1
|
Nie J, Yang H, Liu X, Deng W, Fu B. Identification and validation of shared gene signature of kidney renal clear cell carcinoma and COVID-19. PeerJ 2024; 12:e16927. [PMID: 38464749 PMCID: PMC10921934 DOI: 10.7717/peerj.16927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 01/22/2024] [Indexed: 03/12/2024] Open
Abstract
Background COVID-19 is a severe infectious disease caused by the SARS-CoV-2 virus, and previous studies have shown that patients with kidney renal clear cell carcinoma (KIRC) are more susceptible to SARS-CoV-2 infection than the general population. Nevertheless, their co-pathogenesis remains incompletely elucidated. Methods We obtained shared genes between these two diseases based on public datasets, constructed a prognostic risk model consisting of hub genes, and validated the accuracy of the model using internal and external validation sets. We further analyzed the immune landscape of the prognostic risk model, investigated the biological functions of the hub genes, and detected their expression in renal cell carcinoma cells using qPCR. Finally, we searched the candidate drugs associated with hub gene-related targets from DSigDB and CellMiner databases. Results We obtained 156 shared genes between KIRC and COVID-19 and constructed a prognostic risk model consisting of four hub genes. Both shared genes and hub genes were highly enriched in immune-related functions and pathways. Hub genes were significantly overexpressed in COVID-19 and KIRC. ROC curves, nomograms, etc., showed the reliability and robustness of the risk model, which was validated in both internal and external datasets. Moreover, patients in the high-risk group showed a higher proportion of immune cells, higher expression of immune checkpoint genes, and more active immune-related functions. Finally, we identified promising drugs for COVID-19 and KIRC, such as etoposide, fulvestrant, and topotecan. Conclusion This study identified and validated four shared genes for KIRC and COVID-19. These genes are associated with immune functions and may serve as potential prognostic biomarkers for KIRC. The shared pathways and genes may provide new insights for further mechanistic research and treatment of comorbidities.
Collapse
Affiliation(s)
- Jianqiang Nie
- First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Hailang Yang
- First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiaoqiang Liu
- First Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Institute of Urology, Nanchang, China
| | - Wen Deng
- First Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Institute of Urology, Nanchang, China
| | - Bin Fu
- First Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Institute of Urology, Nanchang, China
| |
Collapse
|
2
|
Chen Y, Markov N, Gigon L, Hosseini A, Yousefi S, Stojkov D, Simon HU. The BK Channel Limits the Pro-Inflammatory Activity of Macrophages. Cells 2024; 13:322. [PMID: 38391935 PMCID: PMC10886595 DOI: 10.3390/cells13040322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 01/31/2024] [Accepted: 02/07/2024] [Indexed: 02/24/2024] Open
Abstract
Macrophages play a crucial role in the innate immune response, serving as key effector cells in the defense against pathogens. Although the role of the large-conductance voltage and calcium-activated potassium channel, also known as the KCa1.1 or BK channel, in regulating neurotransmitter release and smooth muscle contraction is well known, its potential involvement in immune regulation remains unclear. We employed BK-knockout macrophages and noted that the absence of a BK channel promotes the polarization of macrophages towards a pro-inflammatory phenotype known as M1 macrophages. Specifically, the absence of the BK channel resulted in a significant increase in the secretion of the pro-inflammatory cytokine IL-6 and enhanced the activity of extracellular signal-regulated kinases 1 and 2 (Erk1/2 kinases), Ca2+/calmodulin-dependent protein kinase II (CaMKII), and the transcription factor ATF-1 within M1 macrophages. Additionally, the lack of the BK channel promoted the activation of the AIM2 inflammasome without affecting the activation of the NLRC4 and NLRP3 inflammasomes. To further investigate the role of the BK channel in regulating AIM2 inflammasome activation, we utilized BK channel inhibitors, such as paxilline and iberiotoxin, along with the BK channel activator NS-11021. Pharmacological inactivation of the BK channel increased, and its stimulation inhibited IL-1β production following AIM2 inflammasome activation in wild-type macrophages. Moreover, wild-type macrophages displayed increased calcium influx when activated with the AIM2 inflammasome, whereas BK-knockout macrophages did not due to the impaired extracellular calcium influx upon activation. Furthermore, under conditions of a calcium-free medium, IL-1β production following AIM2 inflammasome activation was increased in both wild-type and BK-knockout macrophages. This suggests that the BK channel is required for the influx of extracellular calcium in macrophages, thus limiting AIM2 inflammasome activation. In summary, our study reveals a regulatory role of the BK channel in macrophages under inflammatory conditions.
Collapse
Affiliation(s)
- Yihe Chen
- Institute of Pharmacology, University of Bern, 3010 Bern, Switzerland
| | - Nikita Markov
- Institute of Pharmacology, University of Bern, 3010 Bern, Switzerland
| | - Lea Gigon
- Institute of Pharmacology, University of Bern, 3010 Bern, Switzerland
| | - Aref Hosseini
- Institute of Pharmacology, University of Bern, 3010 Bern, Switzerland
| | - Shida Yousefi
- Institute of Pharmacology, University of Bern, 3010 Bern, Switzerland
| | - Darko Stojkov
- Institute of Pharmacology, University of Bern, 3010 Bern, Switzerland
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, 3010 Bern, Switzerland
- Institute of Biochemistry, Brandenburg Medical School, 16816 Neuruppin, Germany
| |
Collapse
|
3
|
Chen F, Jiang J, Liu D, Li H, Dong L, Song Y, Zhang Y, Wang J, Qin Y, Zhao G. The lncRNA lnc-TSI antagonizes sorafenib resistance in hepatocellular carcinoma via downregulating miR-4726-5p expression and upregulating KCNMA1 expression. J Mol Histol 2024; 55:83-96. [PMID: 38165571 DOI: 10.1007/s10735-023-10173-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 10/25/2023] [Indexed: 01/04/2024]
Abstract
Acquired drug resistance is a main reason for limiting the application of sorafenib in HCC treatment. This study aimed to explore the role and mechanisms of a novel long non-coding RNA (lncRNA), lnc-TSI, in sorafenib resistance of HCC. The interaction between lnc-TSI and miR-4726-5p, and miR-4726-5p and KCNMA1 were predicted using bioinformatic tools. Expression of the molecules in the lnc-TSI/miR-4726-5p/KCNMA1 axis in clinical samples and cell lines, as well as the sorafenib resistant HCC cell lines, was determined using qRT-PCR or western blotting. Expressions of lnc-TSI, miR-4726-5p, and KCNMA1 were manipulated in HepG2 and Huh7 cells through plasmid transfection or lentivirus infection. The CCK-8, flow cytometry, and Tunel assays were employed to determine the role of this axis on sorafenib resistance of HCC. A xenograft model was established using sorafenib-resistant HepG2 and Huh7 cells followed by in vivo sorafenib treatments to confirm the in vitro findings. Lnc-TSI and KCNMA1 expressions were significantly downregulated in HCC clinical samples and cell lines, especially in sorafenib resistance ones, while mi-4726-5p presented a reversed expression pattern. Lnc-TSI interacted with miR-4726-5p, and Lnc-TSI acts as a ceRNA via sponging miR-4726-5p in HCC cells. Overexpression of lnc-TSI and KCNMA1 promoted apoptosis and decreased cell viability of sorafenib-treated HCC cells, thus alleviated sorafenib resistance. miR-4726-5p mimic reversed the KCNMA1-mediated sorafenib sensitivity-promoting effect, while additional overexpression of lnc-TSI reversed the effect of miR-4726-5p. In vivo analysis also showed that overexpression of ln-TSI diminished sorafenib resistance in mice inoculated with sorafenib-resistant HCC cells via increasing KCNMA1 expression and decreasing miR-4726-5p expression. The lnc-TSI/miR-4726-5p/KCNMA1 axis plays a critical role in regulating the resistance of HCC to sorafenib, and might serve as a therapeutic target to manage sorafenib resistance of HCC in clinic.
Collapse
Affiliation(s)
- Fengrong Chen
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 Xi Wu Road, Xi'an, 710003, Shaanxi, China
| | - Jiong Jiang
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 Xi Wu Road, Xi'an, 710003, Shaanxi, China
| | - Dong Liu
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 Xi Wu Road, Xi'an, 710003, Shaanxi, China
| | - Hong Li
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 Xi Wu Road, Xi'an, 710003, Shaanxi, China
| | - Lei Dong
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 Xi Wu Road, Xi'an, 710003, Shaanxi, China
| | - Yahua Song
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 Xi Wu Road, Xi'an, 710003, Shaanxi, China
| | - Ying Zhang
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 Xi Wu Road, Xi'an, 710003, Shaanxi, China
| | - Jing Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 Xi Wu Road, Xi'an, 710003, Shaanxi, China
| | - Yun Qin
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 Xi Wu Road, Xi'an, 710003, Shaanxi, China
| | - Gang Zhao
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 Xi Wu Road, Xi'an, 710003, Shaanxi, China.
| |
Collapse
|
4
|
Fan T, Xiao C, Liu H, Liu Y, Wang L, Tian H, Li C, He J. CXXC finger protein 1 (CFP1) bridges the reshaping of genomic H3K4me3 signature to the advancement of lung adenocarcinoma. Signal Transduct Target Ther 2023; 8:369. [PMID: 37735441 PMCID: PMC10514036 DOI: 10.1038/s41392-023-01612-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 08/08/2023] [Accepted: 08/15/2023] [Indexed: 09/23/2023] Open
Abstract
Histone H3 lysine 4 trimethylation (H3K4me3) is a canonical chromatin modification associated with active gene transcription, playing a pivotal role in regulating various cellular functions. Components of the H3K4me3 methyltransferase complex, known as the proteins associated with SET1 (COMPASS), have been implicated in exerting cancer-protective or cancer-inhibitory effects through inducive H3K4me3 modification. However, the role of the indispensable non-catalytic component of COMPASS CXXC-type zinc finger protein 1 (CFP1) in malignant progression remains unclear. We have unveiled that CFP1 promote lung adenocarcinoma (LUAD) cell proliferation, migration, and invasion while impairing cell apoptosis through in vitro and in vivo models. In addition, high CFP1 expression was identified as emerged as an adverse prognostic indicator across multiple public and in-house LUAD datasets. Notably, CFP1 deficiency led to dual effects on cancer cell transcriptome including extensive inactivation of cancer-promoting as well as activation of cancer repressors. Combining this with the chromatin immunoprecipitation sequencing (ChIP-seq) analysis, we showed that CFP1 ablation reshaped the genomic H3K4me3 distribution signature, with prominent effects on TGF-β and WNT signaling pathways. Collectively, our study proposes that CFP1 mediates tumorigenesis by genomic histone methylation reprogramming, offering insights for future investigations into epigenetic modifications in cancer progression and potential therapeutic advancements.
Collapse
Affiliation(s)
- Tao Fan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chu Xiao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Hengchang Liu
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu Liu
- Department of Intervention, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Liyu Wang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - He Tian
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chunxiang Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
5
|
Jiang L, Li J, Reilly S, Xin H, Guo N, Zhang X. Role of organellar Ca2+-activated K+ channels in disease development. Life Sci 2023; 316:121433. [PMID: 36708987 DOI: 10.1016/j.lfs.2023.121433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 01/16/2023] [Accepted: 01/19/2023] [Indexed: 01/27/2023]
Abstract
The organellar Ca2+-activated K+ channels share a similar ability to transfer the alteration of Ca2+ concentration to membrane conductance of potassium. Multiple effects of Ca2+-activated K+ channels on cell metabolism and complex signaling pathways during organ development have been explored. The organellar Ca2+-activated K+ channels are able to control the ionic equilibrium and are always associated with oxidative stress in different organelles and the whole cells. Some drugs targeting Ca2+-activated K+ channels have been tested for various diseases in clinical trials. In this review, the known roles of organellar Ca2+-activated K+ channels were described, and their effects on different diseases, particularly on diabetes, cardiovascular diseases, and neurological diseases were discussed. It was attempted to summarize the currently known operational modes with the involvement of organellar Ca2+-activated K+ channels. This review may assist scholars to more comprehensively understand organellar Ca2+-activated K+ channels and related diseases.
Collapse
Affiliation(s)
- Lan Jiang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Jiawei Li
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Svetlana Reilly
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Hong Xin
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Nan Guo
- Department of Pharmacy, Minhang hospital, Fudan University, Shanghai, China.
| | - Xuemei Zhang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China.
| |
Collapse
|
6
|
Xie S, Xu C, Wu C, Lou Y, Duan J, Sang R, Lou Z, Hou J, Ge W, Xi Y, Yang X. Co-dependent regulation of p-BRAF and potassium channel KCNMA1 levels drives glioma progression. Cell Mol Life Sci 2023; 80:61. [PMID: 36763212 PMCID: PMC9918570 DOI: 10.1007/s00018-023-04708-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 01/03/2023] [Accepted: 01/23/2023] [Indexed: 02/11/2023]
Abstract
BRAF mutations have been found in gliomas which exhibit abnormal electrophysiological activities, implying their potential links with the ion channel functions. In this study, we identified the Drosophila potassium channel, Slowpoke (Slo), the ortholog of human KCNMA1, as a critical factor involved in dRafGOF glioma progression. Slo was upregulated in dRafGOF glioma. Knockdown of slo led to decreases in dRafGOF levels, glioma cell proliferation, and tumor-related phenotypes. Overexpression of slo in glial cells elevated dRaf expression and promoted cell proliferation. Similar mutual regulations of p-BRAF and KCNMA1 levels were then recapitulated in human glioma cells with the BRAF mutation. Elevated p-BRAF and KCNMA1 were also observed in HEK293T cells upon the treatment of 20 mM KCl, which causes membrane depolarization. Knockdown KCNMA1 in these cells led to a further decrease in cell viability. Based on these results, we conclude that the levels of p-BRAF and KCNMA1 are co-dependent and mutually regulated. We propose that, in depolarized glioma cells with BRAF mutations, high KCNMA1 levels act to repolarize membrane potential and facilitate cell growth. Our study provides a new strategy to antagonize the progression of gliomas as induced by BRAF mutations.
Collapse
Affiliation(s)
- Shanshan Xie
- The Women's Hospital, Institute of Genetics, Zhejiang Provincial Key Laboratory of Genetic and Development Disorders, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Chengyan Xu
- Department of Neurosurgery, The Children's Hospital Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Cheng Wu
- The Women's Hospital, Institute of Genetics, Zhejiang Provincial Key Laboratory of Genetic and Development Disorders, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Yuhan Lou
- The Women's Hospital, Institute of Genetics, Zhejiang Provincial Key Laboratory of Genetic and Development Disorders, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Jingwei Duan
- The Women's Hospital, Institute of Genetics, Zhejiang Provincial Key Laboratory of Genetic and Development Disorders, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Rong Sang
- The Women's Hospital, Institute of Genetics, Zhejiang Provincial Key Laboratory of Genetic and Development Disorders, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Ziwei Lou
- The Women's Hospital, Institute of Genetics, Zhejiang Provincial Key Laboratory of Genetic and Development Disorders, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Jiaru Hou
- The Women's Hospital, Institute of Genetics, Zhejiang Provincial Key Laboratory of Genetic and Development Disorders, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Wanzhong Ge
- The Women's Hospital, Institute of Genetics, Zhejiang Provincial Key Laboratory of Genetic and Development Disorders, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Yongmei Xi
- The Women's Hospital, Institute of Genetics, Zhejiang Provincial Key Laboratory of Genetic and Development Disorders, School of Medicine, Zhejiang University, Hangzhou, 310058, China.
| | - Xiaohang Yang
- The Women's Hospital, Institute of Genetics, Zhejiang Provincial Key Laboratory of Genetic and Development Disorders, School of Medicine, Zhejiang University, Hangzhou, 310058, China.
- Joint Institute of Genetics and Genomic Medicine Between Zhejiang University and the University of Toronto, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
7
|
Kuang Y, He Z, Li L, Wang C, Cheng X, Shi Q, Fu G, Ying J, Tao Q, Hu X. The developmental regulator HAND1 inhibits gastric carcinogenesis through enhancing ER stress apoptosis via targeting CHOP and BAK which is augmented by cisplatin. Int J Biol Sci 2023; 19:120-136. [PMID: 36594085 PMCID: PMC9760445 DOI: 10.7150/ijbs.76345] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 10/02/2022] [Indexed: 11/24/2022] Open
Abstract
Epigenetic disruption of tumor suppressor genes, particularly aberrant CpG methylation, plays a crucial role in gastric cancer (GC) pathogenesis. Through CpG methylome and expression profiling, a developmental transcription factor - Hand-And-Neural-crest-Derivative-expressed 1 (HAND1), was identified methylated and downregulated in GC. However, its role and underlying mechanisms in GC progression are poorly understood. Here, we show that HAND1 was frequently downregulated in GC by promoter methylation, and significantly correlated with tumor progression and poor prognosis of GC patients. High expression of HAND1 in GC patients was associated with significantly higher 5-year overall survival rates. Ectopic expression of HAND1 inhibited GC cell growth and migration in vitro and in vivo. HAND1 expression increased ROS levels and cytosolic Ca2+ concentration, enhanced cisplatin-induced apoptosis through endoplasmic reticulum (ER) stress/mitochondria-mediated apoptosis. Knockdown of CHOP and BAK attenuated HAND1-induced cell apoptosis. Overexpression of CHOP increased BAK expression. HAND1 interacts with CHOP, also directly binds to CHOP and BAK promoters and positively regulates BAK transcription. Thus, the present study demonstrates that HAND1 is a tumor suppressor gene methylated in GC, induces ER stress and apoptosis via CHOP and BAK, which is augmented by cisplatin. Low HAND1 expression is an independent poor prognostic factor for GC. The tumor-specific methylation of HAND1 promoter could be a candidate biomarker for GC.
Collapse
Affiliation(s)
- Yeye Kuang
- Biomedical Research Center, Sir Run Shaw Hospital, Zhejiang University, Hangzhou, China.,Department of Pathology, Sir Run Shaw Hospital, Zhejiang University, Hangzhou, China.,Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Hangzhou 310016, Zhejiang, China
| | - Zhanglian He
- Biomedical Research Center, Sir Run Shaw Hospital, Zhejiang University, Hangzhou, China.,Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Hangzhou 310016, Zhejiang, China
| | - Lili Li
- Cancer Epigenetics Laboratory, Department of Clinical Oncology, State Key Laboratory of Translational Oncology, Sir YK Pao Center for Cancer and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Chan Wang
- Biomedical Research Center, Sir Run Shaw Hospital, Zhejiang University, Hangzhou, China.,Department of Pathology, Sir Run Shaw Hospital, Zhejiang University, Hangzhou, China.,Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Hangzhou 310016, Zhejiang, China
| | - Xiaoqing Cheng
- Department of Pathology, Sir Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Qinglan Shi
- Biomedical Research Center, Sir Run Shaw Hospital, Zhejiang University, Hangzhou, China.,Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Hangzhou 310016, Zhejiang, China
| | - Guoxiang Fu
- Department of Pathology, Sir Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Jianming Ying
- Cancer Epigenetics Laboratory, Department of Clinical Oncology, State Key Laboratory of Translational Oncology, Sir YK Pao Center for Cancer and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Qian Tao
- Cancer Epigenetics Laboratory, Department of Clinical Oncology, State Key Laboratory of Translational Oncology, Sir YK Pao Center for Cancer and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong.,✉ Corresponding authors: X Hu () or Q Tao ()
| | - Xiaotong Hu
- Biomedical Research Center, Sir Run Shaw Hospital, Zhejiang University, Hangzhou, China.,Department of Pathology, Sir Run Shaw Hospital, Zhejiang University, Hangzhou, China.,Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Hangzhou 310016, Zhejiang, China.,✉ Corresponding authors: X Hu () or Q Tao ()
| |
Collapse
|
8
|
Wen H, Feng Z, Ma Y, Liu R, Ou Q, Guo Q, Shen Y, Wu X, Shao Y, Bao H, Wu X. Homologous recombination deficiency in diverse cancer types and its correlation with platinum chemotherapy efficiency in ovarian cancer. BMC Cancer 2022; 22:550. [PMID: 35578198 PMCID: PMC9109318 DOI: 10.1186/s12885-022-09602-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 04/24/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Homologous recombination deficiency (HRD) is a molecular biomarker for administrating PARP inhibitor (PARPi) or platinum-based (Pt) chemotherapy. The most well-studied mechanism of causing HRD is pathogenic BRCA1/2 mutations, while HRD phenotype is also present in patients without BRCA1/2 alterations, suggesting other unknown factors. METHODS The targeted next-generation sequencing (GeneseeqPrime® HRD) was used to evaluate the HRD scores of 199 patients (Cohort I). In Cohort II, a total of 85 Pt-chemotherapy-treated high-grade serous ovarian cancer (HGSOC) patients were included for investigating the role of HRD score in predicting treatment efficacy. The concurrent genomic features analyzed along HRD score evaluation were studied in a third cohort with 416 solid tumor patients (Cohort III). RESULTS An HRD score ≥ 38 was predefined as HRD-positive by analyzing Cohort I (range: 0-107). Over 95% of the BRCA1/2-deficient cases of Cohort I were HRD-positive under this threshold. In Cohort II, Pt-sensitive patients have significantly higher HRD scores than Pt-resistant patients (median: 54 vs. 34, p = 0.031) and a significantly longer PFS was observed in HRD-positive patients (median: 548 vs. 343 days, p = 0.003). Furthermore, TP53, NCOR1, and PTK2 alterations were enriched in HRD-positive patients. In Cohort III, impaired homologous recombination repair pathway was more frequently observed in HRD-positive patients without BRCA1/2 pathogenic mutations. The alteration enrichment of TP53, NCOR1, and PTK2 observed in Cohort II was also validated by the ovarian subgroup in Cohort III. CONCLUSIONS Using an in-house HRD evaluation method, our findings show that overall HRR gene mutations account for a significant part of HRD in the absence of BRCA1/2 aberrations, and suggest that HRD positive status might be a predictive biomarker of Pt-chemotherapy.
Collapse
Affiliation(s)
- Hao Wen
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, 270 Dongan Road, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, 130 Dongan Road, Shanghai, 200032, China
| | - Zheng Feng
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, 270 Dongan Road, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, 130 Dongan Road, Shanghai, 200032, China
| | - Yutong Ma
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc, No. 128 Huakang Road, Pukou District, Nanjing, Jiangsu, 210000, China
| | - Rui Liu
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc, No. 128 Huakang Road, Pukou District, Nanjing, Jiangsu, 210000, China
| | - Qiuxiang Ou
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc, No. 128 Huakang Road, Pukou District, Nanjing, Jiangsu, 210000, China
| | - Qinhao Guo
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, 270 Dongan Road, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, 130 Dongan Road, Shanghai, 200032, China
| | - Yi Shen
- R&D, Nanjing Geneseeq Technology Inc, No. 128 Huakang Road, Pukou District, Nanjing, Jiangsu, 210000, China
| | - Xue Wu
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc, No. 128 Huakang Road, Pukou District, Nanjing, Jiangsu, 210000, China
| | - Yang Shao
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc, No. 128 Huakang Road, Pukou District, Nanjing, Jiangsu, 210000, China.,School of Public Health, Nanjing Medical University, Jiangning District, 101 Longmian Avenue, Nanjing, Jiangsu, 211166, China
| | - Hua Bao
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc, No. 128 Huakang Road, Pukou District, Nanjing, Jiangsu, 210000, China.
| | - Xiaohua Wu
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, 270 Dongan Road, Shanghai, 200032, China. .,Department of Oncology, Shanghai Medical College, Fudan University, 130 Dongan Road, Shanghai, 200032, China.
| |
Collapse
|
9
|
He L, Qian X, Ge P, Fan D, Ma X, Wu Q, Sun J, Yang L, Shen J, Xu L. NOL6 Regulates the Proliferation and Apoptosis of Gastric Cancer Cells via Regulating TP53I3, CDK4 and MCM7 Expression. Front Oncol 2022; 12:708081. [PMID: 35494047 PMCID: PMC9039204 DOI: 10.3389/fonc.2022.708081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 03/03/2022] [Indexed: 11/20/2022] Open
Abstract
Background Gastric cancer (GC) is a prevalent cancer with high mortality and strong invasiveness, and the entire regulatory networks of GC is still unclear. Objective The aim of this study was to explore the specific mechanism of the effect of nucleolar protein 6 (NOL6) on the proliferation and apoptosis of GC cells. Methods The human gastric adenocarcinoma cell line HGC-27 and AGS were cultured. qRT-PCR was used to verify the expression level of NOL6 in GC cells; MTT and EdU were used to test cell proliferation; TUNEL staining and Flow cytometry were used to detect cell apoptosis; The downstream genes and pathways following NOL6 knockdown were explored through the microarray assay and ingenuity pathway analysis, and the downstream genes were finally verified by qRT-PCR and Western blotting. The xenograft mice were used to investigate the effect of NOL6 on GC in vivo. Results TCGA data analysis showed that NOL6 expression level was higher in GC cells than adjacent normal cells. Over-expression of NOL6 increased proliferation and colony formation, and inhibited the apoptotic rate in AGS and HGC-27 cells, while NOL6 knockdown induced the opposite effects. Through microarray assay and IPA analysis, NOL6-related downstream genes and critical signaling pathways were found. And we verified the relationship between downstream genes and GC. Additionally, NOL6 knockdown could decrease the weight and volume of tumor in the mice. Conclusion NOL6 knockdown could inhibit cell proliferation and induce cell apoptosis of GC, suggesting that NOL6 may serve as a potential therapeutic target for treating GC.
Collapse
Affiliation(s)
- Lei He
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaohan Qian
- Medical Center for Digestive Disease, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Pingping Ge
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Dong Fan
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiang Ma
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qiong Wu
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jin Sun
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lihua Yang
- Medical Center for Digestive Disease, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jian Shen
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lijian Xu
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
10
|
Hernández-Gómez C, Hernández-Lemus E, Espinal-Enríquez J. The Role of Copy Number Variants in Gene Co-Expression Patterns for Luminal B Breast Tumors. Front Genet 2022; 13:806607. [PMID: 35432489 PMCID: PMC9010943 DOI: 10.3389/fgene.2022.806607] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 03/03/2022] [Indexed: 12/20/2022] Open
Abstract
Gene co-expression networks have become a usual approach to integrate the vast amounts of information coming from gene expression studies in cancer cohorts. The reprogramming of the gene regulatory control and the molecular pathways depending on such control are central to the characterization of the disease, aiming to unveil the consequences for cancer prognosis and therapeutics. There is, however, a multitude of factors which have been associated with anomalous control of gene expression in cancer. In the particular case of co-expression patterns, we have previously documented a phenomenon of loss of long distance co-expression in several cancer types, including breast cancer. Of the many potential factors that may contribute to this phenomenology, copy number variants (CNVs) have been often discussed. However, no systematic assessment of the role that CNVs may play in shaping gene co-expression patterns in breast cancer has been performed to date. For this reason we have decided to develop such analysis. In this study, we focus on using probabilistic modeling techniques to evaluate to what extent CNVs affect the phenomenon of long/short range co-expression in Luminal B breast tumors. We analyzed the co-expression patterns in chromosome 8, since it is known to be affected by amplifications/deletions during cancer development. We found that the CNVs pattern in chromosome 8 of Luminal B network does not alter the co-expression patterns significantly, which means that the co-expression program in this cancer phenotype is not determined by CNV structure. Additionally, we found that region 8q24.3 is highly dense in interactions, as well as region p21.3. The most connected genes in this network belong to those cytobands and are associated with several manifestations of cancer in different tissues. Interestingly, among the most connected genes, we found MAF1 and POLR3D, which may constitute an axis of regulation of gene transcription, in particular for non-coding RNA species. We believe that by advancing on our knowledge of the molecular mechanisms behind gene regulation in cancer, we will be better equipped, not only to understand tumor biology, but also to broaden the scope of diagnostic, prognostic and therapeutic interventions to ultimately benefit oncologic patients.
Collapse
Affiliation(s)
| | - Enrique Hernández-Lemus
- Computational Genomics Division, National Institute of Genomic Medicine, Mexico City, Mexico
- Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México, Mexico City, Mexico
- *Correspondence: Jesús Espinal-Enríquez, ; Enrique Hernández-Lemus,
| | - Jesús Espinal-Enríquez
- Computational Genomics Division, National Institute of Genomic Medicine, Mexico City, Mexico
- Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México, Mexico City, Mexico
- *Correspondence: Jesús Espinal-Enríquez, ; Enrique Hernández-Lemus,
| |
Collapse
|
11
|
Disruption of the CCDC43-FHL1 interaction triggers apoptosis in gastric cancer cells. Exp Cell Res 2022; 415:113107. [DOI: 10.1016/j.yexcr.2022.113107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 03/09/2022] [Accepted: 03/11/2022] [Indexed: 11/18/2022]
|
12
|
Ding H, Zhang L, Zhang C, Song J, Jiang Y. Screening of Significant Biomarkers Related to Prognosis of Cervical Cancer and Functional Study Based on lncRNA-associated ceRNA Regulatory Network. Comb Chem High Throughput Screen 2021; 24:472-482. [PMID: 32729415 DOI: 10.2174/1386207323999200729113028] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 05/28/2020] [Accepted: 06/15/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Cervical cancer (CESC), which threatens the health of women, has a very high recurrence rate. PURPOSES This study aimed to identify the signature long non-coding RNAs (lncRNAs) associated with the prognosis of CESC and predict the prognostic survival rate with the clinical risk factors. METHODS The CESC gene expression profiling data were downloaded from TCGA database and NCBI Gene Expression Omnibus. Afterwards, the differentially expressed RNAs (DERs) were screened using limma package of R software. R package "survival" was then used to screen the signature lncRNAs associated with independently recurrence prognosis, and a nomogram recurrence rate model based on these signature lncRNAs was constructed to predict the 3-year and 5-year survival probability of CESC. Finally, a competing endogenous RNAs (ceRNA) regulatory network was proposed to study the functions of these genes. RESULTS We obtained 305 DERs significantly associated with prognosis. Afterwards, a risk score (RS) prediction model was established using the screened 5 signature lncRNAs associated with independently recurrence prognosis (DLEU1, LINC01119, RBPMS-AS1, RAD21-AS1 and LINC00323). Subsequently, a nomogram recurrence rate model, proposed with Pathologic N and RS model status, was found to have a good prediction ability for CESC. In ceRNA regulatory network, LINC00323 and DLEU1 were hub nodes which targeted more miRNAs and mRNAs. After that, 15 GO terms and 3 KEGG pathways were associated with recurrence prognosis and showed that the targeted genes PTK2, NRP1, PRKAA1 and HMGCS1 might influence the prognosis of CESC. CONCLUSION The signature lncRNAs can help improve our understanding of the development and recurrence of CESC and the nomogram recurrence rate model can be applied to predict the survival rate of CESC patients in clinical practice.
Collapse
Affiliation(s)
- Haiyan Ding
- Department of Obstetrics and Gynecology, Second Hospital of Jilin University, Changchun, Jilin Province 130041, China
| | - Li Zhang
- Department of Emergency Medicine, Second Hospital of Jilin University, Changchun, Jilin Province 130041, China
| | - Chunmiao Zhang
- Department of Obstetrics and Gynecology, Second Hospital of Jilin University, Changchun, Jilin Province 130041, China
| | - Jie Song
- Department of Hepatobiliary and Pancreatic Medicine, Second Hospital of Jilin University, Changchun, Jilin Province 130041, China
| | - Ying Jiang
- Department of Obstetrics and Gynecology, Second Hospital of Jilin University, Changchun, Jilin Province 130041, China
| |
Collapse
|
13
|
Wang H, Huang H, Wang L, Liu Y, Wang M, Zhao S, Lu G, Kang X. Cancer-associated fibroblasts secreted miR-103a-3p suppresses apoptosis and promotes cisplatin resistance in non-small cell lung cancer. Aging (Albany NY) 2021; 13:14456-14468. [PMID: 33999859 PMCID: PMC8202839 DOI: 10.18632/aging.103556] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 06/04/2020] [Indexed: 12/28/2022]
Abstract
BACKGROUND The cisplatin resistance of non-small cell lung cancer (NSCLC) patients results in low response rate and overall survival rate. Exosomes contribute to pathological processes of multiple cancers. OBJECTIVE In this study, we explored the function and mechanisms of exosomal miR-103a-3p derived from cancer-associated fibroblast (CAF) in cisplatin resistance in NSCLC. RESULTS MiR-103a-3p was highly expressed in CAFs and CAF exosomes, and exosomal miR-103a-3p derived from CAFs in NSCLC. CAFs exosomes co-cultured with NSCLC cells promoted miR-103a-3p expression both in NSCLC cells and its exosomes. Functional experiments showed that exo-miR-103a-3p derived from CAFs promoted cisplatin resistance and inhibited apoptosis in NSCLC cells. Pumilio2 (Pum2) bound with miR-103a-3p in cytoplasm and nucleus, and facilitated packaging into CAF-derived exosomes in NSCLC cells. Further analysis showed Bak1 was a direct target of miR-103a-3p, and miR-103a-3p accelerated cisplatin resistance in NSCLC cells via Bak1 downregulation. In vivo tumorigenesis assay showed CAF-derived exosomal miR-103a-3p enhanced cisplatin resistance and inhibited cell apoptosis in NSCLC. CONCLUSION Our study revealed that CAFs-derived exosomal miR-103a-3p promoted cisplatin resistance by suppressing apoptosis via targeting Bak1, which provided a potential therapeutic target for cisplatin resistance in NSCLC.
Collapse
Affiliation(s)
- Haifeng Wang
- Third Ward of Oncology Department, The First Affiliated Hospital of Xinxiang Medical University, Weihui 453100, Henan, China
| | - Haibo Huang
- Third Ward of Oncology Department, The First Affiliated Hospital of Xinxiang Medical University, Weihui 453100, Henan, China
| | - Lijiang Wang
- Respiratory Ward 2, The First Affiliated Hospital of Xinxiang Medical University, Weihui 453100, Henan, China
| | - Yan Liu
- Respiratory Ward 2, The First Affiliated Hospital of Xinxiang Medical University, Weihui 453100, Henan, China
| | - Ming Wang
- Respiratory Intensive Care Unit, The First Affiliated Hospital of Xinxiang Medical University, Weihui 453100, Henan, China
| | - Shasha Zhao
- Respiratory Intensive Care Unit, The First Affiliated Hospital of Xinxiang Medical University, Weihui 453100, Henan, China
| | - Guangjian Lu
- Clinical Laboratory, The First Affiliated Hospital of Xinxiang Medical University, Weihui 453100, Henan, China
| | - Xiaohong Kang
- First Ward of Oncology Department, The First Affiliated Hospital of Xinxiang Medical University, Weihui 453100, Henan, China
| |
Collapse
|
14
|
Shiozaki A, Marunaka Y, Otsuji E. Roles of Ion and Water Channels in the Cell Death and Survival of Upper Gastrointestinal Tract Cancers. Front Cell Dev Biol 2021; 9:616933. [PMID: 33777930 PMCID: PMC7991738 DOI: 10.3389/fcell.2021.616933] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 02/22/2021] [Indexed: 02/06/2023] Open
Abstract
Ion and water channels were recently shown to be involved in cancer cell functions, and various transporter types have been detected in upper gastrointestinal tract (UGI) cancers. Current information on the expression and roles of these channels and transporters in the death and survival of UGI cancer cells was reviewed herein, and the potential of their regulation for cancer management was investigated. Esophageal cancer (EC) and gastric cancer (GC) cells and tissues express many different types of ion channels, including voltage-gated K+, Cl-, and Ca2+, and transient receptor potential (TRP) channels, which regulate the progression of cancer. Aquaporin (AQP) 1, 3, and 5 are water channels that contribute to the progression of esophageal squamous cell carcinoma (ESCC) and GC. Intracellular pH regulators, including the anion exchanger (AE), sodium hydrogen exchanger (NHE), and vacuolar H+-ATPases (V-ATPase), also play roles in the functions of UGI cancer cells. We have previously conducted gene expression profiling and revealed that the regulatory mechanisms underlying apoptosis in ESCC cells involved various types of Cl- channels, Ca2+ channels, water channels, and pH regulators (Shimizu et al., 2014; Ariyoshi et al., 2017; Shiozaki et al., 2017, 2018a; Kobayashi et al., 2018; Yamazato et al., 2018; Konishi et al., 2019; Kudou et al., 2019; Katsurahara et al., 2020, 2021; Matsumoto et al., 2021; Mitsuda et al., 2021). We have also previously demonstrated the clinicopathological and prognostic significance of their expression in ESCC patients, and shown that their pharmacological blockage and gene silencing had an impact on carcinogenesis, indicating their potential as targets for the treatment of UGI cancers. A more detailed understanding of the molecular regulatory mechanisms underlying cell death and survival of UGI cancers may result in the application of cellular physiological methods as novel therapeutic approaches.
Collapse
Affiliation(s)
- Atsushi Shiozaki
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yoshinori Marunaka
- Department of Molecular Cell Physiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan.,Research Institute for Clinical Physiology, Kyoto Industrial Health Association, Kyoto, Japan.,Research Center for Drug Discovery and Pharmaceutical Development Science, Research Organization of Science and Technology, Ritsumeikan University, Kusatsu, Japan
| | - Eigo Otsuji
- Division of Digestive Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
15
|
Jiang M, Zhang X. Antiangiogenesis Combined with Immunotherapy to Treat Advanced Small-Cell Carcinoma of the Esophagus Resistant to Chemotherapy: According to the Guidance of Next-Generation Sequencing. Onco Targets Ther 2021; 14:1613-1621. [PMID: 33688208 PMCID: PMC7936705 DOI: 10.2147/ott.s293733] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 02/13/2021] [Indexed: 12/24/2022] Open
Abstract
A 64-year-old woman admitted to our hospital with the chief complaint of swallowing obstruction was diagnosed as relapsed small-cell carcinoma of the esophagus. Complete remission (CR) was observed after six cycles of irinotecan plus cisplatin therapy. According to the results of a next-generation sequencing analysis of the tumor specimen, anlotinib (12 mg PO q3w) was recommended. After 1 month of anlotinib treatment, the tumor decreased significantly according to computed tomography scan and gastroscopy. However, the disease progressed after 2 months of therapy. A gene analysis of the new puncture sample showed microsatellite instability and a high tumor mutation burden. Immunohistochemistry indicated positive programmed death ligand-1 expression (>1%). Because of these results, the patient was treated with anlotinib (12 mg PO q3w) in combination with toripalimab (240 mg IV drip q3w). After 3 months of therapy, CR was achieved, although progression-free survival had not been reached at the time of publication.
Collapse
Affiliation(s)
- Man Jiang
- Cancer Precision Medical Center, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266003, People's Republic of China
| | - Xiaochun Zhang
- Cancer Precision Medical Center, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266003, People's Republic of China
| |
Collapse
|
16
|
Zhu M, Liang Q, Chen T, Kong Q, Ye G, Yu S, Li X, He Q, Liu H, Hu Y, Yu J, Li G. Identification and validation of methylated differentially expressed miRNAs and immune infiltrate profile in EBV-associated gastric cancer. Clin Epigenetics 2021; 13:22. [PMID: 33514440 PMCID: PMC7845045 DOI: 10.1186/s13148-020-00989-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 12/01/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The recent discovery of cancer/tissue specificity of miRNA has indicated its great potential as a therapeutic target. In Epstein-Barr virus-associated gastric cancer (EBVaGC), host genes are affected by extensive DNA methylation, including miRNAs. However, the role of methylated miRNA in the development of EBVaGC and immune cell infiltration has largely remained elusive. RESULTS After crossmatching the DNA methylation and expression profile of miRNA and mRNA in the Gene Expression Omnibus (GEO) and the Cancer Genome Atlas Research Network (TCGA), we discovered that miR-129-2-3p was significantly suppressed due to hypermethylation on its enhancer in EBVaGC. The differentially expressed genes (DEGs) added up to 30, among which AKAP12 and LARP6 were predicted to be the target genes of miR-129-2-3p and negatively correlated with patients' survival. Accordingly, miR-129-2-3p was significantly down-regulated in tumor samples in 26 (65%) out of 40 cases in our cohort (P < 0.0001). The proliferation, migration and invasion functions of GC cells were significantly promoted when transfected with miR-129-2-3p inhibitor and suppressed when transfected with mimics or treated with 5-aza-2'-deoxycytidine. Moreover, a comprehensive regulation network was established by combining the putative transcription factors, miRNA-mRNA and protein-protein interaction (PPI) analysis. Pathway enrichment analysis showed that cytokine activity, especially CCL20, was the most prominent biological process in EBVaGC development. Immune cell infiltration analysis demonstrated CD4+ T cell, macrophage and dendritic cell infiltrates were significantly enriched for the prognostic-indicated hub genes. CONCLUSION This study has provided a comprehensive analysis of differentially expressed miRNAs and mRNAs associated with genome-wide DNA methylation by integrating multi-source data including transcriptome, methylome and clinical data from GEO and TCGA, QPCR of tumor samples and cell function assays. It also gives a hint on the relationships between methylated miRNA, DEGs and the immune infiltration. Further experimental and clinical investigations are warranted to explore the underlying mechanism and validate our findings.
Collapse
Affiliation(s)
- Mansheng Zhu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, 510515, China
| | - Qixiang Liang
- Department of Stomatology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Tao Chen
- Department of General Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, 510515, China.
| | - Qian Kong
- Department of Pediatrics, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Gengtai Ye
- Department of General Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, 510515, China
| | - Shitong Yu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, 510515, China
| | - Xunjun Li
- Department of General Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, 510515, China
| | - Qinglie He
- Department of General Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, 510515, China
| | - Hao Liu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, 510515, China
| | - Yanfeng Hu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, 510515, China
| | - Jiang Yu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, 510515, China.
| | - Guoxin Li
- Department of General Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, 510515, China.
| |
Collapse
|
17
|
Huo Q, Ma Y, Yin Y, Qin G. Biomarker Identification for Liver Hepatocellular Carcinoma and Cholangiocarcinoma Based on Gene Regulatory Network Analysis. Curr Bioinform 2021. [DOI: 10.2174/1574893615666200317115609] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Background:
Liver hepatocellular carcinoma (LIHC) and cholangiocarcinoma (CHOL)
are two main histological subtypes of primary liver cancer with a unified molecular landscape, and
feed-forward loops (FFLs) have been shown to be relevant in these complex diseases.
Objective:
To date, there has been no comparative analysis of the pathogenesis of LIHC and CHOL
based on regulatory relationships. Therefore, we investigated the common and distinct regulatory
properties of LIHC and CHOL in terms of gene regulatory networks.
Method:
Based on identified FFLs and an analysis of pathway enrichment, we constructed pathway-specific co-expression networks and further predicted biomarkers for these cancers by network clustering.
Resul:
We identified 20 and 36 candidate genes for LIHC and CHOL, respectively. The literature
from PubMed supports the reliability of our results.
Conclusion:
Our results indicated that the hsa01522-Endocrine resistance pathway was associated
with both LIHC and CHOL. Additionally, six genes (SPARC, CTHRC1, COL4A1, EDIL3, LAMA4
and OLFML2B) were predicted to be highly associated with both cancers, and COL4A2, CSPG4,
GJC1 and ADAMTS7 were predicted to be potential biomarkers of LIHC, and COL6A3, COL1A2,
FAP and COL8A1 were predicted to be potential biomarkers of CHOL. In addition, we inferred that
the Collagen gene family, which appeared more frequently in our overall prediction results, might be
closely related to cancer development.
Collapse
Affiliation(s)
- Qiuyan Huo
- School of Computer Science and Technology, Xidian University, Xi’an,China
| | - Yuying Ma
- School of Computer Science and Technology, Xidian University, Xi’an,China
| | - Yu Yin
- School of Computer Science and Technology, Xidian University, Xi’an,China
| | - Guimin Qin
- School of Computer Science and Technology, Xidian University, Xi’an,China
| |
Collapse
|
18
|
Li R, Shui L, Jia J, Wu C. Construction and Validation of Novel Diagnostic and Prognostic DNA Methylation Signatures for Hepatocellular Carcinoma. Front Genet 2020; 11:906. [PMID: 32922438 PMCID: PMC7456968 DOI: 10.3389/fgene.2020.00906] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 07/22/2020] [Indexed: 12/20/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most prevalent life-threatening human cancers and the leading cause of cancer-related mortality, with increased global incidence within the last decade. Identification of effective diagnostic and prognostic biomarkers would enable reliable risk stratification and efficient screening of high-risk patients, thereby facilitating clinical decision-making. Herein, we performed a comprehensive, robust DNA methylation analysis based on genome-wide DNA methylation profiling. We constructed a diagnostic signature with five DNA methylation markers, which precisely distinguished HCC patients from normal controls. Cox regression and LASSO analysis were applied to construct a prognostic signature with four DNA methylation markers. A one-to-one correlation analysis was carried out between genes of the whole genome and our prognostic signature. Exploration of the biological function and the role of the underlying significantly correlated genes was conducted. A mixed dataset of 463 HCC patients and 253 normal controls, derived from six independent datasets, was used to valid the diagnostic signature. Results showed a specificity of 96.84% and sensitivity of 96.77%. Class scores for the diagnostic signature were significantly different between normal controls, individuals with liver diseases, and HCC patients. The present signature has the potential to serve as a biomarker to monitor health in normal controls. Additionally, HCC patients were successfully separated into low-risk and high-risk groups by the prognostic signature, with a better prognosis for patients in the low-risk group. Kaplan-Meier and ROC analysis confirmed that the prognostic signature performed well. We found eight of the top ten genes to positively correlate with risk scores of the prognostic signature, and to be involved in cell cycle regulation. This eight-gene panel also served as a prognostic signature. The robust evidence presented in this study therefore demonstrates the effectiveness of the prognostic signature. In summary, we constructed diagnostic and prognostic signatures, which have potential for use in diagnosis, surveillance, and prognostic prediction for HCC patients. Eight genes that were significantly and positively correlated with the prognostic signature were strongly associated with cell cycle processes. Therefore, the prognostic signature can be used as a guide by which to measure responsiveness to cell-cycle-targeting agents.
Collapse
Affiliation(s)
- Ran Li
- Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Liyan Shui
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Junling Jia
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.,Innovation Center for Precision Medicine, Zhongtong-Lanbo Diagnostic Ltd, Beijing, China
| | - Chao Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
19
|
Integrated Transcriptomic Analysis Reveals the Molecular Mechanism of Meningiomas by Weighted Gene Coexpression Network Analysis. BIOMED RESEARCH INTERNATIONAL 2020; 2020:4927547. [PMID: 32596316 PMCID: PMC7303753 DOI: 10.1155/2020/4927547] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 04/28/2020] [Accepted: 05/09/2020] [Indexed: 02/04/2023]
Abstract
Meningiomas are the most common primary intracranial tumor in adults. However, to date, systemic coexpression analyses for meningiomas fail to explain its pathogenesis. The aim of the present study was to construct coexpression modules and identify potential biomarkers associated with meningioma progression. Weighted gene coexpression network analysis (WGCNA) was performed based on GSE43290, and module preservation was tested by GSE74385. Functional annotations were performed to analyze biological significance. Hub genes were selected for efficacy evaluations and correlation analyses using two independent cohorts. A total of 14 coexpression modules were identified, and module lightcyan was significantly associated with WHO grades. Functional enrichment analyses of module lightcyan were associated with tumor pathogenesis. The top 10 hub genes were extracted. Ten biomarkers, particularly AHCYL2, FGL2, and KCNMA1, were significantly related to grades and prognosis of meningioma. These findings not only construct coexpression modules leading to the better understanding of its pathogenesis but also provide potential biomarkers that represent specific on tumor grades and identify recurrence, predicting prognosis and progression of meningiomas.
Collapse
|
20
|
Feng L, Jian R, Wang Y, Yao Y, Gao M, Du H. A close relationship between Helicobacter pylori infection and the serum level of CD47 in adults. Scand J Gastroenterol 2020; 55:640-645. [PMID: 32493095 DOI: 10.1080/00365521.2020.1772863] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Objectives: Although Helicobacter pylori (H. pylori) infection is one of the most important risk factors for gastric cancer, the molecular mechanisms underlying the progression of H. pylori-induced gastric cancer remain unclear. Previous studies have demonstrated that Integrin-associated protein (CD47) plays an important role in the development of gastric cancer. Hence, the aim of this study was to preliminarily explore the relationship between CD47 and H. pylori infection.Methods: A total of 417 adults who underwent health checkups at a hospital were recruited in 2018. Serum levels of CD47 and tumor necrosis factor-α (TNF-α) were determined using an enzyme-linked immunosorbent assay. 13C urea breath test (13C-UBT) was carried out to diagnose H. pylori infection in all participants.Results: Compared with H. pylori-negative participants, H. pylori-positive participants have higher levels of serum CD47 and TNF-α. H. pylori infection, the levels of serum TNF-α and low density lipoprotein (LDL) are the independent predictors of serum level of CD47 in adults. In addition, a potential diagnostic value of serum CD47 level for H. pylori infection has been demonstrated in our study.Conclusion: H. pylori infection is closely associated with the serum level of CD47 in adults, suggesting that H. pylori may promote gastric cancer progression by activating CD47-mediated oncogenic pathways.
Collapse
Affiliation(s)
- Lei Feng
- The Division of Gastroenterology and Hepatology, Suining Central Hospital, Suining City, Sichuan Province, China
| | - Rong Jian
- The Division of Gastroenterology and Hepatology, Suining Central Hospital, Suining City, Sichuan Province, China
| | - Yan Wang
- The Division of Gastroenterology and Hepatology, Suining Central Hospital, Suining City, Sichuan Province, China
| | - Yong Yao
- The Division of Gastroenterology and Hepatology, Suining Central Hospital, Suining City, Sichuan Province, China
| | - Mingsheng Gao
- The Division of Gastroenterology and Hepatology, Suining Central Hospital, Suining City, Sichuan Province, China
| | - Heng Du
- The Division of Gastroenterology and Hepatology, Suining Central Hospital, Suining City, Sichuan Province, China
| |
Collapse
|
21
|
Mu HQ, Liang ZQ, Xie QP, Han W, Yang S, Wang SB, Zhao C, Cao YM, He YH, Chen J. Identification of potential crucial genes associated with the pathogenesis and prognosis of prostate cancer. Biomark Med 2020; 14:353-369. [PMID: 32253914 DOI: 10.2217/bmm-2019-0318] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: Prostate cancer (PCa) is the sixth leading cause of cancer-related deaths in men throughout the world. This study aimed to investigate genes associated with the pathogenesis and prognosis of PCa. Materials & methods: Data of PCa cases were obtained from public datasets and were analyzed using an integrated bioinformatics strategy. Results: A total of 969 differential expression genes were identified. Moreover, GSE16560 and The Cancer Genome Atlas (TCGA) data showed a prognostic prompt function of the nine-gene signature, as well as in PCa with Gleason 7. Finally, majority of the nine hub genes were associated with drug sensitivity, mutational landscape, immune infiltrates and clinical characteristics of PCa. Conclusion: The nine-gene signature was correlated with drug sensitivity, mutational landscape, immune infiltrates, clinical characteristics and survival from PCa.
Collapse
Affiliation(s)
- Hai-Qi Mu
- Department of Urology, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhi-Qiang Liang
- Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Shanghai TCM-Integrated Institute of Vascular Anomalies, Shanghai, China
| | - Qi-Peng Xie
- Department of Laboratory Medicine, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wei Han
- Cancer Research Institute, Southern Medical University, Guangzhou, Guangdong, China
| | - Sen Yang
- Department of Urology, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shuai-Bin Wang
- Department of Urology, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Cheng Zhao
- Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Shanghai TCM-Integrated Institute of Vascular Anomalies, Shanghai, China
| | - Ye-Min Cao
- Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Shanghai TCM-Integrated Institute of Vascular Anomalies, Shanghai, China
| | - You-Hua He
- Department of Urology, The Second Affiliated Hospital & Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jian Chen
- Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Shanghai TCM-Integrated Institute of Vascular Anomalies, Shanghai, China
| |
Collapse
|
22
|
Li C, Zheng Y, Pu K, Zhao D, Wang Y, Guan Q, Zhou Y. A four-DNA methylation signature as a novel prognostic biomarker for survival of patients with gastric cancer. Cancer Cell Int 2020; 20:88. [PMID: 32206039 PMCID: PMC7085204 DOI: 10.1186/s12935-020-1156-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 02/26/2020] [Indexed: 12/24/2022] Open
Abstract
Background Gastric cancer (GC) is the fifth most frequently diagnosed cancer and the third leading cause of cancer-related mortality. Lack of prognostic indicators for patient survival hinders GC treatment and survival. Methods and results Methylation profile data of patients with GC obtained from The Cancer Genome Atlas (TCGA) database were analyzed to identify methylation sites as biomarkers for GC prognosis. The cohort was divided into training and validation sets. Univariate Cox, LASSO regression,and multivariate Cox analyses revealed a close correlation of a four-DNA methylation signature as a risk score model with the overall survival of patients with GC. The survival between high-risk and low-risk score patients with GC was significantly different. Analyses of receiver operating characteristics revealed a high prognostic accuracy of the four-DNA methylation signature in patients with GC. The subgroup analysis indicated that the accuracy included that for anatomical region, histologic grade, TNM stage, pathological stage, and sex. The GC prognosis based on the four-DNA methylation signature was more precise than that based on known biomarkers. Conclusions The four-DNA methylation signature could serve as a novel independent prognostic factor that could be an important tool to predict the prognostic outcome of GC patients. This potential must be verified in a large-scale population cohort study and through basic research studies.
Collapse
Affiliation(s)
- Chunmei Li
- 1Key Laboratory for Gastrointestinal Diseases, Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China.,2Department of Oncology, The First Hospital of Lanzhou University, Lanzhou, China
| | - Ya Zheng
- 1Key Laboratory for Gastrointestinal Diseases, Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China.,3Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
| | - Ke Pu
- 1Key Laboratory for Gastrointestinal Diseases, Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China.,3Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
| | - Da Zhao
- 2Department of Oncology, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yuping Wang
- 1Key Laboratory for Gastrointestinal Diseases, Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China.,3Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
| | - Quanlin Guan
- 4Department of Oncology Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yongning Zhou
- 1Key Laboratory for Gastrointestinal Diseases, Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China.,3Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
23
|
A DNA methylation signature to improve survival prediction of gastric cancer. Clin Epigenetics 2020; 12:15. [PMID: 31959204 PMCID: PMC6972030 DOI: 10.1186/s13148-020-0807-x] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 01/02/2020] [Indexed: 02/08/2023] Open
Abstract
Background The current Union International Committee on Cancer or the American Joint Committee on Cancer TNM stage system has shown valuable but insufficient estimation for subsets of gastric cancer and prediction for prognosis patients. Thus, there is an urgent need to identify diagnostic, prognostic, and predictive biomarkers to improve patients’ outcomes. Our aim was to perform an integrative analysis on publicly available datasets to identify epigenetic changes that may play key role in the initiation and progression of gastric cancer, based on which we set to develop a DNA methylation signature to improve survival prediction of gastric cancer. Results A total of 340 methylation-related differentially expression genes (mrDEGs) were screened in gastric cancer patients from The Cancer Genome Atlas (TCGA) project. Pathway enrichment analysis revealed that they were involved in the biological process related to initiation and progression of gastric cancer. Based on the mrDEGs identified, we developed a DNA methylation signature consisting of ten gene members (SCNN1B, NFE2L3, CLDN2, RBPMS2, JPH2, GBP6, COL4A5, SMKR1, PPP1R14A, and ARL4D) according to their methylation β value. This innovative DNA methylation signature was associated with cancer recurrence, while it showed independence of cancer recurrence and TNM stage for survival prediction. Combination of this DNA methylation signature and TNM stage improved overall survival prediction in the receiver operating characteristic analysis. We also verified that two individual genes (PPP1R14A and SCNN1B) of the identified prognostic signature were regulated by promoter region methylation in a panel of gastric cell lines. Conclusions This study presents a powerful DNA methylation signature by performing analyses integrating multi-source data including transcriptome, methylome, and clinical outcome of gastric cancer patients from TCGA. The identified DNA methylation signature may be used to refine the current prognostic model and facilitate further stratification of patients in the future clinical trials. Further experimental studies are warranted to unveil the regulatory mechanism and functional role of all the individual genes of the DNA methylation signature. Also, clinical investigations in large GC patient cohorts are greatly needed to validate our findings.
Collapse
|
24
|
Qin G, Yang L, Ma Y, Liu J, Huo Q. The exploration of disease-specific gene regulatory networks in esophageal carcinoma and stomach adenocarcinoma. BMC Bioinformatics 2019; 20:717. [PMID: 31888440 PMCID: PMC6936086 DOI: 10.1186/s12859-019-3230-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Feed-forward loops (FFLs), consisting of miRNAs, transcription factors (TFs) and their common target genes, have been validated to be important for the initialization and development of complex diseases, including cancer. Esophageal Carcinoma (ESCA) and Stomach Adenocarcinoma (STAD) are two types of malignant tumors in the digestive tract. Understanding common and distinct molecular mechanisms of ESCA and STAD is extremely crucial. RESULTS In this paper, we presented a computational framework to explore common and distinct FFLs, and molecular biomarkers for ESCA and STAD. We identified FFLs by combining regulation pairs and RNA-seq data. Then we constructed disease-specific co-expression networks based on the FFLs identified. We also used random walk with restart (RWR) on disease-specific co-expression networks to prioritize candidate molecules. We identified 148 and 242 FFLs for these two types of cancer, respectively. And we found that one TF, E2F3 was related to ESCA, two genes, DTNA and KCNMA1 were related to STAD, while one TF ESR1 and one gene KIT were associated with both of the two types of cancer. CONCLUSIONS This proposed computational framework predicted disease-related biomolecules effectively and discovered the correlation between two types of cancers, which helped develop the diagnostic and therapeutic strategies of Esophageal Carcinoma and Stomach Adenocarcinoma.
Collapse
Affiliation(s)
- Guimin Qin
- School of Computer Science and Technology, Xidian University, Xi'an, 710071, China
| | - Luqiong Yang
- School of Computer Science and Technology, Xidian University, Xi'an, 710071, China
| | - Yuying Ma
- School of Computer Science and Technology, Xidian University, Xi'an, 710071, China
| | - Jiayan Liu
- School of Computer Science and Technology, Xidian University, Xi'an, 710071, China
| | - Qiuyan Huo
- School of Computer Science and Technology, Xidian University, Xi'an, 710071, China.
| |
Collapse
|
25
|
Xiong J, Tu Y, Feng Z, Li D, Yang Z, Huang Q, Li Z, Cao Y, Jie Z. Epigenetics mechanisms mediate the miR-125a/BRMS1 axis to regulate invasion and metastasis in gastric cancer. Onco Targets Ther 2019; 12:7513-7525. [PMID: 31571904 PMCID: PMC6753057 DOI: 10.2147/ott.s210376] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 08/17/2019] [Indexed: 12/24/2022] Open
Abstract
Purpose Altered expression of breast cancer metastasis suppressor 1 (BRMS1), is a tumor suppressor, which is found in many types of cancers, including gastric cancer (GC), but the mechanism by which BRMS1 inhibits invasion and metastasis in GC is unknown. The aim of the study was to investigate the molecular mechanisms of miR-125a/BRMS1 in GC. Materials and methods The expression of BRMS1 and miR-125a were detected by quantitative real-time PCR (qRT-PCR) and analyzed by bioinformatics. BSP and MSP were used to detecte the methylation status of miR-125a and BRMS1 which was treated by 5-Aza or not. Western Blot and qRT-PCR were used to analyze the expression of BRMS1 and EZH2. Transwell was performed to explore the invasion and metastasis ability of GC cells. The nude mice were used for the tumor formation assay. Results BRMS1 may be regulated by copy number variation (CNV), methylation and miR-125a-5p. As one of the essential components of PRC2, EZH2 is an important regulatory factor resulting in the low expression of miR-125a. An epigenetic mechanism mediates the miR-125a/BRMS1 axis to inhibit the invasion and metastasis of GC cells. In vivo experiments, it is also showed that BRMS1 is involved in invasion and metastasis but not the proliferation in GC. Conclusion These studies shed light on the mechanism of BRMS1 inhibition of GC invasion and metastasis and the development of new drugs targeting the miR-125a/BRMS1 axis, which will be a promising therapeutic strategy for GC and other human cancers.
Collapse
Affiliation(s)
- Jianbo Xiong
- Department of Gastrointestinal Surgery, First Affiliated Hospital, Nanchang University, Nanchang 330006, Jiangxi Province, People's Republic of China
| | - Yi Tu
- Department of Pathology, First Affiliated Hospital, Nanchang University, Nanchang 330006, Jiangxi Province, People's Republic of China
| | - Zongfeng Feng
- Department of Gastrointestinal Surgery, First Affiliated Hospital, Nanchang University, Nanchang 330006, Jiangxi Province, People's Republic of China
| | - Daojiang Li
- Department of General Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei Province, People's Republic of China
| | - Zhouwen Yang
- Department of Gastrointestinal Surgery, First Affiliated Hospital, Nanchang University, Nanchang 330006, Jiangxi Province, People's Republic of China
| | - Qiuxia Huang
- Department of Nursing, First Affiliated Hospital, Nanchang University, Nanchang 330006, Jiangxi Province, People's Republic of China
| | - Zhengrong Li
- Department of Gastrointestinal Surgery, First Affiliated Hospital, Nanchang University, Nanchang 330006, Jiangxi Province, People's Republic of China
| | - Yi Cao
- Department of Gastrointestinal Surgery, First Affiliated Hospital, Nanchang University, Nanchang 330006, Jiangxi Province, People's Republic of China
| | - Zhigang Jie
- Department of Gastrointestinal Surgery, First Affiliated Hospital, Nanchang University, Nanchang 330006, Jiangxi Province, People's Republic of China
| |
Collapse
|
26
|
Moinova HR, LaFramboise T, Lutterbaugh JD, Chandar AK, Dumot J, Faulx A, Brock W, De la Cruz Cabrera O, Guda K, Barnholtz-Sloan JS, Iyer PG, Canto MI, Wang JS, Shaheen NJ, Thota PN, Willis JE, Chak A, Markowitz SD. Identifying DNA methylation biomarkers for non-endoscopic detection of Barrett's esophagus. Sci Transl Med 2019; 10:10/424/eaao5848. [PMID: 29343623 DOI: 10.1126/scitranslmed.aao5848] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 11/08/2017] [Indexed: 12/17/2022]
Abstract
We report a biomarker-based non-endoscopic method for detecting Barrett's esophagus (BE) based on detecting methylated DNAs retrieved via a swallowable balloon-based esophageal sampling device. BE is the precursor of, and a major recognized risk factor for, developing esophageal adenocarcinoma. Endoscopy, the current standard for BE detection, is not cost-effective for population screening. We performed genome-wide screening to ascertain regions targeted for recurrent aberrant cytosine methylation in BE, identifying high-frequency methylation within the CCNA1 locus. We tested CCNA1 DNA methylation as a BE biomarker in cytology brushings of the distal esophagus from 173 individuals with or without BE. CCNA1 DNA methylation demonstrated an area under the curve of 0.95 for discriminating BE-related metaplasia and neoplasia cases versus normal individuals, performing identically to methylation of VIM DNA, an established BE biomarker. When combined, the resulting two biomarker panel was 95% sensitive and 91% specific. These results were replicated in an independent validation cohort of 149 individuals who were assayed using the same cutoff values for test positivity established in the training population. To progress toward non-endoscopic esophageal screening, we engineered a well-tolerated, swallowable, encapsulated balloon device able to selectively sample the distal esophagus within 5 min. In balloon samples from 86 individuals, tests of CCNA1 plus VIM DNA methylation detected BE metaplasia with 90.3% sensitivity and 91.7% specificity. Combining the balloon sampling device with molecular assays of CCNA1 plus VIM DNA methylation enables an efficient, well-tolerated, sensitive, and specific method of screening at-risk populations for BE.
Collapse
Affiliation(s)
- Helen R Moinova
- Department of Medicine, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Thomas LaFramboise
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA.,Department of Genetics and Genome Sciences, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - James D Lutterbaugh
- Department of Medicine, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Apoorva Krishna Chandar
- Department of Medicine, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - John Dumot
- Department of Medicine, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Ashley Faulx
- Department of Medicine, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Wendy Brock
- Department of Medicine, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | | | - Kishore Guda
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Jill S Barnholtz-Sloan
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Prasad G Iyer
- Barrett's Esophagus Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| | - Marcia I Canto
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins Medical Institutions, Baltimore, MD 21205, USA
| | - Jean S Wang
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Nicholas J Shaheen
- Center for Esophageal Diseases and Swallowing, Division of Gastroenterology and Hepatology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Prashanti N Thota
- Digestive Disease and Surgery Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Joseph E Willis
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA. .,Department of Pathology, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA.,University Hospitals Seidman Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Amitabh Chak
- Department of Medicine, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA. .,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA.,University Hospitals Seidman Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Sanford D Markowitz
- Department of Medicine, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA. .,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA.,Department of Genetics and Genome Sciences, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA.,University Hospitals Seidman Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| |
Collapse
|
27
|
Ma G, Liu H, Du M, Zhang G, Lin Y, Ge Y, Wang M, Jin G, Zhao Q, Chu H, Gong W, Zhang Z. A genetic variation in the CpG island of pseudogene GBAP1 promoter is associated with gastric cancer susceptibility. Cancer 2019; 125:2465-2473. [PMID: 30951202 DOI: 10.1002/cncr.32081] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 02/22/2019] [Accepted: 02/27/2019] [Indexed: 01/31/2023]
Abstract
BACKGROUND Previous genome-wide association studies (GWASs) have identified that several single nucleotide polymorphisms (SNPs) are implicated in gastric cancer (GC) risk. However, the multiple statistical comparisons of GWASs may reject some true biological positives with subthreshold P values. METHODS This study annotated the genomic locations of all CpG islands in the genome using the Encyclopedia of DNA Elements (ENCODE). The SNPs in the regions were then genotyped using the Illumina 660W Quad chip. The effects of the prominent variations on GC risk were further confirmed in the other independent cohorts. RESULTS SNP rs2990245, which is located in the promoter of pseudogene GBAP1, was associated with GC risk using GWASs data. An additional cohort of 1275 GC patients and 1424 controls validated that individuals with the CC genotype had a 62% decreased risk of GC compared with those who carried the TT genotype (P = 2.01E-04) in the codominant model. The significant association was observed in the additive, dominant, and recessive models. A meta-analysis combining the results from the GWASs and replication studies revealed that rs2990245 was significantly associated with decreased GC risk (P = 5.59E-12). Importantly, rs2990245 can regulate the expression of GBAP1 by influencing the methylation status of the GBAP1 promoter. GBAP1 can act as a competing endogenous RNA by binding competitively with micro-RNA-212-3p and then promoting GBA expression. CONCLUSION rs2990245 is significantly associated with a decreased risk of GC. Pseudogene GBAP1 contributes to the development and progression of GC by sequestering the miR-212-3p from binding to GBA.
Collapse
Affiliation(s)
- Gaoxiang Ma
- Department of Environmental Genomics, Center for Global Health, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center For Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.,Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China.,School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Hanting Liu
- Department of Environmental Genomics, Center for Global Health, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center For Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.,Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Mulong Du
- Department of Environmental Genomics, Center for Global Health, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center For Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.,Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China.,Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Gang Zhang
- Department of Environmental Genomics, Center for Global Health, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center For Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.,Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Yadi Lin
- Department of Environmental Genomics, Center for Global Health, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center For Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.,Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Yuqiu Ge
- Department of Environmental Genomics, Center for Global Health, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center For Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.,Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Meilin Wang
- Department of Environmental Genomics, Center for Global Health, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center For Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.,Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Guangfu Jin
- Department of Epidemiology and Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Qinghong Zhao
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Haiyan Chu
- Department of Environmental Genomics, Center for Global Health, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center For Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.,Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Weida Gong
- Department of General Surgery, Yixing Cancer Hospital, Yixing, China
| | - Zhengdong Zhang
- Department of Environmental Genomics, Center for Global Health, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center For Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.,Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| |
Collapse
|
28
|
Basile MS, Fagone P, Mangano K, Mammana S, Magro G, Salvatorelli L, Li Destri G, La Greca G, Nicoletti F, Puleo S, Pesce A. KCNMA1 Expression is Downregulated in Colorectal Cancer via Epigenetic Mechanisms. Cancers (Basel) 2019; 11:cancers11020245. [PMID: 30791468 PMCID: PMC6406553 DOI: 10.3390/cancers11020245] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 02/14/2019] [Accepted: 02/16/2019] [Indexed: 02/07/2023] Open
Abstract
KCNMA1 is a gene located at 10q22 that encodes the pore-forming α-subunit of the large-conductance Ca2+-activated K⁺ channel. KCNMA1 is down-regulated in gastric carcinoma tumors, through hypermethylation of its promoter. In the present study, we have evaluated the expression levels of KCNMA1 both in a mouse model of Colorectal Cancer (CRC) and in human CRC samples. Additionally, epigenetic mechanisms of KCNMA1 gene regulation were investigated. We observed a significant down-regulation of KCNMA1 both in a human and mouse model of CRC. No differences in KCNMA1 levels were, however, observed at different TNM stages. We also wanted to determine whether the modulation in KCNMA1 was dependent on epigenetic mechanisms. A statistically significant inverse correlation between KCNMA1 expression and mir-17-5p levels was observed in patients with CRC. Furthermore, in the tumor samples, we found a significant hypermethylation of the promoter, in the loci cg24113782 and cg25655799, compared to healthy tissue. Overall, our data suggest the possible use of KCNMA1 as a therapeutic target in the early stages of CRC.
Collapse
Affiliation(s)
- Maria Sofia Basile
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy.
| | - Paolo Fagone
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy.
| | - Katia Mangano
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy.
| | - Santa Mammana
- IRCCS Centro Neurolesi "Bonino-Pulejo", Strada Statale 113, C.da Casazza, 98124 Messina, Italy.
| | - Gaetano Magro
- Department of Medical and Surgical Sciences and Advanced Technology "G.F. Ingrassia", University of Catania, Via Santa Sofia 86, 95123 Catania, Italy.
| | - Lucia Salvatorelli
- Department of Medical and Surgical Sciences and Advanced Technology "G.F. Ingrassia", University of Catania, Via Santa Sofia 86, 95123 Catania, Italy.
| | - Giovanni Li Destri
- Department of Medical and Surgical Sciences and Advanced Technology "G.F. Ingrassia", University of Catania, Via Santa Sofia 86, 95123 Catania, Italy.
| | - Gaetano La Greca
- Department of Medical and Surgical Sciences and Advanced Technology "G.F. Ingrassia", University of Catania, Via Santa Sofia 86, 95123 Catania, Italy.
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 89, 95123 Catania, Italy.
| | - Stefano Puleo
- Department of Medical and Surgical Sciences and Advanced Technology "G.F. Ingrassia", University of Catania, Via Santa Sofia 86, 95123 Catania, Italy.
| | - Antonio Pesce
- Department of Medical and Surgical Sciences and Advanced Technology "G.F. Ingrassia", University of Catania, Via Santa Sofia 86, 95123 Catania, Italy.
| |
Collapse
|
29
|
Wang F, Chen Q, Huang G, Guo X, Li N, Li Y, Li B. BKCa participates in E2 inducing endometrial adenocarcinoma by activating MEK/ERK pathway. BMC Cancer 2018; 18:1128. [PMID: 30445932 PMCID: PMC6240221 DOI: 10.1186/s12885-018-5027-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 10/31/2018] [Indexed: 11/10/2022] Open
Abstract
Background The large-conductance, voltage-gated, calcium (Ca (2+))-activated potassium channel (BKCa) plays an important role in regulating Ca (2+) signaling and cell physiological function, and is aberrantly expressed in some types of cancers. The present study focuses on identifying the oncogenic potential and clinical significance of BKCa in endometrial adenocarcinoma, as well as exploring the mechanistic relevance by 17β -estradiol (E2) inducing aberrant activation of MEK1/2 and ERK1/2 via BKCa. Methods The expression of BKCa, ERK1/2 and p-ERK1/2 were examined by immunohistochemical staining in 263 cases, including 185 primary types I endometrial cancer tissues, 38 atypical endometrial hyperplasia tissues and 40 normal endometrium tissues. Cell growth, cycle, apoptosis rate, migration and invasion was separately tested in Ishikawa cells using siRNA-BKCa and/or E2 treatment, as well as the expression of these interested proteins by western blot analysis. Results We showed that expression of BKCa is significantly elevated in 185 types I endometrial adenocarcinoma tissues compared to those of the normal endometrium and atypical endometrial hyperplasia tissues. Furthermore, in vitro observations revealed that down-regulation of BKCa expression inhibited cell growth by both enhancing apoptosis and blocking G1/S transition, suppressed cell migration and invasion in Ishakiwa cells, and decreased the expression of p-MEK1/2 and p-ERK1/2. Additionally, RNAi-mediated knockdown of BKCa attenuated the increased cellular growth and invasion, as well as the elevated expression of p-MEK1/2 and p-ERK1/2 proteins, induced by E2 stimulation. More importantly, the aberrant expression of BKCa and p-ERK1/2 were closely related with poor prognostic factors in type I endometrial cancer, and up-regulated expression of p-ERK1/2 was significantly associated with shorter disease-free survival (DFS) and overall survival (OS) and was an independent prognostic factor in type I endometrial cancer patients. Conclusion Our results demonstrated that BKCa and the key downstream effectors p-ERK1/2 could be involved in important signaling pathways in initiation and development of endometrial adenocarcinoma and may provide a new therapeutic approach for women with endometrial cancer. Electronic supplementary material The online version of this article (10.1186/s12885-018-5027-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Fenfen Wang
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University, School of Medicine, Xueshi Road 1, Hangzhou, Zhejiang, 310006, People's Republic of China
| | - Qin Chen
- Department of Pathology, Women's Hospital, Zhejiang University, School of Medicine, Xueshi Road 1, Hangzhou, Zhejiang, 310006, People's Republic of China
| | - Genping Huang
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University, School of Medicine, Xueshi Road 1, Hangzhou, Zhejiang, 310006, People's Republic of China
| | - Xuedong Guo
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University, School of Medicine, Xueshi Road 1, Hangzhou, Zhejiang, 310006, People's Republic of China
| | - Na Li
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University, School of Medicine, Xueshi Road 1, Hangzhou, Zhejiang, 310006, People's Republic of China
| | - Yang Li
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University, School of Medicine, Xueshi Road 1, Hangzhou, Zhejiang, 310006, People's Republic of China
| | - Baohua Li
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University, School of Medicine, Xueshi Road 1, Hangzhou, Zhejiang, 310006, People's Republic of China.
| |
Collapse
|
30
|
Sawaki K, Kanda M, Kodera Y. Review of recent efforts to discover biomarkers for early detection, monitoring, prognosis, and prediction of treatment responses of patients with gastric cancer. Expert Rev Gastroenterol Hepatol 2018; 12:657-670. [PMID: 29902383 DOI: 10.1080/17474124.2018.1489233] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Gastric cancer (GC) is the leading cause of cancer-related death worldwide. Despite recent advances in diagnosis and therapy, the prognosis of patients with GC is poor. Many patients have inoperable disease upon diagnosis or experience recurrent disease after curative gastrectomy. Unfortunately, tumor markers for GC, such as serum carcinoembryonic antigen and carbohydrate antigen 19-9, lack sufficient sensitivity and specificity. Therefore, effective biomarkers are required to detect early GC and to predict tumor recurrence and chemosensitivity. Areas covered: Here we aimed to review recent developments in techniques that improve the detection of aberrant expression of GC-associated molecules, including protein coding genes, microRNAs, long noncoding RNAs, and methylated promoter DNAs. Expert commentary: Detection of genetic and epigenetic alterations in gastric tissue or in the circulation will likely improve the diagnosis and management of GC to achieve significantly improved outcomes.
Collapse
Affiliation(s)
- Koichi Sawaki
- a Department of Gastroenterological Surgery (Surgery II) , Nagoya University Graduate School of Medicine , Nagoya , Japan
| | - Mitsuro Kanda
- a Department of Gastroenterological Surgery (Surgery II) , Nagoya University Graduate School of Medicine , Nagoya , Japan
| | - Yasuhiro Kodera
- a Department of Gastroenterological Surgery (Surgery II) , Nagoya University Graduate School of Medicine , Nagoya , Japan
| |
Collapse
|