1
|
Zhou T, Niu Y, Li Y. Advances in research on malignant tumors and targeted agents for TOP2A (Review). Mol Med Rep 2025; 31:50. [PMID: 39670307 DOI: 10.3892/mmr.2024.13415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 11/28/2024] [Indexed: 12/14/2024] Open
Abstract
The DNA topoisomerase isoform topoisomerase IIα (TOP2A) is essential for the condensation and segregation of cellular mitotic chromosomes and the structural maintenance. It has been demonstrated that TOP2A is highly expressed in various malignancies, including lung adenocarcinoma (LUAD), hepatocellular carcinoma (HCC) and breast cancer (BC), associating with poor prognosis and aggressive tumor behavior. Additionally, TOP2A has emerged as a promising target for cancer therapy, with widespread clinical application of associated chemotherapeutic agents. The present study explored the impact of TOP2A on malignant tumor growth and the advancements in research on its targeted drugs. The fundamental mechanisms of TOP2A have been detailed, its specific roles in tumor cells are analyzed, and its potential as a biomarker for tumor prognosis and therapeutic targeting is highlighted. Additionally, the present review compiles findings from the latest clinical trials of relevant targeted agents, information on newly developed inhibitors, and discusses future research directions and clinical application strategies in cancer therapy, aiming to propose novel ideas and methods.
Collapse
Affiliation(s)
- Tao Zhou
- Department of Hepatobiliary Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi 030032, P.R. China
| | - Yiting Niu
- Department of Hepatobiliary Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi 030032, P.R. China
| | - Yanjun Li
- Department of Hepatobiliary Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi 030032, P.R. China
| |
Collapse
|
2
|
Pan Y, Ma Y, Guan H, Dai G. Pre-treatment of hyponatremia as a biomarker for poor immune prognosis in advanced or metastatic gastric cancer: A retrospective case analysis. Hum Vaccin Immunother 2024; 20:2414546. [PMID: 39411929 PMCID: PMC11486141 DOI: 10.1080/21645515.2024.2414546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 09/22/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
Hyponatremia, a prevalent electrolyte imbalance among tumor patients, has often been overlooked regarding its prognostic significance for immunotherapy. In this study, we delved into the prognostic ramifications of hyponatremia in advanced gastric cancer (AGC) patients undergoing immunotherapy. Enrolling AGC patients diagnosed between December 2014 and May 2021, we extracted pertinent data from electronic medical records, with a median follow-up of 35.8 months. Kaplan-Meier curves illuminated patients' progression-free survival (PFS) and overall survival (OS), while survival disparities were tested using the Mantel-Haenszel log rank test. COX and logistic regressions were employed to scrutinize the correlation between serum sodium levels and prognosis in 268 AGC patients, both at baseline and during treatment. Notably, patients with hyponatremia exhibited shorter PFS (4.7 vs 2.1 months, p = .001*) and OS (12.5 vs 3.9 months, p < .001*). Serum sodium emerged as an independent prognostic factor for both PFS (HR = 1.773; 95% CI 1.067-2.945; p = .001*) and OS (HR = 1.773; 95% CI 1.067-2.945; p = .003*). Subgroup analysis revealed that AGC patients with hyponatremia derived no benefit from immunotherapy in terms of PFS and OS. Strikingly, a decrease in serum sodium during immunotherapy was associated with early relapse and mortality. Based on these findings, we hypothesize that hyponatremia portends poor prognostic outcomes in AGC patients treated with immunotherapy and may serve as a valuable prognostic biomarker. However, further large-scale prospective studies are warranted to validate these observations.
Collapse
Affiliation(s)
- Yuting Pan
- Department of Medical Oncology, Medical School of Chinese PLA, Beijing, China
- Department of Medical Oncology, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Yue Ma
- Department of Medical Oncology, Medical School of Chinese PLA, Beijing, China
- Department of Medical Oncology, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Huafang Guan
- External Relations Office, Yingtan City People’s Hospital, Yingtan, China
| | - Guanghai Dai
- Department of Medical Oncology, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
3
|
Qin G, Wu R, Wang Q, Sun M, Li Y, Duan S, Xu FJ. Injectable Hyaluronic Acid-Based Hydrogels for Rapid Endoscopic Submucosal Dissection. ACS Biomater Sci Eng 2024; 10:7657-7666. [PMID: 39563065 DOI: 10.1021/acsbiomaterials.4c01703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
Endoscopic submucosal dissection (ESD) is a widely used procedure for the treatment of early and precancerous gastrointestinal lesions and has become the standard treatment. In this procedure, the commonly used materials have a short retention time and a limited lifting capacity, which will prolong the duration of the ESD procedure. Furthermore, these liquids tend to diffuse after ESD surgery, failing to adequately protect the wound. Therefore, we designed and developed injectable hydrogels based on hyaluronic acid. A series of oxidized hyaluronic acid (OHA) and hydrazide hyaluronic acid (AHA) were synthesized, and 16 kinds of injectable hydrogels were fabricated to investigate the effects of molecular structures on the properties of the hydrogels. Among these, the O1A3 hydrogel exhibited a suitable injection performance, gelation time, and mechanical properties, along with good blood and cell compatibility in vitro. Subsequently, in a porcine model of the ESD procedure, the results demonstrated that the O1A3 hydrogel exhibited a good retention time and lifting performance while also significantly reducing the operation time from 1-2 h to ∼10 min. Furthermore, the adhesive property of the O1A3 hydrogel on small bleeding spots and wounds could be observed, which was beneficial in protecting the wound from the complex environment of the gastrointestinal tract. The present work of injectable hyaluronic acid-based hydrogels could be promising to improve the efficiency of ESD surgery.
Collapse
Affiliation(s)
- Geng Qin
- Department of Gastroenterology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Ruonan Wu
- State Key Laboratory of Chemical Resource Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology), Ministry of Education, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
| | - Qianqian Wang
- Department of Gastroenterology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Meizhou Sun
- State Key Laboratory of Chemical Resource Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology), Ministry of Education, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
| | - Yang Li
- State Key Laboratory of Chemical Resource Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology), Ministry of Education, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
| | - Shun Duan
- State Key Laboratory of Chemical Resource Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology), Ministry of Education, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
| | - Fu-Jian Xu
- State Key Laboratory of Chemical Resource Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology), Ministry of Education, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
| |
Collapse
|
4
|
Zhang G, Shi A, Ding X, Wang J. The value of a nomogram based on 18F-FDG PET/CT metabolic parameters and metabolic heterogeneity in predicting distant metastasis in gastric cancer. Jpn J Clin Oncol 2024:hyae169. [PMID: 39657166 DOI: 10.1093/jjco/hyae169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 11/23/2024] [Indexed: 12/17/2024] Open
Abstract
OBJECTIVE To investigate the value of metabolic parameters and metabolic heterogeneity from pretreatment deoxy-2-[fluorine-18]-fluoro-D-glucose positron emission tomography/computed tomography (18F-FDG PET/CT) in predicting distant metastasis in gastric cancer. METHODS Eighty-six patients with pathologically confirmed gastric adenocarcinoma were included in this study. All patients underwent a whole-body 18F-FDG PET/CT scan before treatment. Clinicopathologic and imaging data were collected, including metabolic parameters such as maximum standardized uptake value (SUVmax), mean standardized uptake value (SUVmean), metabolic tumor volume (MTV), and total lesion glycolysis (TLG) of the primary gastric cancer lesions. Heterogeneity index (HI)-1 was expressed as the absolute value of the linear regression slopes between the MTVs at different SUVmax thresholds (40% × SUVmax, 80% × SUVmax), while HI-2 was expressed as the difference between SUVmax and SUVmean. Patients were randomly divided into training and validation cohorts at a 7:3 ratio. The correlation between the above parameters and distant metastasis in gastric cancer was analyzed using the training cohort. A nomogram prediction model was then established and later verified with the validation cohort. Finally, decision curve analysis was used to evaluate the clinical utility of the model. RESULTS This study included 86 patients with gastric cancer, with 60 (69.8%) in the training cohort and 26 (30.2%) in the validation cohort. There was no significant difference in the balanced comparison between both cohorts (all P > .05). Among all patients, 31 (36.0%) developed distant metastasis, while 55 (64.0%) did not. In patients who developed distant tumor metastasis, carcinoembryonic antigen, carbohydrate antigen (CA)12-5, CA19-9, CA72-4, MTV, TLG, and HI-1 were significantly higher than in patients without distant metastasis (all P < .05). Multivariate logistic regression analysis identified CA72-4 (OR: 1.151, 95% CI: 1.020-1.300, P = .023) and HI-1 (OR: 1.647, 95% CI: 1.063-2.553, P = .026) as independent risk factors for predicting distant metastasis in gastric cancer. The nomogram constructed from this analysis exhibited high predictive efficacy in the training (AUC: 0.874, 95% CI: 0.766-0.983) and validation (AUC: 0.915, 95% CI: 0.790-1.000) cohorts, providing a net clinical benefit for patients. CONCLUSION HI-1 is an independent risk factor for predicting distant metastasis in gastric cancer. A comprehensive prediction model combining HI-1 with the tumor marker CA72-4 can increase the net clinical benefit for patients.
Collapse
Affiliation(s)
- Guanjie Zhang
- Department of Nuclear Medicine, Second Affiliated Hospital of Fujian Medical University, Donghai Street No. 950, Fengze District, Quanzhou 362018, PR China
- Second Clinical School, Second Affiliated Hospital of Fujian Medical University, Donghai Street No. 950, Fengze District, Quanzhou 362018, PR China
| | - Aiqi Shi
- Department of Nuclear Medicine, Lanzhou University Second Hospital, Cuiyingmen No. 82, Chengguan District, Lanzhou 730030, PR China
| | - Xiaofang Ding
- PET-CT Center of Wuwei Tumor Hospital, Weisheng Lane No. 31, Liangzhou District, Wuwei 733000, PR China
| | - Jianlin Wang
- Department of Nuclear Medicine, Second Affiliated Hospital of Fujian Medical University, Donghai Street No. 950, Fengze District, Quanzhou 362018, PR China
- Second Clinical School, Second Affiliated Hospital of Fujian Medical University, Donghai Street No. 950, Fengze District, Quanzhou 362018, PR China
| |
Collapse
|
5
|
Lin L, Xiao X, Guo X, Zhong C, Zhuang M, Xu J, Wang Y, Chen F. AKR1C3 mediates gastric cancer cell invasion and metastasis via the AKT and JNK/p-NF-κB signaling pathways. Sci Rep 2024; 14:30263. [PMID: 39632995 PMCID: PMC11618362 DOI: 10.1038/s41598-024-82039-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024] Open
Abstract
Gastric cancer (GC) is globally recognized as the fifth most common cancer and the third leading cause of cancer-related mortality. Early metastasis in GC significantly contributes to its high mortality and unfavorable prognosis. However, the underlying mechanisms of this phenomenon remain largely unexplored. Among the various factors involved, AKR1C3 has emerged as a crucial component in the pathways of tumorigenesis and metastasis across multiple cancer types. Yet, the precise significance of AKR1C3 in GC patients' prognosis and its role in GC progression remain elusive. This study illuminated the significant downregulation of AKR1C3 in GC tissues, linking it to an aggressive phenotype and poor prognosis. Interestingly, while AKR1C3 overexpression did not affect the proliferation of GC cells, it significantly inhibited their ability to invade and metastasize. The underlying mechanism appears to involve AKR1C3's inhibition of the p-JNK pathway, which leads to reduced phosphorylation of IKKα/β and IKBα, lowering p-NF-κB levels and hindering its movement into the nucleus, thereby stifling the epithelial-mesenchymal transition (EMT) process in GC cells. These insights reveal AKR1C3's tumor-suppressive effects in GC and suggest its potential as a diagnostic and prognostic biomarker, offering new avenues for targeted therapies in gastric cancer management.
Collapse
Affiliation(s)
- Liying Lin
- Department of Gastroenterology and Fujian Institute of Digestive Disease, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, Fujian, China
| | - Xinzhu Xiao
- Department of Infectious disease, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian, China
- Binhai Campus of the First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, Fuzhou, 350212, Fujian, China
| | - Xiaoxiong Guo
- Department of Gastroenterology and Fujian Institute of Digestive Disease, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, Fujian, China
| | - Canmei Zhong
- Department of Gastroenterology and Fujian Institute of Digestive Disease, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, Fujian, China
| | - Mingkai Zhuang
- Department of Gastroenterology and Fujian Institute of Digestive Disease, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, Fujian, China
| | - Jie Xu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, Fujian, China
| | - Yin Wang
- Department of Gastroenterology, Tongan Ward of the First Affiliated Hospital of Xiamen University, Xiamen, 361026, Fujian, China
| | - Fenglin Chen
- Department of Gastroenterology and Fujian Institute of Digestive Disease, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, 350001, Fujian, China.
| |
Collapse
|
6
|
Wang Z, Chen H, Sun L, Wang X, Xu Y, Tian S, Liu X. Uncovering the potential of APOD as a biomarker in gastric cancer: A retrospective and multi-center study. Comput Struct Biotechnol J 2024; 23:1051-1064. [PMID: 38455068 PMCID: PMC10918487 DOI: 10.1016/j.csbj.2024.02.015] [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: 10/21/2023] [Revised: 02/16/2024] [Accepted: 02/16/2024] [Indexed: 03/09/2024] Open
Abstract
Gastric cancer (GC) poses a significant health challenge worldwide, necessitating the identification of predictive biomarkers to improve prognosis. Dysregulated lipid metabolism is a well-recognized hallmark of tumorigenesis, prompting investigation into apolipoproteins (APOs). In this study, we focused on apolipoprotein D (APOD) following comprehensive analyses of APOs in pan-cancer. Utilizing data from the TCGA-STAD and GSE62254 cohorts, we elucidated associations between APOD expression and multiple facets of GC, including prognosis, tumor microenvironment (TME), cancer biomarkers, mutations, and immunotherapy response, and identified potential anti-GC drugs. Single-cell analyses and immunohistochemical staining confirmed APOD expression in fibroblasts within the GC microenvironment. Additionally, we independently validated the prognostic significance of APOD in the ZN-GC cohort. Our comprehensive analyses revealed that high APOD expression in GC patients was notably associated with unfavorable clinical outcomes, reduced microsatellite instability and tumor mutation burden, alterations in the TME, and diminished response to immunotherapy. These findings provide valuable insights into the potential prognostic and therapeutic implications of APOD in GC.
Collapse
Affiliation(s)
- Zisong Wang
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei Province, China
- School of Basic Medical Sciences, Wuhan University, Wuhan 430071, Hubei Province, China
| | - Hongshan Chen
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei Province, China
| | - Le Sun
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei Province, China
| | - Xuanyu Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei Province, China
| | - Yihang Xu
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei Province, China
| | - Sufang Tian
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei Province, China
| | - Xiaoping Liu
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan 430071, Hubei Province, China
| |
Collapse
|
7
|
Yu L, Bao S, Zhu F, Xu Y, Liu Y, Jiang R, Yang C, Cao F, Chen W, Li P. Association between branched-chain amino acid levels and gastric cancer risk: large-scale prospective cohort study. Front Nutr 2024; 11:1479800. [PMID: 39634548 PMCID: PMC11614650 DOI: 10.3389/fnut.2024.1479800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 10/29/2024] [Indexed: 12/07/2024] Open
Abstract
Background Gastric cancer (GC) remains a malignancy with high incidence and mortality rates worldwide. Although branched-chain amino acids (BCAAs) play a crucial role in various physiological and pathological processes, their specific relationship with risk of GC remains unclear. Methods We conducted a large-scale prospective cohort from UK Biobank database. We evaluated the relationship between BCAA levels and risk of GC using Cox regression, Kaplan-Meier survival curves, the accelerated failure time (AFT) model, and restricted cubic spline (RCS) analysis. Results During the follow-up of 12 years, 247,753 participants were included in the study. And the Cox regression analysis revealed that higher levels of isoleucine (HR = 0.65, 95% CI 0.48-0.89; p = 0.007), leucine (HR = 0.57, 95% CI 0.42-0.79; p < 0.001), valine (HR = 0.53, 95% CI 0.39-0.73; p < 0.001), and total BCAAs were associated with a reduced risk of GC (HR = 0.51, 95% CI 0.37-0.70; p < 0.001). Kaplan-Meier curves and the AFT model confirmed that elevated BCAA levels significantly delayed the onset of GC. Additionally, RCS analysis identified nonlinear dose-response relationships between BCAAs and risk of GC. Stratified analyses indicated that the protective effect of BCAAs was consistent across various subgroups, with a more pronounced impact in older individuals without chronic diseases. Conclusion Elevated BCAA levels are significantly associated with a reduced risk of GC, particularly in older adults. This finding highlights the potential of BCAAs in GC prevention and suggests that future research and clinical practice should emphasize regulating BCAA levels.
Collapse
Affiliation(s)
- Liang Yu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei City, China
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei City, China
| | - Shiming Bao
- Department of Emergency Surgery, Tongling People's Hospital, Tongling, China
| | - Feng Zhu
- Department of General Surgery, Tongling People's Hospital, Tongling, China
| | - Yanyan Xu
- Department of Emergency Surgery, Tongling People's Hospital, Tongling, China
| | - Yanwei Liu
- Department of Emergency Surgery, Tongling People's Hospital, Tongling, China
| | - Runben Jiang
- Department of Emergency Surgery, Tongling People's Hospital, Tongling, China
| | - Chuang Yang
- Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Feng Cao
- Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Wei Chen
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei City, China
| | - Pengtao Li
- Department of Emergency Medicine, No.2 People's Hospital of Fuyang City, Fuyang City, Anhui Province, China
- Department of Emergency Medicine, Fuyang Infectious Disease Clinical College of Anhui Medical University, Hefei City, China
| |
Collapse
|
8
|
Zhong Y, Liu J, Sun K, Wang G, Li B, Liu Y, Chen N, Chen Z. Gastric SMARCA4-Deficient Undifferentiated Carcinoma: A Report of 4 Patients and Literature Review. Int J Surg Pathol 2024:10668969241291884. [PMID: 39563524 DOI: 10.1177/10668969241291884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
SMARCA4-deficient undifferentiated carcinoma (SMARCA4-DUC) of the stomach is a rare gastric tumor that has been receiving increased attention in recent years. With its varied pathological presentations, accurate diagnosis can be challenging. In order to improve our understanding of this aggressive neoplasm, we have carefully documented and analyzed 4 patients with gastric SMARCA4-DUC, adding to the overall knowledge of this uncommon malignancy. The patients were all men, with an average age of 69 years (range 57-76 years). The tumors were located in the gastric antrum (2 patients), the gastric body (1 patient), and the cardia (1 patient). Microscopically, the 3 surgical specimens exhibited similar morphological features: large to medium sized tumor cells, round, irregular to epithelioid undifferentiated cells in solid sheets or discohesive nests, with no obvious stromal reaction. In one patient, short spindle cells arranged in reticular, pseudopapillary, and disorderly patterns were observed amidst extensive interstitial edema. Some areas displayed a lymphoma-like starry sky phenomenon. The biopsy specimens showed large cells diffusely distributed in the submucosa, presenting pale pink cytoplasm, fine chromatin, and prominent large red nucleoli resembling melanoma. All 4 patients demonstrated BRG1 (SMARCA4) loss, INI1 (SMARCB1) retention, and focal positivity for epithelial markers (AE1/AE3 or epithelial membrane antigen). Three patients had clinically advanced (stages IIIC-IV) disease. Follow-up revealed that 1 patient deceased within 4.8 months. This article expands the morphological spectrum of this cancer, providing new insights for accurate cancer diagnosis.
Collapse
Affiliation(s)
- Yuanli Zhong
- Department of Pathology, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Zhejiang Province, China
| | - Jin Liu
- Department of Pathology, Huzhou Central Hospital, Zhejiang Province, China
| | - Ke Sun
- Department of Pathology, the First Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang Province, China
| | - Gangping Wang
- Department of Pathology, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Zhejiang Province, China
| | - Baizhou Li
- Department of Pathology, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Zhejiang Province, China
| | - Yuqing Liu
- Department of Pathology, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Zhejiang Province, China
| | - Nuo Chen
- Department of Pathology, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Zhejiang Province, China
| | - Zhenwei Chen
- Department of Pathology, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Zhejiang Province, China
| |
Collapse
|
9
|
Zhao Z, Li L, Liu Y, Shi L, Yuan M, Shi H, Ji Q, Liu G, Sun J. Prognostic value and immunomodulatory role of DNM1L in gastric adenocarcinoma. Front Oncol 2024; 14:1453795. [PMID: 39507763 PMCID: PMC11540555 DOI: 10.3389/fonc.2024.1453795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 09/30/2024] [Indexed: 11/08/2024] Open
Abstract
Background Mitochondrial fusion and fission are critical for the morphology and function of cells. DNM1L encodes dynamin-related protein 1 (DRP1), a key protein mediating mitochondrial fission, which is upregulated in a variety of cancers and is strongly associated with tumorigenesis. We aim to investigate the relationship between DNM1L and the prognosis of gastric cancer, as well as to explore the function and mechanism of DNM1L in gastric cancer (GC). Materials and methods In this study, we analyzed the expression differences of DNM1L in gastric cancer tissues and paracancerous tissues using The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO) database. This was followed by validation through tissue microarrays. We then utilized the cohort information from these microarrays to explore the relationship between DNM1L expression and gastric cancer prognosis. Furthermore, we conducted enrichment analysis to investigate the function and mechanisms of DNM1L in gastric cancer, and lastly, we performed immune cell infiltration analysis using the CIBERSORT algorithm. Results We discovered that the expression of DNM1L is elevated in GC tissues. TCGA data showed that the overexpression of DNM1L was positively correlated with the T-stage of GC but not with lymph node metastasis, which was also corroborated by our immunohistochemistry experiments. Based on the Kaplan-Meier curves, the high DNM1L expression was remarkably correlated with poor overall survival in patients with GC. In addition, results of COX regression analysis indicated that high DNM1L expression was an independent prognostic factor in patients with GC. Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Gene set enrichment analysis (GSEA) showed that DNM1L was closely associated with multiple signaling pathways and immune responses. CIBERSORT analysis indicated that increased DNM1L expression may affect the infiltration of immune cells in the tumor microenvironment. Conclusion The results of this study indicate that DNM1L is upregulated in gastric cancer (GC) and positively correlates with the T-stage and poor prognosis of GC patients, and it plays an important role in tumor immune infiltration.
Collapse
Affiliation(s)
- Zhuo Zhao
- Department of Peripheral Vascular Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lingxia Li
- Department of Clinical Laboratory, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yan Liu
- Department of Clinical Laboratory, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lei Shi
- Department of Clinical Laboratory, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Meijie Yuan
- Department of Peripheral Vascular Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hongshuo Shi
- Department of Peripheral Vascular Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qing Ji
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guobin Liu
- Department of Peripheral Vascular Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jian Sun
- Department of Peripheral Vascular Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
10
|
Zhu C, Da M, Wu Z, Ma J, Zhu C, An X, Ji D, Xu C. Meta-analysis of FTO expression on the clinicopathologic characteristics and prognosis of gastric cancer. Medicine (Baltimore) 2024; 103:e35714. [PMID: 39331892 PMCID: PMC11441928 DOI: 10.1097/md.0000000000035714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 09/28/2023] [Indexed: 09/29/2024] Open
Abstract
BACKGROUND Fat mass and obesity-related gene (FTO) is aberrantly expressed in various cancers including highly expressed in gastric cancer tissues. The aim of this meta-analysis was to explore the effect of FTO expression on clinicopathological and prognostic outcome of gastric cancer. METHODS China National Knowledge Infrastructure (CNK), Wanfang database, VIP database, Chinese biomedical literature database (CBM), PubMed, Web of Science, the Cochrane library and EMBASE database were searched to screen the literatures according to the inclusion criteria. The search time was the database establishment until May 2023. The two researchers independently searched and screened the literature, extracted pathological data, and conducted The Newcastle-Ottawa scale (NOS) quality evaluation. Analyze the correlation between FTO and pathological indicators of gastric cancer patients and the impact on prognosis, use and Stata 12.0, software for Meta-analysis. RESULTS A total of 1619 patients were studied in this study. The results of the Meta-analysis showed that higher expression levels of FTO were associated with TMN stage (OR = 1.83, 95% CI: 1.11-3.03, P = .019), liver metastases (OR = 3.73, 95% CI: 1.49-9.31, P = .005), vascular invasion (OR = 2.22, 95% CI: 1.36-3.61, P = .001), poorer overall survival (OS) (HR = 0.46, 95% CI: 0.34-0.58, P < .001) and recurrence-free survival (HR = 0.56, 95% CI: 0.40-0.73, P < .001) in gastric cancer patients. There was no significant relationship with the degree of differentiation (OR = 1.08, 95% CI: 0.49-2.35, P = .852), age (OR = 0.89, 95% CI: 0.71-1.11, P = .306), and gender (OR = 0.92, 95% CI: 0.74-1.14, P = .432). CONCLUSION High expression of FTO was associated with risk of distant metastases and poor prognosis for patients with gastric cancer. FTO may be a potential prognostic biomarker for gastric cancer, but due to the limited number of literature, the above results need further research.
Collapse
Affiliation(s)
- Ciba Zhu
- The First Clinical Medicine College, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Mingxu Da
- The First Clinical Medicine College, Gansu University of Traditional Chinese Medicine, Lanzhou, China
- Department of Oncology Surgery, Gansu Provincial Hospital, Lanzhou, China
| | - Ziyao Wu
- The First Clinical Medicine College, Lanzhou University, Lanzhou, China
| | - Jichun Ma
- The First Clinical Medicine College, Lanzhou University, Lanzhou, China
| | - Chenglou Zhu
- The First Clinical Medicine College, Lanzhou University, Lanzhou, China
| | - Xinqiao An
- The First Clinical Medicine College, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Dandan Ji
- The First Clinical Medicine College, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Chunling Xu
- Department of Hepatobiliary and Hernia Surgery, Wuwei Liangzhou Hospital, Wuwei, China
| |
Collapse
|
11
|
Mazurek M, Szewc M, Sitarz MZ, Dudzińska E, Sitarz R. Gastric Cancer: An Up-to-Date Review with New Insights into Early-Onset Gastric Cancer. Cancers (Basel) 2024; 16:3163. [PMID: 39335135 PMCID: PMC11430327 DOI: 10.3390/cancers16183163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/06/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Gastric cancer (GC) is the fifth most frequently diagnosed cancer and the fifth most common cause of cancer death in the world. Regarding the age at which the diagnosis was made, GC is divided into early-onset gastric cancer (EOGC-up to 45 years of age) and conventional GC (older than 45). EOGC constitutes approximately 10% of all GCs. Numerous reports indicate that EOGC is more aggressive than conventional GC and is often discovered at an advanced tumor stage, which has an impact on the five-year survival rate. The median survival rate for advanced-stage GC is very poor, amounting to less than 12 months. Risk factors for GC include family history, alcohol consumption, smoking, Helicobacter pylori, and Epstein-Barr virus infection. It has been shown that a proper diet and lifestyle can play a preventive role in GC. However, research indicates that risk factors for conventional GC are less correlated with EOGC. In addition, the unclear etiology of EOGC and the late diagnosis of this disease limit the possibilities of effective treatment. Genetic factors are considered a likely cause of EOGC, as young patients are less exposed to environmental carcinogens. Research characterizing GC in young patients is scarce. This comprehensive study presents all aspects: epidemiology, risk factors, new treatment strategies, and future directions.
Collapse
Affiliation(s)
- Marek Mazurek
- Department of Surgical Oncology, Masovian Cancer Hospital, 05-135 Wieliszew, Poland;
| | - Monika Szewc
- Department of Normal, Clinical and Imaging Anatomy, Medical University of Lublin, 20-950 Lublin, Poland;
| | - Monika Z. Sitarz
- Department of Conservative Dentistry with Endodontics, Medical University of Lublin, 20-950 Lublin, Poland;
| | - Ewa Dudzińska
- Department of Dietetics and Nutrition Education, Medical University of Lublin, 20-950 Lublin, Poland;
| | - Robert Sitarz
- Department of Normal, Clinical and Imaging Anatomy, Medical University of Lublin, 20-950 Lublin, Poland;
- Department of Surgical Oncology, St. John’s Cancer Center, 20-090 Lublin, Poland
| |
Collapse
|
12
|
Ren C, Cao Z, Liu Y, Wang R, Lin C, Wang Z. Medicinal chemistry aspects of fat mass and obesity associated protein: structure, function and inhibitors. Future Med Chem 2024; 16:1705-1726. [PMID: 39101588 PMCID: PMC11370915 DOI: 10.1080/17568919.2024.2380245] [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: 02/04/2024] [Accepted: 07/09/2024] [Indexed: 08/06/2024] Open
Abstract
Adiposity and obesity-related proteins (FTO), the earliest identified mRNA N6-methyladenosine (m6A) demethylases, are known to play crucial roles in several biological processes. Therefore, FTO is a promising target for anticancer treatment. Understanding the biological functions and regulatory mechanisms of FTO targets can serve as guidelines for drug development. Despite significant efforts to develop FTO inhibitors, no specific small-molecule inhibitors have entered clinical trials so far. In this manuscript, we review the relationship between FTO and various cancers, the small-molecule inhibitors developed against FTO targets from the perspective of medicinal chemistry and other fields, and describe their structural optimization process and structure-activity relationship, providing clues for their future development direction.
Collapse
Affiliation(s)
- Changyu Ren
- Department of Pharmacy, Chengdu Fifth People's Hospital, Chengdu, 611130, China
| | - Zhi Cao
- Medical Quality Control & Evaluation Department, Chengdu Fifth People's Hospital, Chengdu, 611130, China
| | - Yang Liu
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, China
| | - Rui Wang
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, China
| | - Congcong Lin
- Department of Pharmaceutics, College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Zishu Wang
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, China
| |
Collapse
|
13
|
Pu X, Fu Y, Sun Q, Li L, Kwasi A, Ma Z, Fan X, Sun B. NTRK gene alterations were enriched in hepatoid or enteroblastic differentiation type of gastric cancer. J Clin Pathol 2024; 77:608-613. [PMID: 37451841 PMCID: PMC11474252 DOI: 10.1136/jcp-2023-208865] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 06/28/2023] [Indexed: 07/18/2023]
Abstract
Aims Currently, the clinicopathological characteristics of gastric cancer (GC) with oncogenic NTRK alterations are not well known. Although NTRK fusion has been identified as prevalent in DNA mismatch repair protein deficient (dMMR) colorectal cancer (CRC), the relationship between NTRK alterations and dMMR protein expression in GC has not been previously explored. METHODS Our study comprised 51 cases of EBV(Epstein-barr virus)-associated gastric carcinomas, 94 cases of dMMR GC, 90 cases of gastric adenocarcinoma with hepatoid or enteroblastic differentiation (GAHED) and 256 cases of conventional GC. Furthermore, to investigate the connection between NTRK fusion and dMMR proteins, we collected dMMR tumours of various types, including 21 cases of duodenal adenocarcinomas, 46 endometrioid carcinomas and 82 CRCs. NTRK fusion and amplification were screened in GC and various types of dMMR tumours using fluorescence in situ hybridisation (FISH), while cases positive for FISH translocation underwent next-generation sequencing testing. RESULTS Our findings revealed the existence of two cases each of NTRK fusions and NTRK amplifications, which were all enriched in case of GAHED. Additionally, following an analysis of several types of cancers, we discovered that NTRK gene alterations were only present in dMMR CRC. CONCLUSIONS Our results indicate that NTRK gene alterations are not enriched in GC with dMMR but are specifically enriched in cases of GAHED.
Collapse
Affiliation(s)
- Xiaohong Pu
- Department of Pathology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yao Fu
- Department of Pathology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Qi Sun
- Department of Pathology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Lin Li
- Department of Pathology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | | | - Ziyan Ma
- New York University, New York City, New York, USA
| | - Xiangshan Fan
- Department of Pathology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Beicheng Sun
- Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
14
|
Marashi A, Hasany S, Moghimi S, Kiani R, Mehran Asl S, Dareghlou YA, Lorestani P, Varmazyar S, Jafari F, Ataeian S, Naghavi K, Sajjadi SM, Haratian N, Alinezhad A, Azhdarimoghaddam A, Sadat Rafiei SK, Anar MA. Targeting gut-microbiota for gastric cancer treatment: a systematic review. Front Med (Lausanne) 2024; 11:1412709. [PMID: 39170038 PMCID: PMC11337614 DOI: 10.3389/fmed.2024.1412709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 07/17/2024] [Indexed: 08/23/2024] Open
Abstract
Background Preclinical research has identified the mechanisms via which bacteria influence cancer treatment outcomes. Clinical studies have demonstrated the potential to modify the microbiome in cancer treatment. Herein, we systematically analyze how gut microorganisms interact with chemotherapy and immune checkpoint inhibitors, specifically focusing on how gut bacteria affect the pharmacokinetics and pharmacodynamics of cancer treatment. Method This study searched Web of Science, Scopus, and PubMed until August 2023. Studies were screened by their title and abstract using the Rayyan intelligent tool for systematic reviews. Quality assessment of studies was done using the JBI critical appraisal tool. Result Alterations in the gut microbiome are associated with gastric cancer and precancerous lesions. These alterations include reduced microbial alpha diversity, increased bacterial overgrowth, and decreased richness and evenness of gastric bacteria. Helicobacter pylori infection is associated with reduced richness and evenness of gastric bacteria, while eradication only partially restores microbial diversity. The gut microbiome also affects the response to cancer treatments, with higher abundances of Lactobacillus associated with better response to anti-PD-1/PD-L1 immunotherapy and more prolonged progression-free survival. Antibiotic-induced gut microbiota dysbiosis can reduce the anti-tumor efficacy of 5-Fluorouracil treatment, while probiotics did not significantly enhance it. A probiotic combination containing Bifidobacterium infantis, Lactobacillus acidophilus, Enterococcus faecalis, and Bacillus cereus can reduce inflammation, enhance immunity, and restore a healthier gut microbial balance in gastric cancer patients after partial gastrectomy. Conclusion Probiotics and targeted interventions to modulate the gut microbiome have shown promising results in cancer prevention and treatment efficacy.Systematic review registration: https://osf.io/6vcjp.
Collapse
Affiliation(s)
- Amir Marashi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Saina Hasany
- Student Research Committee, Islamic Azad University Tehran Medical Sciences, Tehran, Iran
| | - Sadra Moghimi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Kiani
- Student Research Committee, Islamic Azad University Tehran Medical Sciences, Mashhad, Iran
| | - Sina Mehran Asl
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Student Research Committee, Islamic Azad University Tehran Medical Sciences, Tehran, Iran
| | | | - Parsa Lorestani
- School of Medicine, Shahroud Azad University of Medical Sciences, Shahroud, Iran
| | - Shirin Varmazyar
- Student Research Committee, School of Medicine, Alborz University of Medical Sciences, Alborz, Iran
| | - Fatemeh Jafari
- School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Shakiba Ataeian
- School of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Kiana Naghavi
- Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Negar Haratian
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arman Alinezhad
- Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | | - Mahsa Asadi Anar
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
15
|
Li H, Zhu Q, Liu L, Zou H, Gu D, Wu C, Li W. Preliminary differentiation of benign and malignant gastric wall thickening using dual-layer spectral-detector CT. Acta Radiol 2024; 65:879-888. [PMID: 39051549 DOI: 10.1177/02841851241260873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
BACKGROUND Dual-layer spectral-detector computed tomography (DLCT) may have the potential to evaluate gastric wall thickening. PURPOSE To evaluate the efficacy of DLCT quantitative parameters in differentiating between benign and malignant thickening of the gastric wall. MATERIAL AND METHODS A total of 58 patients with "gastric wall thickening" who underwent multi-phase abdominal enhanced DLCT scans were included in this study. Of these patients, 33 were malignant and 25 were benign. Parameters such as iodine concentration (IC), effective atomic number (Zeff), and attenuation of the lesions were measured during the arterial phase (AP) and venous phase (VP). Binary logistic regression was employed to calculate the combined prediction probabilities. The accuracy of the DLCT parameters was assessed using receiver operating characteristic (ROC) curves. RESULTS The values of IC, nIC, Zeff, normalized Zeff, and attenuation in the AP and VP were significantly higher (all P < 0.05) in the malignant group compared to the benign group. The ROC curves revealed that the IC, Zeff, and attenuation in the VP exhibited high diagnostic performance, with area under the ROC curve (AUC) values of 0.864, 0.862, and 0.840, respectively. The new combination of these three factors and gastric wall thickness had an AUC of 0.884, and the sensitivity and specificity were determined to be 81.8% and 92.0%, respectively. CONCLUSION Spectral CT parameters, particularly the combination of gastric wall thickness, attenuation, IC, and Zeff in VP, have value in distinguishing between benign and malignant gastric wall thickening.
Collapse
Affiliation(s)
- Hongjian Li
- Department of Radiology, Shenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University, Shenzhen, PR China
- Shantou University Medical College, Shantou, PR China
| | - Qianni Zhu
- Department of Radiology, Shenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University, Shenzhen, PR China
| | - Linjiang Liu
- Department of Radiology, Shenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University, Shenzhen, PR China
| | - Haijun Zou
- Department of Pharmacy, Shenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University, Shenzhen, PR China
| | - Dayong Gu
- Department of Laboratory Medicine, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, PR China
| | - Cheng Wu
- Department of Radiology, Shenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University, Shenzhen, PR China
| | - Weihua Li
- Department of Radiology, Shenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University, Shenzhen, PR China
| |
Collapse
|
16
|
Lu W, Aihaiti A, Abudukeranmu P, Liu Y, Gao H. Arachidonic acid metabolism as a novel pathogenic factor in gastrointestinal cancers. Mol Cell Biochem 2024:10.1007/s11010-024-05057-2. [PMID: 38963615 DOI: 10.1007/s11010-024-05057-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 06/25/2024] [Indexed: 07/05/2024]
Abstract
Gastrointestinal (GI) cancers are a major global health burden, representing 20% of all cancer diagnoses and 22.5% of global cancer-related deaths. Their aggressive nature and resistance to treatment pose a significant challenge, with late-stage survival rates below 15% at five years. Therefore, there is an urgent need to delve deeper into the mechanisms of gastrointestinal cancer progression and optimize treatment strategies. Increasing evidence highlights the active involvement of abnormal arachidonic acid (AA) metabolism in various cancers. AA is a fatty acid mainly metabolized into diverse bioactive compounds by three enzymes: cyclooxygenase, lipoxygenase, and cytochrome P450 enzymes. Abnormal AA metabolism and altered levels of its metabolites may play a pivotal role in the development of GI cancers. However, the underlying mechanisms remain unclear. This review highlights a unique perspective by focusing on the abnormal metabolism of AA and its involvement in GI cancers. We summarize the latest advancements in understanding AA metabolism in GI cancers, outlining changes in AA levels and their potential role in liver, colorectal, pancreatic, esophageal, gastric, and gallbladder cancers. Moreover, we also explore the potential of targeting abnormal AA metabolism for future therapies, considering the current need to explore AA metabolism in GI cancers and outlining promising avenues for further research. Ultimately, such investigations aim to improve treatment options for patients with GI cancers and pave the way for better cancer management in this area.
Collapse
Affiliation(s)
- Weiqin Lu
- General Surgery, Cancer Center, Department of Vascular Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | | | | | - Yajun Liu
- Aksu First People's Hospital, Xinjiang, China
| | - Huihui Gao
- Cancer Center, Department of Hospital Infection Management and Preventive Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
17
|
Ji Y, Qin Y, Tan Q, Qiu Y, Han S, Qi X. Development of a chemiluminescence assay for tissue plasminogen activator inhibitor complex and its applicability to gastric cancer. BMC Biotechnol 2024; 24:30. [PMID: 38720310 PMCID: PMC11080135 DOI: 10.1186/s12896-024-00850-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 04/17/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Venous thromboembolism (VTE), is a noteworthy complication in individuals with gastric cancer, but the current diagnosis and treatment methods lack accuracy. In this study, we developed a t-PAIC chemiluminescence kit and employed chemiluminescence to detect the tissue plasminogen activator inhibitor complex (t-PAIC), thrombin-antithrombin III complex (TAT), plasmin-α2-plasmin inhibitor complex (PIC) and thrombomodulin (TM), combined with D-dimer and fibrin degradation products (FDP), to investigate their diagnostic potential for venous thrombosis in gastric cancer patients. The study assessed variations in six indicators among gastric cancer patients at different stages. RESULTS The t-PAIC reagent showed LOD is 1.2 ng/mL and a linear factor R greater than 0.99. The reagents demonstrated accurate results, with all accuracy deviations being within 5%. The intra-batch and inter-batch CVs for the t-PAIC reagent were both within 8%. The correlation coefficient R between this method and Sysmex was 0.979. Gastric cancer patients exhibited elevated levels of TAT, PIC, TM, D-D, FDP compared to the healthy population, while no significant difference was observed in t-PAIC. In the staging of gastric cancer, patients in III-IV stages exhibit higher levels of the six markers compared to those in I-II stages. The ROC curve indicates an enhancement in sensitivity and specificity of the combined diagnosis of four or six indicators. CONCLUSION Our chemiluminescence assay performs comparably to Sysmex's method and at a reduced cost. The use of multiple markers, including t-PAIC, TM, TAT, PIC, D-D, and FDP, is superior to the use of single markers for diagnosing VTE in patients with malignant tumors. Gastric cancer patients should be screened for the six markers to facilitate proactive prophylaxis, determine the most appropriate treatment timing, ameliorate their prognosis, decrease the occurrence of venous thrombosis and mortality, and extend their survival.
Collapse
Affiliation(s)
- Yu Ji
- Department of Pathology, Affiliated Hospital of Jiangnan University, No.1000, Hefeng Road, Wuxi, 214122, Jiangsu, China
- Wuxi Medical College, Jiangnan University, Wuxi, Jiangsu, China
| | - Yan Qin
- Department of Pathology, Affiliated Hospital of Jiangnan University, No.1000, Hefeng Road, Wuxi, 214122, Jiangsu, China
- Wuxi Medical College, Jiangnan University, Wuxi, Jiangsu, China
| | - Qi Tan
- Department of Pathology, Affiliated Hospital of Jiangnan University, No.1000, Hefeng Road, Wuxi, 214122, Jiangsu, China
- Wuxi Medical College, Jiangnan University, Wuxi, Jiangsu, China
| | - Yanru Qiu
- Wuxi Medical College, Jiangnan University, Wuxi, Jiangsu, China
| | - Shuang Han
- Department of Pathology, Affiliated Hospital of Jiangnan University, No.1000, Hefeng Road, Wuxi, 214122, Jiangsu, China.
- Wuxi Medical College, Jiangnan University, Wuxi, Jiangsu, China.
| | - Xiaowei Qi
- Department of Pathology, Affiliated Hospital of Jiangnan University, No.1000, Hefeng Road, Wuxi, 214122, Jiangsu, China.
- Wuxi Medical College, Jiangnan University, Wuxi, Jiangsu, China.
| |
Collapse
|
18
|
Hosseinzadeh S, Imani M, Pourfarzi F, Jafari N, AbedianKenari S, Safarzadeh E. Combination of IFN-gamma with STING agonist and PD-1 immune checkpoint blockade: a potential immunotherapy for gastric cancer. Med Oncol 2024; 41:110. [PMID: 38592576 DOI: 10.1007/s12032-024-02326-4] [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: 12/30/2023] [Accepted: 02/05/2024] [Indexed: 04/10/2024]
Abstract
Suppression of the cGAS-STING pathway is an immune escape mechanism in cancer cells. The critical role of this pathway in gastric cancer (GC) is not fully understood. Herein, we evaluated the effect of the interferon-gamma (IFN-gamma), STING agonist, PD-1 immune checkpoint blockade, and their combination on the cGAS-STING pathway in GC. Expression of cGAS and STING in tumor tissue samples and adjacent normal tissue (ANT) biopsies of fifty new GC patients was evaluated by quantitative real-time PCR (qRT-PCR). Moreover, cGAS and STING expression levels were examined in Peripheral Blood Mononuclear Cells (PBMC) samples of forty GC patients and twenty-five healthy subjects. The apoptosis rate of cancer cells was analyzed by Annexin V-FITC/PI. Cell proliferation was measured by the BrdU assay. Also, IFN-β levels were evaluated in the supernatants of the treated groups. The cGAS expression was decreased in patients with distant metastasis. Co-cultures treated with IFN-gamma showed an elevated level of cGAS and STING expressions in PBMC and cancer cells. The rate of apoptosis increased in all the treatment groups. In addition, the rate of proliferation in PBMCs increased in different treated groups. The main role of PBMCs in cytotoxicity was determined by a comparative analysis of the viability of cells treated with all treatments, both with and without PBMCs. The production of IFN-β was elevated in all treated groups. The current study suggests that a combination therapy using IFN-gamma, STING agonist, and anti-PD-1 antibody can provide a promising approach to the treatment of GC.
Collapse
Affiliation(s)
- Shahnaz Hosseinzadeh
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
- Immunogenetics Research Center, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mahsa Imani
- Faculty of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Farhad Pourfarzi
- Digestive Disease Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Narjes Jafari
- Immunogenetics Research Center, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Saeid AbedianKenari
- Immunogenetics Research Center, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
- Department of Immunology, Faculty of Medicine, School of Medicine, Mazandaran University of Medical Sciences, Sari, 4816978741, Mazandaran, Iran.
| | - Elham Safarzadeh
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
- Department of Microbiology, Parasitology and Immunology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, 5166614711, Iran.
| |
Collapse
|
19
|
Wang SY, Wang YX, Shen A, Yang XQ, Liang CC, Huang RJ, Jian R, An N, Xiao YL, Wang LS, Zhao Y, Lin C, Wang CP, Yuan ZP, Yuan SQ. Construction of a gene model related to the prognosis of patients with gastric cancer receiving immunotherapy and exploration of COX7A1 gene function. Eur J Med Res 2024; 29:180. [PMID: 38494472 PMCID: PMC11337786 DOI: 10.1186/s40001-024-01783-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 03/10/2024] [Indexed: 03/19/2024] Open
Abstract
BACKGROUND GC is a highly heterogeneous tumor with different responses to immunotherapy, and the positive response depends on the unique interaction between the tumor and the tumor microenvironment (TME). However, the currently available methods for prognostic prediction are not satisfactory. Therefore, this study aims to construct a novel model that integrates relevant gene sets to predict the clinical efficacy of immunotherapy and the prognosis of GC patients based on machine learning. METHODS Seven GC datasets were collected from the Gene Expression Omnibus (GEO) database, The Cancer Genome Atlas (TCGA) database and literature sources. Based on the immunotherapy cohort, we first obtained a list of immunotherapy related genes through differential expression analysis. Then, Cox regression analysis was applied to divide these genes with prognostic significancy into protective and risky types. Then, the Single Sample Gene Set Enrichment Analysis (ssGSEA) algorithm was used to score the two categories of gene sets separately, and the scores differences between the two gene sets were used as the basis for constructing the prognostic model. Subsequently, Weighted Correlation Network Analysis (WGCNA) and Cytoscape were applied to further screen the gene sets of the constructed model, and finally COX7A1 was selected for the exploration and prediction of the relationship between the clinical efficacy of immunotherapy for GC. The correlation between COX7A1 and immune cell infiltration, drug sensitivity scoring, and immunohistochemical staining were performed to initially understand the potential role of COX7A1 in the development and progression of GC. Finally, the differential expression of COX7A1 was verified in those GC patients receiving immunotherapy. RESULTS First, 47 protective genes and 408 risky genes were obtained, and the ssGSEA algorithm was applied for model construction, showing good prognostic discrimination ability. In addition, the patients with high model scores showed higher TMB and MSI levels, and lower tumor heterogeneity scores. Then, it is found that the COX7A1 expressions in GC tissues were significantly lower than those in their corresponding paracancerous tissues. Meanwhile, the patients with high COX7A1 expression showed higher probability of cancer invasion, worse clinical efficacy of immunotherapy, worse overall survival (OS) and worse disease-free survival (DFS). CONCLUSIONS The ssGSEA score we constructed can serve as a biomarker for GC patients and provide important guidance for individualized treatment. In addition, the COX7A1 gene can accurately distinguish the prognosis of GC patients and predict the clinical efficacy of immunotherapy for GC patients.
Collapse
Affiliation(s)
- Si-Yu Wang
- Department of Oncology, The First People's Hospital of Yibin, No. 65, Wenxing Street, Cuiping District, Yibin, 644000, China
| | - Yu-Xin Wang
- The First Hospital of Jilin University, Changchun, 130000, China
| | - Ao Shen
- 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, China
| | - Xian-Qi Yang
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Cheng-Cai Liang
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Run-Jie Huang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Rui Jian
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Nan An
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Yu-Long Xiao
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Li-Shuai Wang
- Department of Oncology, The First People's Hospital of Yibin, No. 65, Wenxing Street, Cuiping District, Yibin, 644000, China
| | - Yin Zhao
- Department of Oncology, The First People's Hospital of Yibin, No. 65, Wenxing Street, Cuiping District, Yibin, 644000, China
| | - Chuan Lin
- Department of Oncology, The First People's Hospital of Yibin, No. 65, Wenxing Street, Cuiping District, Yibin, 644000, China
| | - Chang-Ping Wang
- Department of Oncology, The First People's Hospital of Yibin, No. 65, Wenxing Street, Cuiping District, Yibin, 644000, China
| | - Zhi-Ping Yuan
- Department of Oncology, The First People's Hospital of Yibin, No. 65, Wenxing Street, Cuiping District, Yibin, 644000, China
| | - Shu-Qiang Yuan
- Department of Gastric Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China.
| |
Collapse
|
20
|
Jiang T, Xu L, Qu X, Li R, Cheng Y, He H. Hsa_circ_0014606 Derived from Exosomes Promotes Gastric Carcinoma Tumorigenesis and Proliferation by Sponging miR-514b-3p to Upregulate HNRNPC. Dig Dis Sci 2024; 69:811-820. [PMID: 38217675 DOI: 10.1007/s10620-023-08254-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 12/14/2023] [Indexed: 01/15/2024]
Abstract
Gastric cancer is a common malignant tumor, and due to its insidious onset and limited screening methods, most patients are diagnosed with advanced disease and have a poor prognosis. The circRNA in exosomes has an essential role in cancer diagnosis and treatment. However, the part of hsa_circ_0014606 within exosomes in gastric cancer progression is unclear. Firstly, we extracted exosomes from the serum of gastric cancer patients and healthy individuals by ultracentrifugation and analyzed the expression of hsa_circ_0014606 in both exosomes; then knocked down hsa_circ_0014606 in vivo and in vitro, respectively, to observe its effect on the physiological function of gastric cancer cells; finally, we used bioinformatics to screen hsa_circ_0014606 targeting miRNAs and mRNAs, and experiments were performed to verify the interrelationship between the three. The results showed that the level of hsa_circ_0014606 in the serum exosomes of gastric cancer patients was significantly higher than that of the healthy population. The knockdown of hsa_circ_0014606 slowed the proliferation of gastric cancer cells, significantly reduced migration and invasion ability, accelerated apoptosis, and reduced tumor size in mice. In addition, the expression of hsa_circ_0014606 was negatively correlated with the expression of miR-514b-3p and positively correlated with the expression of heterogeneous nuclear ribonucleoprotein C (HNRNPC). In conclusion, hsa_circ_0014606 exerted a pro-cancer effect indirectly through miR-514b-3p targeting gene HNRNPC, and this study provides a new potential target for treating gastric cancer.
Collapse
Affiliation(s)
- Tao Jiang
- Department of Otolaryngology, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| | - Lingling Xu
- Department of Medical Oncology, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Xiaona Qu
- Department of Medical Oncology, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Rui Li
- Department of Otolaryngology, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| | - Ye Cheng
- Department of Medical Oncology, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Hongmei He
- Department of Medical Oncology, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China.
| |
Collapse
|
21
|
Ha GW, Hwang HP, Cho YG, Park J. Clinical and Genetic Characteristics of Early and Advanced Gastric Cancer. Curr Issues Mol Biol 2024; 46:1208-1218. [PMID: 38392195 PMCID: PMC10887908 DOI: 10.3390/cimb46020077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/24/2024] Open
Abstract
Gastric cancer (GC) persists as the fourth most prevalent cause of global cancer-related mortality, presenting a challenge due to the scarcity of available therapeutic strategies. Precision medicine is crucial not only in the treatment but also in the management of GC. We performed gene panel sequencing with Oncomine focus assay comprising 52 cancer-associated genes and MSI analysis in 100 case-matched gastric cancer cases. A comprehensive analysis of clinical and genetic characteristics was conducted on these genetic results and clinicopathological findings. Upon comparison of clinicopathological characteristics, significant differences between early gastric cancer (EGC) and advanced gastric cancer (AGC) were observed in tumor location (p = 0.003), Lauren classification (p = 0.015), T stage (p = 0.000), and N stage (p = 0.015). The six most frequently mutated genes were PIK3CA (29%, 10/35), ERBB2 (17%, 6/35), KRAS (14%, 5/35), ALK (6%, 2/35), ESR1 (6%, 2/35), and FGFR3 (6%, 2/35). Regarding genetic variation, there was a tendency for the N stage to be higher in GC patients with mutated genes (p = 0.014). The frequency of mutations in GC patients was statistically significantly higher in AGC (n = 24) compared to EGC (n = 11) (odds ratio, 2.792; 95% confidence interval, 1.113 to 7.007; p = 0.026). Six of the ten GC patients carrying mutated genes and exhibiting MSI were classified into intestinal-type and undifferentiated GC, with the location of the tumor being in the lower-third. Among these patients, five harbored mutated PIK3CA, while the remaining patient had a mutation in ALK. Conclusions: AGC patients more frequently exhibited alterations of PIK3CA, KRAS, and ERBB2 as somatic oncogenic drivers, and displayed a higher prevalence of cumulative genetic events, including increased rates of PIK3CA mutations, enhanced detection of immunotherapy biomarkers, and mutations of the ESR1 gene.
Collapse
Affiliation(s)
- Gi Won Ha
- Department of Surgery, Jeonbuk National University Medical School and Hospital, Jeonju 54907, Republic of Korea
| | - Hong Pil Hwang
- Department of Surgery, Jeonbuk National University Medical School and Hospital, Jeonju 54907, Republic of Korea
| | - Yong Gon Cho
- Department of Laboratory Medicine, Jeonbuk National University Medical School and Hospital, Jeonju 54907, Republic of Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju 54907, Republic of Korea
| | - Joonhong Park
- Department of Laboratory Medicine, Jeonbuk National University Medical School and Hospital, Jeonju 54907, Republic of Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju 54907, Republic of Korea
- Department of Laboratory Medicine, Daejeon St. Mary's Hospital, Daejeon 34943, Republic of Korea
| |
Collapse
|
22
|
Zhang S, Ren D, Hou H, Yao L, Yuan H. M-CSF secreted by gastric cancer cells exacerbates the progression of gastric cancer by increasing the expression of SHP2 in tumor-associated macrophages. Aging (Albany NY) 2023; 15:15525-15534. [PMID: 38159254 PMCID: PMC10781482 DOI: 10.18632/aging.205390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 10/23/2023] [Indexed: 01/03/2024]
Abstract
OBJECTIVE To investigate the effect of Src homology 2 domain-containing tyrosine phosphatase-2 (SHP2) in tumor-associated macrophages (TAMs), which is mediated by macrophage colony-stimulating factor (M-CSF) secreted by gastric cancer cells, on the development of gastric cancer and its molecular mechanism. METHODS The progression of gastric cancer was detected by nude mouse tumor-bearing experiments. Colony formation assay and cell counting kit-8 (CCK8) assay were used to detect the proliferation capacity of gastric cancer cells. The migration capacity of gastric cancer cells was examined by wound healing assay. Transwell migration and invasion assays were performed on gastric cancer cells. Detection of relevant protein expression using western blotting. RESULTS Overexpression of SHP2 could promote the progression of gastric cancer in nude mice. The results of colony formation assay and CCK8 assay showed that overexpression of SHP2 could enhance the proliferation of gastric cancer cells. It was found by wound healing assay and Transwell assay that overexpression of SHP2 could facilitate the migration and invasion of gastric cancer cells. The results of Western blotting revealed that overexpression of SHP2 could increase the expressions of p-STAT3, s-PD-1, p-Src, p-Lyn, p-PI3K, p-AKT, Arginase-1, MMP1 and MMP3 but decrease the expressions of TBK1 and SOCS1 in TAMs, and also increase the expressions of CD9, TSG101 and s-PD-1 in exosomes. CONCLUSION M-CSF secreted by gastric cancer cells can promote the proliferation, invasion and migration of gastric cancer cells by increasing the expression of SHP2 in TAMs.
Collapse
Affiliation(s)
- Shaohua Zhang
- Eighth People’s Hospital of Hebei Province, Shijiazhuang 050000, China
| | - Dongfei Ren
- Eighth People’s Hospital of Hebei Province, Shijiazhuang 050000, China
| | - Huiyu Hou
- HeBei General Hospital, Shijiazhuang 050000, China
| | - Li Yao
- Handan Central Hospital, Handan 056000, China
| | - Hufang Yuan
- The Fourth Hospital of Hebei Medical University, Shijiazhuang 050000, China
| |
Collapse
|
23
|
Zhang H, Shi J, Xie H, Liu X, Ruan G, Lin S, Ge Y, Liu C, Chen Y, Zheng X, Song M, Yang M, Zhang X, Shi HP. Superiority of CRP-albumin-lymphocyte index as a prognostic biomarker for patients with gastric cancer. Nutrition 2023; 116:112191. [PMID: 37716090 DOI: 10.1016/j.nut.2023.112191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 06/05/2023] [Accepted: 08/06/2023] [Indexed: 09/18/2023]
Abstract
OBJECTIVES The new C-reactive protein (CRP)-albumin-lymphocyte (CALLY) index is an immune nutrition scoring system based on serum CRP) serum albumin, and lymphocyte counts. The aim of this study was to verify the prognostic value of the CALLY index in patients with gastric cancer and to evaluate the superiority of this new system. METHODS We retrospectively analyzed the data of patients with gastric cancer who were followed up from the INSCOC database between May 2013 and December 2018. Through simple random sampling, patients with gastric cancer were placed into one of two groups: the training group (n = 684) or the verification group (n = 290) in a ratio of 7:3. Correlation analysis, Kaplan-Meier method, and cubic spline function were used to analyze the relationship between the CALLY index and overall survival (OS) in these patients. Based on the results of Cox regression analysis of the training cohort, a nomogram model for predicting 1 -, 2 -, 3-, and 5-y OS was established and verified internally. The prediction accuracy and benefit of the nomogram in gastric cancer were evaluated by calibration and clinical decision curve and compared with the traditional TNM gastric cancer staging system. RESULTS The CALLY index was negatively correlated with the age of patients with gastric cancer (men, r = -0.1; women, r = -0.1), but positively correlated with body mass index (BMI; men, r = 0.063; women, r = 0.058), and the cutoff value of the CALLY index was determined as 1.12. The OS of patients with gastric cancer and a CALLY index >1.12 was significantly higher than that of patients with gastric cancer and a CALLY index ≤1.12 (P < 0.0001). There was an L-shaped dose-response relationship between the CALLY index and OS in patients with gastric cancer, and age, TNM stage, surgical treatment, chemotherapy, BMI, and the CALLY index were significantly correlated with the prognosis of patients with gastric cancer. Tumor TNM stage, BMI, and the CALLY index were independent risk factors affecting the prognosis of patients with gastric cancer. The CALLY index was a protective factor in the following patient factors: diagnosis of gastric cancer; <65 y of age; male; TNM 3 stage; BMI 18.5 to 23.9 kg/m2; smoker; consumer of alcohol; no radio- or chemotherapy; surgery; presence of diabetes, hypertension, or both; no family history of cancer; experienced a significant interaction with chemotherapy and surgery. A nomogram based on TNM staging, BMI, and the CALLY index has good predictive ability and clinical application value. Compared with traditional TNM staging systems, the nomogram has better resolution and accuracy in predicting 1 -, 2 -, 3-, and 5-year OS. CONCLUSION The CALLY index can be used as an independent prognostic factor for patients with gastric cancer, and constructs a nomogram prediction model combining TNM staging, BMI, and CALLY index, which yields better predictions than traditional TNM staging.
Collapse
Affiliation(s)
- Heyang Zhang
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, China; Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, China; Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, China
| | - Jinyu Shi
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, China; Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, China; Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, China
| | - Hailun Xie
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, China; Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, China; Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, China
| | - Xiaoyue Liu
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, China; Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, China; Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, China
| | - Guotian Ruan
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, China; Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, China; Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, China
| | - Shiqi Lin
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, China; Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, China; Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, China
| | - Yizhong Ge
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, China; Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, China; Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, China
| | - Chenan Liu
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, China; Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, China; Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, China
| | - Yue Chen
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, China; Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, China; Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, China
| | - Xin Zheng
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, China; Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, China; Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, China
| | - Mengmeng Song
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, China; Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, China; Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, China
| | - Ming Yang
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, China; Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, China; Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, China
| | - Xiaowei Zhang
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, China; Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, China; Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, China
| | - Han-Ping Shi
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, China; Beijing International Science and Technology Cooperation Base for Cancer Metabolism and Nutrition, Beijing, China; Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, China.
| |
Collapse
|
24
|
Zhang Z, Li Q, Sun S, Li Z, Cui Z, Liu Q, Zhang Y, Xiong S, Zhang S. Expression of SMARCA2 and SMARCA4 in gastric adenocarcinoma and construction of a nomogram prognostic model. Int J Clin Oncol 2023; 28:1487-1500. [PMID: 37634210 DOI: 10.1007/s10147-023-02403-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 08/06/2023] [Indexed: 08/29/2023]
Abstract
BACKGROUND Aberrant expression of SWI/SNF complex subunits is closely associated with tumorigenesis. The clinicopathological and prognostic significance of altered SMARCA2 and SMARCA4 subunits has not been well evaluated in gastric adenocarcinoma. METHODS We collected 1271 postoperative cases of gastric adenocarcinoma and then constructed tissue microarrays (TMA), from which we obtained the immunohistochemistry expression of SMARCA2 and SMARCA4. Next, we screened the variables related to the loss of SMARCA2 and SMARCA4 by univariate correlation analysis and multivariate logistic regression analysis. Then, we identified the variables related to prognosis by univariate and multivariate Cox regression analysis. Finally, we constructed a nomogram prognostic model and evaluated it. RESULTS The loss of SMARCA2 and SMARCA4 occurred in 236 (18.57%) and 86 (6.77%) cases, respectively, including 26 cases of co-loss. After multivariate logistic regression, variables independently associated with SMARCA2 loss were T stage, differentiation status, WHO histological classification, and EBER. Variables independently associated with SMARCA4 loss were differentiation status, WHO histological classification, PD-L1, and MMR. Survival analysis revealed that the SMARCA2 and SMARCA4 lost groups showed worse survival than the corresponding present groups (P = 0.032 and P = 0.0048, respectively). Univariate and multivariate Cox analyses identified independent prognostic factors, including age, T stage, N stage, M stage, SMARCA2, and chemotherapy. CONCLUSION The loss of SMARCA2 and SMARCA4 correlated with poor differentiation, leading to a worse prognosis. SMARCA2, as an independent prognostic factor, combined with other clinicopathological variables, established a novel nomogram prognostic model, which outperformed the AJCC TNM model.
Collapse
Affiliation(s)
- Zhenkun Zhang
- Weihai Municipal Hospital, Shandong University, Weihai, 264200, Shandong, China
- Department of Oncology, Shouguang People's Hospital, Weifang, 262700, Shandong, China
| | - Qiujing Li
- Department of Pathology, Weihai Municipal Hospital, Shandong University, No. 70 Heping Road, Huancui District, Weihai, 264200, Shandong, China
| | - Shanshan Sun
- Department of Oncology, Weihai Municipal Hospital, Shandong University, Weihai, 264200, Shandong, China
| | - Zhe Li
- Weifang Medical College, Weifang, 261053, Shandong, China
| | - ZhengGuo Cui
- Department of Environmental Health, University of Fukui School of Medical Sciences, 23-3 Matsuoka Shimoaizuki, Eiheiji, Fukui, 910-1193, Japan
| | - Qian Liu
- Department of Pathology, Weihai Municipal Hospital, Shandong University, No. 70 Heping Road, Huancui District, Weihai, 264200, Shandong, China
| | - Yujie Zhang
- Department of Pathology, Weihai Municipal Hospital, Shandong University, No. 70 Heping Road, Huancui District, Weihai, 264200, Shandong, China
| | - Sili Xiong
- Weifang Medical College, Weifang, 261053, Shandong, China
| | - Shukun Zhang
- Department of Pathology, Weihai Municipal Hospital, Shandong University, No. 70 Heping Road, Huancui District, Weihai, 264200, Shandong, China.
| |
Collapse
|
25
|
Cao F, Liu Y, Cheng Y, Wang Y, He Y, Xu Y. Multi-omics characteristics of tumor-associated macrophages in the tumor microenvironment of gastric cancer and their exploration of immunotherapy potential. Sci Rep 2023; 13:18265. [PMID: 37880233 PMCID: PMC10600170 DOI: 10.1038/s41598-023-38822-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 07/15/2023] [Indexed: 10/27/2023] Open
Abstract
The incidence and mortality rate of gastric cancer (GC) have remained high worldwide. Although some progress has been made in immunotargeted therapy, the treatment effect remains limited. With more attention has been paid to the immune potential of tumor-associated macrophages (TAMs), but the specific mechanisms of tumor immunity are still unclear. Thus, we screened marker genes in TAMs differentiation (MDMs) through single-cell RNA sequencing, and combined with GC transcriptome data from TCGA and GEO databases, the clinical and TME characteristics, prognostic differences, immune infiltration, and drug sensitivity among different subtypes of patients with GC in different data sets were analyzed. A prognostic model of GC was constructed to evaluate the prognosis and immunotherapy response of patients with GC. In this study, we extensively studied the mutations in MDMs such as CGN, S100A6, and C1QA, and found differences in the infiltration of immune cells and immune checkpoints including M2 TAMs, T cells, CD274, and CTLA4 in different GC subtypes. In the model, we constructed a predictive scoring system with high accuracy and screened out key MDMs-related genes associated with prognosis and M2 TAMs, among which VKORC1 may be involved in GC progression and iron death in tumor cells. Therefore, this study explores the therapeutic strategy of TAMs reprogramming in-depth, providing new ideas for the clinical diagnosis, treatment, and prognosis assessment of GC.
Collapse
Affiliation(s)
- Feng Cao
- Department of General Surgery, University Hospital RWTH Aachen, 52074, Aachen, Germany
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Yanwei Liu
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Yunsheng Cheng
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Yong Wang
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230022, China.
| | - Yan He
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230022, China.
| | - Yanyan Xu
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230022, China.
| |
Collapse
|
26
|
De Marco K, Lepore Signorile M, Di Nicola E, Sanese P, Fasano C, Forte G, Disciglio V, Pantaleo A, Varchi G, Del Rio A, Grossi V, Simone C. SMYD3 Modulates the HGF/MET Signaling Pathway in Gastric Cancer. Cells 2023; 12:2481. [PMID: 37887325 PMCID: PMC10605494 DOI: 10.3390/cells12202481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 10/11/2023] [Accepted: 10/13/2023] [Indexed: 10/28/2023] Open
Abstract
Gastric cancer (GC) is the third most deadly cancer worldwide. Considerable efforts have been made to find targetable drivers in order to improve patient outcomes. MET is one of the most important factors involved in GC initiation and progression as it plays a major role in GC invasiveness and is related to cancer stemness. Unfortunately, treatment strategies targeting MET are still limited, with a proportion of patients responding to therapy but later developing resistance. Here, we showed that MET is a molecular partner of the SMYD3 methyltransferase in GC cells. Moreover, we found that SMYD3 pharmacological inhibition affects the HGF/MET downstream signaling pathway. Extensive cellular analyses in GC models indicated that EM127, a novel active site-selective covalent SMYD3 inhibitor, can be used as part of a synergistic approach with MET inhibitors in order to enhance the targeting of the HGF/MET pathway. Importantly, our data were confirmed in a 3D GC cell culture system, which was used as a surrogate to evaluate stemness characteristics. Our findings identify SMYD3 as a promising therapeutic target to impair the HGF/MET pathway for the treatment of GC.
Collapse
Affiliation(s)
- Katia De Marco
- Medical Genetics, National Institute for Gastroenterology—IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (K.D.M.); (M.L.S.); (E.D.N.); (P.S.); (C.F.); (G.F.); (V.D.); (A.P.)
| | - Martina Lepore Signorile
- Medical Genetics, National Institute for Gastroenterology—IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (K.D.M.); (M.L.S.); (E.D.N.); (P.S.); (C.F.); (G.F.); (V.D.); (A.P.)
| | - Elisabetta Di Nicola
- Medical Genetics, National Institute for Gastroenterology—IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (K.D.M.); (M.L.S.); (E.D.N.); (P.S.); (C.F.); (G.F.); (V.D.); (A.P.)
| | - Paola Sanese
- Medical Genetics, National Institute for Gastroenterology—IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (K.D.M.); (M.L.S.); (E.D.N.); (P.S.); (C.F.); (G.F.); (V.D.); (A.P.)
| | - Candida Fasano
- Medical Genetics, National Institute for Gastroenterology—IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (K.D.M.); (M.L.S.); (E.D.N.); (P.S.); (C.F.); (G.F.); (V.D.); (A.P.)
| | - Giovanna Forte
- Medical Genetics, National Institute for Gastroenterology—IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (K.D.M.); (M.L.S.); (E.D.N.); (P.S.); (C.F.); (G.F.); (V.D.); (A.P.)
| | - Vittoria Disciglio
- Medical Genetics, National Institute for Gastroenterology—IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (K.D.M.); (M.L.S.); (E.D.N.); (P.S.); (C.F.); (G.F.); (V.D.); (A.P.)
| | - Antonino Pantaleo
- Medical Genetics, National Institute for Gastroenterology—IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (K.D.M.); (M.L.S.); (E.D.N.); (P.S.); (C.F.); (G.F.); (V.D.); (A.P.)
| | - Greta Varchi
- Institute for Organic Synthesis and Photoreactivity (ISOF), National Research Council of Italy (CNR), 40129 Bologna, Italy; (G.V.); (A.D.R.)
| | - Alberto Del Rio
- Institute for Organic Synthesis and Photoreactivity (ISOF), National Research Council of Italy (CNR), 40129 Bologna, Italy; (G.V.); (A.D.R.)
- Innovamol Consulting Srl, 41126 Modena, Italy
| | - Valentina Grossi
- Medical Genetics, National Institute for Gastroenterology—IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (K.D.M.); (M.L.S.); (E.D.N.); (P.S.); (C.F.); (G.F.); (V.D.); (A.P.)
| | - Cristiano Simone
- Medical Genetics, National Institute for Gastroenterology—IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (K.D.M.); (M.L.S.); (E.D.N.); (P.S.); (C.F.); (G.F.); (V.D.); (A.P.)
- Medical Genetics, Department of Precision and Regenerative Medicine and Jonic Area (DiMePRe-J), University of Bari Aldo Moro, 70124 Bari, Italy
| |
Collapse
|
27
|
Wang Z, Ding J, Xiao Y, Xiao K, Su P, Dong Z, Zhang Y. Serum extracellular vesicles with NSD1 and FBXO7 mRNA as novel biomarkers for gastric cancer. Clin Biochem 2023; 120:110653. [PMID: 37742869 DOI: 10.1016/j.clinbiochem.2023.110653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/05/2023] [Accepted: 09/21/2023] [Indexed: 09/26/2023]
Abstract
BACKGROUND Messenger RNAs (mRNAs) in serum extracellular vesicles (EVs) are effective non-invasive biomarkers for various types of cancer, however, their role as biomarkers for gastric cancer is yet to be investigated. Therefore, the current study was designed to explore their potential as novel biomarkers for gastric cancer. METHODS The mRNAs in serum EVs from four patients with gastric cancer and four healthy controls were investigated. mRNAs in serum EVs were extracted for high-throughput RNA sequencing (RNA-seq). Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were performed to predict cancer-related genes. Candidate mRNAs were validated using reverse transcription-quantitative polymerase chain reaction. The diagnostic and prognostic values of mRNAs for gastric cancer were evaluated by receiver operating characteristic (ROC) curves and Kaplan-Meier analysis, respectively. RESULTS RNA-seq revealed 13,229 upregulated and 7,079 downregulated mRNAs in serum EVs. GO and KEGG analyses showed that certain mRNAs were associated with tumorigenesis and progression. From these, 10 were selected according to our criteria (|Fold Change| > 10, P < 0.05). NSD1 was upregulated and FBXO7 was downregulated in patients with gastric cancer compared with the healthy controls. The area under the ROC curves of these two mRNAs combined was 0.84, with a sensitivity of 78 % and a specificity of 92 %. NSD1 and FBXO7 were also associated with tumor size, distal metastasis, and TNM stage. Furthermore, NSD1 expression was strongly associated with prognosis, as revealed from our follow-up studies and online database analysis. However, FBXO7 was only significantly associated with prognosis in our follow-up data. CONCLUSIONS NSD1 and FBXO7 in serum EVs have important roles in gastric cancer and may be useful biomarkers for its diagnosis and prognosis.
Collapse
Affiliation(s)
- Zhen Wang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan 250012, Shandong, China; Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, 107 Wenhuaxi Road, Jinan 250012, Shandong, China
| | - Juan Ding
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan 250012, Shandong, China; Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, 107 Wenhuaxi Road, Jinan 250012, Shandong, China
| | - Yilei Xiao
- Department of Neurosurgery, Liaocheng People's Hospital, 67 Dongchangxi Road, Liaocheng 252000, Shandong, China
| | - Ke Xiao
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan 250012, Shandong, China; Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, 107 Wenhuaxi Road, Jinan 250012, Shandong, China
| | - Ping Su
- National Administration of Health Data, Jinan 250000, Shandong, China
| | - Zhaogang Dong
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan 250012, Shandong, China; Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, 107 Wenhuaxi Road, Jinan 250012, Shandong, China.
| | - Yi Zhang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan 250012, Shandong, China; Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, 107 Wenhuaxi Road, Jinan 250012, Shandong, China.
| |
Collapse
|
28
|
Dong S, Wang Z, Xiong W. POFUT1 promotes gastric cancer progression through Notch/Wnt dual signaling pathways dependent on the parafibromin-NICD1-β-catenin complex. J Chin Med Assoc 2023; 86:806-817. [PMID: 37501238 DOI: 10.1097/jcma.0000000000000957] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/29/2023] Open
Abstract
BACKGROUND Aberrant glycosylation performed by glycosyltransferases is a leading cause of gastric cancer (GC). Protein O-fucosyltransferase 1 (POFUT1) expression is increased in GC specimens and cells. In this study, the biological effects and mechanisms of POFUT1 underlying the development of GC were investigated. METHODS POFUT1 downregulated and upregulated GC cells were established. The effects of POFUT1 on cell proliferation, metastasis and apoptosis were examined using cell counting kit-8 (CCK8) assay, transwell assay, and flow cytometry. Subcutaneous xenograft tumor models were established followed by immunohistochemistry staining of resected tumors. Facilitating modulators and transcription factors were detected by western blot, immunofluorescence, luciferase reporter assay, and co-immunoprecipitation. RESULTS POFUT1 played a pro-oncogenic role both in vivo and in vitro, which promoted proliferation and metastasis, as well as inhibited apoptosis in GC cells. POFUT1 promoted Cyclin D3 expression and inhibited the expression of apoptotic proteins, such as Bcl-2-associated X protein (Bax) and cleaved caspase 3, facilitating tumor growth. Moreover, POFUT1 accelerated matrix metalloproteases expression and attenuated E-cadherin expression, contributing to GC metastasis. In addition, POFUT1 expression promoted the expression and nuclear translocation of Notch1 intracellular domain (NICD1) and β-catenin and inhibited β-catenin phosphorylation degradation, accompanied by the activation of recombination signal binding protein-Jκ (RBP-J) and T-cell factor (TCF) transcription factors, respectively. It is notable that parafibromin integrated NICD1 and β-catenin, enabling the concerted activation of Wnt and Notch signaling targeted proteins. CONCLUSION These observations indicated that POFUT1 promoted GC development through activation of Notch and Wnt signaling pathways, which depended on the parafibromin-NICD1-β-catenin complex. This work provides new evidence for the further diagnosis and treatment of GC.
Collapse
Affiliation(s)
- Shuang Dong
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhirong Wang
- Department of Gastroenterology, Shanghai Tongji Hospital Affiliated to Tongji University, Shanghai, China
| | - Wujun Xiong
- Department of Gastroenterology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| |
Collapse
|
29
|
Nie D, Zheng H, An G, Li J. Development and validation of a novel nomogram for postoperative overall survival of patients with primary gastric signet-ring cell carcinoma: a population study based on SEER database. J Cancer Res Clin Oncol 2023; 149:8593-8603. [PMID: 37097391 DOI: 10.1007/s00432-023-04796-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 04/15/2023] [Indexed: 04/26/2023]
Abstract
BACKGROUND Gastric signet ring cell carcinoma (GSRCC) is a highly malignant subtype of gastric cancer. We tried to establish and validate a nomogram using common clinical variables to achieve more personalized management. METHODS We analyzed patients with GSRCC in the Surveillance, Epidemiology, and End Results database between 2004 and 2017. The survival curve was calculated by the Kaplan-Meier method, and the difference in survival curve was tested by log-rank test. We used the cox proportional hazard model to evaluate independent factors of prognosis, and established a nomogram to predict 1-, 3- and 5- overall survival (OS). Harrell's consistency index and calibration curve were used to measure the discrimination and calibration of the nomogram. In addition, we used decision curve analysis (DCA) to compare the net clinical benefits of the nomogram and American Joint Committee on Cancer (AJCC) staging system. RESULTS The prognosis nomogram predicting 1-, 3- and 5-years OS for patients with GSRCC is established for the first time. The C-index and AUC of nomogram were higher than that of the American Joint Committee on Cancer (AJCC) staging system in the training set. Our model also shows better performance than the AJCC staging system in the validation set, and importantly, DCA shows that our model has a better net benefit than the AJCC stage. CONCLUSIONS We have developed and validated a new nomogram and risk classification system, which is better than the AJCC staging system. It will help clinicians manage postoperative patients with GSRCC more accurately.
Collapse
Affiliation(s)
- Duorui Nie
- Department of Oncology, The First Hospital of Hunan University of Chinese Medicine, Yuhua District, Changsha, 410007, Hunan, China
| | - Hao Zheng
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Guilin An
- School of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan, China
| | - Jing Li
- Department of Oncology, The First Hospital of Hunan University of Chinese Medicine, Yuhua District, Changsha, 410007, Hunan, China.
| |
Collapse
|
30
|
Zhang H, Zhuo H, Hou J, Cai J. Machine learning models predict the mTOR signal pathway-related signature in the gastric cancer involving 2063 samples of 7 centers. Aging (Albany NY) 2023; 15:6152-6162. [PMID: 37341987 PMCID: PMC10373976 DOI: 10.18632/aging.204817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 05/17/2023] [Indexed: 06/22/2023]
Abstract
Gastric cancer, as a tumor with poor prognosis, has been widely studied. Distinguishing the types of gastric cancer is helpful. Using the transcriptome data of gastric cancer in our study, relevant proteins of mTOR signaling pathway were screened to identify key genes by four machine learning models, and the models were validated in external datasets. Through correlation analysis, we explored the relationship between five key genes and immune cells and immunotherapy. By inducing cellular senescence in gastric cancer cells with bleomycin, we investigated changes in the expression levels of HRAS through western blot. By PCA clustering analysis, we used the five key genes for gastric cancer typing and explored differences in drug sensitivity and enrichment pathways between different clustering groups. We found that the SVM machine learning model was superior, and the five genes (PPARA, FNIP1, WNT5A, HRAS, HIF1A) were highly correlated with different immune cells in multiple databases. These five key genes have a significant impact on immunotherapy. Using the five genes for gastric cancer gene typing, four genes were expressed higher in group 1 and were more sensitive to drugs in group 2. These results suggest that subtype-specific markers can improve the treatment and provide precision drugs for gastric cancer patients.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361004, Fujian, China
- Institute of Gastrointestinal Oncology, Medical College of Xiamen University, Xiamen 361004, Fujian, China
- Xiamen Municipal Key Laboratory of Gastrointestinal Oncology, Xiamen 361004, Fujian, China
| | - Huiqin Zhuo
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361004, Fujian, China
- Institute of Gastrointestinal Oncology, Medical College of Xiamen University, Xiamen 361004, Fujian, China
- Xiamen Municipal Key Laboratory of Gastrointestinal Oncology, Xiamen 361004, Fujian, China
| | - Jingjing Hou
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361004, Fujian, China
- Institute of Gastrointestinal Oncology, Medical College of Xiamen University, Xiamen 361004, Fujian, China
- Xiamen Municipal Key Laboratory of Gastrointestinal Oncology, Xiamen 361004, Fujian, China
| | - Jianchun Cai
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361004, Fujian, China
- Institute of Gastrointestinal Oncology, Medical College of Xiamen University, Xiamen 361004, Fujian, China
- Xiamen Municipal Key Laboratory of Gastrointestinal Oncology, Xiamen 361004, Fujian, China
| |
Collapse
|
31
|
Wei G, Wang Y, Yang P, Peng S, Duan S, Hu X, Yuan L, Bao G. Enhancing Vulnerability of Afatinib using Erastin via xCT-mediated ROS/P38MAPK Signaling Feedback Loop in Gastric Cancer Cells. Gene 2023; 873:147468. [PMID: 37169154 DOI: 10.1016/j.gene.2023.147468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 05/01/2023] [Accepted: 05/04/2023] [Indexed: 05/13/2023]
Abstract
Ferroptosis, being classified as a form of regulated cell death, was driven by the oxidative injury induced by lipid peroxidation(LPO). Recently, ferroptosis has been confirmed to exert a critical effect in the pathogenesis and treatment of various tumors, including gastric cancer (GC). Erastin, as a frequently used ferroptosis inducer, caused ferroptosis by downregulating the xCT expression resulting in increasing reactive oxygen species (ROS) and aggravating the LPO. However, the mechanisms of Erastin in ferroptosis regulation, especially in GC, remain largely elusive. This work firstly demonstrated that Erastin inhibited cell growth and promoted apoptosis and ferroptosis in AGS and BGC823 cells. Then, based on Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis of Erastin-related targets screened by using PharmMapper Web, the P38MAPK signaling was explored and validated in AGS and BGC-823 cells. Besides, the Fer-1 and p38 inhibitor were performed to investigate the mechanisms of ferroptosis induced by Erastin in depth. This work revealed a feedback mode among xCT, ROS and the P38MAPK pathway, which affected each other. It meant that Erastin regulated ferroptosis through the xCT-mediated ROS/P38MAPK signaling feedback loop. In addition, it was noticed that in co-operation with Erastin, the cytotoxic effects of Afatinib on cells were aggravated by further strengthening ferroptosis with activation of the P38MAPK pathway. In summary, those works provided evidence that Erastin plays an important role in increasing the cytotoxic effect on GC cells treated with Afitinib. Furthermore, the Erastin-induced ferroptosis via the xCT-mediated ROS/P38MAPK pathway feedback loop provides new strategies for GC comprehensive treatment.
Collapse
Affiliation(s)
- Gang Wei
- Shaanxi University of Traditional Chinese Medicine, Xianyang, Shannxi, China; Department of General Surgery, The Second Affiliated Hospital of Air Force Medical University, Xi'an, Shannxi, China
| | - Yan Wang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Air Force Medical University, Xi'an, Shannxi, China
| | - Ping Yang
- Department of General Surgery, The Second Affiliated Hospital of Air Force Medical University, Xi'an, Shannxi, China
| | - Shujia Peng
- Department of General Surgery, The Second Affiliated Hospital of Air Force Medical University, Xi'an, Shannxi, China
| | - Sensen Duan
- Department of General Surgery, The Second Affiliated Hospital of Air Force Medical University, Xi'an, Shannxi, China
| | - Xi'e Hu
- Department of General Surgery, The Second Affiliated Hospital of Air Force Medical University, Xi'an, Shannxi, China
| | - Lijuan Yuan
- Department of General Surgery, The Second Affiliated Hospital of Air Force Medical University, Xi'an, Shannxi, China.
| | - Guoqiang Bao
- Department of General Surgery, The Second Affiliated Hospital of Air Force Medical University, Xi'an, Shannxi, China.
| |
Collapse
|
32
|
Doshi K, James E. A Rare Case of Metastasis of Gastric Adenocarcinoma to the Leptomeninges. Cureus 2023; 15:e37120. [PMID: 37153327 PMCID: PMC10159219 DOI: 10.7759/cureus.37120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2023] [Indexed: 04/07/2023] Open
Abstract
Leptomeningeal carcinomatosis (LMC) is defined as the diffuse infiltration of malignant cells throughout the pia and arachnoid membrane. LMC is commonly observed in patients with leukemia, lymphoma, and breast and lung cancer. The prevalence of LMC spread in patients with primary gastric malignancy is very rare. Due to its devastating neurological complications and high mortality, it is difficult to assess the associated clinical features, treatment outcomes, and prognostic factors. Current treatment options include intra-thecal chemotherapy, radiotherapy, and supportive care with a median survival of three to four months. LMC is a rare manifestation of gastric cancer and is an extremely fatal disease. Therefore, it is difficult to distinguish LMC from other neurological etiologies. We present a unique case of an individual who presented with headaches and was found to have LMC.
Collapse
|
33
|
Nie G, Zhang H, Yan J, Xie D, Zhang H, Li X. Construction and validation of a novel nomogram to predict cancer-specific survival in patients with gastric adenocarcinoma. Front Oncol 2023; 13:1114847. [PMID: 36845677 PMCID: PMC9948249 DOI: 10.3389/fonc.2023.1114847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 01/20/2023] [Indexed: 02/11/2023] Open
Abstract
Background and aims Adenocarcinoma is one of the most common pathological types of gastric cancer. The aims of this study were to develop and validate prognostic nomograms that could predict the probability of cancer-specific survival (CSS) for gastric adenocarcinoma (GAC) patients at 1, 3, and 5 years. Methods In total, 7747 patients with GAC diagnosed between 2010 and 2015, and 4591 patients diagnosed between 2004 and 2009 from the Surveillance, Epidemiology, and End Results (SEER) database were included in this study. The 7747 patients were used as a prognostic cohort to explore GAC-related prognostic risk factors. Moreover, the 4591 patients were used for external validation. The prognostic cohort was also divided into a training and internal validation sets for construction and internal validation of the nomogram. CSS predictors were screened using least absolute shrinkage and selection operator regression analysis. A prognostic model was built using Cox hazard regression analysis and provided as static and dynamic network-based nomograms. Results The primary site, tumor grade, surgery of the primary site, T stage, N stage, and M stage were determined to be independent prognostic factors for CSS and were subsequently included in construction of the nomogram. CSS was accurately estimated using the nomogram at 1, 3, and 5 years. The areas under the curve (AUCs) for the training group at 1, 3, and 5 years were 0.816, 0.853, and 0.863, respectively. Following internal validation, these values were 0.817, 0.851, and 0.861. Further, the AUC of the nomogram was much greater than that of American Joint Committee on Cancer (AJCC) or SEER staging. Moreover, the anticipated and actual CSS values were in good agreement based on decision curves and time-calibrated plots. Then, patients from the two subgroups were divided into high- and low-risk groups based on this nomogram. The survival rate of high-risk patients was considerably lower than that of low-risk patients, according to Kaplan-Meier (K-M) curves (p<0.0001). Conclusions A reliable and convenient nomogram in the form of a static nomogram or an online calculator was constructed and validated to assist physicians in quantifying the probability of CSS in GAC patients.
Collapse
Affiliation(s)
- Guole Nie
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Honglong Zhang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Jun Yan
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China,Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China,Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
| | - Danna Xie
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Haijun Zhang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Xun Li
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China,Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China,Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China,*Correspondence: Xun Li,
| |
Collapse
|
34
|
Comprehensive Molecular Analyses of Notch Pathway-Related Genes to Predict Prognosis and Immunotherapy Response in Patients with Gastric Cancer. JOURNAL OF ONCOLOGY 2023; 2023:2205083. [PMID: 36733672 PMCID: PMC9889149 DOI: 10.1155/2023/2205083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/06/2022] [Accepted: 11/24/2022] [Indexed: 01/26/2023]
Abstract
Gastric cancer (GC) is a highly molecular heterogeneous tumor with unfavorable outcomes. The Notch signaling pathway is an important regulator of immune cell differentiation and has been associated with autoimmune disorders, the development of tumors, and immunomodulation caused by tumors. In this study, by developing a gene signature based on genes relevant to the Notch pathway, we could improve our ability to predict the outcome of patients with GC. From the TCGA database, RNA sequencing data of GC tumors and associated normal tissues were obtained. Microarray data were collected from GEO datasets. The Molecular Signature Database (MSigDB) was accessed in order to retrieve sets of human Notch pathway-related genes (NPRGs). The LASSO analysis performed on the TCGA cohort was used to generate a multigene signature based on prognostic NPRGs. In order to validate the gene signature, the GEO cohort was utilized. Using the CIBERSORT method, we were able to determine the amounts of immune cell infiltration in the GC. In this study, a total of 21 differentially expressed NPRGs were obtained between GC specimens and nontumor specimens. The construction of a prognostic prediction model for patients with GC involved the identification and selection of three different NPRGs. According to the appropriate cutoff value, the patients with GC were divided into two groups: those with a low risk and those with a high risk. The time-dependent ROC curves demonstrated that the new model had satisfactory performance when it came to prognostic prediction. Multivariate assays confirmed that the risk score was an independent marker that may be used to predict the outcome of GC. In addition, the generated nomogram demonstrated a high level of predictive usefulness. Moreover, the scores of immunological infiltration of the majority of immune cells were distinctly different between the two groups, and the low-risk group responded to immunotherapy in a significantly greater degree. According to the results of a functional enrichment study of candidate genes, there are multiple pathways and processes associated with cancer. Taken together, a new gene model associated with the Notch pathway may be utilized for the purpose of predicting the prognosis of GC. One potential method of treatment for GC is to focus on NPRGs.
Collapse
|
35
|
Rossi G, Petrone MC, Healey AJ, Arcidiacono PG. Gastric cancer in 2022: Is there still a role for endoscopic ultrasound? World J Gastrointest Endosc 2023; 15:1-9. [PMID: 36686065 PMCID: PMC9846830 DOI: 10.4253/wjge.v15.i1.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/07/2022] [Accepted: 12/21/2022] [Indexed: 01/09/2023] Open
Abstract
Gastric cancer (GC) represents the fourth leading cause of cancer death worldwide and many factors can influence its development (diet, geographic area, genetic, Helicobacter pylori or Epstein-Barr virus infections). High quality endoscopy represents the modality of choice for GC diagnosis. The correct morphologic classification during a high-resolution endoscopy is fundamental for oncologic diagnosis, staging and therapeutic decisions. Since its initial introduction in clinical practice the endoscopic ultrasound (EUS) has been considered a valuable tool for tumor (T-) and lymph nodes (N-) staging also in GC, in order to establish the best therapeutic strategy for the patient (e.g., upfront surgery vs neoadjuvant treatments). EUS tools as elastography, Doppler and contrast administration can improve diagnosis mainly in case of malignant lymph node evaluation. EUS has a marginal role in disease staging but has a fundamental role in case of a pre-endoscopic resection management and in the new era of endoscopic mucosal resection or submucosal dissection as minimally invasive surgery. Diagnosis and locoregional staging of GC with EUS are a method of inarguable value for the assessment of gastric wall involvement and presence of infiltrated paragastric lymph nodes. EUS can also have a role in disease restaging in those patients who have undergone neoadjuvant treatment. EUS can also have a role in the advanced phases of the disease, in facilitating palliative, minimally-invasive treatments, such as gastroenterostomy or biliary drainages. This review intends to discuss the modern role of EUS in GC topic.
Collapse
Affiliation(s)
- Gemma Rossi
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute IRCCS, Vita Salute San Raffaele University, Milan 20132, Italy
| | - Maria Chiara Petrone
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute IRCCS, Vita Salute San Raffaele University, Milan 20132, Italy
| | - Andrew J Healey
- Department of Clinical Surgery, University of Edinburgh, Royal Infirmary of Edinburgh, Edinburgh EH16 4SA, United Kingdom
| | - Paolo Giorgio Arcidiacono
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute IRCCS, Vita Salute San Raffaele University, Milan 20132, Italy
| |
Collapse
|
36
|
Malek Abbaslou E, Farsad-Akhtar N, Rajabi A, Rasoolnezhad M, Safaralizadeh R. Overexpression of linc RNA-POU3F3 in gastric cancer tissues compared to adjacent non-tumor tissues and its association with clinicopathological characteristics. GENE REPORTS 2022. [DOI: 10.1016/j.genrep.2022.101696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
37
|
Chen M, Li C, Sun M, Li Y, Sun X. Recent developments in PD-1/PD-L1 blockade research for gastroesophageal malignancies. Front Immunol 2022; 13:1043517. [PMID: 36505480 PMCID: PMC9731511 DOI: 10.3389/fimmu.2022.1043517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 11/03/2022] [Indexed: 11/25/2022] Open
Abstract
Gastroesophageal cancers (GECs) comprise malignancies in the stomach, esophagus, and gastroesophageal junction. Despite ongoing improvements in chemoradiotherapy, the clinical outcomes of GEC have not significantly improved over the years, and treatment remains challenging. Immune checkpoint inhibitors (ICIs) have been the subject of clinical trials worldwide for several years. Encouraging results have been reported in different countries, but further research is required to apply ICIs in the clinical care of patients with GEC. This review summarizes completed and ongoing clinical trials with programmed death 1 (PD-1)/programmed death-ligand 1 (PD-L1) pathway blockers in GEC and current biomarkers used for predicting PD-1/PD-L1 blockade efficacy. This review captures the main findings of PD-1/PD-L1 antibodies combined with chemotherapy as an effective first-line treatment and a monotherapy in second-line or more treatment and in maintenance therapy. This review aims to provide insight that will help guide future research and clinical trials, thereby improving the outcomes of patients with GEC.
Collapse
Affiliation(s)
- Meng Chen
- Department of Gastroenterology, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Chenyan Li
- Department of Endocrinology and Metabolism, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Mingjun Sun
- Department of Gastroenterology, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yiling Li
- Department of Gastroenterology, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xuren Sun
- Department of Gastroenterology, First Affiliated Hospital of China Medical University, Shenyang, China,*Correspondence: Xuren Sun,
| |
Collapse
|
38
|
Yang J, Wang Y, Ge R, Jia X, Ge C, Cen Y, Pan D. Overexpression of Copines‐1 is associated with clinicopathological parameters and poor outcome in gastric cancer. J Clin Lab Anal 2022; 36. [DOI: 10.1002/jcla.24744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/23/2022] [Accepted: 10/09/2022] [Indexed: 11/06/2022] Open
Affiliation(s)
- Jun Yang
- Department of Cytopathology Ningbo Clinical Pathology Diagnosis Center Ningbo China
- Department of Pathology Ningbo Medical Center Lihuili Hospital Ningbo China
| | - Yingjing Wang
- Department of Histopathology Ningbo Clinical Pathology Diagnosis Center Ningbo China
| | - Rong Ge
- Department of Histopathology Ningbo Clinical Pathology Diagnosis Center Ningbo China
| | - Xiupeng Jia
- Department of Histopathology Ningbo Clinical Pathology Diagnosis Center Ningbo China
| | - Congshan Ge
- Department of Cytopathology Ningbo Clinical Pathology Diagnosis Center Ningbo China
- Department of Pathology Ningbo Medical Center Lihuili Hospital Ningbo China
| | - Youqing Cen
- Department of Cytopathology Ningbo Clinical Pathology Diagnosis Center Ningbo China
| | - Deng Pan
- Department of Cytopathology Ningbo Clinical Pathology Diagnosis Center Ningbo China
- Department of Pathology Ningbo Medical Center Lihuili Hospital Ningbo China
| |
Collapse
|
39
|
Evaluation of Tumor Necrosis Factor-Alpha Gene (−308 G/A, −238 G/A and −857 C/T) Polymorphisms and the Risk of Gastric Cancer in Eastern Indian Population. GASTROENTEROLOGY INSIGHTS 2022. [DOI: 10.3390/gastroent13040034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Introduction: Gastric cancer (GC) is one of the leading causes of cancer-related decimations worldwide. The gastric infection at both the stomach and duodenum with Helicobacter pylori causes inflammation by the tumor necrosis factor-alpha (TNF-α). The aim of the study was to associate and evaluate the three TNF-α gene polymorphisms at positions −308 G/A, −238 G/A, and −857 C/T with the risk of GC. Methods: A total of 156 individuals (consecutively diagnosed 95 GC patients and 61 controls) above the age of 18 years were enrolled in the study. Healthy individuals with normal upper gastrointestinal endoscopy (UGIE) irrespective of their family history of GC or peptic ulcer were included as controls. The cited three TNF-α gene polymorphisms were evaluated using polymerase chain reaction-restriction fragment length polymorphism (RFLP). Results: There was no significant difference in the distribution of gene polymorphisms as genetic factors, TNF-α−308 GA/AA (22.1% vs. 14.8%, p = 0.2), TNF-α−238 GA/AA (21% vs. 19.6%, p = 0.8), and TNF-α−857 CT/TT (8.4% vs. 11.5%, p = 0.5), between GC cases and healthy controls. A subgroup analysis of H. pylori-positive patients showed that there was no significant difference in the distribution of GA/AA polymorphisms in TNF-α−308 (15(45.5%) vs. 3(23%); p = 0.17) and −238 (12(36.3%) vs. 2(15.4%); p = 0.17), and the distribution of TT/CT −857 CT/TT (13(39.4%) vs. 2(15.4%); p = 0.13), among the GC cases and controls. Conclusion: The statistical comparisons of GA/AA vs. GG genotypes at −308 (with OR = 1.6, 95% CI: 0.6–3.8), −238 (OR = 1.09, 95% CI: 0.4–2.4) and TT/CT vs. CC genotypes at −857 (OR = 0.7, 95% CI: 0.2–2.1) did not suggest any association of TNF-α with GC in the population herein. Hence, the TNF-α (−308 G/A, −238 G/A and −857 C/T) may not be the associating factor for GC incidence determined by the PCR–RFLP method.
Collapse
|
40
|
Shen X, Zhu X, Hu P, Ji T, Qin Y, Zhu J. Knockdown circZNF131 Inhibits Cell Progression and Glycolysis in Gastric Cancer Through miR-186-5p/PFKFB2 Axis. Biochem Genet 2022; 60:1567-1584. [PMID: 35059934 DOI: 10.1007/s10528-021-10165-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 12/06/2021] [Indexed: 11/02/2022]
Abstract
Gastric cancer (GC) is a prevalent and heterogeneous malignancy in the digestive system. Increasing studies have suggested that circular RNAs are implicated in GC pathogenesis. This study aimed to explore the biological role and underlying mechanism of circRNA zinc finger protein 131 (circZNF131) in GC. The expression pattern of circZNF131, microRNA-186-5p (miR-186-5p), and 6-phosphofructo-2-kinase/fructose-2, 6-bisphosphatase 2 (PFKFB2) mRNA in GC tissues and cells was detected by quantitative real-time polymerase chain reaction. The stability of circZNF131 was verified using ribonuclease R assay. Functional experiments were performed by colony formation assay for cloning ability analysis, transwell assay and wounding healing assay for cell metastasis, and flow cytometry for cell apoptosis. Glycolysis metabolism was investigated by determining the levels of glucose uptake and lactate production. The protein detection of apoptosis- or glycolysis-associated markers, PFKFB2, and Ki-67 was implemented by western blot or immunohistochemistry. Dual-luciferase reporter assay was conducted to identify the interaction between miR-186-5p and circZNF131 or PFKFB2. The role of circZNF131 on tumor growth in nude mice was investigated via xenograft tumor assay. Expression analysis indicated that circZNF131 was upregulated in GC tissues and cells in a stable structure. Functional analyses showed that circZNF131 knockdown suppressed GC cell colony formation ability, migration, invasion and glycolysis metabolism, and induced cell apoptosis. Mechanically, miR-186-5p was a target of circZNF131, and miR-186-5p could bind to PFKFB2. Rescue experiments presented that miR-186-5p inhibition reversed the effects of circZNF131 knockdown on GC cell growth and glycolysis, and PFKFB2 overexpression abolished the impacts of miR-186-5p restoration on GC cell progression. Moreover, circZNF131 could positively modulate PFKFB2 expression via sponging miR-186-5p. In vivo, circZNF131 knockdown hindered GC tumor growth by regulating the miR-186-5p/PFKFB2 axis. circZNF131 could exert an oncogenic role in GC malignant development through the miR-186-5p/PFKFB2 axis, which might provide novel targets for GC treatment.
Collapse
Affiliation(s)
- Xingjie Shen
- Department of Gastroenterology, Jinan Central Hospital Affiliated to Shandong First Medical University, No.105 Jiefang Road, Lixia District, Jinan City, 250013, Shandong Province, China.
| | - Xiaoyan Zhu
- Department of Gastroenterology, Jinan Central Hospital Affiliated to Shandong First Medical University, No.105 Jiefang Road, Lixia District, Jinan City, 250013, Shandong Province, China
| | - Peixin Hu
- Department of Gastroenterology, Jinan Central Hospital Affiliated to Shandong First Medical University, No.105 Jiefang Road, Lixia District, Jinan City, 250013, Shandong Province, China
| | - Tingting Ji
- Department of Gastroenterology, Jinan Central Hospital Affiliated to Shandong First Medical University, No.105 Jiefang Road, Lixia District, Jinan City, 250013, Shandong Province, China
| | - Ying Qin
- Department of Gastroenterology, Jinan Central Hospital Affiliated to Shandong First Medical University, No.105 Jiefang Road, Lixia District, Jinan City, 250013, Shandong Province, China
| | - Jingyu Zhu
- Department of Gastroenterology, Jinan Central Hospital Affiliated to Shandong First Medical University, No.105 Jiefang Road, Lixia District, Jinan City, 250013, Shandong Province, China
| |
Collapse
|
41
|
Long non-coding RNA SNHG1/microRNA-195-5p/Yes-associated protein axis affects the proliferation and metastasis of gastric cancer via the Hippo signaling pathway. Funct Integr Genomics 2022; 22:1043-1055. [PMID: 35819551 DOI: 10.1007/s10142-022-00876-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 06/10/2022] [Accepted: 06/10/2022] [Indexed: 01/18/2023]
Abstract
Long non-coding RNA (lncRNA) small nucleolar RNA host gene 1 (SNHG1) has been found to be highly expressed in gastric cancer (GC). However, the study for exploring the effects of SNHG1 and microRNA (miR)-195-5p on GC is limited. This research commits to unravel the regulatory effects of SNHG1, miRNA-195-5p, and Yes-associated protein 1 (YAP1) on GC. SNHG1, miR-195-5p and YAP1 levels in GC tissues and GC cells were detected. The GC cells were treated with various constructs altering SNHG1 or miR-195-5p expression to determine the biological activities of GC cell in vitro. The effect of SNHG1 inhibition on subcutaneous tumorigenesis of GC cells in a nude mouse model in vivo was detected. The binding relation among SNHG1, miR-195-5p, and YAP1 was validated. SNHG1 and YAP1 levels were elevated and miR-195-5p level was reduced in GC. Reduction of SNHG1 or elevation of miR-195-5p retarded GC cell biological activity in vitro. Downregulated SNHG1 suppressed tumor growth in vivo. SNHG1 bound to miR-195-5p, and miR-195-5p directly targeted YAP1. The downregulated SNHG1 hinders the biological behaviors of GC cells via the modulation of the miR-195-5p/YAP1 axis.
Collapse
|
42
|
SMYD3 regulates gastric cancer progression and macrophage polarization through EZH2 methylation. Cancer Gene Ther 2022; 30:575-581. [PMID: 36127410 DOI: 10.1038/s41417-022-00535-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 08/21/2022] [Accepted: 09/06/2022] [Indexed: 11/12/2022]
Abstract
SET and MYND domain-containing protein 3 (SMYD3), a known histone methyltransferase, was reported to regulate cancer pathogenesis. However, its role in gastric development and progression remains unclear. EZH2 methylation had been associated with cancer metastasis, but the EZH2 methylation status in gastric cancer (GC) is unknown. Here, we report that EZH2 K421 methylation was responsible for gastric cancer cell soft agar colony formation, in vivo metastasis, and macrophage polarization. Mechanically, we identified SMYD3 as the methyltransferase of EZH2 at K421 residue which accelerates EZH2 Ubiquitin proteasome degradation. Cell harboring non-methylated EZH2 mutants promotes gastric cancer cell metastasis. Taken together, our results showed that SMYD3-EZH2 axis restricts gastric cancer metastasis via integrating epigenetic signaling.
Collapse
|
43
|
Chen J, Guo Y, Fang M, Yuan Y, Zhu Y, Xin Y, Zhang L. Neoadjuvant chemoradiotherapy for resectable gastric cancer: A meta-analysis. Front Oncol 2022; 12:927119. [PMID: 35992846 PMCID: PMC9388908 DOI: 10.3389/fonc.2022.927119] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 06/28/2022] [Indexed: 02/03/2023] Open
Abstract
Objectives To evaluate the clinical curative effects and toxicity of neoadjuvant chemoradiotherapy for resectable gastric cancer compared to those of neoadjuvant chemotherapy. Methods A systematic review and meta-analysis of the randomized controlled trials (RCTs) of neoadjuvant chemoradiotherapy versus neoadjuvant chemotherapy were performed in patients with resectable gastric cancer. Results Seven RCTs were included (601 patients; 302 in the neoadjuvant chemoradiotherapy group and 299 in the neoadjuvant chemotherapy group). The neoadjuvant chemoradiotherapy group had an increased number of patients with a complete response [odds ratio (OR) = 3.79, 95% confidence interval (CI): 1.68–8.54, p = 0.001] and improved objective response rate (OR = 2.78, 95% CI: 1.69–4.57, p < 0.0001), 1-year (OR = 3.51, 95% CI: 1.40–8.81, p = 0.007) and 3-year (OR = 2.14, 95% CI: 1.30–3.50, p = 0.003) survival rates, R0 resection rate (OR = 2.21, 95% CI: 1.39–3.50, p = 0.0008), and complete pathologic response (OR = 4.39, 95% CI: 1.59–12.14, p = 0.004). Regarding the incidence of adverse effects after neoadjuvant therapy, only the occurrence rate of gastrointestinal reaction in the neoadjuvant chemoradiotherapy group was higher than that in the neoadjuvant chemotherapy group (OR = 1.76, 95% CI: 1.09–2.85, p = 0.02), and there was no significant difference in other adverse effects. There was no difference in the incidence of postoperative complications between the two groups. Conclusion Neoadjuvant chemoradiotherapy for resectable gastric cancer has several advantages in terms of efficacy and safety compared to neoadjuvant chemotherapy. Therefore, neoadjuvant chemoradiotherapy has great potential as an effective therapy for resectable gastric cancers. Systematic Review Registration https://inplasy.com/inplasy-2022-3-0164, registration number INPLASY202230164.
Collapse
Affiliation(s)
- Jiuzhou Chen
- Department of Radiation, The Affiliated Hospital of Xuzhou Medical University, Jiangsu, China
- Department of Cancer Institute, Xuzhou Medical University, Jiangsu, China
| | - Yaru Guo
- Department of Radiation, The Affiliated Hospital of Xuzhou Medical University, Jiangsu, China
- Department of Cancer Institute, Xuzhou Medical University, Jiangsu, China
| | - Miao Fang
- Department of Radiation, The Affiliated Hospital of Xuzhou Medical University, Jiangsu, China
- Department of Cancer Institute, Xuzhou Medical University, Jiangsu, China
| | - Yan Yuan
- Department of Radiation, The Affiliated Hospital of Xuzhou Medical University, Jiangsu, China
- Department of Cancer Institute, Xuzhou Medical University, Jiangsu, China
| | - Youqi Zhu
- Department of Radiation, The Affiliated Hospital of Xuzhou Medical University, Jiangsu, China
- Department of Cancer Institute, Xuzhou Medical University, Jiangsu, China
| | - Yong Xin
- Department of Radiation, The Affiliated Hospital of Xuzhou Medical University, Jiangsu, China
- Department of Cancer Institute, Xuzhou Medical University, Jiangsu, China
- *Correspondence: Yong Xin, ; Longzhen Zhang,
| | - Longzhen Zhang
- Department of Radiation, The Affiliated Hospital of Xuzhou Medical University, Jiangsu, China
- Department of Cancer Institute, Xuzhou Medical University, Jiangsu, China
- *Correspondence: Yong Xin, ; Longzhen Zhang,
| |
Collapse
|
44
|
Wang S, Wu J, Wang Z, Gong Z, Liu Y, Wang Z. Emerging Roles of Circ-ZNF609 in Multiple Human Diseases. Front Genet 2022; 13:837343. [PMID: 35938040 PMCID: PMC9353708 DOI: 10.3389/fgene.2022.837343] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 06/03/2022] [Indexed: 12/11/2022] Open
Abstract
Circular RNAs (circRNAs) are a special type of endogenous RNAs with extensive roles in multiple human diseases. They are formed by back-splicing of partial sequences of the parental precursor mRNAs. Unlike linear RNAs, their covalently closed loop structure without a 5′ cap and a 3′ polyadenylated tail confers on them high stability and they are difficult to be digested by RNase R. Increasing evidence has proved that aberrant expressions of many circRNAs are detected and that circRNAs exert essential biological functions in disease development and progression via acting as a molecular sponge of microRNA, interacting with proteins as decoys or scaffolds, or self-encoding small peptides. Circular RNA zinc finger protein 609 (circ-ZNF609) originates from exon2 of ZNF609, which is located at chromosome 15q22.31, and it has recently been proved that it can translate into a protein. Being aberrantly upregulated in various diseases, it could promote malignant progression of human tumors, as well as tumor cell proliferation, migration, and invasion. Here in this review, we concluded the biological functions and potential mechanisms of circ-ZNF609 in multiple diseases, which could be further explored as a targetable molecule in future accurate diagnosis and prognosis.
Collapse
Affiliation(s)
| | | | | | | | - Yiyang Liu
- *Correspondence: Yiyang Liu, ; Zengjun Wang,
| | | |
Collapse
|
45
|
Su Z, Shu K, Li G. Increased ANXA5 expression in stomach adenocarcinoma infers a poor prognosis and high level of immune infiltration. Cancer Biomark 2022; 35:155-165. [PMID: 35912732 DOI: 10.3233/cbm-210482] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND: The prognostic role of annexin A5 (ANXA5) in stomach adenocarcinoma (STAD) has not been studied, and its relationship with immune infiltration is still unclear. OBJECTIVE: This investigation aimed at exploring the role of ANXA5 in STAD using an integrated bioinformatics analysis. METHODS: The expression of ANXA5 in STAD and the correlations between the effect of ANXA5 and survival of STAD patients were investigated using database. The clusterProfiler package in R software was used to perform enrichment analysis on the top 100 co-expressed genes of ANXA5 from the COXPRESdb online database. Correlations between ANXA5 and immune cell infiltrates were analyzed using the TIMER database. RESULTS: In STAD, ANXA5 expression was significantly upregulated and increased ANXA5 expression was significantly correlated with poor overall survival (P< 0.05). In multivariate analysis, upregulated ANXA5 expression was an independent predictive factors of poor prognosis (P< 0.05). The co-expressed genes were involved in extracellular matrix (ECM)-related processes. In STAD, ANXA5 expression was significantly correlated with various infiltrating immune cells (P< 0.05). CONCLUSIONS: Together with our findings, ANXA5 could serve as a potential biomarker to assess prognosis and immune infiltration level in STAD.
Collapse
Affiliation(s)
- Zhaoran Su
- Department of Gastrointestinal Surgery, People’s Hospital of Tongling City, Tongling, Anhui, China
- Department of Digestive Endoscopy, People’s Hospital of Tongling City, Tongling, Anhui, China
| | - Kuanshan Shu
- Department of Gastrointestinal Surgery, People’s Hospital of Tongling City, Tongling, Anhui, China
- Department of Digestive Endoscopy, People’s Hospital of Tongling City, Tongling, Anhui, China
| | - Guangyao Li
- Department of Gastrointestinal Surgery, The Second People’s Hospital of Wuhu, Wuhu, Anhui, China
| |
Collapse
|
46
|
Najafi S, Khatami SH, Khorsand M, Jamali Z, Shabaninejad Z, Moazamfard M, Majidpoor J, Aghaei Zarch SM, Movahedpour A. Long non-coding RNAs (lncRNAs); roles in tumorigenesis and potentials as biomarkers in cancer diagnosis. Exp Cell Res 2022; 418:113294. [PMID: 35870535 DOI: 10.1016/j.yexcr.2022.113294] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 07/11/2022] [Accepted: 07/16/2022] [Indexed: 12/15/2022]
Abstract
New research has indicated that long non-coding RNAs (lncRNAs) play critical roles in a broad range of biological processes, including the pathogenesis of many complex human diseases, including cancer. The detailed regulation mechanisms of many lncRNAs in cancer initiation and progression have yet to be discovered, even though a few of lncRNAs' functions in cancer have been characterized. In the present study, we summarize recent advances in the mechanisms and functions of lncRNAs in cancer. We focused on the roles of newly-identified lncRNAs as oncogenes and tumor suppressors, as well as the potential pathways these molecules could play. The paper also discusses their potential uses as biomarkers for the diagnosis and prognosis of cancer.
Collapse
Affiliation(s)
- Sajad Najafi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Seyyed Hossein Khatami
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Marjan Khorsand
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zeinab Jamali
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Shabaninejad
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Jamal Majidpoor
- Department of Anatomy, Faculty of Medicine, Infectious Disease Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Seyed Mohsen Aghaei Zarch
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
47
|
Gao W, Yang M. Identification by Bioinformatics Analysis of Potential Key Genes Related to the Progression and Prognosis of Gastric Cancer. Front Oncol 2022; 12:881015. [PMID: 35600357 PMCID: PMC9114743 DOI: 10.3389/fonc.2022.881015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 03/25/2022] [Indexed: 12/12/2022] Open
Abstract
Objective Despite increasingly sophisticated medical technology, the prognosis of patients with advanced gastric cancer is still not objectively certain. Therefore, it is urgent to identify new diagnostic and prognostic biomarkers. To identify potential critical genes related to gastric cancer’s staging mechanism and to the prognosis of gastric cancer. Methods Dynamic trend analysis was conducted to find genes with similar trends in gastric cancer staging in order to explore the differentially expressed genes in gastric cancer and identify the intersection of the results of the dynamic trend analysis. Functional predictive analysis were performed on the obtained genes to observe the expression of prognostic genes in gastric cancer and in gastric cancer stages as well as the correlation with tumor immune cell infiltration. Gastric cancer samples were collected and sequenced for follow-up analysis based on the results of the Cancer Genome Atlas (TCGA) database analysis. Results The expression of genes enriched in module 0 had a similar trend in gastric cancer staging. 3213 differential genes were screened. A total of 50 intersection genes were obtained among genes with similar trends, of which only 10 genes have prognostic significance in gastric cancer. These 10 genes were correlated with macrophage infiltration in varying degrees. In addition, we found that AGT was significantly abnormally expressed in the results of sample sequencing. AGT was related to the occurrence of gastric cancer and interacted with brd9, golph3, nom1, klhl25, and psmd11. Conclusion AGT has prominent abnormal expression in gastric cancer and may promote gastric cancer progression. This study provides a new direction for further exploring potential biomarkers and molecular targeted gastric cancer therapy.
Collapse
Affiliation(s)
- Wencang Gao
- Department of Oncology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Min Yang
- Department of Oncology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
48
|
Wang X, Song Z, Meng Q, Xia S, Wang C, Huang X. Circular RNA circ_0006089 regulates the IGF1R expression by targeting miR-143-3p to promote gastric cancer proliferation, migration and invasion. Cell Cycle 2022:1-14. [PMID: 35545863 DOI: 10.1080/15384101.2022.2075197] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 04/13/2022] [Accepted: 05/03/2022] [Indexed: 02/08/2023] Open
Abstract
Circular RNAs (circRNAs) figure prominently in regulating the progression of a variety of human malignancies. This study was performed to probe how circ_0006089 functioned in gastric cancer (GC). CircRNA expression profile GSE83521 was downloaded from Gene Expression Omnibus (GEO) database, and circRNAs and analyzed. Quantitative real-time polymerase chain reaction (qRT-PCR) was used to measure circ_0006089, microRNA-143-3p (miR-143-3p) and insulin-like growth factor 1 receptor (IGF1R) mRNA expressions in GC tissues and cell lines. Kaplan-Meier curves were used to detect the relationship between circ_0006089 expression and overall survival time of GC patients. Cell counting kit-8 (CCK-8) and 5-bromo-2-deoxyuridine (BrdU) assays were employed to detect the proliferative ability of GC cells after circ_0006089 was overexpressed or knocked down. Wound healing assay and Transwell assay were executed to examine the migration and invasion ability of GC cells. Western blot was adopted to detect IGF1R protein expressions. Circ_0006089 expression was up-regulated in GC samples and cell lines. And high circ_0006089 expression was associated with shorter survival time in GC patients. Circ_0006089 overexpression in GC cells significantly accelerated GC cell proliferation, migration and invasion, whereas circ_0006089 knockdown resulted in the opposite effects. Additionally, miR-143-3p was validated as a downstream target of circ_0006089, and circ_0006089 could positively regulate IGF1R expression via repressing miR-143-3p. Circ_0006089 is highly expressed in GC, and it promotes the malignancy of GC cells via modulating miR-143-3p/IGF1R axis, suggesting that circ_0006089 may serve as a promising therapeutic target for GC.
Collapse
Affiliation(s)
- Xian Wang
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Chinese PLA Medical School, Beijing, China
| | - Zhou Song
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Qingyu Meng
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Shaoyou Xia
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Chunxi Wang
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xiaohui Huang
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
49
|
Anisi Stellati Fructus, a Significant Traditional Chinese Medicine (TCM) Herb and Its Bioactivity against Gastric Cancer. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:4071489. [PMID: 35586683 PMCID: PMC9110155 DOI: 10.1155/2022/4071489] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 03/21/2022] [Accepted: 03/30/2022] [Indexed: 01/19/2023]
Abstract
Anisi stellati fructus (ASF) is the fruit of Illicium verum Hook F. (Chinese star anise), which is native to many countries, and is a significant Chinese medicinal herb. Gastric cancer (GC) is one of the major fatal types of cancers with multiple stages and a poor prognosis. The present review aims to discuss the bioactive properties of ASF and its phytocompounds against GC, with a particular insight into the molecular mechanisms and signaling pathways involved in its anti-GC mechanism. Furthermore, it highlights the potential mechanism of action of major phytocompounds of ASF against GC. Clinical studies (in vitro and in vivo) regarding the action of ASF and its major bioactive compounds such as quercetin, luteolin, kaempferol, d-limonene, and honokiol against GC were reviewed. For this review, search of literature was performed in Science, PubMed, Google Scholar, Web of Science, and Scopus related to ASF and its phytocompounds, from which only relevant studies were chosen. Major bioactive compounds of ASF and their extracts have proven to be effective against GC due to the mechanistic action of these compounds involving signaling pathways that target cancer cell apoptosis, proliferation, and tumor metastasis in GC cells. Existing reports of these compounds and their combinatory effects with other modern anticancer agents have also been reviewed. From its traditional use to its role as an anticancer agent, ASF and its bioactive phytocompounds have been observed to be effective in modern research, specifically against GC. However, further studies are required for the identification of molecular targets and pharmacokinetic potential and for the formulation of anti-GC drugs.
Collapse
|
50
|
Abnormal phenotype of Nrf2 is associated with poor prognosis through hypoxic/VEGF-A-Rap1b/VEGFR2 pathway in gastric cancer. Aging (Albany NY) 2022; 14:3293-3312. [PMID: 35417854 PMCID: PMC9037254 DOI: 10.18632/aging.204013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 11/11/2021] [Indexed: 12/15/2022]
Abstract
Metastasis is the major cause of death in gastric cancer patients and altered expression of Nrf2 is associated with cancer development. This study assessed Nrf2 and HO-1 expression and hypoxia-induced Nrf2 expression in the promotion of metastatic potential of gastric cancer cells, the relationship of Rap1b and Nrf2 was also discussed. Nrf2 and HO-1 expression were significantly associated with clinicopathological characteristic and were independent prognostic predictors in gastric cancer patients. Hypoxia up-regulated the expression of Nrf2, HO-1 and HIF-1α, whereas knockdown of Nrf2 inhibited cell invasion capacity and reduced the expression of Nrf2, HO-1 and HIF-1α. Patients in the Rap1b (+) Nrf2 (+) group had worst overall survival compared with those from other groups. Knockdown of Rap1b and Nrf2 significantly inhibited cell invasion capacity in the common group compared with the other groups. Hypoxia or VEGF-A facilitated the nuclear translocation of Nrf2 through Rap1b or VEGFR2. Hypoxia or VEGF-A did not induce the phosphorylation of P-Erk1/2 and P-Akt after knockdown of Rap1b or VEGFR2. Hypoxia promoted the gastric cancer malignant behavior through the upregulation of Rap1b and Nrf2. Hypoxia/VEGF-A-Rap1b/VEGFR2 facilitated the nuclear translocation of Nrf2. Targeting Rap1b and Nrf2 may be a novel therapeutic strategy for gastric cancer.
Collapse
|