, 3004, Australia
| | - Yuanyuan Zhao
- Department of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Huihua Xiong
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Tal Sneh
- Center for Molecular Design and Biomimetics, The Biodesign Institute, Arizona State University, Tempe, AZ, 85281, USA
| | - Yang Fan
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Jianhua Wang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Xiao Zhou
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Chen Gong
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China.
| |
Collapse
|
23
|
Saeidi J, Motaghipur R, Sepehrian A, Mohtashami M, Forooghi Nia F, Ghasemi A. Dietary fats promote inflammation in Wistar rats as well as induce proliferation, invasion of SKOV3 ovarian cancer cells. J Food Biochem 2020; 44:e13177. [PMID: 32157714 DOI: 10.1111/jfbc.13177] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 01/22/2020] [Accepted: 01/30/2020] [Indexed: 11/30/2022]
Abstract
The role of high fat diet (HFD) in ovarian cancer and its underlying mechanisms are poorly known. In current investigation, we investigated inflammatory and oncogenic effect of dietary fats in female Wistar rats and ovarian cancer cell line (SKOV3). The ELISA kits were used for adipokines and inflammatory factors analyses in sera collected from rats fed with high fat diet (SR-HFD). Cell growth, proliferation, apoptosis, migration, and invasion were measured in SKOV3 cells treated with the SR-HFD and FA mix. IL6, IL1β, TNFα, NF-kβ, and p53 expression were measured in cells incubated with the mentioned treatments. Leptin and inflammatory factors increased, while adiponectin decreased in SR-HFD. Moreover, FA mix significantly induced proliferation, migration, and invasion, promoted the expression of inflammatory factors and NF-κB and inhibited apoptosis markers in SKOV3 cells. Taken together, our findings revealed that diet might be a crucial factor in ovarian cancer progression through altering the inflammatory factors. PRACTICAL APPLICATIONS: The HFD-mediated obesity promotes cancer progression in various tissues. This study highlights the progression of inflammation in female Wistar rats and the growth of ovarian cancer cells by dietary fats. Thus, dietary factors can be considered as key factors for the prevention of ovarian cancer.
Collapse
Affiliation(s)
- Jafar Saeidi
- Department of Physiology, School of Basic Science, Neyshabur Branch, Islamic Azad University, Neyshabur, Iran
| | - Reza Motaghipur
- Department of Genetic, School of Basic Science, Neyshabur Branch, Islamic Azad University, Neyshabur, Iran
| | - Atefe Sepehrian
- Department of Genetic, School of Basic Science, Neyshabur Branch, Islamic Azad University, Neyshabur, Iran
| | - Mahnaz Mohtashami
- Department of Biology, School of Basic Science, Neyshabur Branch, Islamic Azad University, Neyshabur, Iran
| | - Fatemeh Forooghi Nia
- Department of Biology, School of Basic Science, Shiraz Branch, Islamic Azad University, Shiraz, Iran
| | - Ahmad Ghasemi
- Non-communicable Disease Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran.,Department of Basic Sciences, Neyshabur University of Medical Sciences, Neyshabur, Iran
| |
Collapse
|
24
|
Tavares V, Pinto R, Assis J, Pereira D, Medeiros R. Venous thromboembolism GWAS reported genetic makeup and the hallmarks of cancer: Linkage to ovarian tumour behaviour. Biochim Biophys Acta Rev Cancer 2020; 1873:188331. [DOI: 10.1016/j.bbcan.2019.188331] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 11/01/2019] [Accepted: 11/01/2019] [Indexed: 12/14/2022]
|
25
|
Chandra A, Pius C, Nabeel M, Nair M, Vishwanatha JK, Ahmad S, Basha R. Ovarian cancer: Current status and strategies for improving therapeutic outcomes. Cancer Med 2019; 8:7018-7031. [PMID: 31560828 PMCID: PMC6853829 DOI: 10.1002/cam4.2560] [Citation(s) in RCA: 212] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 08/22/2019] [Accepted: 08/24/2019] [Indexed: 12/14/2022] Open
Abstract
Of all the gynecologic tumors, ovarian cancer (OC) is known to be the deadliest. Advanced‐stages of OC are linked with high morbidity and low survival rates despite the immense amount of research in the field. Shortage of promising screening tools for early‐stage detection is one of the major challenges linked with the poor survival rate for patients with OC. In OC, therapeutic management is used with multidisciplinary approaches that includes debulking surgery, chemotherapy, and (rarely) radiotherapy. Recently, there is an increasing interest in using immunomodulation for treating OC. Relapse rates are high in this malignancy and averages around every 2‐years. Further treatments after the relapse are more intense, increasing the toxicity, resistance to chemotherapy drugs, and financial burden to patients with poor quality‐of‐life. A procedure that has been studied to help reduce the morbidity rate involves pre‐sensitizing cancer cells with standard therapy in order to produce optimal results with minimum dosage. Utilizing such an approach, platinum‐based agents are effective due to their increased response to platinum‐based chemotherapy in relapsed cases. These chemo‐drugs also help address the issue of drug resistance. After conducting an extensive search with available literature and the resources for clinical trials, information is precisely documented on current research, biomarkers, options for treatment and clinical trials. Several schemes for enhancing the therapeutic responses for OC are discussed systematically in this review with an attempt in summarizing the recent developments in this exciting field of translational/clinical research.
Collapse
Affiliation(s)
- Ashwin Chandra
- Texas College of Osteopathic Medicine, UNT Health Science Center, Fort Worth, TX, USA
| | - Cima Pius
- Miami Medical School, Miami, FL, USA
| | - Madiha Nabeel
- Graduate School of Biomedical Sciences, UNT Health Science Center, Fort Worth, TX, USA
| | - Maya Nair
- Graduate School of Biomedical Sciences, UNT Health Science Center, Fort Worth, TX, USA
| | - Jamboor K Vishwanatha
- Graduate School of Biomedical Sciences, UNT Health Science Center, Fort Worth, TX, USA
| | | | - Riyaz Basha
- Texas College of Osteopathic Medicine, UNT Health Science Center, Fort Worth, TX, USA.,Graduate School of Biomedical Sciences, UNT Health Science Center, Fort Worth, TX, USA
| |
Collapse
|
26
|
Matsuoka A, Mizumoto Y, Ono M, Kagami K, Obata T, Terakawa J, Maida Y, Nakamura M, Daikoku T, Fujiwara H. Novel strategy of ovarian cancer implantation: Pre-invasive growth of fibrin-anchored cells with neovascularization. Cancer Sci 2019; 110:2658-2666. [PMID: 31199029 PMCID: PMC6676109 DOI: 10.1111/cas.14098] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 05/23/2019] [Accepted: 05/27/2019] [Indexed: 01/23/2023] Open
Abstract
Although direct adhesion of cancer cells to the mesothelial cell layer is considered to be a key step for peritoneal invasion of ovarian cancer cell masses (OCM), we recently identified a different strategy for the peritoneal invasion of OCM. In 6 out of 20 cases of ovarian carcinoma, extraperitoneal growth of the OCM was observed along with the neovascularization of feeding vessels, which connect the intraperitoneal host stroma and extraperitoneal lesions through the intact mesothelial cell layer. As an early step, the OCMs anchor in the extraperitoneal fibrin networks and then induce the migration of CD34-positive and vascular endothelial growth factor A (VEGF-A)-positive endothelial cells, constructing extraperitoneal vascular networks around the OCM. During the extraperitoneal growth of OCM, podoplanin-positive and α smooth muscle actin (αSMA)-positive cancer-associated fibroblasts (CAF) appears. In more advanced lesions, the boundary line of mesothelial cells disappears around the insertion areas of feeding vessels and then extraperitoneal and intraperitoneal stroma are integrated, enabling the OCM to invade the host stroma, being associated with CAF. In addition, tissue factors (TF) are strongly detected around these peritoneal implantation sites and their levels in ascites were higher than that in blood. These findings demonstrate the presence of neovascularization around fibrin net-anchored OCMs on the outer side of the intact peritoneal surface, suggesting a novel strategy for peritoneal invasion of ovarian cancer and TF-targeted intraperitoneal anti-cancer treatment. We observed and propose a novel strategy for peritoneal implantation of ovarian cancer. The strategy includes the preinvasive growth of fibrin-anchored cancer cells along with neovascularization on the outer side of the intact peritoneal surface.
Collapse
Affiliation(s)
- Ayumi Matsuoka
- Department of Obstetrics and GynecologyGraduate School of Medical SciencesKanazawa UniversityKanazawaJapan
| | - Yasunari Mizumoto
- Department of Obstetrics and GynecologyGraduate School of Medical SciencesKanazawa UniversityKanazawaJapan
| | - Masanori Ono
- Department of Obstetrics and GynecologyGraduate School of Medical SciencesKanazawa UniversityKanazawaJapan
| | - Kyosuke Kagami
- Department of Obstetrics and GynecologyGraduate School of Medical SciencesKanazawa UniversityKanazawaJapan
| | - Takeshi Obata
- Department of Obstetrics and GynecologyGraduate School of Medical SciencesKanazawa UniversityKanazawaJapan
| | - Junpei Terakawa
- Institute for Experimental AnimalsAdvanced Science Research CenterKanazawa UniversityKanazawaJapan
| | - Yoshiko Maida
- Department of NursingCollege of Medical, Pharmaceutical, and Health SciencesKanazawa UniversityKanazawaJapan
| | - Mitsuhiro Nakamura
- Department of Obstetrics and GynecologyGraduate School of Medical SciencesKanazawa UniversityKanazawaJapan
| | - Takiko Daikoku
- Institute for Experimental AnimalsAdvanced Science Research CenterKanazawa UniversityKanazawaJapan
| | - Hiroshi Fujiwara
- Department of Obstetrics and GynecologyGraduate School of Medical SciencesKanazawa UniversityKanazawaJapan
| |
Collapse
|
27
|
Kawakami E, Tabata J, Yanaihara N, Ishikawa T, Koseki K, Iida Y, Saito M, Komazaki H, Shapiro JS, Goto C, Akiyama Y, Saito R, Saito M, Takano H, Yamada K, Okamoto A. Application of Artificial Intelligence for Preoperative Diagnostic and Prognostic Prediction in Epithelial Ovarian Cancer Based on Blood Biomarkers. Clin Cancer Res 2019; 25:3006-3015. [DOI: 10.1158/1078-0432.ccr-18-3378] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 01/08/2019] [Accepted: 02/18/2019] [Indexed: 12/20/2022]
|
28
|
Wang Q, Yu C, Shi S, Su X, Zhang J, Ding Y, Sun Y, Liu M, Li C, Zhao X, Jiang W, Wei T. An analysis of plasma reveals proteins in the acute phase response pathway to be candidate diagnostic biomarkers for depression. Psychiatry Res 2019; 272:404-410. [PMID: 30611956 DOI: 10.1016/j.psychres.2018.11.069] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 10/14/2018] [Accepted: 11/30/2018] [Indexed: 12/28/2022]
Abstract
Globally, depression is one of the most serious debilitating psychiatric mental disorders. In this study, we validated the expression levels of fibrinogen alpha (FGA), fibrinogen beta (FGB), fibrinogen gamma (FGG), Complement factor B (CFB) and serpin family D member 1(SERPIND1) in the acute phase response signaling pathway in plasma samples using enzyme-linked immunosorbent assay (ELISA).Then illuminate the roles of FGA, FGB, FGG, CFB, SERPIND1 in depression using microarray data. Gene expression dataset GSE98793 was downloaded from the Gene Expression Omnibus database. There were 128 whole blood samples included 64 patients with major depressed patients and 64 healthy controls. Differentially expressed genes (DEGs) were identified, and then protein-protein interaction (PPI) network was constructed to screen crucial genes associated with FGA, FGB, FGG, CFB and SERPIND1. Moreover, gene ontology (GO) biological processes analyses was performed. The ELISA data showed that the expression levels of FGA, FGB, FGG, CFB and SERPIND1 were up-regulated in depressed patients. The enriched GO terms were predominantly associated with the biological processes including more genes were inflammation related. The PPI network was found these five genes interacted with 11 genes. FGA, FGB, FGG, CFB and SERPIND1 may be important in the pathogenesis of depression.
Collapse
Affiliation(s)
- Qi Wang
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University-Daqing, Daqing, Heilongjiang 163000, PR China
| | - Chunyue Yu
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University-Daqing, Daqing, Heilongjiang 163000, PR China
| | - Shanshan Shi
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University-Daqing, Daqing, Heilongjiang 163000, PR China
| | - Xiaojie Su
- Department of Biochemistry and molecular biology, College of Medical laboratory and technology, Harbin Medical University-Daqing, Daqing, Heilongjiang 163000, PR China
| | - Jian Zhang
- College of Medical Informatics, Harbin Medical University-Daqing, Daqing, Heilongjiang 163000, PR China
| | - Yongqing Ding
- Department of Women's Psychological Clinic, Fifth Affiliated Hospital of Harbin Medical University, Daqing, Heilongjiang 163000, PR China
| | - Yanan Sun
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University-Daqing, Daqing, Heilongjiang 163000, PR China
| | - Min Liu
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University-Daqing, Daqing, Heilongjiang 163000, PR China
| | - Chunquan Li
- College of Medical Informatics, Harbin Medical University-Daqing, Daqing, Heilongjiang 163000, PR China
| | - Xiwu Zhao
- Department of Neurology, The Third People's Hospital of Daqing, Daqing, Heilongjiang 163000, PR China
| | - Wenhai Jiang
- Department of Neurology, The Third People's Hospital of Daqing, Daqing, Heilongjiang 163000, PR China
| | - Taiming Wei
- Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University-Daqing, Daqing, Heilongjiang 163000, PR China.
| |
Collapse
|
29
|
Peltier J, Roperch JP, Audebert S, Borg JP, Camoin L. Activation peptide of the coagulation factor XIII (AP-F13A1) as a new biomarker for the screening of colorectal cancer. Clin Proteomics 2018; 15:15. [PMID: 29657559 PMCID: PMC5890357 DOI: 10.1186/s12014-018-9191-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 04/02/2018] [Indexed: 01/06/2023] Open
Abstract
Background Colorectal cancer (CRC) remains a major cause of cancer fatalities in developed countries. The risk of death is correlated to the stage of CRC during the primary diagnosis. Early diagnosis is closely associated with enhanced survival rate. We therefore investigated the AP-F13A1 as a potential protein marker of CRC. Methods The protein expression of FXIII in 40 serum samples was evaluated by enzyme-linked immunosorbent assays. Additionally, targeted proteomic assays (LC-PRM) were used to evaluate the expression of the activation peptide of F13A1 (AP-F13A1) in a further 113 serum samples. Results were analyzed by the Wilcoxon test and receiver operating characteristic curves generated to assess statistical differences and diagnostic factors between CRC patients and controls. Results AP-F13A1 was quantified in human serum samples using calibration curves with excellent linearity. AP-F13A1 was reduced in CRC patients using PRM assays from two distinct biobanks. The AUC for AP-F13A1 were 0.95 and 0.93. Sensitivity/specificity values for the two sets of patients were 75%/95% and 71%/95% respectively. Conclusion We have presented the proof of principle that in vivo release of AP-F13A1 can be measured by PRM-based strategies in CRC serum samples. AP-F13A1 may be an effective serological biomarker as part of a screening program of CRC detection. Electronic supplementary material The online version of this article (10.1186/s12014-018-9191-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Julien Peltier
- 1Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille Protéomique, Marseille, France, Aix Marseille Univ, 27 Bd Leï Roure, BP30059, 13273 Marseille Cedex 9, France.,PROFILOME SAS, 24 Rue du Faubourg Saint-Jacques, 75014 Paris, France.,3Present Address: Institute for Cell and Molecular Biosciences, Faculty of Medical Sciences, Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle Upon Tyne, NE2 4HH UK
| | - Jean-Pierre Roperch
- PROFILOME SAS, 24 Rue du Faubourg Saint-Jacques, 75014 Paris, France.,Present Address: OncoDiag, Pépinière scientifique, Rue de Pacy, Miserey, France
| | - Stéphane Audebert
- 1Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille Protéomique, Marseille, France, Aix Marseille Univ, 27 Bd Leï Roure, BP30059, 13273 Marseille Cedex 9, France
| | - Jean-Paul Borg
- 1Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille Protéomique, Marseille, France, Aix Marseille Univ, 27 Bd Leï Roure, BP30059, 13273 Marseille Cedex 9, France
| | - Luc Camoin
- 1Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille Protéomique, Marseille, France, Aix Marseille Univ, 27 Bd Leï Roure, BP30059, 13273 Marseille Cedex 9, France
| |
Collapse
|
30
|
Xu WY, Zhang HH, Yang XB, Bai Y, Lin JZ, Long JY, Xiong JP, Zhang JW, Sang XT, Zhao HT. Prognostic significance of combined preoperative fibrinogen and CA199 in gallbladder cancer patients. World J Gastroenterol 2018; 24:1451-1463. [PMID: 29632426 PMCID: PMC5889825 DOI: 10.3748/wjg.v24.i13.1451] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 03/07/2018] [Accepted: 03/10/2018] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate the prognostic value of the combination of preoperative plasma fibrinogen and CA199 in patients with gallbladder carcinoma (GBC).
METHODS The clinicopathological data of 154 GBC patients were retrospectively reviewed after surgery. A receiver operating characteristic (ROC) curve was plotted to verify the optimum cut-off values for plasma fibrinogen and CA199. Univariate and multivariate survival analyses were performed to identify the factors associated with GBC prognosis. Based on the HRs calculated via multivariate survival analyses, patients with elevated plasma fibrinogen and CA199 levels were allocated a score of 2.1; those with an elevated plasma fibrinogen level only were allocated a score of 1, those with an elevated CA199 level only were allocated a score of 1.1, and those with neither of these abnormalities were allocated a score of 0.
RESULTS ROC curve analysis showed that the optimum cut-off values for preoperative plasma fibrinogen and CA199 were 3.47 g/L and 25.45 U/mL, respectively. Multivariate analysis indicated that elevated preoperative plasma fibrinogen and CA199 levels were significantly correlated with worse overall survival (OS) (HR = 1.711, 95%CI: 1.114-2.627, P = 0.014, and HR = 1.842, 95%CI: 1.111-3.056, P = 0.018). When we combined these two parameters, the area under the ROC curve increased from 0.735 (for preoperative plasma fibrinogen only) and 0.729 (for preoperative CA199 only) to 0.765. When this combined variable was added to the multivariate analysis, the combination of plasma fibrinogen and CA199 (P < 0.001), resection margin (P < 0.001) and TNM stage (P = 0.010) were independent prognostic factors for GBC.
CONCLUSION The combination of plasma fibrinogen and CA199 may serve as a more efficient independent prognostic biomarker for postoperative GBC patients than either parameter alone.
Collapse
Affiliation(s)
- Wei-Yu Xu
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Hao-Hai Zhang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Xiao-Bo Yang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Yi Bai
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Jian-Zhen Lin
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Jun-Yu Long
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Jian-Ping Xiong
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Jun-Wei Zhang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Xin-Ting Sang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Hai-Tao Zhao
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
31
|
Bekos C, Grimm C, Brodowicz T, Petru E, Hefler L, Reimer D, Koch H, Reinthaller A, Polterauer S, Polterauer M. Prognostic role of plasma fibrinogen in patients with uterine leiomyosarcoma - a multicenter study. Sci Rep 2017; 7:14474. [PMID: 29101329 PMCID: PMC5670177 DOI: 10.1038/s41598-017-13934-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 10/03/2017] [Indexed: 02/07/2023] Open
Abstract
Fibrinogen has an important pathophysiological role in tumor cell progression and development of metastases in different types of cancer. The present study aimed to evaluate the role of pre-treatment fibrinogen plasma concentrations as a biomarker for tumor biology and prognosis in patients with uterine leiomyosarcoma (ULMS). Clinical data of patients with ULMS were assessed in this multi-center study Pre-therapeutic fibrinogen plasma concentrations were evaluated. We investigated the association between fibrinogen plasma levels and clinico-pathological parameters and performed univariate and multivariable survival analyses. In total, 70 women with ULMS were included into the analysis. Mean (SD) pre-treatment fibrinogen plasma levels were 480.2 (172.3) mg/dL. Patients with advanced tumor stage, increased tumor size and higher histological grading had higher fibrinogen levels (p = 0.02, p = 0.013, and p = 0.029, respectively). In ULMS patients with increased fibrinogen levels 5-year overall survival (OS) rates were 25.0% compared to 52.9% in ULMS patients with normal fibrinogen, respectively. Univariate survival analyses revealed that elevated fibrinogen plasma levels (p = 0.030), advanced tumor stage (p < 0.001) and undifferentiated histology (p = 0.003) showed association with unfavorable OS. In multivariable analysis, histological grade (p = 0.03) and tumor stage (0.02) were independently associated with survival. Elevated fibrinogen plasma levels were associated with aggressive tumor biology and poor prognosis in women with ULMS. Fibrinogen might be useful as a novel biomarker in ULMS.
Collapse
Affiliation(s)
- Christine Bekos
- Department of General Gynaecology and Gynaecological Oncology, Gynecologic Cancer Unit - Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Christoph Grimm
- Department of General Gynaecology and Gynaecological Oncology, Gynecologic Cancer Unit - Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Thomas Brodowicz
- Clinical Division of Oncology, Department of Medicine 1, Comprehensive Cancer Center - Medical University Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Edgar Petru
- Department of Obstetrics and Gynaecology of the Medical University of Graz, Graz, Austria
| | - Lukas Hefler
- Department of Gynaecology, Barmherzige Schwestern Hospital Linz, Linz, Austria
| | - Daniel Reimer
- Department of Obstetrics and Gynaecology of the Medical University of Innsbruck, Tirol, Austria
| | - Horst Koch
- Department of Obstetrics and Gynaecology of the Medical University of Salzburg, Salzburg, Austria
| | - Alexander Reinthaller
- Department of General Gynaecology and Gynaecological Oncology, Gynecologic Cancer Unit - Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.,Karl Landsteiner Institute for General Gynecology and Experimental Gynecologic Oncology, Vienna, Austria
| | - Stephan Polterauer
- Department of General Gynaecology and Gynaecological Oncology, Gynecologic Cancer Unit - Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria. .,Karl Landsteiner Institute for General Gynecology and Experimental Gynecologic Oncology, Vienna, Austria.
| | - Mariella Polterauer
- Department of General Gynaecology and Gynaecological Oncology, Gynecologic Cancer Unit - Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| |
Collapse
|
32
|
Thuwajit C, Ferraresi A, Titone R, Thuwajit P, Isidoro C. The metabolic cross-talk between epithelial cancer cells and stromal fibroblasts in ovarian cancer progression: Autophagy plays a role. Med Res Rev 2017; 38:1235-1254. [PMID: 28926101 PMCID: PMC6032948 DOI: 10.1002/med.21473] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 08/16/2017] [Accepted: 08/23/2017] [Indexed: 12/17/2022]
Abstract
Cancer and stromal cells, which include (cancer‐associated) fibroblasts, adipocytes, and immune cells, constitute a mixed cellular ecosystem that dynamically influences the behavior of each component, creating conditions that ultimately favor the emergence of malignant clones. Ovarian cancer cells release cytokines that recruit and activate stromal fibroblasts and immune cells, so perpetuating a state of inflammation in the stroma that hampers the immune response and facilitates cancer survival and propagation. Further, the stroma vasculature impacts the metabolism of the cells by providing or limiting the availability of oxygen and nutrients. Autophagy, a lysosomal catabolic process with homeostatic and prosurvival functions, influences the behavior of cancer cells, affecting a variety of processes such as the survival in metabolic harsh conditions, the invasive growth, the development of immune and chemo resistance, the maintenance of stem‐like properties, and dormancy. Further, autophagy is involved in the secretion and the signaling of promigratory cytokines. Cancer‐associated fibroblasts can influence the actual level of autophagy in ovarian cancer cells through the secretion of pro‐inflammatory cytokines and the release of autophagy‐derived metabolites and substrates. Interrupting the metabolic cross‐talk between cancer cells and cancer‐associated fibroblasts could be an effective therapeutic strategy to arrest the progression and prevent the relapse of ovarian cancer.
Collapse
Affiliation(s)
- Chanitra Thuwajit
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Alessandra Ferraresi
- Laboratory of Molecular Pathology and Nanobioimaging, Department of Health Sciences, Università del Piemonte Orientale "A. Avogadro", Novara, Italy
| | - Rossella Titone
- Laboratory of Molecular Pathology and Nanobioimaging, Department of Health Sciences, Università del Piemonte Orientale "A. Avogadro", Novara, Italy
| | - Peti Thuwajit
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Ciro Isidoro
- Laboratory of Molecular Pathology and Nanobioimaging, Department of Health Sciences, Università del Piemonte Orientale "A. Avogadro", Novara, Italy.,Visiting Professor at Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
33
|
Lee S, Huh SJ, Oh SY, Koh MS, Kim SH, Lee JH, Han JY, Choi HJ, Kim SJ, Kim HJ. Clinical significance of coagulation factors in operable colorectal cancer. Oncol Lett 2017; 13:4669-4674. [PMID: 28599468 PMCID: PMC5452961 DOI: 10.3892/ol.2017.6058] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 02/17/2017] [Indexed: 12/27/2022] Open
Abstract
Abnormal hemostasis in cancer patients has prev iously been studied. The primary objective of the present study was to evaluate the association between preoperative hemostasis markers and clinicopathological parameters, and to identify a hemostasis marker affecting survival in patients following curative resection for colorectal cancer. A total of 170 patients who underwent curative surgery for colorectal carcinoma were evaluated. Preoperative coagulation tests included platelet, prothrombin time (PT), activated partial thromboplastin time (aPTT), fibrinogen, D-dimer and fibrinogen degradation product (FDP). The clinicopathological variables, including age, gender, tumor location (rectum/colon), tumor size (≥5 cm vs. <5 cm), depth of tumor invasion, lymph node metastasis, stage, lymphovascular invasion, margin involvement and histological differentiation were analyzed. The median age of analyzed patients was 63 years (range, 28-84). The male to female ratio was 62:38. Increased levels of plasma fibrinogen, PT and platelet count (PLT) were associated with larger tumor size (P<0.001, P=0.015 and P=0.002, respectively). Increased plasma fibrinogen levels were significantly associated with depth of tumor invasion and stage (P=0.014 and P=0.048, respectively). Increased plasma D-dimer and FDP levels were significantly associated with tumor node metastasis stage (P=0.031 and P=0.002, respectively). Prolonged PT level (≥11.7 sec), hyper-fibrinogenemia (≥327 mg/dl), high D-dimer level (≥1.3 µg/ml) and increased FDP level (≥2.7 µg/ml) were the prognostic factors associated with shorter survival. Preoperative plasma fibrinogen level was significantly associated with tumor size and depth of tumor invasion. Preoperative plasma prolonged PT level, hyperfibrinogenemia, high D-dimer level and increased FDP level may function as hemostasis markers that predict overall survival in operable patients with colorectal cancer.
Collapse
Affiliation(s)
- Suee Lee
- Department of Internal Medicine, Dong-A University College of Medicine, Busan 49201, Republic of Korea
| | - Seok Jae Huh
- Department of Internal Medicine, Dong-A University College of Medicine, Busan 49201, Republic of Korea
| | - Sung Yong Oh
- Department of Internal Medicine, Dong-A University College of Medicine, Busan 49201, Republic of Korea
| | - Myeong Seok Koh
- Department of Internal Medicine, Dong-A University College of Medicine, Busan 49201, Republic of Korea
| | - Sung-Hyun Kim
- Department of Internal Medicine, Dong-A University College of Medicine, Busan 49201, Republic of Korea
| | - Ji Hyun Lee
- Department of Internal Medicine, Dong-A University College of Medicine, Busan 49201, Republic of Korea
| | - Jin Young Han
- Department of Laboratory Medicine, Dong-A University College of Medicine, Busan 49201, Republic of Korea
| | - Hong Jo Choi
- Department of Surgery, Dong-A University College of Medicine, Busan 49201, Republic of Korea
| | - Su Jin Kim
- Department of Pathology, Dong-A University College of Medicine, Busan 49201, Republic of Korea
| | - Hyo-Jin Kim
- Department of Internal Medicine, Dong-A University College of Medicine, Busan 49201, Republic of Korea
| |
Collapse
|
34
|
Zhou Z, Zeng F, Yuan J, Tang J, Colditz GA, Tworoger SS, Trabert B, Su X. Pelvic inflammatory disease and the risk of ovarian cancer: a meta-analysis. Cancer Causes Control 2017; 28:415-428. [PMID: 28342087 DOI: 10.1007/s10552-017-0873-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 02/14/2017] [Indexed: 02/04/2023]
Abstract
PURPOSE Previous studies on pelvic inflammatory disease (PID) and the risk of ovarian cancer have found inconsistent results. We performed an updated meta-analysis to summarize the evidence of this association. METHODS PubMed, Embase, and ISI web of science databases were searched through October 2016 for studies that investigated the PID and ovarian cancer association. Summary risk estimates were calculated using random-effects meta-analysis. RESULT Thirteen studies were eligible for analysis, which included six cohort studies and seven case-control studies. PID was associated with an increased risk of ovarian cancer overall [relative risk (RR) 1.24, 95% CI 1.06-1.44; I 2 = 58.8%]. In analyses stratified by race, a significant positive association was observed in studies conducted among Asian women (RR 1.69, 95% CI 1.22-2.34; I 2 = 0%), but marginally significant among Caucasians (RR 1.18, 95% CI 1.00-1.39; I 2 = 60.7%).Risk estimates were elevated in both cohort (RR1.32; 95% CI 1.05-1.66; I 2 = 64.7%) and case-control studies (RR 1.17; 95% CI 0.93-1.49; I 2 = 57.6%), albeit not statistically significant in case-control studies. CONCLUSIONS Our results suggested that PID might be a potential risk factor of ovarian cancer, with pronounced associations among Asian women. Large and well-designed studies with objective assessment methods, such as hospital records, are needed to confirm the findings of this meta-analysis.
Collapse
Affiliation(s)
- Zhiyi Zhou
- Faculty of Medicine, School of Public Health and Primary Care, The Chinese University of Hong Kong, Room 508, 5/F, School of Public Health, Prince of Wales Hospital, Shatin, N.T., Hong Kong, China.,Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Fangfang Zeng
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jianhui Yuan
- Shenzhen Center for Disease Control and Prevention, Shenzhen, China
| | - Jinling Tang
- Faculty of Medicine, School of Public Health and Primary Care, The Chinese University of Hong Kong, Room 508, 5/F, School of Public Health, Prince of Wales Hospital, Shatin, N.T., Hong Kong, China.,The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, 518057, China
| | - Graham A Colditz
- Alvin J. Siteman Cancer Center and Department of Surgery, Washington University School of Medicine, 660 S Euclid Ave, St Louis, MO, 63110, USA
| | - Shelley S Tworoger
- Channing Division of Network Medicine, Harvard Medical School and the Brigham and Women's Hospital, 181 Longwood Ave., 3rd Fl., Boston, MA, 02115, USA.,Department of Epidemiology, Harvard T. H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA, 02115, USA
| | - Britton Trabert
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, NCI Shady Grove, 9609 Medical Center Drive, MSC 9774, Bethesda, MD, 20892, USA.
| | - Xuefen Su
- Faculty of Medicine, School of Public Health and Primary Care, The Chinese University of Hong Kong, Room 508, 5/F, School of Public Health, Prince of Wales Hospital, Shatin, N.T., Hong Kong, China. .,The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, 518057, China.
| |
Collapse
|
35
|
Luo Y, Kim HS, Kim M, Lee M, Song YS. Elevated plasma fibrinogen levels and prognosis of epithelial ovarian cancer: a cohort study and meta-analysis. J Gynecol Oncol 2017; 28:e36. [PMID: 28382799 PMCID: PMC5391395 DOI: 10.3802/jgo.2017.28.e36] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 01/06/2017] [Accepted: 02/08/2017] [Indexed: 01/02/2023] Open
Abstract
Objective To evaluate the effect of elevated plasma fibrinogen levels on the prognosis of epithelial ovarian cancer (EOC). Methods We reviewed the data of 217 patients with advanced-stage EOC between 2000 and 2012, and investigated the prognostic role of elevated plasma fibrinogen levels compared with serum CA-125 levels, neutrophil to lymphocyte ratio (NLR) and platelet to lymphocyte ratio (PLR). For further evaluation, we performed a meta-analysis using 5 cohort studies published to July 2015, including our cohort study after a literature review. Results Among the four biomarkers, only plasma fibrinogen levels >485.2 mg/dL were correlated with impaired progression-free survival (PFS) and overall survival (OS) (median, 13.9 vs. 20.3 months and 42.2 vs. 55.4 months; p<0.010). Elevated plasma fibrinogen levels were an independent factor for poor PFS with marginal significance and OS (adjusted hazard ratios [HRs]=1.389 and 1.581; 95% confidence intervals [CIs]=0.979–1.972 and 1.032–2.423, respectively). Furthermore, crude and subgroup meta-analyses demonstrated that elevated plasma fibrinogen levels were associated with impaired PFS and OS in patients with all stage EOC. Conclusion Elevated plasma fibrinogen levels be more important for predicting survival than serum CA-125 levels, NLR and PLR in patients with EOC, in particular, advanced-stage disease. Moreover, it may be related to poor prognosis of EOC.
Collapse
Affiliation(s)
- Yanlin Luo
- Department of Gynecologic Oncology, Affiliated Cancer Hospital of Zhengzhou University (Henan Cancer Hospital), Zhengzhou, China
| | - Hee Seung Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| | - Miseon Kim
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seoungnam, Korea
| | - Maria Lee
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| | - Yong Sang Song
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
36
|
Ebrahimi S, Rahmani F, Behnam-Rassouli R, Hoseinkhani F, Parizadeh MR, Keramati MR, Khazaie M, Avan A, Hassanian SM. Proinflammatory signaling functions of thrombin in cancer. J Cell Physiol 2017; 232:2323-2329. [PMID: 28004386 DOI: 10.1002/jcp.25753] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 12/20/2016] [Indexed: 12/19/2022]
Abstract
Thrombin-induced activation of protease-activated receptors (PARs) represents a link between inflammation and cancer. Proinflammatory signaling functions of thrombin are associated with several inflammatory diseases including neurodegenerative, cardiovascular, and of special interest in this review cancer. Thrombin-induced inflammatory responses up-regulates expression of cytokines, adhesion molecules, angiogenic factors, and matrix-degrading proteases that facilitate tumor cells proliferation, angiogenesis, invasion, and metastasis. This review summarizes the current knowledge about the mechanisms of thrombin-mediated proinflammatory responses in cancer pathology for a better understanding and hence a better management of this disease.
Collapse
Affiliation(s)
- Safieh Ebrahimi
- Department of Medical Biochemistry, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farzad Rahmani
- Department of Medical Biochemistry, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Fatemeh Hoseinkhani
- Department of Medical Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Parizadeh
- Department of Medical Biochemistry, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Metabolic Syndrome Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Reza Keramati
- Cancer Molecular Pathology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Khazaie
- Department of Medical Physiology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Metabolic Syndrome Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Molecular Medicine Group, Department of Modern Sciences and Technologies, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Department of Medical Biochemistry, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Metabolic Syndrome Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Microanatomy Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
37
|
Basha R, Mohiuddin Z, Rahim A, Ahmad S. Ovarian Cancer and Resistance to Therapies: Clinical and Laboratory Perspectives. DRUG RESISTANCE IN BACTERIA, FUNGI, MALARIA, AND CANCER 2017:511-537. [DOI: 10.1007/978-3-319-48683-3_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
38
|
Alexander ET, Minton AR, Peters MC, van Ryn J, Gilmour SK. Thrombin inhibition and cisplatin block tumor progression in ovarian cancer by alleviating the immunosuppressive microenvironment. Oncotarget 2016; 7:85291-85305. [PMID: 27852034 PMCID: PMC5356737 DOI: 10.18632/oncotarget.13300] [Citation(s) in RCA: 27] [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: 05/27/2016] [Accepted: 10/26/2016] [Indexed: 12/11/2022] Open
Abstract
Cancer is often associated with an increased risk of thrombotic complications which can be aggravated by treatment with chemotherapeutics such as cisplatin. Multiple lines of evidence suggest that thrombin activity promotes tumor growth and metastasis. We examined the effect of co-treatment with dabigatran etexilate, a direct thrombin inhibitor, and cisplatin using the murine ID8 ovarian cancer model. Mice receiving co-treatment with both dabigatran etexilate and low dose cisplatin had significantly smaller tumors, developed less ascites and had lower levels of circulating activated platelets and tissue factor (TF) positive microparticles than those treated with dabigatran etexilate or cisplatin alone. Co-treatment with dabigatran etexilate and cisplatin significantly decreased the number of Gr1+/CD11b+ myeloid derived suppresser cells and CD11b+/CD11c+ dendritic cells in the ascites of ID8 tumor-bearing mice. Co-treatment also significantly reduced levels of pro-tumorigenic cytokines including TGF-β, VEGF, IL-6, IL-10, and MCP-1 in the ascites while increasing IFN-γ production by CD8+ effector T cells in the tumor ascites. These results demonstrate that co-treatment with dabigatran etexilate significantly augments the anti-tumor activity of cisplatin in ovarian tumor progression by alleviating the immunosuppressive microenvironment, suggesting that thrombin may be a potential therapeutic target for treatment of ovarian cancer.
Collapse
Affiliation(s)
| | | | - Molly C. Peters
- Lankenau Institute for Medical Research, Wynnewood, PA 19096, USA
| | - Joanne van Ryn
- Boehringer Ingelheim Pharma GmbH & Co. KG, 88397 Biberach an der Riss, Germany
| | - Susan K. Gilmour
- Lankenau Institute for Medical Research, Wynnewood, PA 19096, USA
| |
Collapse
|
39
|
Swier N, Versteeg HH. Reciprocal links between venous thromboembolism, coagulation factors and ovarian cancer progression. Thromb Res 2016; 150:8-18. [PMID: 27988375 DOI: 10.1016/j.thromres.2016.12.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 11/23/2016] [Accepted: 12/03/2016] [Indexed: 02/06/2023]
Abstract
Ovarian cancer is the most lethal gynecological malignancy, which is due to late presentation. Treating advanced stage ovarian cancer is difficult, and tumor recurrence and chemoresistance frequently occur. In addition, early detection remains a major challenge as there are no early warning signs and no appropriate biomarkers. To reduce mortality rates of ovarian cancer patients, novel drug targets and biomarkers are needed. We postulate that hemostatic keyplayers are of importance when combatting ovarian cancer. The majority of ovarian cancer patients have abnormal hemostatic blood serum marker levels, which indicate an activated coagulation system. This makes patients more prone to experiencing venous thromboembolism (VTE), and the occurrence of VTE in ovarian cancer patients adversely affects survival. Coagulation activation also promotes tumor progression as it influences tumor biology at several stages and the decreased survival rates associated with ovarian cancer-associated thrombosis are more likely due to cancer metastasis rather than to fatal thromboembolic events. In this review, we will discuss; (1) Population studies that address the bidirectional relationship between VTE and ovarian cancer, and the most important risk factors involved; (2) The mechanisms of coagulation factors and platelets that are critically involved in the development of VTE, and the progression of ovarian cancer; (3) Roles and future directions of coagulation factors in ovarian cancer therapy, and in diagnosis and prognosis of ovarian cancer as biomarkers.
Collapse
Affiliation(s)
- Nathalie Swier
- Department of Internal Medicine, Thrombosis and Hemostasis Division, Leiden University Medical Center, Albinusdreef 2, 2333, ZA, Leiden, The Netherlands
| | - Henri H Versteeg
- Department of Internal Medicine, Thrombosis and Hemostasis Division, Leiden University Medical Center, Albinusdreef 2, 2333, ZA, Leiden, The Netherlands.
| |
Collapse
|
40
|
Jammal MP, Martins-Filho A, Silveira TP, Murta EFC, Nomelini RS. Cytokines and Prognostic Factors in Epithelial Ovarian Cancer. Clin Med Insights Oncol 2016; 10:71-6. [PMID: 27512342 PMCID: PMC4973765 DOI: 10.4137/cmo.s38333] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Revised: 07/07/2016] [Accepted: 07/09/2016] [Indexed: 12/19/2022] Open
Abstract
INTRODUCTION Ovarian cancer has a high mortality and delayed diagnosis. Inflammation is a risk factor for ovarian cancer, and the inflammatory response is involved in almost all stages of tumor development. Immunohistochemical staining in stroma and epithelium of a panel of cytokines in benign and malignant ovarian neoplasm was evaluated. In addition, immunostaining was related to prognostic factors in malignant tumors. METHOD The study group comprised 28 ovarian benign neoplasias and 28 ovarian malignant neoplasms. A panel of cytokines was evaluated by immunohistochemistry (Th1: IL-2 and IL-8; Th2: IL-5, IL-6, and IL-10; and TNFR1). Chi-square test with Yates' correction was used, which was considered significant if less than 0.05. RESULTS TNFR1, IL-5, and IL-10 had more frequent immunostaining 2/3 in benign neoplasms compared with malignant tumors. Malignant tumors had more frequent immunostaining 2/3 for IL-2 in relation to benign tumors. The immunostaining 0/1 of IL 8 was more frequent in the stroma of benign neoplasms compared with malignant neoplasms. Evaluation of the ovarian cancer stroma showed that histological grade 3 was significantly correlated with staining 2/3 for IL-2 (P = 0.004). Women whose disease-free survival was less than 2.5 years had TNFR1 stromal staining 2/3 (P = 0.03) more frequently. CONCLUSION IL-2 and TNFR1 stromal immunostaining are related prognostic factors in ovarian cancer and can be the target of new therapeutic strategies.
Collapse
Affiliation(s)
- Millena Prata Jammal
- Research Institute of Oncology (IPON)/Discipline of Gynecology and Obstetrics, Federal University of Triângulo Mineiro, Uberaba, MG, Brazil
| | - Agrimaldo Martins-Filho
- Research Institute of Oncology (IPON)/Discipline of Gynecology and Obstetrics, Federal University of Triângulo Mineiro, Uberaba, MG, Brazil
| | - Thales Parenti Silveira
- Discipline of Special Pathology, Federal University of Triângulo Mineiro, Uberaba, MG, Brazil
| | - Eddie Fernando Candido Murta
- Research Institute of Oncology (IPON)/Discipline of Gynecology and Obstetrics, Federal University of Triângulo Mineiro, Uberaba, MG, Brazil
| | - Rosekeila Simões Nomelini
- Research Institute of Oncology (IPON)/Discipline of Gynecology and Obstetrics, Federal University of Triângulo Mineiro, Uberaba, MG, Brazil
| |
Collapse
|
41
|
Berger MD, Heini AD, Seipel K, Mueller B, Angelillo-Scherrer A, Pabst T. Increased fibrinogen levels at diagnosis are associated with adverse outcome in patients with acute myeloid leukemia. Hematol Oncol 2016; 35:789-796. [DOI: 10.1002/hon.2307] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 04/11/2016] [Accepted: 04/21/2016] [Indexed: 11/06/2022]
Affiliation(s)
- Martin D. Berger
- Department of Medical Oncology; University Hospital and University of Berne; Berne Switzerland
| | - Alexander D. Heini
- Department of Medical Oncology; University Hospital and University of Berne; Berne Switzerland
| | - Katja Seipel
- Department of Clinical Research; University Hospital and University of Berne; Berne Switzerland
| | - Beatrice Mueller
- Department of Clinical Research; University Hospital and University of Berne; Berne Switzerland
| | | | - Thomas Pabst
- Department of Medical Oncology; University Hospital and University of Berne; Berne Switzerland
| |
Collapse
|
42
|
Feng Z, Wen H, Bi R, Duan Y, Yang W, Wu X. Thrombocytosis and hyperfibrinogenemia are predictive factors of clinical outcomes in high-grade serous ovarian cancer patients. BMC Cancer 2016; 16:43. [PMID: 26817451 PMCID: PMC4730624 DOI: 10.1186/s12885-016-2070-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 01/18/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Over 20% of ovarian cancer patients have preoperative thrombocytosis or hyperfibrinogenemia. We aimed to demonstrate the clinical and prognostic significance of thrombocytosis and hyperfibrinogenemia in high-grade serous ovarian cancer (HGSC). METHODS We retrospectively investigated HGSC patients who underwent primary staging or debulking surgery between April 2005 and June 2013 in our institution. None of these patients had received neoadjuvant chemotherapy. Data, including age, performance status, FIGO stage, serum CA125, platelet count, fibrinogen level, and surgical residual disease, were collected. Thrombocytosis was defined as a platelet count greater than 450 × 10(9)/L, and hyperfibrinogenemia was defined as a fibrinogen level higher than 4.00 g/L. Progression-free survival (PFS) and overall survival (OS) were analyzed with the Kaplan-Meier method and log-rank tests for univariate analyses. For the multivariate analyses, Cox regression analysis was used to evaluate the effects of the prognostic factors, which are expressed as hazard ratios (HRs). RESULTS A total of 875 consecutive HGSC patients were identified. The median follow-up time was 29 (1-115) months. The median (interquartile range, IQR) preoperative platelet count was 301 (235-383) × 10(9)/L, and 121 (13.8%) women had thrombocytosis. The median (IQR) preoperative fibrinogen level was 3.85 (3.19-4.45) g/L, and 332 (45.9%) of the patients had hyperfibrinogenemia. Both preoperative thrombocytosis and hyperfibrinogenemia were associated with an advanced FIGO stage (p = 0.008 and <0.001, respectively), an increased CA125 level (p = 0.004 and 0.001, respectively), more extensive ascites (p < 0.001 and <0.001, respectively), more extensive residual disease (p < 0.001 and <0.001, respectively) and chemosensitivity (p = 0.043 and <0.001, respectively). In the univariate analyses, hyperfibrinogenemia was associated with reduced PFS (p < 0.001) and OS (p < 0.001). However, thrombocytosis was not found to be a potential predictor of PFS (P = 0.098) or OS (p = 0.894). In the multivariate analyses, hyperfibrinogenemia was an independent predictor of OS (p = 0.014) but not PFS (p = 0.062). CONCLUSION Preoperative thrombocytosis and hyperfibrinogenemia reflected tumor burden to some extent and thus influenced treatment outcomes, and the fibrinogen level was found to be useful as a prognostic predictor in the HGSC patients.
Collapse
Affiliation(s)
- Zheng Feng
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, 270 Dong-an Road, Shanghai, 200032, China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Hao Wen
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, 270 Dong-an Road, Shanghai, 200032, China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Rui Bi
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China. .,Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
| | - Yachen Duan
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, 270 Dong-an Road, Shanghai, 200032, China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Wentao Yang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China. .,Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
| | - Xiaohua Wu
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, 270 Dong-an Road, Shanghai, 200032, China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
43
|
Li H, Zhao T, Ji X, Liang S, Wang Z, Yang Y, Yin J, Wang R. Hyperfibrinogenemia predicts poor prognosis in patients with advanced biliary tract cancer. Tumour Biol 2015; 37:3535-42. [PMID: 26453118 DOI: 10.1007/s13277-015-4184-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 10/01/2015] [Indexed: 12/11/2022] Open
Abstract
Hyperfibrinogenemia reportedly predicts poor prognosis in several cancers but has not been reviewed for biliary tract cancer (BTC). The aim of the present study was to evaluate associations between baseline plasma fibrinogen concentrations, clinicopathological characteristics, and survival parameters in patients with BTC. Data for 127 patients with BTC diagnosed at the Zhongshan Affiliated Hospital of Dalian University (Liaoning, China) from January 2011 to December 2014 were retrospectively evaluated. Associations between baseline fibrinogen concentrations, selected clinicopathological characteristics, and the prognostic value were examined using SPSS software. Data for 37 patients (29.1 % of study cohort) who had undergone curative intent surgery and 90 (70.9 %) with advanced biliary tract cancer (ABTC) were analyzed. The mean plasma fibrinogen concentration 4.0 ± 0.9 g/L for the entire cohort. The percentages with hyperfibrinogenemia (>4 g/L) were 45.7, 37.8, and 48.9 % overall and in the surgical and ABTC groups, respectively. Hyperfibrinogenemia was associated with performance status (PS) and neutrophil/lymphocyte ratio in the entire cohort but not with other relevant clinicopathological factors. Log-rank test indicated that baseline hyperfibrinogenemia was associated with decreased progression-free survival (PFS) and overall survival (OS) for patients with unresectable ABTC (P > 0.05). Multivariate analysis showed that poor PS and baseline hyperfibrinogenemia were independently associated with worse survival (HR: 1.39, 95 % CI: 1.02-1.90, P = 0.04; HR: 1.75.95 %, 95 % CI: 1.01-3.01, P = 0.04, respectively). Baseline hyperfibrinogenemia is an independent predictor of poor prognosis in patients with ABTC. Baseline plasma fibrinogen concentrations may be a readily available and inexpensive prognostic biomarker in patients with ABTC; this needs further validation in large prospective clinical trials.
Collapse
Affiliation(s)
- Heming Li
- Department of Oncology, Zhongshan Hospital of Dalian University, No. 6, Liberation Street, Zhongshan District, Dalian, 116001, People's Republic of China
| | - Tong Zhao
- Department of Oncology, Zhongshan Hospital of Dalian University, No. 6, Liberation Street, Zhongshan District, Dalian, 116001, People's Republic of China
| | - Xuening Ji
- Department of Oncology, Zhongshan Hospital of Dalian University, No. 6, Liberation Street, Zhongshan District, Dalian, 116001, People's Republic of China
| | - Shanshan Liang
- Department of Oncology, Zhongshan Hospital of Dalian University, No. 6, Liberation Street, Zhongshan District, Dalian, 116001, People's Republic of China
| | - Zhe Wang
- Department of Oncology, Zhongshan Hospital of Dalian University, No. 6, Liberation Street, Zhongshan District, Dalian, 116001, People's Republic of China
| | - Yulong Yang
- Department of Oncology, Zhongshan Hospital of Dalian University, No. 6, Liberation Street, Zhongshan District, Dalian, 116001, People's Republic of China
| | - Jiajun Yin
- Department of Oncology, Zhongshan Hospital of Dalian University, No. 6, Liberation Street, Zhongshan District, Dalian, 116001, People's Republic of China
| | - Ruoyu Wang
- Department of Oncology, Zhongshan Hospital of Dalian University, No. 6, Liberation Street, Zhongshan District, Dalian, 116001, People's Republic of China.
| |
Collapse
|
44
|
Awan FM, Naz A, Obaid A, Ali A, Ahmad J, Anjum S, Janjua HA. Identification of Circulating Biomarker Candidates for Hepatocellular Carcinoma (HCC): An Integrated Prioritization Approach. PLoS One 2015; 10:e0138913. [PMID: 26414287 PMCID: PMC4586137 DOI: 10.1371/journal.pone.0138913] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 09/06/2015] [Indexed: 12/14/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the world's third most widespread cancer. Currently available circulating biomarkers for this silently progressing malignancy are not sufficiently specific and sensitive to meet all clinical needs. There is an imminent and pressing need for the identification of novel circulating biomarkers to increase disease-free survival rate. In order to facilitate the selection of the most promising circulating protein biomarkers, we attempted to define an objective method likely to have a significant impact on the analysis of vast data generated from cutting-edge technologies. Current study exploits data available in seven publicly accessible gene and protein databases, unveiling 731 liver-specific proteins through initial enrichment analysis. Verification of expression profiles followed by integration of proteomic datasets, enriched for the cancer secretome, filtered out 20 proteins including 6 previously characterized circulating HCC biomarkers. Finally, interactome analysis of these proteins with midkine (MDK), dickkopf-1 (DKK-1), current standard HCC biomarker alpha-fetoprotein (AFP), its interacting partners in conjunction with HCC-specific circulating and liver deregulated miRNAs target filtration highlighted seven novel statistically significant putative biomarkers including complement component 8, alpha (C8A), mannose binding lectin (MBL2), antithrombin III (SERPINC1), 11β-hydroxysteroid dehydrogenase type 1 (HSD11B1), alcohol dehydrogenase 6 (ADH6), beta-ureidopropionase (UPB1) and cytochrome P450, family 2, subfamily A, polypeptide 6 (CYP2A6). Our proposed methodology provides a swift assortment process for biomarker prioritization that eventually reduces the economic burden of experimental evaluation. Further dedicated validation studies of potential putative biomarkers on HCC patient blood samples are warranted. We hope that the use of such integrative secretome, interactome and miRNAs target filtration approach will accelerate the selection of high-priority biomarkers for other diseases as well, that are more amenable to downstream clinical validation experiments.
Collapse
Affiliation(s)
- Faryal Mehwish Awan
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12 Islamabad, Pakistan
| | - Anam Naz
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12 Islamabad, Pakistan
| | - Ayesha Obaid
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12 Islamabad, Pakistan
| | - Amjad Ali
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12 Islamabad, Pakistan
| | - Jamil Ahmad
- Research Center for Modeling and Simulation (RCMS), National University of Sciences and Technology (NUST), H-12 Islamabad, Pakistan
| | - Sadia Anjum
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12 Islamabad, Pakistan
| | - Hussnain Ahmed Janjua
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12 Islamabad, Pakistan
| |
Collapse
|
45
|
Jin D, Lee H. A computational approach to identifying gene-microRNA modules in cancer. PLoS Comput Biol 2015; 11:e1004042. [PMID: 25611546 PMCID: PMC4303261 DOI: 10.1371/journal.pcbi.1004042] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 11/16/2014] [Indexed: 11/21/2022] Open
Abstract
MicroRNAs (miRNAs) play key roles in the initiation and progression of various cancers by regulating genes. Regulatory interactions between genes and miRNAs are complex, as multiple miRNAs can regulate multiple genes. In addtion, these interactions vary from patient to patient and even among patients with the same cancer type, as cancer development is a heterogeneous process. These relationships are more complicated because transcription factors and other regulatory molecules can also regulate miRNAs and genes. Hence, it is important to identify the complex relationships between genes and miRNAs in cancer. In this study, we propose a computational approach to constructing modules that represent these relationships by integrating the expression data of genes and miRNAs with gene-gene interaction data. First, we used a biclustering algorithm to construct modules consisting of a subset of genes and a subset of samples to incorporate the heterogeneity of cancer cells. Second, we combined gene-gene interactions to include genes that play important roles in cancer-related pathways. Then, we selected miRNAs that are closely associated with genes in the modules based on a Gaussian Bayesian network and Bayesian Information Criteria. When we applied our approach to ovarian cancer and glioblastoma (GBM) data sets, 33 and 54 modules were constructed, respectively. In these modules, 91% and 94% of ovarian cancer and GBM modules, respectively, were explained either by direct regulation between genes and miRNAs or by indirect relationships via transcription factors. In addition, 48.4% and 74.0% of modules from ovarian cancer and GBM, respectively, were enriched with cancer-related pathways, and 51.7% and 71.7% of miRNAs in modules were ovarian cancer-related miRNAs and GBM-related miRNAs, respectively. Finally, we extensively analyzed significant modules and showed that most genes in these modules were related to ovarian cancer and GBM. A microRNA (miRNA) is a small RNA molecule that regulates the expression of mRNA genes. A miRNA can regulate multiple genes, and a gene can be regulated by multiple miRNAs. The regulation of genes by miRNAs may vary from patient to patient, even if they suffer from the same type of cancer. In this study, we identify the relationships between genes and miRNAs in cancer patients using expression data. Because these relationships are complicated by the involvement of transcription factors, which are among the most influential regulators of genes, we also attempt to explain the triple relationship among genes, miRNAs, and transcription factors. We constructed modules consisting of a set of genes and miRNAs, in which the expression levels are highly correlated. In most of these modules, genes and miRNAs are related to specific cancer types; their relationships are explained both by direct regulation of genes by miRNAs and by indirect relationships via transcription factors.
Collapse
Affiliation(s)
- Daeyong Jin
- School of Information and Communications, Gwangju Institute of Science and Technology, Gwangju, South Korea
| | - Hyunju Lee
- School of Information and Communications, Gwangju Institute of Science and Technology, Gwangju, South Korea
- * E-mail:
| |
Collapse
|
46
|
Leszczak V, Popat KC. Improved in vitro blood compatibility of polycaprolactone nanowire surfaces. ACS APPLIED MATERIALS & INTERFACES 2014; 6:15913-24. [PMID: 25184556 PMCID: PMC4173746 DOI: 10.1021/am503508r] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 09/03/2014] [Indexed: 05/27/2023]
Abstract
There are a multitude of polymeric materials currently utilized to prepare a variety of blood-contacting implantable medical devices. These devices include tissue grafts, coronary artery and vascular stents, and orthopedic implants. The thrombogenic nature of such materials can cause serious complications in patients, and ultimately lead to functional failure. To date, there is no truly hemocompatible biomaterial surface. Nanostructured surfaces improve cellular interactions but there is a limited amount of information regarding their blood compatibility. In this study, the in vitro blood compatibility of four different surfaces (control, PCL; nanowire, NW; collagen immobilized control, cPCL; collagen immobilized nanowire, cNW) were investigated for their use as interfaces for blood-contacting implants. The results presented here indicate enhanced in vitro blood compatibility of nanowire surfaces compared control surfaces. Although there were no significant differences in leukocyte adhesion, there was a decrease in platelet adhesion on NW surfaces. Scanning electron microscopy images showed a decrease in platelet/leukocyte complexes on cNW surfaces and no apparent complexes were formed on NW surfaces compared to PCL and cPCL surfaces. The increase in these complexes likely contributed to a higher expression of specific markers for platelet and leukocyte activation on PCL and cPCL surfaces. No significant differences were found in contact and complement activation on any surface. Further, thrombin antithrombin complexes were significantly reduced on NW surfaces. A significant increase in hemolysis and fibrinogen adsorption was identified on PCL surfaces likely caused by its hydrophobic surface. This work shows the improved blood-compatibility of nanostructured surfaces, identifying this specific nanoarchitecture as a potential interface for promoting the long-term success of blood-contacting biomaterials.
Collapse
Affiliation(s)
- Victoria Leszczak
- Department
of Mechanical Engineering, School of Biomedical Engineering, Colorado State University, Fort
Collins, Colorado 80523, United States
| | - Ketul C. Popat
- Department
of Mechanical Engineering, School of Biomedical Engineering, Colorado State University, Fort
Collins, Colorado 80523, United States
| |
Collapse
|
47
|
The impact of plasma Epstein-Barr virus DNA and fibrinogen on nasopharyngeal carcinoma prognosis: an observational study. Br J Cancer 2014; 111:1102-11. [PMID: 25051405 PMCID: PMC4453843 DOI: 10.1038/bjc.2014.393] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 06/04/2014] [Accepted: 06/18/2014] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The impact of combining plasma fibrinogen levels with Epstein-Barr Virus DNA (EBV DNA) levels on the prognosis for patients with nasopharyngeal carcinoma (NPC) was evaluated. METHODS In this observational study, 2563 patients with non-metastatic NPC were evaluated for the effects of circulating plasma fibrinogen and EBV DNA levels on disease-free survival (DFS), distant metastasis-free survival (DMFS), and overall survival (OS). RESULTS Compared with the bottom biomarker tertiles, TNM stage-adjusted hazard ratios (HR, 95% confidence intervals (CIs)) for predicting DFS in fibrinogen tertiles 2 to 3 were 1.26 (1.00 to 1.60) and 1.81 (1.45 to 2.26), respectively; HR for EBV DNA tertiles 2 to 3 were 1.49 (1.12 to 1.98) and 4.24 (3.27 to 5.49), respectively. After additional adjustment for established risk factors, both biomarkers were still associated (P for trend <0.001) with reduced DFS (HR: 1.79, 95% CI, 1.43 to 2.25 for top fibrinogen tertiles; HR: 4.04, 95% CI: 3.10 to 5.27 for top EBV DNA tertiles compared with the bottom tertiles). For patients with advanced-stage disease, those with high fibrinogen levels (3.34 g l(-1)) presented with worse DFS, regardless of EBV DNA 4000 or <4000 copies ml(-1) subgroup. Similar findings were observed for DMFS and OS. CONCLUSIONS Circulating fibrinogen and EBV DNA significantly correlate with NPC patients survival. Combined fibrinogen and EBV DNA data lead to improved prognostic prediction in advanced-stage disease.
Collapse
|
48
|
Kristjánsdóttir B, Partheen K, Fung ET, Yip C, Levan K, Sundfeldt K. Early inflammatory response in epithelial ovarian tumor cyst fluids. Cancer Med 2014; 3:1302-12. [PMID: 24947406 PMCID: PMC4302680 DOI: 10.1002/cam4.282] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2014] [Revised: 05/06/2014] [Accepted: 05/07/2014] [Indexed: 01/21/2023] Open
Abstract
Mortality rates for epithelial ovarian cancer (EOC) are high, mainly due to late-stage diagnosis. The identification of biomarkers for this cancer could contribute to earlier diagnosis and increased survival rates. Given that chronic inflammation plays a central role in cancer initiation and progression, we selected and tested 15 cancer-related cytokines and growth factors in 38 ovarian cyst fluid samples. We used ovarian cyst fluid since it is found in proximity to the pathology and mined it for inflammatory biomarkers suitable for early detection of EOC. Immunoprecipitation and high-throughput sample fractionation were obtained by using tandem antibody libraries bead and mass spectrometry. Two proteins, monocyte chemoattractant protein-1 (MCP-1/CCL2) and interleucin-8 (IL-8/CXCL8), were significantly (P < 0.0001) higher in the malignant (n = 16) versus benign (n = 22) tumor cysts. Validation of MCP-1, IL-8, and growth-regulated protein-α (GROα/CXCL1) was performed with ELISA in benign, borderline, and malignant cyst fluids (n = 256) and corresponding serum (n = 256). CA125 was measured in serum from all patients and used in the algorithms performed. MCP-1, IL-8, and GROα are proinflammatory cytokines and promoters of tumor growth. From 5- to 100-fold higher concentrations of MCP-1, IL-8 and GROα were detected in the cyst fluids compared to the serum. Significant (P < 0.001) cytokine response was already established in borderline cyst fluids and stage I EOC. In serum a significant (P < 0.01) increase of IL-8 and GROα was found, but not until stage I and stage III EOC, respectively. These findings confirm that early events in tumorigenesis can be analyzed and detected in the tumor environment and we conclude that ovarian cyst fluid is a promising source in the search for new biomarkers for early ovarian tumors.
Collapse
Affiliation(s)
- Björg Kristjánsdóttir
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences, University of Gothenburg, Gothenburg, Sweden
| | | | | | | | | | | |
Collapse
|
49
|
Abstract
Growing understanding of the role of thrombocytosis, high platelet turnover, and the presence of activated platelets in the circulation in cancer progression and metastasis has brought megakaryocytes into focus. Platelet biology is essential to hemostasis, vascular integrity, angiogenesis, inflammation, innate immunity, wound healing, and cancer biology. However, before megakaryocyte/platelet-directed therapies can be considered for clinical use, understanding of the mechanism and biology of paraneoplastic thrombocytosis in malignancy is required. Here, we provide an overview of the clinical implications, biological significance, and mechanisms of paraneoplastic thrombocytosis in the context of ovarian cancer.
Collapse
Affiliation(s)
- Ashley N Davis
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Vahid Afshar-Kharghan
- Department of Benign Hematology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX; Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TTX; Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX.
| |
Collapse
|
50
|
Kuan AS, Teng CJ, Wu HH, Su VYF, Chen YT, Chien SH, Yeh CM, Hu LY, Chen TJ, Tzeng CH, Liu CJ. Risk of ischemic stroke in patients with ovarian cancer: a nationwide population-based study. BMC Med 2014; 12:53. [PMID: 24661584 PMCID: PMC4022213 DOI: 10.1186/1741-7015-12-53] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 02/25/2014] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Cancer patients are at risk of thromboembolism. However, studies investigating the relationship between ovarian cancer and ischemic stroke are lacking. The objectives of this study were to assess the association between ovarian cancer and ischemic stroke, and to determine the predictive risk factors. METHODS Ovarian cancer patients aged 20 years and older without antecedent cerebrovascular events and who were followed up for more than 1 year between 1 January 2003 and 31 December 2011 were recruited from the Taiwan National Health Insurance database. Hazard ratios (HRs) of stroke risk for ovarian cancer patients compared with an age- and comorbidity-matched cohort were calculated by Cox proportional regression analysis. The difference in cumulative ischemic stroke incidence between ovarian cancer patients and the matched cohort was analyzed with the Kaplan-Meier method and tested with the log-rank test. RESULTS Each cohort (ovarian cancer and matched cohort) consisted of 8,810 individuals, with a median age of 49 years. After a median follow-up of 2.68 and 3.85 years, respectively, the ischemic stroke incidence was 1.38-fold higher in the ovarian cancer cohort than in the comparison cohort (9.4 versus 6.8 per 1,000 person-years), with an age- and comorbidity-adjusted HR of 1.49 (P <0.001). The ischemic stroke risk imposed by ovarian cancer was more prominent in patients under 50 years old (HR 2.28; P <0.001) compared with patients 50 years and older (HR 1.33; P = 0.005). Significant risk factors predicting stroke development were age 50 years and older (HR 2.21; P <0.001), hypertension (HR 1.84; P <0.001), diabetes mellitus (HR 1.71; P <0.001), and treatment with chemotherapy (HR 1.45; P = 0.017), especially platinum-based regimens. CONCLUSIONS Ovarian cancer patients were at an increased risk of developing ischemic stroke. Age, hypertension, diabetes, and chemotherapy treatment were independent risk factors.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Chia-Jen Liu
- School of Medicine, National Yang-Ming University, Taipei, Taiwan.
| |
Collapse
|