14951
|
Intarajak T, Udomchaiprasertkul W, Bunyoo C, Yimnoon J, Soonklang K, Wiriyaukaradecha K, Lamlertthon W, Sricharunrat T, Chaiwiriyawong W, Siriphongpreeda B, Sutheeworapong S, Kusonmano K, Kittichotirat W, Thammarongtham C, Jenjaroenpun P, Wongsurawat T, Nookaew I, Auewarakul C, Cheevadhanarak S. Genetic Aberration Analysis in Thai Colorectal Adenoma and Early-Stage Adenocarcinoma Patients by Whole-Exome Sequencing. Cancers (Basel) 2019; 11:E977. [PMID: 31336886 PMCID: PMC6679221 DOI: 10.3390/cancers11070977] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 05/31/2019] [Accepted: 06/03/2019] [Indexed: 02/06/2023] Open
Abstract
Colorectal adenomas are precursor lesions of colorectal adenocarcinoma. The transition from adenoma to carcinoma in patients with colorectal cancer (CRC) has been associated with an accumulation of genetic aberrations. However, criteria that can screen adenoma progression to adenocarcinoma are still lacking. This present study is the first attempt to identify genetic aberrations, such as the somatic mutations, copy number variations (CNVs), and high-frequency mutated genes, found in Thai patients. In this study, we identified the genomic abnormality of two sample groups. In the first group, five cases matched normal-colorectal adenoma-colorectal adenocarcinoma. In the second group, six cases matched normal-colorectal adenomas. For both groups, whole-exome sequencing was performed. We compared the genetic aberration of the two sample groups. In both normal tissues compared with colorectal adenoma and colorectal adenocarcinoma analyses, somatic mutations were observed in the tumor suppressor gene APC (Adenomatous polyposis coli) in eight out of ten patients. In the group of normal tissue comparison with colorectal adenoma tissue, somatic mutations were also detected in Catenin Beta 1 (CTNNB1), Family With Sequence Similarity 123B (FAM123B), F-Box And WD Repeat Domain Containing 7 (FBXW7), Sex-Determining Region Y-Box 9 (SOX9), Low-Density Lipoprotein Receptor-Related Protein 5 (LRP5), Frizzled Class Receptor 10 (FZD10), and AT-Rich Interaction Domain 1A (ARID1A) genes, which are involved in the Wingless-related integration site (Wnt) signaling pathway. In the normal tissue comparison with colorectal adenocarcinoma tissue, Kirsten retrovirus-associated DNA sequences (KRAS), Tumor Protein 53 (TP53), and Ataxia-Telangiectasia Mutated (ATM) genes are found in the receptor tyrosine kinase-RAS (RTK-RAS) signaling pathway and p53 signaling pathway, respectively. These results suggest that APC and TP53 may act as a potential screening marker for colorectal adenoma and early-stage CRC. This preliminary study may help identify patients with adenoma and early-stage CRC and may aid in establishing prevention and surveillance strategies to reduce the incidence of CRC.
Collapse
Affiliation(s)
- Thoranin Intarajak
- Bioinformatics and Systems Biology Program, School of Bioresources and Technology and School of Information Technology, King Mongkut's University of Technology Thonburi, Bangkok 10150, Thailand
- Bioinformatics Unit for Genomic Analysis, Division of Research and International Relations, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok 10210, Thailand
- Systems Biology and Bioinformatics Research Group, Pilot Plant Development and Training Institute, King Mongkut's University of Technology Thonburi, Bangkok 10150, Thailand
| | - Wandee Udomchaiprasertkul
- Molecular Biology and Genomic Laboratory, Division of Research and International Relations, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok 10210, Thailand
| | - Chakrit Bunyoo
- Bioinformatics Unit for Genomic Analysis, Division of Research and International Relations, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok 10210, Thailand
| | - Jutamas Yimnoon
- Cytogenetics Unit, Central Research Laboratory, Division of Research and International Relations, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok 10210, Thailand
| | - Kamonwan Soonklang
- Data Management Unit, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok 10210, Thailand
| | - Kriangpol Wiriyaukaradecha
- Molecular Biology and Genomic Laboratory, Division of Research and International Relations, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok 10210, Thailand
| | - Wisut Lamlertthon
- Faculty of Medicine and Public Health, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok 10210, Thailand
| | - Thaniya Sricharunrat
- Pathology Laboratory Unit, Chulabhorn Hospital, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok 10210, Thailand
| | - Worawit Chaiwiriyawong
- Department of Medical Oncology, Chulabhorn Hospital, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok 10210, Thailand
| | - Bunchorn Siriphongpreeda
- Faculty of Medicine and Public Health, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok 10210, Thailand
| | - Sawannee Sutheeworapong
- Bioinformatics and Systems Biology Program, School of Bioresources and Technology and School of Information Technology, King Mongkut's University of Technology Thonburi, Bangkok 10150, Thailand
- Systems Biology and Bioinformatics Research Group, Pilot Plant Development and Training Institute, King Mongkut's University of Technology Thonburi, Bangkok 10150, Thailand
| | - Kanthida Kusonmano
- Bioinformatics and Systems Biology Program, School of Bioresources and Technology and School of Information Technology, King Mongkut's University of Technology Thonburi, Bangkok 10150, Thailand
- Systems Biology and Bioinformatics Research Group, Pilot Plant Development and Training Institute, King Mongkut's University of Technology Thonburi, Bangkok 10150, Thailand
- School of Bioresources and Technology, King Mongkut's University of Technology Thonburi, Bangkok 10150, Thailand
| | - Weerayuth Kittichotirat
- Bioinformatics and Systems Biology Program, School of Bioresources and Technology and School of Information Technology, King Mongkut's University of Technology Thonburi, Bangkok 10150, Thailand
- Systems Biology and Bioinformatics Research Group, Pilot Plant Development and Training Institute, King Mongkut's University of Technology Thonburi, Bangkok 10150, Thailand
| | - Chinae Thammarongtham
- Biochemical Engineering and Systems Biology research group, National Center for Genetic Engineering and Biotechnology (BIOTEC) at King Mongkut's University of Technology Thonburi, Bangkhuntien, Bangkok 10150, Thailand
| | - Piroon Jenjaroenpun
- Department of Biomedical Informatics, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Thidathip Wongsurawat
- Department of Biomedical Informatics, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Intawat Nookaew
- Department of Biomedical Informatics, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Department of Biology and Biological Engineering, Chalmers University of Technology, SE-412 96 Gothenburg, Sweden
- Department of Physiology and Biophysics, College of Medicine, The University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Chirayu Auewarakul
- Faculty of Medicine and Public Health, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok 10210, Thailand.
| | - Supapon Cheevadhanarak
- Systems Biology and Bioinformatics Research Group, Pilot Plant Development and Training Institute, King Mongkut's University of Technology Thonburi, Bangkok 10150, Thailand.
- School of Bioresources and Technology, King Mongkut's University of Technology Thonburi, Bangkok 10150, Thailand.
| |
Collapse
|
14952
|
Huang M, Yan C, Xiao J, Wang T, Ling R. Relevance and clinicopathologic relationship of BRAF V600E, TERT and NRAS mutations for papillary thyroid carcinoma patients in Northwest China. Diagn Pathol 2019; 14:74. [PMID: 31300059 PMCID: PMC6626378 DOI: 10.1186/s13000-019-0849-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 06/20/2019] [Indexed: 01/05/2023] Open
Abstract
Background To determine the relevance of the single or combination mutations of BRAF V600E, TERT, and NRAS genes and the clinicopathologic relationship in papillary thyroid cancer (PTC). Methods Patients with PTC were enrolled into the study between February 2018 and April 2019. Based on the number of mutant genes, we classified the participants into single BRAF V600E mutation group, double mutations group and no mutation group. Single factor and multiple logistic regression analyses were applied to explore the independent factors. Review Manager 5.3 was used for meta-analysis to review the clinical efficacy of gene co-mutations. Results Finally, 483 patients were enrolled into the study and 419 (86.7%) of them harbored BRAF V600E mutation. TERT or NRAS mutation was likely to coexist with BRAF V600E mutation in PTC. BRAF V600E and NRAS promoter co-mutations was identified in 6 patients, with a prevalence of 1.2%. Prevalence of BRAF V600E and TERT coexistence in PTC was 2.1%. Significant differences were found among age, pathology, multifocality, bilateral lesions, lymph node metastasis, and 131I radiotherapy, P < 0.01. Multiple logistic regression analyses demonstrated that age [odds ratio (OR) = 1.044, 95% confidence interval (CI) = 1.013–1.076; P = 0.006], lymph node metastasis [OR = 0.094, 95% CI = 0.034–0.264; P < 0.001], 131I radiotherapy [OR = 7.628, 95% CI = 2.721–21.378; P < 0.001] were risk factors for BRAF V600E mutation. Besides, age [OR = 1.135, 95% CI = 1.069–1.205; P < 0.001], multiple leisions [OR = 4.128, 95% CI = 1.026–16.614; P = 0.046], pathology [OR = 3.954, 95% CI = 1.235–12.654; P = 0.021] were independent factors for combination mutations. Meta-analysis showed significant association of BRAF V600E+/TERT+ co-mutations with lymph node metastasis, multifocality, distant metastasis, tumor recurrence, extrathyroidal extension, and dead of disease. Conclusions Prevalence of BRAF V600E mutation in Northwest China was higher than other areas. Age, multiple lesions, and pathology were independent factors for double mutation of BRAF V600E/TERT or BRAF V600E/NRAS. Coexistence of BRAF V600E and TERT promoter mutations was significantly correlated with poor outcome.
Collapse
Affiliation(s)
- Meiling Huang
- Department of Thyroid, Breast, and Vascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Changjiao Yan
- Department of Thyroid, Breast, and Vascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Jingjing Xiao
- Department of Thyroid, Breast, and Vascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Ting Wang
- Department of Thyroid, Breast, and Vascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China.
| | - Rui Ling
- Department of Thyroid, Breast, and Vascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
14953
|
Jia X, Zhao Q, Zhang Y, Dong Y, Lei L, Williamson RA, Lei Y, Tan X, Zhang D, Hu J. Identification of a Five-CpG Signature with Diagnostic Value in Thyroid Cancer. J Comput Biol 2019; 26:1409-1417. [PMID: 31290678 DOI: 10.1089/cmb.2019.0165] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Thyroid cancer (TC) ranks as the most common endocrine malignancy, and its incidence and mortality rates continue to rise annually. Increasing evidence have shown that DNA methylation, a kind of stable epigenetic modification, is associated with carcinogenesis, suggesting its potential as biomarkers for the early detection of tumors. With the aim of exploring likely DNA methylation biomarkers for TC diagnosis, we conducted a synthetic analysis of DNA methylation profiles based on 789 samples from the Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) databases. In the discovery phase, we identified five CpG probes (cg11228682, cg01291854, cg06778183, cg01668008, and cg01702055) on the condition of DNA methylation data from GSE86961 (n = 82) and constructed a five-CpG signature-based diagnostic model for TC. In addition, we validated the diagnostic score formula in two independent training cohorts, GSE97466 (n = 141) and TCGA (n = 566), as well as the previous developing cohort GSE86961. Receiver operating characteristic analysis revealed that the five-CpG signature had a good diagnostic performance to distinguish TC samples from benign samples. In conclusion, our findings suggest that the five-CpG signature could provide a novel biomarker with useful applications in TC diagnosis.
Collapse
Affiliation(s)
- Xi Jia
- Department of Nuclear Medicine, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Qian Zhao
- Department of Otolaryngology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Yuanyuan Zhang
- Department of Pediatrics, and The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Yiping Dong
- Department of Oncology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Li Lei
- Department of Cell Biology and Genetics, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Ramone A Williamson
- Department of Cell Biology and Genetics, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Yutiantian Lei
- Department of Oncology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Xinyue Tan
- Department of Oncology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Dan Zhang
- Department of Cell Biology and Genetics, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Jinsong Hu
- Department of Cell Biology and Genetics, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, China
| |
Collapse
|
14954
|
Do H, Kim D, Kang J, Son B, Seo D, Youn H, Youn B, Kim W. TFAP2C increases cell proliferation by downregulating GADD45B and PMAIP1 in non-small cell lung cancer cells. Biol Res 2019; 52:35. [PMID: 31296259 PMCID: PMC6625030 DOI: 10.1186/s40659-019-0244-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 07/05/2019] [Indexed: 12/25/2022] Open
Abstract
Background Non-small cell lung cancer (NSCLC) is one of the leading causes of death in the world. NSCLC diagnosed at an early stage can be highly curable with a positive prognosis, but biomarker limitations make it difficult to diagnose lung cancer at an early stage. To identify biomarkers for lung cancer development, we previously focused on the oncogenic roles of transcription factor TFAP2C in lung cancers and revealed the molecular mechanism of several oncogenes in lung tumorigenesis based on TFAP2C-related microarray analysis. Results In this study, we analyzed microarray data to identify tumor suppressor genes and nine genes downregulated by TFAP2C were screened. Among the nine genes, we focused on growth arrest and DNA-damage-inducible beta (GADD45B) and phorbol-12-myristate-13-acetate-induced protein 1 (PMAIP1) as representative TFAP2C-regulated tumor suppressor genes. It was observed that overexpressed TFAP2C resulted in inhibition of GADD45B and PMAIP1 expressions at both the mRNA and protein levels in NSCLC cells. In addition, downregulation of GADD45B and PMAIP1 by TFAP2C promoted cell proliferation and cell motility, which are closely associated with NSCLC tumorigenesis. Conclusion This study indicates that GADD45B and PMAIP1 could be promising tumor suppressors for NSCLC and might be useful as prognostic markers for use in NSCLC therapy. Electronic supplementary material The online version of this article (10.1186/s40659-019-0244-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hyunhee Do
- Department of Science Education, Korea National University of Education, Cheongju-si, Chungbuk, 28173, Republic of Korea
| | - Dain Kim
- Department of Science Education, Korea National University of Education, Cheongju-si, Chungbuk, 28173, Republic of Korea
| | - JiHoon Kang
- Department of Integrated Biological Science, Pusan National University, Busan, 46241, Republic of Korea
| | - Beomseok Son
- Department of Integrated Biological Science, Pusan National University, Busan, 46241, Republic of Korea
| | - Danbi Seo
- Department of Science Education, Korea National University of Education, Cheongju-si, Chungbuk, 28173, Republic of Korea
| | - HyeSook Youn
- Department of Integrative Bioscience and Biotechnology, Sejong University, Seoul, 05006, Republic of Korea
| | - BuHyun Youn
- Department of Integrated Biological Science, Pusan National University, Busan, 46241, Republic of Korea. .,Department of Biological Sciences, Pusan National University, Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan, 46241, Republic of Korea.
| | - Wanyeon Kim
- Department of Science Education, Korea National University of Education, Cheongju-si, Chungbuk, 28173, Republic of Korea. .,Department of Biology Education, Korea National University of Education, 250 Taeseongtabyeon-ro, Gangnae-myeon, Heungdeok-gu, Cheongju-si, Chungbuk, 28173, Republic of Korea.
| |
Collapse
|
14955
|
Quantifying the relationship between age at diagnosis and breast cancer-specific mortality. Breast Cancer Res Treat 2019; 177:713-722. [DOI: 10.1007/s10549-019-05353-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 07/05/2019] [Indexed: 12/18/2022]
|
14956
|
Amirahmadi R, Kumar AJ, Cowan M, Deepak J. Lung Cancer Screening in Patients with COPD-A Case Report. ACTA ACUST UNITED AC 2019; 55:medicina55070364. [PMID: 31336732 PMCID: PMC6681240 DOI: 10.3390/medicina55070364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 06/28/2019] [Accepted: 07/08/2019] [Indexed: 11/16/2022]
Abstract
We present two cases demonstrating the nuances that must be considered when determining if a patient could benefit from low dose computed tomography (LDCT) lung cancer screening. Our case report discusses the available literature, where it exists, on lung cancer screening with special attention to the impact of chronic obstructive pulmonary disease (COPD), and poor functional status. Patients with COPD and concurrent smoking history are at higher risk of lung cancer and may therefore benefit from lung cancer screening. However, this population is at increased risk for complications related to biopsies and lobar resections. Appropriate interventions other than surgical resection exist for COPD patients with poor pulmonary reserve. Risks and benefits of lung cancer screening are unique to each patient and require shared decision-making.
Collapse
Affiliation(s)
- Roxana Amirahmadi
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Avnee J Kumar
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Mark Cowan
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Janaki Deepak
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Division of Pulmonary and Critical Care Medicine, Baltimore VA Medical Health Center, Baltimore, MD 21201, USA
| |
Collapse
|
14957
|
Qiu Y, Jiang J, Zhang M, Qin Y. Positive PD-L1 expression is predictive for patients with advanced EGFR wild-type non-small cell lung cancer treated with gemcitabine and cisplatin. Oncol Lett 2019; 18:161-168. [PMID: 31289485 PMCID: PMC6539442 DOI: 10.3892/ol.2019.10302] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 03/18/2019] [Indexed: 12/26/2022] Open
Abstract
This retrospective study aimed to investigate the association between programmed death ligand-1 (PD-L1) expression and the clinicopathological characteristics of patients with advanced epidermal growth factor receptor (EGFR) wild-type non-small cell lung cancer (NSCLC). The predictive role and cut-off value of PD-L1 expression was subsequently investigated. A total of 172 patients with advanced EGFR wild-type NSCLC were enrolled. All patients received platinum-based doublet chemotherapy (gemcitabine plus cisplatin). PD-L1 expression in lung tissues was assessed using immunohistochemical methods. The χ2 test was used to analyze the association between PD-L1 expression and clinicopathological characteristics. Survival time analysis was performed using the Kaplan-Meier method. The two groups, positive PD-L1 expression and negative PD-L1 expression, were compared using the log-rank test. Multivariate analysis using the Cox proportional hazard regression model was conducted to determine prognostic factors for overall survival (OS) and progression-free survival (PFS) times. Positive PD-L1 expression was observed in 48.3% (84/172), 40.7% (70/172), 21.5% (37/172) and 8.1% (14/172) of patients when using cut-off values of 1, 5, 10 and 50%, respectively. The χ2 test revealed that elevated pretreatment C-reactive protein (CRP) level and cancer stage IV were significantly associated with positive PD-L1 expression. The OS and PFS of positive PD-L1 (1, 5, 10 and 50% cut-off) expression group were shorter compared with the negative PD-L1 (1, 5, 10 and 50% cut-off) expression group. Multivariate survival analysis revealed that PD-L1 expression ≥50% was significantly associated with decreased OS and PFS [OS time, P=0.001; hazard ratio (HR), 2.768; 95% confidence interval (CI), 1.551–4.940; PFS time, P=0.002; HR, 2.537; 95% CI, 1.423–4.524]. These results indicated that positive PD-L1 (50% cut-off) expression was an independent predictor of poor prognosis for patients with advanced NSCLC treated with gemcitabine plus cisplatin. PD-L1 expression was associated with CRP level and cancer stage. The results obtained in the present study suggest that positive PD-L1 expression serves a prognostic role in advanced NSCLC and that the optimal cut-off value may be 50%.
Collapse
Affiliation(s)
- Yajuan Qiu
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Junguang Jiang
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Mingzhi Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yanru Qin
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
14958
|
Zhou C, Qian W, Li J, Ma J, Chen X, Jiang Z, Cheng L, Duan W, Wang Z, Wu Z, Ma Q, Li X. High glucose microenvironment accelerates tumor growth via SREBP1-autophagy axis in pancreatic cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:302. [PMID: 31296258 PMCID: PMC6625066 DOI: 10.1186/s13046-019-1288-7] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Accepted: 06/24/2019] [Indexed: 02/08/2023]
Abstract
Background Diabetes is recognized to be a risk factor of pancreatic cancer, but the mechanism has not been fully elucidated. Sterol regulatory element binding protein 1 (SREBP1) is an important transcription factor involved in both lipid metabolism and tumor progression. However, the relationship between high glucose microenvironment, SREBP1 and pancreatic cancer remains to be explored. Methods Clinical data and surgical specimens were collected. Pancreatic cancer cell lines BxPc-3 and MiaPaCa-2 were cultured in specified medium. Immunohistochemistry (IHC) and western blotting were performed to detect the expression of SREBP1. MTT and colony formation assays were applied to investigate cell proliferation. Immunofluorescence, mRFP-GFP adenoviral vector and transmission electron microscopy were performed to evaluate autophagy. We used streptozotocin (STZ) to establish a high glucose mouse model for the in vivo study. Results We found that high blood glucose levels were associated with poor prognosis in pancreatic cancer patients. SREBP1 was overexpressed in both pancreatic cancer tissues and pancreatic cancer cell lines. High glucose microenvironment promoted tumor proliferation, suppressed apoptosis and inhibited autophagy level by enhancing SREBP1 expression. In addition, activation of autophagy accelerated SREBP1 expression and suppressed apoptosis. Moreover, high glucose promotes tumor growth in vivo by enhancing SREBP1 expression. Conclusion Our results indicate that SREBP1-autophagy axis plays a crucial role in tumor progression induced by high glucose microenvironment. SREBP1 may represent a novel target for pancreatic cancer prevention and treatment.
Collapse
Affiliation(s)
- Cancan Zhou
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Weikun Qian
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Jie Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Jiguang Ma
- Department of Anesthesiology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Xin Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Zhengdong Jiang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Liang Cheng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Wanxing Duan
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Zheng Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Zheng Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Qingyong Ma
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.
| | - Xuqi Li
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.
| |
Collapse
|
14959
|
Feng C, Bai M, Zhang H, Zeng A, Zhang W. Prioritization and comprehensive analysis of genes associated with melanoma. Oncol Lett 2019; 18:127-136. [PMID: 31289481 PMCID: PMC6540330 DOI: 10.3892/ol.2019.10284] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 02/28/2019] [Indexed: 11/10/2022] Open
Abstract
Melanoma is a malignant tumor derived from melanocytes, which occurs mostly in the skin. A major challenge in cancer research is the biological interpretation of the complexity of cancer somatic mutation profiles. The aim of the present study was to obtain a comprehensive understanding of the formation and development of melanoma and to identify its associated genes. In the present study, a pipeline was proposed for investigating key genes associated with melanoma based on the Online Mendelian Inheritance in Man and Search Tool for the Retrieval of Interacting Genes/Proteins databases through a random walk model. Additionally, functional enrichment analysis was performed for key genes associated with melanoma. This identified a total of 17 biological processes and 30 pathways which may be associated with melanoma. In addition, melanoma-specific network analysis followed by Kaplan-Meier analysis along with log-rank tests identified tyrosinase, hedgehog acyltransferase, BRCA1-associated protein 1 and melanocyte inducing transcription factor as potential therapeutic targets for melanoma. In conclusion, the present study increased the knowledge of melanoma progression and may be helpful for improving its prognosis.
Collapse
Affiliation(s)
- Cheng Feng
- Department of Plastic Surgery, Peking Union Medical College Hospital, Beijing 100730, P.R. China
| | - Ming Bai
- Department of Plastic Surgery, Peking Union Medical College Hospital, Beijing 100730, P.R. China
| | - Hailin Zhang
- Department of Plastic Surgery, Peking Union Medical College Hospital, Beijing 100730, P.R. China
| | - Ang Zeng
- Department of Plastic Surgery, Peking Union Medical College Hospital, Beijing 100730, P.R. China
| | - Wenchao Zhang
- Department of Plastic Surgery, Peking Union Medical College Hospital, Beijing 100730, P.R. China
| |
Collapse
|
14960
|
George J, Nihal M, Singh CK, Ahmad N. 4'-Bromo-resveratrol, a dual Sirtuin-1 and Sirtuin-3 inhibitor, inhibits melanoma cell growth through mitochondrial metabolic reprogramming. Mol Carcinog 2019; 58:1876-1885. [PMID: 31292999 DOI: 10.1002/mc.23080] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 06/12/2019] [Accepted: 06/13/2019] [Indexed: 12/24/2022]
Abstract
Sirtuin-1 and -3 (SIRT1 and SIRT3) are important nicotinamide adenine dinucleotide (NAD+ )-dependent deacetylases known to regulate a variety of cellular functions. Studies have shown that SIRT1 and SIRT3 were overexpressed in human melanoma cells and tissues and their inhibition resulted in a significant antiproliferative response in human melanoma cells and antitumor response in a mouse xenograft model of melanoma. In this study, we determined the antiproliferative efficacy of a newly identified dual small molecule inhibitor of SIRT1 and SIRT3, 4'-bromo-resveratrol (4'-BR), in human melanoma cell lines (G361, SK-MEL-28, and SK-MEL-2). Our data demonstrate that 4'-BR treatment of melanoma cells resulted in (a) decrease in proliferation and clonogenic survival; (b) induction of apoptosis accompanied by a decrease in procaspase-3, procaspase-8, and increase in the cleavage of caspase-3 and poly (ADP-ribose) polymerase (PARP); (c) marked downregulation of proliferating cell nuclear antigen (PCNA); and (d) inhibition of melanoma cell migration. Further, 4'-BR caused a G0/G1 phase arrest of melanoma cells that was accompanied by an increase in WAF-1/P21 and decrease in Cyclin D1/Cyclin-dependent kinase 6 protein levels. Furthermore, we found that 4'-BR causes a decrease in lactate production, glucose uptake, and NAD+ /NADH ratio. These responses were accompanied by downregulation in lactate dehydrogenase A and glucose transporter 1 in melanoma cells. Collectively, our data suggest that dual inhibition of SIRT1 and SIRT3 using 4'-BR imparted antiproliferative effects in melanoma cells through a metabolic reprogramming and affecting the cell cycle and apoptosis signaling. Therefore, concomitant pharmacological inhibition of SIRT1 and SIRT3 needs further investigation for melanoma management.
Collapse
Affiliation(s)
- Jasmine George
- Department of Dermatology, Medical Sciences Center, University of Wisconsin, Madison, Wisconsin
| | - Minakshi Nihal
- Department of Dermatology, Medical Sciences Center, University of Wisconsin, Madison, Wisconsin
| | - Chandra K Singh
- Department of Dermatology, Medical Sciences Center, University of Wisconsin, Madison, Wisconsin
| | - Nihal Ahmad
- Department of Dermatology, Medical Sciences Center, University of Wisconsin, Madison, Wisconsin.,Research, William S. Middleton VA Medical Center, Madison, Wisconsin
| |
Collapse
|
14961
|
Blakely AM, Wong P, Chu P, Warner SG, Raoof M, Singh G, Fong Y, Melstrom LG. Intraoperative bile spillage is associated with worse survival in gallbladder adenocarcinoma. J Surg Oncol 2019; 120:603-610. [PMID: 31292970 DOI: 10.1002/jso.25617] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 06/21/2019] [Indexed: 12/24/2022]
Abstract
BACKGROUND Gallbladder adenocarcinoma is often incidentally identified following cholecystectomy. We hypothesized that intraoperative bile spillage would be a negative prognostic factor. METHODS A retrospective review of patients treated at a cancer center with histologically confirmed gallbladder adenocarcinoma, 2009-2017, was performed. Patient, disease, and treatment factors were analyzed in terms of progression-free survival (PFS) and overall survival (OS). RESULTS Sixty-six patients were identified. Tumor stage was T1 (n = 8, 12%), T2 (n = 23, 35%), T3 (n = 35, 53%). Node stage was N0 (n = 22, 33%), N1+ (n = 26, 39%), Nx (n = 18, 27%). Operations included cholecystectomy alone (n = 27, 36%), cholecystectomy and partial hepatectomy (n = 30, 45%), or hepaticojejunostomy (n = 9, 14%). Median PFS was 7 months (interquartile range [IQR], 2-19); median OS was 16 months (IQR, 10-31). Subset multivariate proportional hazards regression of 41 patients who underwent initial cholecystectomy showed decreased PFS was associated with intraoperative spillage (n = 12, 29%; hazard ratio [HR], 5.5; P = .0014); decreased OS was associated with drain placement (n = 21, 51%; HR, 8.1; P = .006). CONCLUSIONS Intraoperative bile spillage and surgical drain placement at initial cholecystectomy are negatively associated with PFS and OS in gallbladder adenocarcinoma. Explicit documentation of spillage and drain placement rationale is critical, possibly indicating locally advanced disease and prompting stronger consideration of systemic therapy before definitive resection.
Collapse
Affiliation(s)
- Andrew M Blakely
- Department of Surgery, City of Hope National Medical Center, Duarte, California
| | - Paul Wong
- Department of Surgery, City of Hope National Medical Center, Duarte, California
| | - Peiguo Chu
- Department of Pathology, City of Hope National Medical Center, Duarte, California
| | - Susanne G Warner
- Department of Surgery, City of Hope National Medical Center, Duarte, California
| | - Mustafa Raoof
- Department of Surgery, City of Hope National Medical Center, Duarte, California
| | - Gagandeep Singh
- Department of Surgery, City of Hope National Medical Center, Duarte, California
| | - Yuman Fong
- Department of Surgery, City of Hope National Medical Center, Duarte, California
| | - Laleh G Melstrom
- Department of Surgery, City of Hope National Medical Center, Duarte, California
| |
Collapse
|
14962
|
Kennedy A, Shipley D, Shpak M, Blakely L, Hemphill B, Shih K, Lane C, Zimmerman L, McKenzie A, Mainwaring M, Peyton JD, Zubkus J, Wright D, Singh J, Bendell JC. Regorafenib Prior to Selective Internal Radiation Therapy Using 90Y-Resin Microspheres for Refractory Metastatic Colorectal Cancer Liver Metastases: Analysis of Safety, Dosimetry, and Molecular Markers. Front Oncol 2019; 9:624. [PMID: 31355141 PMCID: PMC6636394 DOI: 10.3389/fonc.2019.00624] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 06/24/2019] [Indexed: 12/22/2022] Open
Abstract
Background: This Phase II, open-label, study examined the safety of regorafenib followed by selective internal radiation therapy (SIRT) with regorafenib re-initiation in the treatment of metastatic colorectal cancer (mCRC) patients with liver metastases who are not surgical candidates. Methods: Patients received 160 mg regorafenib daily on a 21-day course followed by a 1 week washout prior to SIRT. Liver function was evaluated at 2 and 4 weeks after SIRT, and regorafenib re-initiated if liver function was normal. Patients were evaluated for safety, and restaged at weeks 6 and 12 following SIRT. In addition, protein and cytokine assays of blood were performed to identify candidate molecular biomarkers associated with outcomes. Individual patient voxel-based dosimetry assessment was performed post-SIRT. Results: Twenty-Five patients were enrolled and received a median 11 weeks regorafenib. Three patients received regorafenib, but not SIRT due to disease progression. The remaining 22 patients received SIRT with a median activity delivered to the liver of 38 mCi, mean normal liver dose of 14.98 Gy and tumor mean dose of 29.0 Gy with a tumor to normal ratio mean of 2.42. There were four treatment-related serious AEs and no treatment-related deaths. Median progression-free survival was 3.7 months and the median overall survival was 12.1 months. The relative densities of several biomolecules changed significantly during the course of treatment, most notably post-treatment increases in levels of sex-hormone binding globulin (SHBG) and decreased levels of the cytokine MIG (CXL9). Decreases in von Willebrand factor (VWF), the ankyrin repeat domain (ANKRD26), and MIG were associated with improved survival times. Post-treatment increases in alpha-2-macroglobulin (A2M) and the cytokine intercellular adhesion molecule (ICAM-1) were associated with reduced overall survival time, while increases in Eotaxin (CCL14) predicted longer overall survival times. Conclusions: The treatment of mCRC patients with liver metastases using regorafenib followed by SIRT was tolerable in this patient population. Further efficacy analysis of this treatment schema and analysis of potential molecular biomarkers using larger sample sizes is merited.
Collapse
Affiliation(s)
- Andrew Kennedy
- Sarah Cannon Research Institute, Nashville, TN, United States
| | - Dianna Shipley
- Sarah Cannon Research Institute, Nashville, TN, United States.,Tennessee Oncology, PLLC, Nashville, TN, United States
| | - Max Shpak
- Sarah Cannon Research Institute, Nashville, TN, United States.,Center for Systems and Synthetic Biology, University of Texas, Austin, TX, United States
| | - Laura Blakely
- Sarah Cannon Research Institute, Nashville, TN, United States.,Tennessee Oncology, PLLC, Nashville, TN, United States
| | - Brian Hemphill
- Sarah Cannon Research Institute, Nashville, TN, United States.,Tennessee Oncology, PLLC, Nashville, TN, United States
| | - Kent Shih
- Sarah Cannon Research Institute, Nashville, TN, United States.,Tennessee Oncology, PLLC, Nashville, TN, United States
| | - Cassie Lane
- Sarah Cannon Research Institute, Nashville, TN, United States
| | - Lisa Zimmerman
- Sarah Cannon Research Institute, Nashville, TN, United States
| | - Andrew McKenzie
- Sarah Cannon Research Institute, Nashville, TN, United States
| | - Mark Mainwaring
- Sarah Cannon Research Institute, Nashville, TN, United States.,Tennessee Oncology, PLLC, Nashville, TN, United States
| | - James D Peyton
- Sarah Cannon Research Institute, Nashville, TN, United States.,Tennessee Oncology, PLLC, Nashville, TN, United States
| | - John Zubkus
- Sarah Cannon Research Institute, Nashville, TN, United States.,Tennessee Oncology, PLLC, Nashville, TN, United States
| | - David Wright
- Sarah Cannon Research Institute, Nashville, TN, United States.,Florida Cancer Specialists, Tampa, FL, United States
| | | | - Johanna C Bendell
- Sarah Cannon Research Institute, Nashville, TN, United States.,Tennessee Oncology, PLLC, Nashville, TN, United States
| |
Collapse
|
14963
|
Rajitha B, Malla RR, Vadde R, Kasa P, Prasad GLV, Farran B, Kumari S, Pavitra E, Kamal MA, Raju GSR, Peela S, Nagaraju GP. Horizons of nanotechnology applications in female specific cancers. Semin Cancer Biol 2019; 69:376-390. [PMID: 31301361 DOI: 10.1016/j.semcancer.2019.07.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 06/23/2019] [Accepted: 07/04/2019] [Indexed: 12/20/2022]
Abstract
Female-specific cancers are the most common cancers in women worldwide. Early detection methods remain unavailable for most of these cancers, signifying that most of them are diagnosed at later stages. Furthermore, current treatment options for most female-specific cancers are surgery, radiation and chemotherapy. Although important milestones in molecularly targeted approaches have been achieved lately, current therapeutic strategies for female-specific cancers remain limited, ineffective and plagued by the emergence of chemoresistance, which aggravates prognosis. Recently, the application of nanotechnology to the medical field has allowed the development of novel nano-based approaches for the management and treatment of cancers, including female-specific cancers. These approaches promise to improve patient survival rates by reducing side effects, enabling selective delivery of drugs to tumor tissues and enhancing the uptake of therapeutic compounds, thus increasing anti-tumor activity. In this review, we focus on the application of nano-based technologies to the design of novel and innovative diagnostic and therapeutic strategies in the context of female-specific cancers, highlighting their potential uses and limitations.
Collapse
Affiliation(s)
- Balney Rajitha
- Department of Pathology, WellStar Hospital, Marietta, GA, 30060, USA
| | - Rama Rao Malla
- Department of Biochemistry, GITAM Institute of Science, GITAM University, Visakhapatnam, AP, 530045, India
| | - Ramakrishna Vadde
- Department of Biotechnology and Bioinformatics, Yogi Vemana University, Kadapa, AP, 516003, India
| | - Prameswari Kasa
- Dr. LV Prasad Diagnostics and Research Laboratory, Khairtabad, Hyderabad, TS, 500004, India
| | | | - Batoul Farran
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, 30322, USA
| | - Seema Kumari
- Department of Biochemistry, GITAM Institute of Science, GITAM University, Visakhapatnam, AP, 530045, India
| | - Eluri Pavitra
- Department of Biological Engineering, Biohybrid Systems Research Center (BSRC), Inha University, 100, Inha-ro, Incheon 22212, Republic of Korea
| | - Mohammad Amjad Kamal
- King Fahd Medical Research Center, King Abdulaziz University, P. O. Box 80216, Jeddah 21589, Saudi Arabia; Enzymoics, 7 Peterlee Place, Hebersham, NSW 2770, Australia; Novel Global Community Educational Foundation, Australia
| | - Ganji Seeta Rama Raju
- Department of Energy and Materials Engineering, Dongguk University-Seoul, Seoul 04620, Republic of Korea
| | - Sujatha Peela
- Department of Biotechnology, Dr. B.R. Ambedkar University, Srikakulam, AP, 532410, India
| | - Ganji Purnachandra Nagaraju
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|
14964
|
Endoscopic submucosal dissection (ESD) versus transanal endoscopic microsurgery (TEM) for treatment of rectal tumors: a comparative systematic review and meta-analysis. Surg Endosc 2019; 34:1688-1695. [PMID: 31292744 DOI: 10.1007/s00464-019-06945-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 06/26/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND While multiple studies have evaluated endoscopic submucosal dissection (ESD) and transanal endoscopic microsurgery (TEM) to remove large rectal tumors, there remains a paucity of data to evaluate their comparative efficacy and safety. The primary aim of this study was to perform a structured systematic review and meta-analysis to compare efficacy and safety of ESD versus TEM for the treatment of rectal tumors. METHODS Individualized search strategies were developed from inception through November 2018 in accordance with PRISMA guidelines. Measured outcomes included pooled enbloc resection rates, margin-negative (R0) resection rates, procedure-associated adverse events, and rates of recurrence. This was a cumulative meta-analysis performed by calculating pooled proportions. Heterogeneity was assessed with Cochran Q test and I2 statistics, and publication bias by funnel plot using Egger and Begg tests. RESULTS Three studies (n = 158 patients; 55.22% male) were included in this meta-analysis. Patients with ESD compared to TEM had similar age (P = 0.090), rectal tumor size (P = 0.108), and diagnosis rate of adenoma to cancer (P = 0.53). ESD lesions were more proximal as compared to TEM (8.41 ± 3.49 vs. 5.11 ± 1.43 cm from the anal verge; P < 0.001). Procedure time and hospital stay were shorter for ESD compared to TEM [(79.78 ± 24.45 vs. 116.61 ± 19.35 min; P < 0.001) and (3.99 ± 0.32 vs. 5.83 ± 0.94 days; P < 0.001), respectively]. No significant differences between enbloc resection rates [OR 0.98 (95% CI 0.22-4.33); P = 0.98; I2 = 0.00%] and R0 resection rates [OR 1.16 (95% CI 0.36-3.76); P = 0.80; I2 = 0.00%] were noted between ESD and TEM. ESD and TEM reported similar rates of adverse events [OR 1.15 (95% CI 0.47-2.77); P = 0.80; I2 = 0.00%] and rates of recurrence [OR 0.46 (95% CI 0.07-3.14); P = 0.43; I2 = 0.00%]. CONCLUSION ESD and TEM possess similar rates of resection, adverse events, and recurrence for patients with large rectal tumors; however, ESD is associated with significantly shorter procedure times and duration of hospitalization. Future studies are needed to evaluate healthcare utilization for these two strategies.
Collapse
|
14965
|
Jiang L, Gu Y, Du Y, Liu J. Exosomes: Diagnostic Biomarkers and Therapeutic Delivery Vehicles for Cancer. Mol Pharm 2019; 16:3333-3349. [PMID: 31241965 DOI: 10.1021/acs.molpharmaceut.9b00409] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Exosomes are described as nanoscale extracellular vesicles (EVs) secreted by multiple cell types and extensively distributed in various biological fluids. They contain multifarious bioactive molecules and transfer them to adjoining or distal cells through systemic circulation, participating in intracellular and intercellular communication, and modulating host-tumor cell interactions. Recent research has indicated that exosomes obtained from different biological fluids and their contents (proteins, nucleic acids, glycoconjugates, and lipids) can serve as biomarkers for cancer diagnosis, prognosis, and therapeutic response. Furthermore, the discovery of exosomes as therapeutic delivery vehicles has drawn much attention in antineoplastic drug delivery. They can be utilized for therapeutic delivery of proteins, genetic drugs, and chemotherapeutic drugs. Herein, this review summarizes the biogenesis, structure, and components of exosomes, focusing primarily on their two possible applications as diagnostic biomarkers and therapeutic delivery vehicles for cancers.
Collapse
Affiliation(s)
- Liangdi Jiang
- Department of Pharmacy , Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College , Fudan University , Shanghai 200032 , China.,College of Pharmacy , Shandong University of Traditional Chinese Medicine , Jinan , Shandong 250355 , China
| | - Yongwei Gu
- Department of Pharmacy , Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College , Fudan University , Shanghai 200032 , China
| | - Yue Du
- Department of Pharmacy , Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College , Fudan University , Shanghai 200032 , China.,College of Pharmacy , Shandong University of Traditional Chinese Medicine , Jinan , Shandong 250355 , China
| | - Jiyong Liu
- Department of Pharmacy , Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College , Fudan University , Shanghai 200032 , China
| |
Collapse
|
14966
|
Yan B, Chen B, Min S, Gao Y, Zhang Y, Xu P, Li C, Chen J, Luo G, Liu C. iTRAQ-based Comparative Serum Proteomic Analysis of Prostate Cancer Patients with or without Bone Metastasis. J Cancer 2019; 10:4165-4177. [PMID: 31413735 PMCID: PMC6691707 DOI: 10.7150/jca.33497] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 05/12/2019] [Indexed: 12/18/2022] Open
Abstract
Background: Once prostate cancer developed bone metastasis, the quality of life and prognosis of patients are seriously affected as no effective treatment is currently available. It is necessary to explore the mechanism of bone metastasis and new therapeutic targets. Purpose: To find out the differentially expressed serum proteins in prostate cancer patients with bone metastasis and analyze the expression of key proteins in prostate cancer tissues, serum and prostate cancer cell lines. So as to provide a basis for revealing the mechanism of bone metastasis and designing new therapeutic targets. Methods: iTRAQ-based proteomics method was used to compare serum differential proteins in prostate cancer patients with and without bone metastasis. Three key proteins (CD59, haptoglobin and tetranectin) which had significant fold changes were selected to validate the results of mass spectrometry. Immunohistochemistry and ELISA were applied to tissues and serum samples from prostate cancer patients, respectively, for validation. Finally, western blot, flow cytometry, and immunocytochemistry were used to analyze the expression of the three differentially expressed proteins in the prostate cancer cell lines PC3, LNCap, and Du145. Results: Thirty-two differentially expressed proteins related to bone metastasis of prostate cancer were identified, of which 11 were up-regulated and 21 were down-regulated. CD59 and haptoglobin were up-regulated in prostate cancer with bone metastasis while tetranectin was down-regulated. Tetranectin showed differential expression in epithelial and stromal cells of prostate cancer and hyperplasia tissues.The expression of CD59 was highest in PC3 and lowest in LNCap, while the expression of haptoglobin and tetranectin was the highest in DU145 and lowest in PC3. Conclusion: Mass spectrometry analysis showed that there were more differentially expressed proteins in the serum of patients with bone metastasis than those without metastasis. It has been verified that CD59, haptoglobin and tetranectin are prostate cancer bone metastasis related proteins.
Collapse
Affiliation(s)
- Bo Yan
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Department of Urology, Guizhou Provincial People's Hospital, Medical College of Guizhou University, Guiyang, Guizhou, China
| | - Binshen Chen
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Shaoju Min
- Department of Clinical Laboratory, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Yubo Gao
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yiming Zhang
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Peng Xu
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Chaoming Li
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiasheng Chen
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Guangheng Luo
- Department of Urology, Guizhou Provincial People's Hospital, Medical College of Guizhou University, Guiyang, Guizhou, China
| | - Chunxiao Liu
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
14967
|
Ünver HM, Ayan E. Skin Lesion Segmentation in Dermoscopic Images with Combination of YOLO and GrabCut Algorithm. Diagnostics (Basel) 2019; 9:E72. [PMID: 31295856 PMCID: PMC6787581 DOI: 10.3390/diagnostics9030072] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 06/26/2019] [Accepted: 07/08/2019] [Indexed: 01/22/2023] Open
Abstract
Skin lesion segmentation has a critical role in the early and accurate diagnosis of skin cancer by computerized systems. However, automatic segmentation of skin lesions in dermoscopic images is a challenging task owing to difficulties including artifacts (hairs, gel bubbles, ruler markers), indistinct boundaries, low contrast and varying sizes and shapes of the lesion images. This paper proposes a novel and effective pipeline for skin lesion segmentation in dermoscopic images combining a deep convolutional neural network named as You Only Look Once (YOLO) and the GrabCut algorithm. This method performs lesion segmentation using a dermoscopic image in four steps: 1. Removal of hairs on the lesion, 2. Detection of the lesion location, 3. Segmentation of the lesion area from the background, 4. Post-processing with morphological operators. The method was evaluated on two publicly well-known datasets, that is the PH2 and the ISBI 2017 (Skin Lesion Analysis Towards Melanoma Detection Challenge Dataset). The proposed pipeline model has achieved a 90% sensitivity rate on the ISBI 2017 dataset, outperforming other deep learning-based methods. The method also obtained close results according to the results obtained from other methods in the literature in terms of metrics of accuracy, specificity, Dice coefficient, and Jaccard index.
Collapse
Affiliation(s)
- Halil Murat Ünver
- Department of Computer Engineering, Kırıkkale University, 71451 Kırıkkale, Turkey
| | - Enes Ayan
- Department of Computer Engineering, Kırıkkale University, 71451 Kırıkkale, Turkey.
| |
Collapse
|
14968
|
Wu X, Wu Q, Zhou X, Huang J. SphK1 functions downstream of IGF-1 to modulate IGF-1-induced EMT, migration and paclitaxel resistance of A549 cells: A preliminary in vitro study. J Cancer 2019; 10:4264-4269. [PMID: 31413745 PMCID: PMC6691691 DOI: 10.7150/jca.32646] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 05/25/2019] [Indexed: 12/28/2022] Open
Abstract
Insulin-like growth factor-1 (IGF-1) -induced epithelial-mesenchymal transition (EMT) plays a key role in the metastasis and drug resistance of non-small cell lung cancer (NSCLC). Sphingosine kinase-1 (SphK1) is also involved in EMT of NSCLC. However, the interaction between SphK1 and IGF-1 in the EMT of NSCLC is largely unknown. To clarify this issue, we examined the involvement of SphK1 in IGF-1-induced EMT using human lung cancer cell line A549, and its paclitaxel-resistant subline. Cell viability was evaluated by cell counting kit-8 assay; Migratory ability was examined using scratch wound healing test; Protein expression levels of SphK1, vimentin, fibronectin, N-cadherin and E-cadherin were detected by western blot analysis, respectively. The results showed that, IGF-1 treatment of A549 cells stimulated the expression of SphK1, the activation of ERK and AKT, the cell migration, and the expression of EMT hallmark proteins, while inhibition of SphK1 by its specific inhibitor SKI-II suppressed all the above changes and increased the sensitivity of A549 cells to paclitaxel. Our data demonstrate that SphK1 acts as a downstream effector of IGF-1 and plays a critical role in IGF-1-induced EMT, cell migration and paclitaxel resistance of A549 cells, suggesting that SphK1 might be a potential therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Xingping Wu
- Department of Respiratory Medicine, the First Affiliated Hospital of Soochow University, Jiangsu, P.R. China.,State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, P.R. China.,Department of Respirology, the First People's Hospital of Lianyungang, Jiangsu, P.R. China
| | - Qibiao Wu
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, P.R. China
| | - Xiqiao Zhou
- Department of Gastroenterology, the First Affiliated Hospital of Nanjing Medical University, Jiangsu, P.R. China
| | - Jianan Huang
- Department of Respiratory Medicine, the First Affiliated Hospital of Soochow University, Jiangsu, P.R. China
| |
Collapse
|
14969
|
Chen H, Li L, Hu J, Zhao Z, Ji L, Cheng C, Zhang G, zhang T, Li Y, Chen H, Pan S, Sun B. UBL4A inhibits autophagy-mediated proliferation and metastasis of pancreatic ductal adenocarcinoma via targeting LAMP1. J Exp Clin Cancer Res 2019; 38:297. [PMID: 31288830 PMCID: PMC6617940 DOI: 10.1186/s13046-019-1278-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Accepted: 06/13/2019] [Indexed: 02/19/2023] Open
Abstract
BACKGROUND Ubiquitin-like protein 4A (UBL4A) plays a significant role in protein metabolism and the maintenance of cellular homeostasis. In cancer, UBL4A represses tumorigenesis and is involved in various signaling pathways. Pancreatic ductal adenocarcinoma (PDAC) is still a major cause of cancer-related death and the underlying molecular mechanism of UBL4A and PDAC remains unknown. METHODS First, the prognostic role of UBL4A and its expression in human PDAC patients and in pancreatic cancer cell lines were detected by survival analysis and qRT-PCR, western blotting, and immunohistochemistry. Next, the effects of UBL4A on proliferation and metastasis in pancreatic cancer were evaluated by functional assays in vitro and in vivo. In addition, chloroquine was introduced to determine the role of autophagy in UBL4A-related tumor proliferation and metastasis. Ultimately, coimmunoprecipitation was used to confirm the interaction between UBL4A and lysosome associated membrane protein-1 (LAMP1), and western blotting was performed to explore the UBL4A mechanism. RESULTS We found that UBL4A was decreased in PDAC and that high levels of UBL4A correlated with a favorable prognosis. We observed that UBL4A inhibited tumor proliferation and metastasis through suppression of autophagy, a critical intracellular catabolic process that reportedly protects cells from nutrient starvation and other stress conditions. UBL4A caused impaired autophagic degradation in vitro, a crucial process in autophagy, by disturbing the function of lysosomes and contributing to autophagosome accumulation. We found a positive correlation between UBL4A and LAMP1. Furthermore, UBL4A caused lysosomal dysfunction by directly interacting with LAMP1, and LAMP1 overexpression reversed the antitumor effects of UBL4A in pancreatic cancer. In addition, we demonstrated that UBL4A suppressed tumor growth and metastasis in a pancreatic orthotopic tumor model. CONCLUSIONS These findings suggest that UBL4A exerts an antitumor effect on autophagy-related proliferation and metastasis in PDAC by directly targeting LAMP1. Herein, we describe a novel mechanism of UBL4A that suppresses the progression of pancreatic cancer. UBL4A might be a promising target for the treatment and prognostication of PDAC.
Collapse
Affiliation(s)
- Hongze Chen
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 23 Youzheng Street, Nangang District, Harbin, 150001 Heilongjiang Province China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, Heilongjiang China
| | - Le Li
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 23 Youzheng Street, Nangang District, Harbin, 150001 Heilongjiang Province China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, Heilongjiang China
| | - Jisheng Hu
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 23 Youzheng Street, Nangang District, Harbin, 150001 Heilongjiang Province China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, Heilongjiang China
| | - Zhongjie Zhao
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 23 Youzheng Street, Nangang District, Harbin, 150001 Heilongjiang Province China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, Heilongjiang China
| | - Liang Ji
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, Heilongjiang China
- Department of Breast Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang China
| | - Chundong Cheng
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 23 Youzheng Street, Nangang District, Harbin, 150001 Heilongjiang Province China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, Heilongjiang China
| | - Guangquan Zhang
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 23 Youzheng Street, Nangang District, Harbin, 150001 Heilongjiang Province China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, Heilongjiang China
| | - Tao zhang
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 23 Youzheng Street, Nangang District, Harbin, 150001 Heilongjiang Province China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, Heilongjiang China
| | - Yilong Li
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 23 Youzheng Street, Nangang District, Harbin, 150001 Heilongjiang Province China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, Heilongjiang China
| | - Hua Chen
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 23 Youzheng Street, Nangang District, Harbin, 150001 Heilongjiang Province China
| | - Shangha Pan
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 23 Youzheng Street, Nangang District, Harbin, 150001 Heilongjiang Province China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, Heilongjiang China
| | - Bei Sun
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 23 Youzheng Street, Nangang District, Harbin, 150001 Heilongjiang Province China
| |
Collapse
|
14970
|
Incidence of Skeletal-Related Events in Patients with Castration-Resistant Prostate Cancer: An Observational Retrospective Cohort Study in the US. Prostate Cancer 2019; 2019:5971615. [PMID: 31360552 PMCID: PMC6652049 DOI: 10.1155/2019/5971615] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 04/09/2019] [Accepted: 05/14/2019] [Indexed: 11/18/2022] Open
Abstract
Background and Objective Skeletal-related events (SREs) are common in men with bone metastases and have negative consequences for patients with castration-resistant prostate cancer (CRPC), including pain, reduced quality of life, and increased mortality. We estimated incidence rates of first SREs in a cohort of men with CRPC in the Surveillance, Epidemiology, and End Results-Medicare database. Methods We included men aged ≥ 65 years with a prostate cancer diagnosis in 2000-2011 if they had no prior malignancy (other than nonmelanoma skin cancer) and had surgical or medical castration with subsequent second-line systemic therapy, which was used to infer castration resistance. The first occurrence of an SRE (fracture, bone surgery, radiation therapy, or spinal cord compression) in Medicare claims was identified. Incidence rates of SREs were estimated in all eligible person-time and, in secondary analyses, stratified by any use of bone-targeted agents (BTAs) and history of SRE. Results Of 2,234 men with CRPC (84% white, mean age = 76.6 years), 896 (40%) had an SRE during follow-up, with 74% occurring within a year after cohort entry. Overall, the incidence rate of SREs was 3.78 (95% CI, 3.53-4.03) per 100 person-months. The incidence rate of SREs before any BTA use was 4.16 (95% CI, 3.71-4.65) per 100 person-months, and after any BTA use was 3.60 (95% CI, 3.32-3.91) per 100 person-months. The incidence rate in patients with no history of SRE was 3.33 (95% CI 3.01-3.68) per 100 person-months, and in patients who had such a history, it was 4.20 (95% CI 3.84-4.58) per 100 person-months. Conclusions In this large cohort of elderly men with CRPC in the US, SREs were common. A decrease in incidence of SREs after starting BTA is suggested, but the magnitude of the effect may be confounded by indication and other factors such as age and prior SRE.
Collapse
|
14971
|
Expression Signatures of Cisplatin- and Trametinib-Treated Early-Stage Medaka Melanomas. G3-GENES GENOMES GENETICS 2019; 9:2267-2276. [PMID: 31101653 PMCID: PMC6643878 DOI: 10.1534/g3.119.400051] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Small aquarium fish models provide useful systems not only for a better understanding of the molecular basis of many human diseases, but also for first-line screening to identify new drug candidates. For testing new chemical substances, current strategies mostly rely on easy to perform and efficient embryonic screens. Cancer, however, is a disease that develops mainly during juvenile and adult stage. Long-term treatment and the challenge to monitor changes in tumor phenotype make testing of large chemical libraries in juvenile and adult animals cost prohibitive. We hypothesized that changes in the gene expression profile should occur early during anti-tumor treatment, and the disease-associated transcriptional change should provide a reliable readout that can be utilized to evaluate drug-induced effects. For the current study, we used a previously established medaka melanoma model. As proof of principle, we showed that exposure of melanoma developing fish to the drugs cisplatin or trametinib, known cancer therapies, for a period of seven days is sufficient to detect treatment-induced changes in gene expression. By examining whole body transcriptome responses we provide a novel route toward gene panels that recapitulate anti-tumor outcomes thus allowing a screening of thousands of drugs using a whole-body vertebrate model. Our results suggest that using disease-associated transcriptional change to screen therapeutic molecules in small fish model is viable and may be applied to pre-clinical research and development stages in new drug discovery.
Collapse
|
14972
|
Jia H, Tian J, Liu B, Meng H, Pan F, Li C. Efficacy and safety of artificial pneumothorax with position adjustment for CT-guided percutaneous transthoracic microwave ablation of small subpleural lung tumors. Thorac Cancer 2019; 10:1710-1716. [PMID: 31290286 PMCID: PMC6669918 DOI: 10.1111/1759-7714.13137] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 06/17/2019] [Accepted: 06/17/2019] [Indexed: 01/28/2023] Open
Abstract
Background To evaluate the efficacy and safety of artificial pneumothorax with position adjustment for computed tomograpy (CT)‐guided percutaneous transthoracic microwave ablation (MWA) of small subpleural lung tumors. Methods Fifty‐six patients with small subpleural lung tumors (< 3.0 cm) entered the study and underwent CT‐guided MWA with (group I: 24 patients with 24 tumors) or without (group II: 32 patients with 34 tumors) the support of artificial pneumothorax. Follow‐up contrast‐enhanced CT scans were reviewed. Pain VAS (visual analog scale) scores at, during, and after ablation were compared between the two groups. Technical success, technique efficacy, local tumor control and complications were compared. Results Creation of the artificial pneumothorax was achieved for 24/24 (100%) in group I and no complication related to the procedure was observed. Technical success of MWA was achieved for all 58 tumors. Primary efficacy of MWA was achieved in 23 of 24 tumors (95.8%) treated in group I, and 32 of 34 tumors (94.1%) treated in group II (P = 0.771). The 12‐month local tumor control was achieved in 87.5% (21/24) in group I compared with 88.2% (30/34) in group II (P = 0.833). Pain VAS scores in group I were significantly decreased after the pneumothorax induction at, during, and after ablation compared with group II (P < 0.05). There was no significant difference in MWA‐related complications (P > 0.05). Conclusion Artificial pneumothorax with position adjustment for CT‐guided MWA is effective and may be safely applied to small subpleural lung tumors. Artificial pneumothorax is a reliable therapy for pain relief.
Collapse
Affiliation(s)
- Haipeng Jia
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, China
| | - Jie Tian
- Department of Respiration, Huantai County Hospital of Traditional Chinese Medicine, Zibo, China
| | - Bo Liu
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, China
| | - Hong Meng
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, China
| | - Fengmin Pan
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, China
| | - Chunhai Li
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
14973
|
Wu S, Liu X, He J, Wang H, Luo Y, Gong W, Li Y, Huang Y, Zhong L, Zhao Y. A Dual Targeting Magnetic Nanoparticle for Human Cancer Detection. NANOSCALE RESEARCH LETTERS 2019; 14:228. [PMID: 31289961 PMCID: PMC6616609 DOI: 10.1186/s11671-019-3049-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 06/12/2019] [Indexed: 06/09/2023]
Abstract
Malignant tumors are a major threat to human life and high lymphatic vessel density is often associated with metastatic tumors. With the discovery of molecules targeted at the lymphatic system such as lymphatic vessel endothelial hyaluronan receptor 1 (LYVE-1) and Podoplanin, many studies have been performed to determine the role of lymphatic endothelial cells (LECs) in tumor metastasis. However, disadvantages such as non-specificity and high cost limit their research and diagnostic applications. In this study, Fe3O4@KCTS, a core-shell type of magnetic nanoparticles, was prepared by activating Fe3O4 with carbodiimide and cross-linking it with α-ketoglutarate chitosan (KCTS). The LYVE-1 and Podoplanin antibodies were then incorporated onto the surface of these magnetic nanoparticles and as a result, dual-targeting magnetic nanoprobes were developed. The experimental tests of this study demonstrated that a dual-targeting magnetic nanoprobe with high-purity LECs from tumor tissues was successfully developed, providing a basis for clinical application of LECs in colorectal cancer treatment as well as in early clinical diagnosis using bimodal imaging.
Collapse
Affiliation(s)
- Siwen Wu
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative InnovationCenter for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, Guangxi, China
- School of Preclinical Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Xiyu Liu
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative InnovationCenter for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Jian He
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative InnovationCenter for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Huiling Wang
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative InnovationCenter for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Yiqun Luo
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative InnovationCenter for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Wenlin Gong
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative InnovationCenter for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Yanmei Li
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative InnovationCenter for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Yong Huang
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative InnovationCenter for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Liping Zhong
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative InnovationCenter for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, Guangxi, China.
| | - Yongxiang Zhao
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative InnovationCenter for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, Guangxi, China.
| |
Collapse
|
14974
|
Sun D, Tan S, Xiong Y, Pu W, Li J, Wei W, Huang C, Wei YQ, Peng Y. MicroRNA Biogenesis is Enhanced by Liposome-Encapsulated Pin1 Inhibitor in Hepatocellular Carcinoma. Am J Cancer Res 2019; 9:4704-4716. [PMID: 31367251 PMCID: PMC6643437 DOI: 10.7150/thno.34588] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 05/05/2019] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is in an urgent need of new, effective therapies to reduce morbidity and mortality. We have previously demonstrated that peptidyl-prolyl cis/trans isomerase Pin1 is a potential target for HCC therapy, due to its pivotal role in HCC development through regulating miRNA biogenesis, and discovered the small molecule API-1 as a novel and specific Pin1 inhibitor. Despite its significant anti-HCC activity, the low water solubility and in vivo bioavailability of API-1 limit its clinical application. To address these issues, we herein developed a liposomal formulation of API-1 to improve API-1 delivery and enhance its anti-HCC efficacy. Methods: We designed and developed a nanoscale liposomal formulation of API-1, named as API-LP. Subsequently, the mean diameter, polydispersity, zeta potential, encapsulation efficiency and thermal properties of the optimization API-LP were characterized. The enhanced anti-HCC activity and the molecular mechanism of API-LP were investigated both in vitro and in vivo. Finally, the safety and pharmacokinetic property of API-LP were evaluated systematically. Results: API-LP had good formulation characteristics and exhibited an enhanced in vitro activity of suppressing proliferation and migration of HCC cells when compared with free API-1. The mechanism study showed that API-LP upregulated miRNA biogenesis via inhibiting Pin1 activity followed by restoring the nucleus-to-cytoplasm export of XPO5. Because of the increased delivery efficiency, API-LP displayed a stronger ability to promote miRNA biogenesis than free API-1. Importantly, API-LP displayed higher systemic exposure than free API-1 in mice without apparent toxicity, resulting in an enhanced tumor inhibition in xenograft mice. Conclusion: The development and assessment of API-LP provide an attractive and safe anti-HCC agent, highlighting the miRNA-based treatment for human cancers.
Collapse
|
14975
|
Wang P, Gao XY, Yang SQ, Sun ZX, Dian LL, Qasim M, Phyo AT, Liang ZS, Sun YF. Jatrorrhizine inhibits colorectal carcinoma proliferation and metastasis through Wnt/β-catenin signaling pathway and epithelial-mesenchymal transition. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:2235-2247. [PMID: 31371920 PMCID: PMC6627180 DOI: 10.2147/dddt.s207315] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 05/21/2019] [Indexed: 01/29/2023]
Abstract
Purpose Jatrorrhizine (JAT) is a natural protoberberine alkaloid, possesses detoxification, bactericidal and hypoglycemic activities. However, its anti-cancer mechanism is not clear. This study aimed to investigate the mechanism of JAT through which inhibits colorectal cancer in HCT-116 and HT-29 cells. Methods MTT assay and colony formation assay were used to check the cell proliferation ability. Cell apoptosis and cell cycle were measured by Hoechst 33342 staining and flow cytometry, respectively. Cell migration and invasion were detected by scratch wound healing assay and trans-well assay, respectively. Further, expression of related proteins was examined via Western blotting and the in vivo anti-cancer effect of JAT was confirmed by nude mice xenograft model. Results The research showed that JAT inhibited the proliferation of HCT-116 and HT-29 cells with IC50 values of 6.75±0.29 μM and 5.29±0.13 μM, respectively, for 72 hrs. It has also showed a time dependently, cell cycle arrested in S phase, promoted cell apoptosis and suppressed cell migration and invasion. In addition, JAT inhibited Wnt signaling pathway by reducing β-catenin and increasing GSK-3β expressions. Increased expression of E-cadherin, while decreased N-cadherin, indicating that JAT treatment suppressed the process of cell epithelial–mesenchymal transition (EMT). In HCT-116 nude mice xenograft model, JAT inhibited tumor growth and metastasis, and induced apoptosis of tumor cells. Conclusion This study demonstrated that JAT efficiently inhibited colorectal cancer cells growth and metastasis, which provides a new point for clinical treatment of colorectal cancer.
Collapse
Affiliation(s)
- Pan Wang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, People's Republic of China
| | - Xiao-Yan Gao
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, People's Republic of China
| | - Si-Qian Yang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, People's Republic of China
| | - Zhi-Xin Sun
- College of Life Sciences, Wenzhou Medical University, Wenzhou 325035, People's Republic of China.,Department of Life Sciences, Zaozhuang No.1 Middle School, Zaozhuang, 277100, People's Republic of China
| | - Lu-Lu Dian
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, People's Republic of China
| | - Muhammad Qasim
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, People's Republic of China.,Institute of Sustainable Halophyte Utilization, University of Karachi, Karachi 75270, Pakistan
| | - Aung Thu Phyo
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, People's Republic of China.,Department of Biotechnology, Mandalay Technological University, Mandalay 05072, Myanmar
| | - Zong-Suo Liang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, People's Republic of China
| | - Yan-Fang Sun
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, People's Republic of China
| |
Collapse
|
14976
|
Zahra MH, Salem TAR, El-Aarag B, Yosri N, El-Ghlban S, Zaki K, Marei AH, Abd El-Wahed A, Saeed A, Khatib A, AlAjmi MF, Shathili AM, Xiao J, Khalifa SAM, El-Seedi HR. Alpinia zerumbet (Pers.): Food and Medicinal Plant with Potential In Vitro and In Vivo Anti-Cancer Activities. Molecules 2019; 24:2495. [PMID: 31288458 PMCID: PMC6651078 DOI: 10.3390/molecules24132495] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 06/30/2019] [Accepted: 07/05/2019] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND/AIM Plants play an important role in anti-cancer drug discovery, therefore, the current study aimed to evaluate the biological activity of Alpinia zerumbet (A. zerumbet) flowers. METHODS The phytochemical and biological criteria of A. zerumbet were in vitro investigated as well as in mouse xenograft model. RESULTS A. zerumbet extracts, specially CH2Cl2 and MeOH extracts, exhibited the highest potent anti-tumor activity against Ehrlich ascites carcinoma (EAC) cells. The most active CH2Cl2 extract was subjected to bioassay-guided fractionation leading to isolatation of the naturally occurring 5,6-dehydrokawain (DK) which was characterized by IR, MS, 1H-NMR and 13C-NMR. A. zerumbet extracts, specially MeOH and CH2Cl2 extracts, exhibited significant inhibitory activity towards tumor volume (TV). Furthermore, A. zerumbet extracts declined the high level of malonaldehyde (MDA) as well as elevated the levels of superoxide dismutase (SOD) and catalase (CAT) in liver tissue homogenate. Moreover, DK showed anti-proliferative action on different human cancer cell lines. The recorded IC50 values against breast carcinoma (MCF-7), liver carcinoma (Hep-G2) and larynx carcinoma cells (HEP-2) were 3.08, 6.8, and 8.7 µg/mL, respectively. CONCLUSION Taken together, these findings open the door for further investigations in order to explore the potential medicinal properties of A. zerumbet.
Collapse
Affiliation(s)
- Maram Hussein Zahra
- Division of Chemistry and Biotechnology, Graduate School of Natural Science and Technology, Okayama University, Okayama 7008530, Japan.
| | - Tarek A R Salem
- Department of Biochemistry, College of Medicine, Qassim University, Al-Qassim 51452, Saudi Arabia.
- Department of Molecular Biology, Genetic Engineering & Biotechnology Institute, University of Sadat City, Sadat City 32958, Egypt.
| | - Bishoy El-Aarag
- Division of Chemistry and Biotechnology, Graduate School of Natural Science and Technology, Okayama University, Okayama 7008530, Japan
- Biochemistry Division, Chemistry Department, Faculty of Science, Menoufia University, Shebin El-Koom 32512, Egypt
| | - Nermeen Yosri
- Department of Chemistry, Faculty of Science, Menoufia University, Shebin El-Koom 32512, Egypt
| | - Samah El-Ghlban
- Biochemistry Division, Chemistry Department, Faculty of Science, Menoufia University, Shebin El-Koom 32512, Egypt
| | - Kholoud Zaki
- Department of Chemistry, Faculty of Science, Menoufia University, Shebin El-Koom 32512, Egypt
| | - Amel H Marei
- Department of Chemistry, Faculty of Science, Menoufia University, Shebin El-Koom 32512, Egypt
| | - Aida Abd El-Wahed
- Department of Chemistry, Faculty of Science, Menoufia University, Shebin El-Koom 32512, Egypt
- Division of Pharmacognosy, Department of Medicinal Chemistry, Uppsala University, Biomedical Centre, Box 574, SE-751 23 Uppsala, Sweden
- Department of Bee Research, Plant Protection Research Institute, Agricultural Research Centre, Giza 12627, Egypt
| | - Aamer Saeed
- Department of Chemistry, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Alfi Khatib
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, International Islamic University Malaysia, Kuantan 25200, Pahang, Malaysia
| | - Mohamed F AlAjmi
- Pharmacognosy Group, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | | | - Jianbo Xiao
- Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau 999078, China
| | - Shaden A M Khalifa
- Clinical Research Centre, Karolinska University Hospital, 14186 Huddinge, Sweden
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE 106 91 Stockholm, Sweden
| | - Hesham R El-Seedi
- Department of Chemistry, Faculty of Science, Menoufia University, Shebin El-Koom 32512, Egypt.
- Division of Pharmacognosy, Department of Medicinal Chemistry, Uppsala University, Biomedical Centre, Box 574, SE-751 23 Uppsala, Sweden.
- Al-Rayan Research and Innovation Center, Al-Rayan Colleges, Medina 42541, Saudi Arabia.
- College of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, China.
| |
Collapse
|
14977
|
Lin S, Yang L, Yao Y, Xu L, Xiang Y, Zhao H, Wang L, Zuo Z, Huang X, Zhao C. Flubendazole demonstrates valid antitumor effects by inhibiting STAT3 and activating autophagy. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:293. [PMID: 31287013 PMCID: PMC6615228 DOI: 10.1186/s13046-019-1303-z] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 06/30/2019] [Indexed: 12/21/2022]
Abstract
Background Signal transducer and activator of transcription 3 (STAT3) is an oncogene, which upregulates in approximately 70% of human cancers. Autophagy is an evolutionarily conserved process which maintains cellular homeostasis and eliminates damaged cellular components. Moreover, the STAT3 signaling pathway, which may be triggered by cancer cells, has been implicated in the autophagic process. Methods In this study, we found that the anthelmintic flubendazole exerts potent antitumor activity in three human colorectal cancer (CRC) cell lines and in the nude mouse model. The inhibition of cell proliferation in vitro by flubendazole was evaluated using a clonogenic assay and the MTT assay. Western blot analysis, flow cytometry analysis, siRNA growth experiment and cytoplasmic and nuclear protein extraction were used to investigate the mechanisms of inhibiting STAT3 signaling and activation of autophagy induced by flubendazole. Additionally, the expression of STAT3 and mTOR was analyzed in paired colorectal cancer and normal tissues collected from clinical patients. Results Flubendazole blocked the IL6-induced nuclear translocation of STAT3, which led to inhibition of the transcription of STAT3 target genes, such as MCL1, VEGF and BIRC5. In addition, flubendazole also reduced the expression of P-mTOR, P62, BCL2, and upregulated Beclin1 and LC3-I/II, which are major autophagy-related genes. These processes induced potent cell apoptosis in CRC cells. In addition, flubendazole displayed a synergistic effect with the chemotherapeutic agent 5-fluorouracil in the treatment of CRC. Conclusions Taken together, these results indicate that flubendazole exerts antitumor activities by blocking STAT3 signaling and inevitably affects the autophagy pathway. Flubendazole maybe a novel anticancer drug and offers a distinctive therapeutic strategy in neoadjuvant chemotherapy of CRC. Electronic supplementary material The online version of this article (10.1186/s13046-019-1303-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shichong Lin
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People's Republic of China.,The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People's Republic of China
| | - Lehe Yang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People's Republic of China.,The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People's Republic of China
| | - Yulei Yao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People's Republic of China.,The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People's Republic of China
| | - Lingyuan Xu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People's Republic of China.,The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People's Republic of China
| | - Youqun Xiang
- The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People's Republic of China
| | - Haiyang Zhao
- The Institute of Life Sciences, Wenzhou University, Wenzhou, Zhejiang, 325035, People's Republic of China
| | - Liangxing Wang
- The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People's Republic of China
| | - Zhigui Zuo
- The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People's Republic of China.
| | - Xiaoying Huang
- The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, People's Republic of China.
| | - Chengguang Zhao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, People's Republic of China.
| |
Collapse
|
14978
|
Neureiter D, Stintzing S, Kiesslich T, Ocker M. Hepatocellular carcinoma: Therapeutic advances in signaling, epigenetic and immune targets. World J Gastroenterol 2019; 25:3136-3150. [PMID: 31333307 PMCID: PMC6626722 DOI: 10.3748/wjg.v25.i25.3136] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 05/02/2019] [Accepted: 05/18/2019] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) remains a global medical burden with rising incidence due to chronic viral hepatitis and non-alcoholic fatty liver diseases. Treatment of advanced disease stages is still unsatisfying. Besides first and second generation tyrosine kinase inhibitors, immune checkpoint inhibitors have become central for the treatment of HCC. New modalities like epigenetic therapy using histone deacetylase inhibitors (HDACi) and cell therapy approaches with chimeric antigen receptor T cells (CAR-T cells) are currently under investigation in clinical trials. Development of such novel drugs is closely linked to the availability and improvement of novel preclinical and animal models and the identification of predictive biomarkers. The current status of treatment options for advanced HCC, emerging novel therapeutic approaches and different preclinical models for HCC drug discovery and development are reviewed here.
Collapse
Affiliation(s)
- Daniel Neureiter
- Institute of Pathology, Cancer Cluster Salzburg, Paracelsus Medical University/Salzburger Landeskliniken (SALK), Salzburg 5020, Austria
| | - Sebastian Stintzing
- Medical Department, Division of Oncology and Hematology, Campus Charité Mitte, Charité University Medicine Berlin, Berlin 10117, Germany
| | - Tobias Kiesslich
- Department of Internal Medicine I, Paracelsus Medical University/Salzburger Landeskliniken (SALK) and Institute of Physiology and Pathophysiology, Paracelsus Medical University, Salzburg 5020, Austria
| | - Matthias Ocker
- Translational Medicine Oncology, Bayer AG, Berlin 13353, Germany
- Charité University Medicine Berlin, Berlin 10117, Germany
| |
Collapse
|
14979
|
Zhou R, Wu Z, Deng X, Chen H. The long non-coding RNA OLC8 enhances gastric cancer by interaction with IL-11. J Clin Lab Anal 2019; 33:e22962. [PMID: 31273847 PMCID: PMC6805327 DOI: 10.1002/jcla.22962] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 06/02/2019] [Accepted: 06/05/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The gastric cancer (GC) represents a common malignancy especially in China. Long non-coding RNAs (lncRNAs) are critically involved in various types of cancer. However, the underlying mechanisms of OLC8 in gastric cancer are still largely unknown. METHODS The lncRNA profiling was used to identify novel lncRNAs associated with GC. The expression of OLC8 was quantified using qRT-PCR. Migration and viability assays were performed to evaluate the in vitro effects. Xenograft tumor models were conducted to investigate the in vivo oncogenic potential. RNA-seq was used to identify IL-11 as OLC8 binding partner. RESULTS In current study, we have identified a novel lncRNA termed OLC8. OLC8 was significantly overexpressed in gastric cancer specimens and cell lines. In vitro experiments showed that OLC8 facilitated migration and viability of MKN1 and AGS cells. As expected, in vivo experiments also confirmed an oncogenic role for OLC8. Mechanistic study indicated that OLC8 associated with IL-11 transcripts. The OLC8-IL-11 binding greatly impaired the degradation of IL-11 mRNAs. Not surprisingly, enhanced expression of IL-11 could increase STAT3 activation to favor gastric cancer development. CONCLUSIONS Our current research has identified OLC8 as a novel oncogenic lncRNA in IL-11/STAT3 signaling, and OLC8 may constitute a potential target for gastric cancer intervention.
Collapse
Affiliation(s)
- Rongjia Zhou
- Department of Gastroenterology, Panyu Central Hospital, Guangzhou, Guangdong, China
| | - Zhanbin Wu
- Department of Gastroenterology, Panyu Central Hospital, Guangzhou, Guangdong, China
| | - Xixiang Deng
- Department of Gastroenterology, Panyu Central Hospital, Guangzhou, Guangdong, China
| | - Haojun Chen
- Department of Gastroenterology, Panyu Central Hospital, Guangzhou, Guangdong, China
| |
Collapse
|
14980
|
Zhang Y, Zhang Y, Xu H. LIMCH1 suppress the growth of lung cancer by interacting with HUWE1 to sustain p53 stability. Gene 2019; 712:143963. [PMID: 31279706 DOI: 10.1016/j.gene.2019.143963] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 06/10/2019] [Accepted: 07/03/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND The aim of this study was to identify the expression of LIM and calponin-homology domains 1 (LIMCH1) in lung cancer and normal tissues, to determine the interaction between LIMCH1 and HUWE1 in regulating p53 stability. METHODS The expression of LIMCH1 was detected by the Oncomine and Cancer Genome Atlas databases. Expression of LIMCH1 mRNA was identified using qRT-PCR. In transfected human lung cancer cells, co-immunoprecipitation experiments were performed. The mechanism that HUWE1 sustained lung cancer malignancy was verified by western blotting. The proliferation of tranfected cells was assessed by CCK-8 assay and colony formation. RESULTS Bioinformatic data and e TCGA database suggested LIMCH1 mRNA levels in tumor tissues were down-regulated compared to tumor adjacent tissues. We found low expression of LIMCH1 mRNA in tumor sites and tumor cell line. Exogenous expression of LIMCH1 interacts with HUWE1 promotes expression of p53. Use of siRNA or shRNA against LIMCH1 resulted in decreased p53 protein levels. LIMCH1 deletion lead to enhance of p53 ubiquitination and protein expression of p53 and substrate p21, puma. Growth curve showed that LIMCH1 deletion significantly promoted the proliferation of A549 cells. CONCLUSIONS LIMCH1 was a negative regulator and indicated a new molecular mechanism for the pathogenesis of lung cancer via modulating HUWE1 and p53.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- Department of Laboratory Medicine, The Affiliated Huaian No.1 People's Hospital of Nanjing, Medical University, Huai'an, Jiangsu, 223300, China.
| | - Yingmei Zhang
- Department of Laboratory Medicine, The Affiliated Huaian No.1 People's Hospital of Nanjing, Medical University, Huai'an, Jiangsu, 223300, China
| | - Haiyan Xu
- Department of Cardiology, The Affiliated Huaian No.1 People's Hospital of Nanjing, Medical University, Huai'an, Jiangsu, 223300, China
| |
Collapse
|
14981
|
Devarakonda S, Govindan R. Untangling the evolutionary roots of lung cancer. Nat Commun 2019; 10:2979. [PMID: 31278259 PMCID: PMC6611778 DOI: 10.1038/s41467-019-10879-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 06/06/2019] [Indexed: 01/01/2023] Open
Abstract
The genomic and host factors that drive the progression of pre-invasive lesions in non-small cell lung cancer are poorly understood. Studying these factors can advance our knowledge of lung cancer biology, aid in the development of better screening strategies and improve patient outcomes.
Collapse
Affiliation(s)
- Siddhartha Devarakonda
- Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, United States.,Siteman Cancer Center, St. Louis, MO, 63110, United States
| | - Ramaswamy Govindan
- Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, United States. .,Siteman Cancer Center, St. Louis, MO, 63110, United States.
| |
Collapse
|
14982
|
Cao C, Zhou JY, Xie SW, Guo XJ, Li GT, Gong YJ, Yang WJ, Li Z, Zhong RH, Shao HH, Zhu Y. Metformin Enhances Nomegestrol Acetate Suppressing Growth of Endometrial Cancer Cells and May Correlate to Downregulating mTOR Activity In Vitro and In Vivo. Int J Mol Sci 2019; 20:E3308. [PMID: 31284427 PMCID: PMC6650946 DOI: 10.3390/ijms20133308] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 06/30/2019] [Accepted: 07/03/2019] [Indexed: 12/12/2022] Open
Abstract
This study investigated the effect of a novel progestin and its combination with metformin on the growth of endometrial cancer (EC) cells. Inhibitory effects of four progestins, including nomegestrol acetate (NOMAC), medroxyprogesterone acetate, levonorgestrel, and cyproterone acetate, were evaluated in RL95-2, HEC-1A, and KLE cells using cell counting kit-8 assay. Flow cytometry was performed to detect cell cycle and apoptosis. The activity of Akt (protein kinase B), mTOR (mammalian target of rapamycin) and its downstream substrates 4EBP1 (4E-binding protein 1) and eIF4G (Eukaryotic translation initiation factor 4G) were assayed by Western blotting. Nude mice were used to assess antitumor effects in vivo. NOMAC inhibited the growth of RL95-2 and HEC-1A cells, accompanied by arresting the cell cycle at G0/G1 phase, inducing apoptosis, and markedly down-regulating the level of phosphorylated mTOR/4EBP1/eIF4G in both cell lines (p < 0.05). Metformin significantly increased the inhibitory effect of and apoptosis induced by NOMAC and strengthened the depressive effect of NOMAC on activity of mTOR and its downstream substrates, compared to their treatment alone (p < 0.05). In xenograft tumor tissues, metformin (100 mg/kg) enhanced the suppressive effect of NOMAC (100 mg/kg) on mTOR signaling and increased the average concentration of NOMAC by nearly 1.6 times compared to NOMAC treatment alone. Taken together, NOMAC suppressing the growth of EC cells likely correlates to down-regulating the activity of the mTOR pathway and metformin could strengthen this effect. Our findings open a new window for the selection of progestins in hormone therapy of EC.
Collapse
Affiliation(s)
- Can Cao
- Pharmacy School, Fudan University, Shanghai 200032, China
- Lab of Reproductive Pharmacology, NHC Key Lab of Reproduction Regulation, Shanghai Institute of Planned Parenthood Research, Fudan University, Shanghai 200032, China
| | - Jie-Yun Zhou
- Lab of Reproductive Pharmacology, NHC Key Lab of Reproduction Regulation, Shanghai Institute of Planned Parenthood Research, Fudan University, Shanghai 200032, China
| | - Shu-Wu Xie
- Lab of Reproductive Pharmacology, NHC Key Lab of Reproduction Regulation, Shanghai Institute of Planned Parenthood Research, Fudan University, Shanghai 200032, China
| | - Xiang-Jie Guo
- Lab of Reproductive Pharmacology, NHC Key Lab of Reproduction Regulation, Shanghai Institute of Planned Parenthood Research, Fudan University, Shanghai 200032, China
| | - Guo-Ting Li
- Lab of Reproductive Pharmacology, NHC Key Lab of Reproduction Regulation, Shanghai Institute of Planned Parenthood Research, Fudan University, Shanghai 200032, China
| | - Yi-Juan Gong
- Lab of Reproductive Pharmacology, NHC Key Lab of Reproduction Regulation, Shanghai Institute of Planned Parenthood Research, Fudan University, Shanghai 200032, China
| | - Wen-Jie Yang
- Lab of Reproductive Pharmacology, NHC Key Lab of Reproduction Regulation, Shanghai Institute of Planned Parenthood Research, Fudan University, Shanghai 200032, China
| | - Zhao Li
- Lab of Reproductive Pharmacology, NHC Key Lab of Reproduction Regulation, Shanghai Institute of Planned Parenthood Research, Fudan University, Shanghai 200032, China
| | - Rui-Hua Zhong
- Lab of Reproductive Pharmacology, NHC Key Lab of Reproduction Regulation, Shanghai Institute of Planned Parenthood Research, Fudan University, Shanghai 200032, China
| | - Hai-Hao Shao
- Lab of Reproductive Pharmacology, NHC Key Lab of Reproduction Regulation, Shanghai Institute of Planned Parenthood Research, Fudan University, Shanghai 200032, China
| | - Yan Zhu
- Lab of Reproductive Pharmacology, NHC Key Lab of Reproduction Regulation, Shanghai Institute of Planned Parenthood Research, Fudan University, Shanghai 200032, China.
| |
Collapse
|
14983
|
PI3K-AKT-mTOR and NFκB Pathways in Ovarian Cancer: Implications for Targeted Therapeutics. Cancers (Basel) 2019; 11:cancers11070949. [PMID: 31284467 PMCID: PMC6679095 DOI: 10.3390/cancers11070949] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 06/10/2019] [Accepted: 06/30/2019] [Indexed: 02/07/2023] Open
Abstract
Ovarian cancer is the most lethal gynecologic malignancy in the United States, with an estimated 22,530 new cases and 13,980 deaths in 2019. Recent studies have indicated that the phosphoinositol 3 kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR), as well as the nuclear factor-κ light chain enhancer of activated B cells (NFκB) pathways are highly mutated and/or hyper-activated in a majority of ovarian cancer patients, and are associated with advanced grade and stage disease and poor prognosis. In this review, we will investigate PI3K/AKT/mTOR and their interconnection with NFκB pathway in ovarian cancer cells.
Collapse
|
14984
|
Kabacaoglu D, Ruess DA, Ai J, Algül H. NF-κB/Rel Transcription Factors in Pancreatic Cancer: Focusing on RelA, c-Rel, and RelB. Cancers (Basel) 2019; 11:E937. [PMID: 31277415 PMCID: PMC6679104 DOI: 10.3390/cancers11070937] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 06/26/2019] [Accepted: 07/02/2019] [Indexed: 02/07/2023] Open
Abstract
Regulation of Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB)/Rel transcription factors (TFs) is extremely cell-type-specific owing to their ability to act disparately in the context of cellular homeostasis driven by cellular fate and the microenvironment. This is also valid for tumor cells in which every single component shows heterogenic effects. Whereas many studies highlighted a per se oncogenic function for NF-κB/Rel TFs across cancers, recent advances in the field revealed their additional tumor-suppressive nature. Specifically, pancreatic ductal adenocarcinoma (PDAC), as one of the deadliest malignant diseases, shows aberrant canonical-noncanonical NF-κB signaling activity. Although decades of work suggest a prominent oncogenic activity of NF-κB signaling in PDAC, emerging evidence points to the opposite including anti-tumor effects. Considering the dual nature of NF-κB signaling and how it is closely linked to many other cancer related signaling pathways, it is essential to dissect the roles of individual Rel TFs in pancreatic carcinogenesis and tumor persistency and progression. Here, we discuss recent knowledge highlighting the role of Rel TFs RelA, RelB, and c-Rel in PDAC development and maintenance. Next to providing rationales for therapeutically harnessing Rel TF function in PDAC, we compile strategies currently in (pre-)clinical evaluation.
Collapse
Affiliation(s)
- Derya Kabacaoglu
- Internal Medicine II, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany
| | - Dietrich A Ruess
- Department of Surgery, Faculty of Medicine, Medical Center, University of Freiburg, 79106 Freiburg, Germany
| | - Jiaoyu Ai
- Internal Medicine II, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany
| | - Hana Algül
- Internal Medicine II, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany.
| |
Collapse
|
14985
|
Boice JD, Held KD, Shore RE. Radiation epidemiology and health effects following low-level radiation exposure. JOURNAL OF RADIOLOGICAL PROTECTION : OFFICIAL JOURNAL OF THE SOCIETY FOR RADIOLOGICAL PROTECTION 2019; 39:S14-S27. [PMID: 31272090 DOI: 10.1088/1361-6498/ab2f3d] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Radiation epidemiology is the study of human disease following radiation exposure to populations. Epidemiologic studies of radiation-exposed populations have been conducted for nearly 100 years, starting with the radium dial painters in the 1920s and most recently with large-scale studies of radiation workers. As radiation epidemiology has become increasingly sophisticated it is used for setting radiation protection standards as well as to guide the compensation programmes in place for nuclear weapons workers, nuclear weapons test participants, and other occupationally exposed workers in the United States and elsewhere. It is known with high assurance that radiation effects at levels above 100-150 mGy can be detected as evidenced in multiple population studies conducted around the world. The challenge for radiation epidemiology is evaluating the effects at low doses, below about 100 mGy of low-linear energy transfer radiation, and assessing the risks following low dose-rate exposures over years. The weakness of radiation epidemiology in directly studying low dose and low dose-rate exposures is that the signal, i.e. the excess numbers of cancers associated with low-level radiation exposure, is so very small that it cannot be seen against the very high background occurrence of cancer in the population, i.e. a lifetime risk of incidence reaching up to about 38% (i.e. 1 in 3 persons will develop a cancer in their lifetime). Thus, extrapolation models are used for the management of risk at low doses and low dose rates, but having adequate information from low dose and low dose-rate studies would be highly desirable. An overview of recently conducted radiation epidemiologic studies which evaluate risk following low-level radiation exposures is presented. Future improvements in risk assessment for radiation protection may come from increasingly informative epidemiologic studies, combined with mechanistic radiobiologic understanding of adverse outcome pathways, with both incorporated into biologically based models.
Collapse
Affiliation(s)
- J D Boice
- National Council on Radiation Protection and Measurements, Bethesda, Maryland, United States of America. Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | | | | |
Collapse
|
14986
|
Matsuo K, Huang Y, Zivanovic O, Shimada M, Machida H, Grubbs BH, Roman LD, Wright JD. Effectiveness of postoperative chemotherapy for stage IC mucinous ovarian cancer. Gynecol Oncol 2019; 154:505-515. [PMID: 31279493 DOI: 10.1016/j.ygyno.2019.06.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 06/17/2019] [Accepted: 06/22/2019] [Indexed: 12/24/2022]
Abstract
OBJECTIVE To examine the association between postoperative chemotherapy and survival of women with stage IC mucinous ovarian cancer (MOC). METHODS Comprehensive nationwide tumor registry data from the Commission on Cancer-accredited facilities in the United States from 2004 to 2014 were retrospectively examined. Women with stage IC MOC who underwent primary surgery followed by postoperative chemotherapy were compared to those who did not receive. Clinico-pathological factors associated with chemotherapy use, and overall survival associated with chemotherapy use were examined with multivariable models and propensity score inverse probability of treatment weighting (IPTW). External validation was performed by examining the Surveillance, Epidemiology, and End Results Program from 1988 to 2014. RESULTS There were 532 (58.5%) women who received postoperative chemotherapy and 377 (41.5%) women who did not. On multivariable analysis, those with moderately-/poorly-differentiated tumors, large tumor size, and who underwent lymphadenectomy were more likely to receive postoperative chemotherapy whereas young women and those with capsule rupture alone were less likely to receive postoperative chemotherapy (all, P < 0.05). After IPTW, there was no difference in overall survival among women who received postoperative chemotherapy versus those who did not on multivariable analysis (adjusted 4-year rates: 85.8% versus 86.3%, adjusted-hazard ratio [HR] 0.88, 95% confidence interval [CI] 0.60-1.31). Similarly, there was no benefit with chemotherapy regardless of patient age, tumor differentiation, performance of nodal dissection, and substage groups. Among 912 cases in the validation cohort (postoperative chemotherapy use, n = 520 [57.0%]), postoperative chemotherapy use was not associated with cause-specific survival (adjusted-HR 1.296, 95% CI 0.846-1.984, P = 0.233) or overall survival (adjusted-HR 1.131, 95% CI 0.849-1.508, P = 0.400). CONCLUSION Postoperative chemotherapy was received by fewer than 60% of women with stage IC MOC, and postoperative chemotherapy was not associated with improved survival.
Collapse
Affiliation(s)
- Koji Matsuo
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Southern California, Los Angeles, CA, USA; Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA.
| | - Yongmei Huang
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Oliver Zivanovic
- Gynecology Service, Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Muneaki Shimada
- Department of Obstetrics and Gynecology, Tohoku University School of Medicine, Miyagi, Japan
| | - Hiroko Machida
- Department of Obstetrics and Gynecology, Tokai University School of Medicine, Kanagawa, Japan
| | - Brendan H Grubbs
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of Southern California, Los Angeles, CA, USA
| | - Lynda D Roman
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Southern California, Los Angeles, CA, USA; Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Jason D Wright
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Columbia University College of Physicians and Surgeons, New York, NY, USA.
| |
Collapse
|
14987
|
Mou K, Zhang X, Mu X, Ge R, Han D, Zhou Y, Wang L. LNMAT1 Promotes Invasion-Metastasis Cascade in Malignant Melanoma by Epigenetically Suppressing CADM1 Expression. Front Oncol 2019; 9:569. [PMID: 31334110 PMCID: PMC6617740 DOI: 10.3389/fonc.2019.00569] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Accepted: 06/12/2019] [Indexed: 12/11/2022] Open
Abstract
The invasion-metastasis cascade is one of the most important factors relating to poor survival and prognosis of malignant melanoma (MM) patients. Long non-coding RNA lymph node metastasis associated transcript 1 (LNMAT1) is a key regulator in lymph node metastasis of multiple cancer types, but the roles and underlying mechanisms of LNMAT1 in the invasion-metastasis cascade of MM remain unclear. In the present study, we aimed to investigate the expression and function of LNMAT1 in MM. Here, we found that LNMAT1 was upregulated in MM tissues and cells, and its expression levels were further enhanced in MM patients with lymph node metastasis and metastatic MM cells. Using loss-of-function assays, we found that LNMAT1 promoted cell migration and invasion and lung metastasis in MM in vitro and in vivo. Moreover, we found that cell adhesion molecule 1 (CADM1), the established tumor suppressor in MM, was the downstream target of LNMAT1. Mechanistically, LNMAT1 epigenetically suppressed CADM1 expression by recruiting EZH2, the key regulator of trimethylation of histone H3 at lysine 27 (H3K27me3), to the CADM1 promoter, resulting in transcriptional inhibition of CADM1. Lastly, rescue assays demonstrated that LNMAT1 promoted cell migration and invasion of MM by suppressing CADM1 expression. Our findings elucidate a new mechanism for LNMAT1-mediated invasion-metastasis cascade in MM and suggest that LNMAT1 may be a new therapeutic target and prognostic predictor for MM.
Collapse
Affiliation(s)
- Kuanhou Mou
- Department of Dermatology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiang Zhang
- Department of Clinical Laboratory Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xin Mu
- Department of Dermatology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Rui Ge
- Department of Dermatology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Dan Han
- Department of Dermatology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yan Zhou
- Department of Dermatology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Lijuan Wang
- Department of Dermatology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
14988
|
Zhuang Y, Bai Y, Hu Y, Guo Y, Xu L, Hu W, Yang L, Zhao C, Li X, Zhao H. Rhein sensitizes human colorectal cancer cells to EGFR inhibitors by inhibiting STAT3 pathway. Onco Targets Ther 2019; 12:5281-5291. [PMID: 31308698 PMCID: PMC6618855 DOI: 10.2147/ott.s206833] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 05/28/2019] [Indexed: 12/12/2022] Open
Abstract
Background Activation of epidermal growth factor receptor (EGFR) has been reported in a variety of cancer types, including colorectal cancer (CRC), and represents a potential chemotherapeutic drug target. EGFR tyrosine kinase inhibitors (EGFR-TKIs) have been increasingly applied in the clinical treatment of CRC, but development of drug resistance during the treatment has greatly limited their application. Signal transducer and activator of transcription 3 (STAT3) and its mediated signal transduction pathway play an important role in the occurrence, development and metastasis of CRC, and are related to the development of EGFR-TKI resistance in CRC. Methods Cell viability, colony formation and cellular morphology were examined to evaluate the potent antiproliferative effect of the STAT3 inhibitor napabucasin, LY5 and rhein on the human CRC cell lines HCT116, SW620, RKO and DLD-1. Flow cytometry-based analysis was employed to determine whether rhein can affect the cell cycle and apoptosis. The expression level of phosphorylated STAT3 (P-STAT3), and cell cycle- and apoptosis-related proteins BCL2, CDC2 BAX, Cyclin D1 and Cyclin B1 were detected by Western blot analysis. Results This study revealed that rhein can significantly reduce cell viability and stimulate apoptosis in human CRC cells in a dose-dependent manner. In addition, rhein induced cell cycle arrest at the G2/M phase in CRC cells and dose-dependently inhibited the expression of cell cycle-related proteins. Additionally, it was found that napabucasin, LY5 and rhein considerably sensitized cells to the EGFR-TKI erlotinib, thus suppressing CRC cell proliferation. Rhein also inhibited the phosphorylation of its downstream target STAT3. Inhibition of STAT3 and EGFR phosphorylation was also observed after treatment with a combination of rhein and EGFR inhibitors. Conclusion This study confirmed the synergistic effect of STAT3 inhibitor and EGFR inhibitor in CRC cell lines. Additionally, we found that rhein sensitizes human CRC cells to EGFR-TKIs by inhibiting STAT3 pathway. When combined with EGFR-TKIs, rhein may be a novel STAT3 inhibitor in CRC.
Collapse
Affiliation(s)
- Yan Zhuang
- The Institute of Life Sciences, Wenzhou University, Wenzhou, Zhejiang 325035, People's Republic of China.,School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Ying Bai
- The Institute of Life Sciences, Wenzhou University, Wenzhou, Zhejiang 325035, People's Republic of China.,School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Yan Hu
- Department of Pharmacy, Taizhou Enze Medical Center (Group), Taizhou Hospital of Zhejiang Province, Taizhou, Zhejiang 317000, People's Republic of China
| | - Yueqin Guo
- The Institute of Life Sciences, Wenzhou University, Wenzhou, Zhejiang 325035, People's Republic of China.,School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Lingyuan Xu
- The Institute of Life Sciences, Wenzhou University, Wenzhou, Zhejiang 325035, People's Republic of China.,School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Wanle Hu
- Department of Coloproctology, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, People's Republic of China
| | - Lehe Yang
- The Institute of Life Sciences, Wenzhou University, Wenzhou, Zhejiang 325035, People's Republic of China.,School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Chengguang Zhao
- The Institute of Life Sciences, Wenzhou University, Wenzhou, Zhejiang 325035, People's Republic of China.,School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Xiaokun Li
- The Institute of Life Sciences, Wenzhou University, Wenzhou, Zhejiang 325035, People's Republic of China.,School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| | - Haiyang Zhao
- The Institute of Life Sciences, Wenzhou University, Wenzhou, Zhejiang 325035, People's Republic of China.,School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, People's Republic of China
| |
Collapse
|
14989
|
Chen S, Tauber G, Langsenlehner T, Schmölzer LM, Pötscher M, Riethdorf S, Kuske A, Leitinger G, Kashofer K, Czyż ZT, Polzer B, Pantel K, Sedlmayr P, Kroneis T, El-Heliebi A. In Vivo Detection of Circulating Tumor Cells in High-Risk Non-Metastatic Prostate Cancer Patients Undergoing Radiotherapy. Cancers (Basel) 2019; 11:E933. [PMID: 31277254 PMCID: PMC6678903 DOI: 10.3390/cancers11070933] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 06/20/2019] [Accepted: 06/28/2019] [Indexed: 12/13/2022] Open
Abstract
High-risk non-metastatic prostate cancer (PCa) has the potential to progress into lethal disease. Treatment options are manifold but, given a lack of surrogate biomarkers, it remains unclear which treatment offers the best results. Several studies have reported circulating tumor cells (CTCs) to be a prognostic biomarker in metastatic PCa. However, few reports on CTCs in high-risk non-metastatic PCa are available. Herein, we evaluated CTC detection in high-risk non-metastatic PCa patients using the in vivo CellCollector CANCER01 (DC01) and CellSearch system. CTC counts were analyzed and compared before and after radiotherapy (two sampling time points) in 51 high-risk non-metastatic PCa patients and were further compared according to isolation technique; further, CTC counts were correlated to clinical features. Use of DC01 resulted in a significantly higher percentage of CTC-positive samples compared to CellSearch (33.7% vs. 18.6%; p = 0.024) and yielded significantly higher CTC numbers (range: 0-15 vs. 0-5; p = 0.006). Matched pair analysis of samples between two sampling time points showed no difference in CTC counts determined by both techniques. CTC counts were not correlated with clinicopathological features. In vivo enrichment using DC01 has the potential to detect CTC at a higher efficiency compared to CellSearch, suggesting that CTC is a suitable biomarker in high-risk non-metastatic PCa.
Collapse
Affiliation(s)
- Shukun Chen
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Department of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, 8010 Graz, Austria
| | - Gerlinde Tauber
- Department of Therapeutic Radiology and Oncology, Medical University of Graz, 8036 Graz, Austria
| | - Tanja Langsenlehner
- Department of Therapeutic Radiology and Oncology, Medical University of Graz, 8036 Graz, Austria
| | - Linda Maria Schmölzer
- Department of Therapeutic Radiology and Oncology, Medical University of Graz, 8036 Graz, Austria
| | - Michaela Pötscher
- Department of Therapeutic Radiology and Oncology, Medical University of Graz, 8036 Graz, Austria
| | - Sabine Riethdorf
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Andra Kuske
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Gerd Leitinger
- Department of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, 8010 Graz, Austria
| | - Karl Kashofer
- Diagnostic and Research Institute of Pathology, Medical University Graz, 8036 Graz, Austria
| | - Zbigniew T Czyż
- Division Personalized Tumor Therapy, Fraunhofer Institute for Toxicology and Experimental Medicine, 93053 Regensburg, Germany
| | - Bernhard Polzer
- Division Personalized Tumor Therapy, Fraunhofer Institute for Toxicology and Experimental Medicine, 93053 Regensburg, Germany
| | - Klaus Pantel
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Peter Sedlmayr
- Department of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, 8010 Graz, Austria
| | - Thomas Kroneis
- Department of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, 8010 Graz, Austria.
| | - Amin El-Heliebi
- Department of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, 8010 Graz, Austria
- Center for Biomarker Research, CBmed, 8010 Graz, Austria
| |
Collapse
|
14990
|
Obesity-Induced Adipose Tissue Inflammation as a Strong Promotional Factor for Pancreatic Ductal Adenocarcinoma. Cells 2019; 8:cells8070673. [PMID: 31277269 PMCID: PMC6678863 DOI: 10.3390/cells8070673] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 06/28/2019] [Accepted: 07/02/2019] [Indexed: 02/07/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is expected to soon become the second leading cause of cancer related deaths in the United States. This may be due to the rising obesity prevalence, which is a recognized risk factor for PDAC. There is great interest in deciphering the underlying driving mechanisms of the obesity–PDAC link. Visceral adiposity has a strong correlation to certain metabolic diseases and gastrointestinal cancers, including PDAC. In fact, our own data strongly suggest that visceral adipose tissue inflammation is a strong promoter for PDAC growth and progression in a genetically engineered mouse model of PDAC and diet-induced obesity. In this review, we will discuss the relationship between obesity-associated adipose tissue inflammation and PDAC development, with a focus on the key molecular and cellular components in the dysfunctional visceral adipose tissue, which provides a tumor permissive environment.
Collapse
|
14991
|
Smith CG, Davenport DL, Gorski J, McDowell A, Burgess BT, Fredericks TI, Baldwin LA, Miller RW, DeSimone CP, Dietrich CS, Gallion HH, Pavlik EJ, van Nagell JR, Ueland FR. Clinical Factors Associated with Longer Hospital Stay Following Ovarian Cancer Surgery. Healthcare (Basel) 2019; 7:E85. [PMID: 31277282 PMCID: PMC6787623 DOI: 10.3390/healthcare7030085] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 06/23/2019] [Accepted: 06/24/2019] [Indexed: 12/18/2022] Open
Abstract
Background: Ovarian cancer (OC) is the leading cause of death from gynecologic malignancy and is treated with a combination of cytoreductive surgery and platinum-based chemotherapy. Extended length of stay (LOS) after surgery can affect patient morbidity, overall costs, and hospital resource utilization. The primary objective of this study was to identify factors contributing to prolonged LOS for women undergoing surgery for ovarian cancer. Methods: The American College of Surgeons National Surgical Quality Improvement Program (ACS-NSQIP) database was queried to identify women from 2012-2016 who underwent hysterectomy for ovarian, fallopian tube and peritoneal cancer. The primary outcome was LOS >50th percentile. Preoperative and intraoperative variables were examined to determine which were associated with prolonged LOS. Results: From 2012-2016, 1771 women underwent elective abdominal surgery for OC and were entered in the ACS-NSQIP database. The mean and median LOS was 4.6 and 4.0 days (IQR 0-38), respectively. On multivariate analysis, factors associated with prolonged LOS included: American Society of Anesthesiologists (ASA) Classification III (aOR 1.71, 95% CI 1.38-2.13) or IV (aOR 1.88, 95% CI 1.44-2.46), presence of ascites (aOR 1.88, 95% CI 1.44-2.46), older age (aOR 1.23, 95% CI 1.13-1.35), platelet count >400,000/mm3 (aOR 1.74, 95% CI 1.29-2.35), preoperative blood transfusion (aOR 11.00, 95% CI 1.28-94.77), disseminated cancer (aOR 1.28, 95% CI 1.03-1.60), increased length of operation (121-180 min, aOR 1.47, 95% CI 1.13-1.91; >180 min, aOR 2.78, 95% CI 2.13-3.64), and postoperative blood transfusion within 72 h of incision (aOR 2.04, 95% CI 1.59-2.62) (p < 0.05 for all). Conclusions: Longer length of hospital stay following surgery for OC is associated with many patient, disease, and treatment-related factors. The extent of surgery, as evidenced by perioperative blood transfusion and length of surgical procedure, is a factor that can potentially be modified to shorten LOS, improve patient outcomes, and reduce hospital costs.
Collapse
Affiliation(s)
- Christopher G Smith
- Department of Obstetrics & Gynecology, University of Kentucky, Lexington, KY 40536-0293, USA.
| | - Daniel L Davenport
- Department of Surgery, University of Kentucky, Lexington, KY 40536-0293, USA
| | - Justin Gorski
- Department of Obstetrics & Gynecology, University of Kentucky, Lexington, KY 40536-0293, USA
- Division of Gynecologic Oncology, Markey Cancer Center, University of Kentucky, Lexington, KY 40536-0293, USA
| | - Anthony McDowell
- Department of Obstetrics & Gynecology, University of Kentucky, Lexington, KY 40536-0293, USA
- Division of Gynecologic Oncology, Markey Cancer Center, University of Kentucky, Lexington, KY 40536-0293, USA
| | - Brian T Burgess
- Department of Obstetrics & Gynecology, University of Kentucky, Lexington, KY 40536-0293, USA
- Division of Gynecologic Oncology, Markey Cancer Center, University of Kentucky, Lexington, KY 40536-0293, USA
| | - Tricia I Fredericks
- Department of Obstetrics & Gynecology, University of Kentucky, Lexington, KY 40536-0293, USA
- Division of Gynecologic Oncology, Markey Cancer Center, University of Kentucky, Lexington, KY 40536-0293, USA
| | - Lauren A Baldwin
- Department of Obstetrics & Gynecology, University of Kentucky, Lexington, KY 40536-0293, USA
- Division of Gynecologic Oncology, Markey Cancer Center, University of Kentucky, Lexington, KY 40536-0293, USA
| | - Rachel W Miller
- Department of Obstetrics & Gynecology, University of Kentucky, Lexington, KY 40536-0293, USA
- Division of Gynecologic Oncology, Markey Cancer Center, University of Kentucky, Lexington, KY 40536-0293, USA
| | - Christopher P DeSimone
- Department of Obstetrics & Gynecology, University of Kentucky, Lexington, KY 40536-0293, USA
- Division of Gynecologic Oncology, Markey Cancer Center, University of Kentucky, Lexington, KY 40536-0293, USA
| | - Charles S Dietrich
- Department of Obstetrics & Gynecology, University of Kentucky, Lexington, KY 40536-0293, USA
- Division of Gynecologic Oncology, Markey Cancer Center, University of Kentucky, Lexington, KY 40536-0293, USA
| | - Holly H Gallion
- Department of Obstetrics & Gynecology, University of Kentucky, Lexington, KY 40536-0293, USA
- Division of Gynecologic Oncology, Markey Cancer Center, University of Kentucky, Lexington, KY 40536-0293, USA
| | - Edward J Pavlik
- Department of Obstetrics & Gynecology, University of Kentucky, Lexington, KY 40536-0293, USA
- Division of Gynecologic Oncology, Markey Cancer Center, University of Kentucky, Lexington, KY 40536-0293, USA
| | - John R van Nagell
- Department of Obstetrics & Gynecology, University of Kentucky, Lexington, KY 40536-0293, USA
- Division of Gynecologic Oncology, Markey Cancer Center, University of Kentucky, Lexington, KY 40536-0293, USA
| | - Frederick R Ueland
- Department of Obstetrics & Gynecology, University of Kentucky, Lexington, KY 40536-0293, USA
- Division of Gynecologic Oncology, Markey Cancer Center, University of Kentucky, Lexington, KY 40536-0293, USA
| |
Collapse
|
14992
|
Utne I, Løyland B, Grov EK, Paul S, Wong ML, Conley YP, Cooper BA, Levine JD, Miaskowski C. Co-occuring symptoms in older oncology patients with distinct attentional function profiles. Eur J Oncol Nurs 2019; 41:196-203. [PMID: 31358253 DOI: 10.1016/j.ejon.2019.07.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 06/30/2019] [Accepted: 07/02/2019] [Indexed: 02/07/2023]
Abstract
PURPOSE Evaluate how subgroups of older adults with distinct attentional function profiles differ on the severity of nine common symptoms and determine demographic and clinical characteristics and symptom severity scores associated with membership in the low and moderate attentional function classes. METHODS Three subgroups of older oncology outpatients were identified using latent profile analysis based on Attentional Function Index (AFI) scores. Symptoms were assessed prior to the second or third cycle of CTX. Logistic regressions evaluated for associations with attentional function class membership. RESULTS For trait anxiety, state anxiety, depression, sleep disturbance, morning fatigue, and evening fatigue scores, differences among the latent classes followed the same pattern (low > moderate > high). For morning and evening energy, compared to high class, patients in low and moderate classes reported lower scores. For pain, compared to moderate class, patients in low class reported higher scores. In the logistic regression analysis, compared to high class, patients with lower income, higher comorbidity, higher CTX toxicity score, and higher levels of state anxiety, depression, and sleep disturbance were more likely to be in low AFI class. Compared to high class, patients with higher comorbidity and trait anxiety and lower morning energy were more likely to be in moderate AFI class. CONCLUSIONS Consistent with the hypothesis that an increased risk for persistent cognitive decline is likely related to a variety of physical and psychological factors, for six of the nine symptoms, a "dose response" effect was observed with higher symptom severity scores associated with a progressive decline in attentional function.
Collapse
Affiliation(s)
- Inger Utne
- Department of Nursing and Health Promotion, Faculty of Health Sciences, OsloMet - Oslo Metropolitan University, Oslo, Norway
| | - Borghild Løyland
- Department of Nursing and Health Promotion, Faculty of Health Sciences, OsloMet - Oslo Metropolitan University, Oslo, Norway
| | - Ellen Karine Grov
- Department of Nursing and Health Promotion, Faculty of Health Sciences, OsloMet - Oslo Metropolitan University, Oslo, Norway
| | - Steven Paul
- School of Nursing, University of California, San Francisco, CA, USA
| | - Melisa L Wong
- School of Medicine, University of California, San Francisco, CA, USA
| | - Yvette P Conley
- School of Nursing, University of Pittsburgh, Pittsburgh, PA, USA
| | - Bruce A Cooper
- School of Nursing, University of California, San Francisco, CA, USA
| | - Jon D Levine
- School of Medicine, University of California, San Francisco, CA, USA
| | | |
Collapse
|
14993
|
Floudas CS, Brar G, Mabry-Hrones D, Duffy AG, Wood B, Levy E, Krishnasamy V, Fioravanti S, Bonilla CM, Walker M, Morelli MP, Kleiner DE, Steinberg SM, Figg WD, Greten TF, Xie C. A Pilot Study of the PD-1 Targeting Agent AMP-224 Used With Low-Dose Cyclophosphamide and Stereotactic Body Radiation Therapy in Patients With Metastatic Colorectal Cancer. Clin Colorectal Cancer 2019; 18:e349-e360. [PMID: 31351862 DOI: 10.1016/j.clcc.2019.06.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 06/18/2019] [Accepted: 06/25/2019] [Indexed: 01/18/2023]
Abstract
BACKGROUND The prognosis of metastatic colorectal cancer (mCRC) is poor. We assessed the feasibility, safety, and efficacy of the anti-programmed cell death 1 fusion protein AMP-224 in combination with low-dose cyclophosphamide and stereotactic body radiation (SBRT) treatment in patients with mCRC refractory to standard chemotherapy. PATIENTS AND METHODS Fifteen patients were enrolled. Six received SBRT 8 Gy on day 0 (dose level 1), whereas 9 received 8 Gy on days -2 to day 0. All received cyclophosphamide 200 mg/m2 intravenously (I.V.) on day 0. On day 1, both groups received AMP-224 10 mg/kg I.V., repeated every 2 weeks for a total of 6 doses. Primary end points were feasibility and safety. RESULTS Ten (67%) patients completed 6 doses of AMP-224; 5 patients (33%) discontinued treatment because of disease progression. No dose-limiting toxicity was observed; 9 patients (60%) experienced treatment-related adverse events, all Grade 1 or 2. No objective response was noted; 3 patients (20%) had stable disease. Median progression-free survival and overall survival were 2.8 months (95% confidence interval [CI], 1.2-2.8 months) and 6.0 months (95% CI, 2.8-9.6 months), respectively. M2 macrophage polarization was present in the pretreatment tumor biopsy samples, but not post-treatment samples. CONCLUSION AMP-224 in combination with SBRT and low-dose cyclophosphamide was well tolerated, however, no significant clinical benefit was observed in patients with mCRC.
Collapse
Affiliation(s)
- Charalampos S Floudas
- Hematology/Oncology Fellowship Program, National Heart, Lung, and Blood Institute/National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Gagandeep Brar
- Hematology/Oncology Fellowship Program, National Heart, Lung, and Blood Institute/National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Donna Mabry-Hrones
- Gastrointestinal Malignancies Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Austin G Duffy
- Gastrointestinal Malignancies Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Bradford Wood
- Radiology and Imaging Sciences, Center for Cancer Research, National Institutes of Health, Bethesda, MD
| | - Elliot Levy
- Radiology and Imaging Sciences, Center for Cancer Research, National Institutes of Health, Bethesda, MD
| | - Venkatesh Krishnasamy
- Radiology and Imaging Sciences, Center for Cancer Research, National Institutes of Health, Bethesda, MD
| | - Suzanne Fioravanti
- Gastrointestinal Malignancies Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Cecilia M Bonilla
- Hematology/Oncology Fellowship Program, National Heart, Lung, and Blood Institute/National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Melissa Walker
- Gastrointestinal Malignancies Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Maria Pia Morelli
- Hematology/Oncology Fellowship Program, National Heart, Lung, and Blood Institute/National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - David E Kleiner
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Seth M Steinberg
- Biostatistics and Data Management Section, Center for Cancer Research, National Institutes of Health, Bethesda, MD
| | - William D Figg
- Clinical Pharmacology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Tim F Greten
- Gastrointestinal Malignancies Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD; National Cancer Institute, Center for Cancer Research, Liver Cancer Program, National Institutes of Health, Bethesda, MD.
| | - Changqing Xie
- Gastrointestinal Malignancies Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD.
| |
Collapse
|
14994
|
Whole Genome Analysis and Prognostic Model Construction Based on Alternative Splicing Events in Endometrial Cancer. BIOMED RESEARCH INTERNATIONAL 2019; 2019:2686875. [PMID: 31355251 PMCID: PMC6634061 DOI: 10.1155/2019/2686875] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 06/04/2019] [Indexed: 12/31/2022]
Abstract
Objectives A growing body of evidence has shown that aberrant alternative splicing (AS) is closely related to the occurrence and development of cancer. However, prior studies mainly have concentrated on a few genes that exhibit aberrant AS. This study aimed to determine AS events through whole genome analysis and construct a prognostic model of endometrial cancer (EC). Methods We downloaded gene expression RNAseq data from UCSC Xena, and seven types of AS events from TCGA SpliceSeq. Univariate Cox regression was employed to analyze the prognostic-related alternative splicing events (PASEs) and splicing factors; multivariate Cox regression was conducted to analyze the effect of risk score (All) and clinicopathological parameters on EC prognosis. An underlying interaction network of PASEs of EC was constructed by Cytoscape Reactome FI, GO, and KEGG pathway enrichment was performed by DAVID. ROC curves and Kaplan-Meir analysis were used to assess the diagnostic value of prognostic model. The correlation between PASEs and splicing factors was analyzed by GraphPad Prism; then a network was constructed using Cytoscape. Results In total, 28,281 AS events in EC were identified, which consisted of 1166 PASEs. RNPS1, NEK2, and CTNNB1 were the hub genes in the network of the top 600 PASEs. The area under the curve (AUC) of risk score (All) reached 0.819. Risk score (All) together with FIGO stage, cancer status, and primary therapy outcome success was risk factors of the prognosis of EC patients. Splicing factors YBX1, HNRNPDL, and HNRNPA1 were significantly related to the overall survival (OS). The splicing network indicated that the expression of splicing factors was significantly correlated with percent-splice-in (PSI) value of PASEs. Conclusion We constructed a model for predicting the prognosis of EC patients based on PASEs using whole genome analysis of AS events and thereby provided a reliable theoretical basis for EC clinical prognosis evaluation.
Collapse
|
14995
|
Tian W, Liu S, Li B. Potential Role of Exosomes in Cancer Metastasis. BIOMED RESEARCH INTERNATIONAL 2019; 2019:4649705. [PMID: 31355262 PMCID: PMC6634128 DOI: 10.1155/2019/4649705] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 04/02/2019] [Accepted: 04/24/2019] [Indexed: 12/20/2022]
Abstract
High cancer mortality is attributed to metastasis to a large extent. However, cancer metastasis remains devoid of dynamic monitoring and early prevention in terms of current advances in diagnostic means and therapeutic modalities. Meanwhile, studies have shown that reciprocal crosstalk among cells via exosomes plays a critical role in maintaining normal physiological state or triggering disease progression, including cancer metastasis. Therefore, in this review, we focus on the latest literature (primarily from 2018) to summarize action mechanisms and experimental studies of exosomes in cancer metastasis and put forward some problems as well as new outlooks of these studies.
Collapse
Affiliation(s)
- Wenjuan Tian
- Department of Clinical Laboratory, Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
- School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Shanshan Liu
- Department of Clinical Laboratory, Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
- School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Burong Li
- Department of Clinical Laboratory, Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| |
Collapse
|
14996
|
Olender J, Wang BD, Ching T, Garmire LX, Garofano K, Ji Y, Knox T, Latham P, Nguyen K, Rhim J, Lee NH. A Novel FGFR3 Splice Variant Preferentially Expressed in African American Prostate Cancer Drives Aggressive Phenotypes and Docetaxel Resistance. Mol Cancer Res 2019; 17:2115-2125. [PMID: 31266816 DOI: 10.1158/1541-7786.mcr-19-0415] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 06/19/2019] [Accepted: 06/21/2019] [Indexed: 01/08/2023]
Abstract
Alternative splicing (AS) has been shown to participate in prostate cancer development and progression; however, a link between AS and prostate cancer health disparities has been largely unexplored. Here we report on the cloning of a novel splice variant of FGFR3 that is preferentially expressed in African American (AA) prostate cancer. This novel variant (FGFR3-S) omits exon 14, comprising 123 nucleotides that encode the activation loop in the intracellular split kinase domain. Ectopic overexpression of FGFR3-S in European American (EA) prostate cancer cell lines (PC-3 and LNCaP) led to enhanced receptor autophosphorylation and increased activation of the downstream signaling effectors AKT, STAT3, and ribosomal S6 compared with FGFR3-L (retains exon 14). The increased oncogenic signaling imparted by FGFR3-S was associated with a substantial gain in proliferative and antiapoptotic activities, as well as a modest but significant gain in cell motility. Moreover, the FGFR3-S-conferred proliferative and motility gains were highly resistant to the pan-FGFR small-molecule inhibitor dovitinib and the antiapoptotic gain was insensitive to the cytotoxic drug docetaxel, which stands in marked contrast with dovitinib- and docetaxel-sensitive FGFR3-L. In an in vivo xenograft model, mice injected with PC-3 cells overexpressing FGFR3-S exhibited significantly increased tumor growth and resistance to dovitinib treatment compared with cells overexpressing FGFR3-L. In agreement with our in vitro and in vivo findings, a high FGFR3-S/FGFR3-L expression ratio in prostate cancer specimens was associated with poor patient prognosis. IMPLICATIONS: This work identifies a novel FGFR3 splice variant and supports the hypothesis that differential AS participates in prostate cancer health disparities.
Collapse
Affiliation(s)
- Jacqueline Olender
- Department of Pharmacology and Physiology, The George Washington University School of Medicine and Health Sciences, GW Cancer Center, Washington, D. C
| | - Bi-Dar Wang
- Department of Pharmaceutical Sciences, School of Pharmacy and Health Professions, University of Maryland Eastern Shore, Princess Anne, Maryland
| | - Travers Ching
- Cancer Epidemiology Program, University of Hawaii, Honolulu, Hawaii
| | - Lana X Garmire
- Department of Computational Medicine and Bioinformatics, School of Medicine, University of Michigan, Ann Arbor, Michigan
| | - Kaitlin Garofano
- Department of Pharmacology and Physiology, The George Washington University School of Medicine and Health Sciences, GW Cancer Center, Washington, D. C
| | - Youngmi Ji
- Adeno-Associated Virus Biology Section, National Institute of Dental and Craniofacial Research, NIH, Bethesda, Maryland
| | - Tessa Knox
- Department of Pharmacology and Physiology, The George Washington University School of Medicine and Health Sciences, GW Cancer Center, Washington, D. C
| | - Patricia Latham
- Department of Pathology, The George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | - Kenneth Nguyen
- Department of Pharmacology and Physiology, The George Washington University School of Medicine and Health Sciences, GW Cancer Center, Washington, D. C
| | - Johng Rhim
- Center for Prostate Disease Research, Department of Surgery, Uniformed Services University of Health Sciences, Bethesda, Maryland
| | - Norman H Lee
- Department of Pharmacology and Physiology, The George Washington University School of Medicine and Health Sciences, GW Cancer Center, Washington, D. C.
| |
Collapse
|
14997
|
Sultana S, Song DY, Lee J. Deformable registration of PET/CT and ultrasound for disease-targeted focal prostate brachytherapy. J Med Imaging (Bellingham) 2019; 6:035003. [PMID: 31528661 PMCID: PMC6739636 DOI: 10.1117/1.jmi.6.3.035003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 08/20/2019] [Indexed: 12/27/2022] Open
Abstract
We propose a deformable registration algorithm for prostate-specific membrane antigen (PSMA) PET/CT and transrectal ultrasound (TRUS) fusion. Accurate registration of PSMA PET to intraoperative TRUS will allow physicians to customize dose planning based on the regions involved. The inputs to the registration algorithm are the PET/CT and TRUS volumes as well as the prostate segmentations. PET/CT and TRUS volumes are first rigidly registered by maximizing the overlap between the segmented prostate binary masks. Three-dimensional anatomical landmarks are then automatically extracted from the boundary as well as within the prostate. Then, a deformable registration is performed using a regularized thin plate spline where the landmark localization error is optimized between the extracted landmarks that are in correspondence. The proposed algorithm was evaluated on 25 prostate cancer patients treated with low-dose-rate brachytherapy. We registered the postimplant CT to TRUS using the proposed algorithm and computed target registration errors (TREs) by comparing implanted seed locations. Our approach outperforms state-of-the-art methods, with significantly lower ( mean ± standard deviation ) TRE of 1.96 ± 1.29 mm while being computationally efficient (mean computation time of 38 s). The proposed landmark-based PET/CT-TRUS deformable registration algorithm is simple, computationally efficient, and capable of producing quality registration of the prostate boundary as well as the internal gland.
Collapse
Affiliation(s)
- Sharmin Sultana
- Johns Hopkins University, Department of Radiation Oncology and Molecular Radiation Sciences, Baltimore, Maryland, United States
| | - Daniel Y. Song
- Johns Hopkins University, Department of Radiation Oncology and Molecular Radiation Sciences, Baltimore, Maryland, United States
| | - Junghoon Lee
- Johns Hopkins University, Department of Radiation Oncology and Molecular Radiation Sciences, Baltimore, Maryland, United States
| |
Collapse
|
14998
|
Said N. Establishing and characterization of human and murine bladder cancer organoids. Transl Androl Urol 2019; 8:S310-S313. [PMID: 31392155 PMCID: PMC6642964 DOI: 10.21037/tau.2019.06.05] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 06/06/2019] [Indexed: 01/01/2023] Open
Affiliation(s)
- Neveen Said
- Cancer Biology Department, Wake Forest Baptist Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Pathology Department, Wake Forest Baptist Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Urology Department, Wake Forest Baptist Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
14999
|
Ou-Yang F, Tsai IH, Tang JY, Yen CY, Cheng YB, Farooqi AA, Chen SR, Yu SY, Kao JK, Chang HW. Antiproliferation for Breast Cancer Cells by Ethyl Acetate Extract of Nepenthes thorellii x ( ventricosa x maxima). Int J Mol Sci 2019; 20:ijms20133238. [PMID: 31266224 PMCID: PMC6651324 DOI: 10.3390/ijms20133238] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 06/25/2019] [Accepted: 06/28/2019] [Indexed: 12/15/2022] Open
Abstract
Extracts from the Nepenthes plant have anti-microorganism and anti-inflammation effects. However, the anticancer effect of the Nepenthes plant is rarely reported, especially for breast cancer cells. Here, we evaluate the antitumor effects of the ethyl acetate extract of Nepenthesthorellii x (ventricosa x maxima) (EANT) against breast cancer cells. Cell viability and flow cytometric analyses were used to analyze apoptosis, oxidative stress, and DNA damage. EANT exhibits a higher antiproliferation ability to two breast cancer cell lines (MCF7 and SKBR3) as compared to normal breast cells (M10). A mechanistic study demonstrates that EANT induces apoptosis in breast cancer cells with evidence of subG1 accumulation and annexin V increment. EANT also induces glutathione (GSH) depletion, resulting in dramatic accumulations of reactive oxygen species (ROS) and mitochondrial superoxide (MitoSOX), as well as the depletion of mitochondrial membrane potential (MMP). These oxidative stresses attack DNA, respectively leading to DNA double strand breaks and oxidative DNA damage in γH2AX and 8-oxo-2′deoxyguanosine (8-oxodG) assays. Overall these findings clearly revealed that EANT induced changes were suppressed by the ROS inhibitor. In conclusion, our results have shown that the ROS-modulating natural product (EANT) has antiproliferation activity against breast cancer cells through apoptosis, oxidative stress, and DNA damage.
Collapse
Affiliation(s)
- Fu Ou-Yang
- Division of Breast Surgery and Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
- Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
| | - I-Hsuan Tsai
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Jen-Yang Tang
- Department of Radiation Oncology, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
| | - Ching-Yu Yen
- Department of Oral and Maxillofacial Surgery Chi-Mei Medical Center, Tainan 71004, Taiwan
- School of Dentistry, Taipei Medical University, Taipei 11050, Taiwan
| | - Yuan-Bin Cheng
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Ammad Ahmad Farooqi
- Institute of Biomedical and Genetic Engineering (IBGE), Islamabad 44000, Pakistan
| | - Shu-Rong Chen
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Szu-Yin Yu
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Jun-Kai Kao
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung 40227, Taiwan.
- Pediatric Department, Children's Hospital, Changhua Christian Hospital, Changhua 50006, Taiwan.
- School of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| | - Hsueh-Wei Chang
- Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan.
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 80424, Taiwan.
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan.
| |
Collapse
|
15000
|
Park JY, Luu HN, Park HY, Lin HY, Radlein S, Di Pietro G, Yeo CD, Kim SJ, Kang N, Antwi S, Sexton WJ, Spiess PE, Dickinson S, Parker A. Telomere length in peripheral blood leukocytes and risk of renal cell carcinoma. Transl Cancer Res 2019; 8:S397-S403. [PMID: 35117117 PMCID: PMC8798989 DOI: 10.21037/tcr.2019.06.36] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 06/12/2019] [Indexed: 11/06/2022]
Abstract
BACKGROUND Telomeres are essential for chromosomal stability and may play a key role in carcinogenesis. Telomere length is suggested as a tentative biomarker of risk for renal cell carcinoma (RCC). However, results of previous association studies between telomere length and risk for RCC are inconsistent. METHODS We evaluated RCC risk in relation to peripheral blood leukocyte telomere length using a hospital-based case-control study of 169 RCC cases and 189 controls. Cases were histologically-confirmed RCC patients who were treated at the Moffitt Cancer Center (Tampa, FL). Controls with no history of cancer underwent a screening exam at the Lifetime Cancer Screening Center at Moffitt Cancer Center to rule out the presence of cancer. Relative telomere length (RTL) was measured by quantitative real-time polymerase chain reaction (PCR) using peripheral blood leukocyte DNA. Logistic regression was used to determine the association between RTL and RCC risk. RESULTS As expected, increasing age was inversely correlated with RTL (Pearson r=-0.213, P=0.003) among controls but not cases. Average RTL was significantly shorter in cases as compared with controls [mean ± standard deviation (SD): 3.18±1.50 and 4.39±1.99, respectively, P<0.001]. In contrast, average RTL was not significantly different by gender, race, smoking status among controls or by clinical stages among RCC cases. In regression analysis, we observed that shorter RTL is significantly associated with RCC risk [odds ratio (OR) =1.48; 95% confidence interval (CI): 1.27-1.71] after adjustment for covariates. CONCLUSIONS We found that shorter RTL is associated with an increased risk for RCC. Our findings suggest that telomere length may be involved in the development of RCC.
Collapse
Affiliation(s)
- Jong Y. Park
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL, USA
| | - Hung N. Luu
- Division of Cancer Control and Population Sciences, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hyun Y. Park
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL, USA
| | - Hui-Yi Lin
- Biostatistics Program, School of Public Health, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Selina Radlein
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL, USA
| | | | - Chang Dong Yeo
- Division of Pulmonology, Department of Internal Medicine, The Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seung Joon Kim
- Division of Pulmonology, Department of Internal Medicine, The Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Nahyeon Kang
- Division of Pulmonology, Department of Internal Medicine, The Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Samuel Antwi
- Department of Health Science Research, Mayo Clinic, Jacksonville, FL, USA
| | - Wade J. Sexton
- Department of Genitourinary Oncology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Philippe E. Spiess
- Department of Genitourinary Oncology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Shohreh Dickinson
- Department of Genitourinary Oncology, Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Alexander Parker
- University of Florida Health Science Center, Jacksonville, FL, USA
| |
Collapse
|