1
|
Huang SC, Chang IYF, Chen TC, Lin HC, Tsai CY, Hsu JT, Yeh CN, Chang SC, Yeh TS. Redefining aberrant P53 expression of gastric cancer and its distinct clinical significance among molecular-histologic subtypes. Asian J Surg 2024; 47:4699-4705. [PMID: 38845323 DOI: 10.1016/j.asjsur.2024.05.121] [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: 03/13/2024] [Revised: 05/08/2024] [Accepted: 05/17/2024] [Indexed: 11/09/2024] Open
Abstract
BACKGROUND Numerous studies have demonstrated a correlation between p53 overexpression and diminished survival in gastric cancer patients. However, conflicting findings exist, and we hypothesize that these discrepancies arise from the cancer's complexity and heterogeneity, coupled with a lack of consensus on aberrant p53 expression. METHODS We enrolled a cohort of 187 patients with surgically resected gastric cancer. Patient categorization was based on Epstein-Barr virus (EBV), microsatellite instability (MSI), and Lauren classification (intestinal, diffuse and mixed). Utilizing an incremental algorithm, we evaluated p53 immunohistochemical (IHC) patterns in all 187 cases, while next-generation sequencing was successfully performed on 152 cases to identify TP53 mutations (mutTP53). RESULTS MutTP53 was identified in 32 % of the 152 cases, comprising 36 missense, 5 nonsense, and 7 frameshift alterations. Missense mutations predominantly correlated with p53 overexpression, while nonsense and frameshifting alterations related to null expression. Trial calculations indicated that null expression and a p53 IHC cutoff at >40 % offered the best prediction of mutTP53 (kappa coefficient, 0.427), with the highest agreement (0.524) observed in diffuse type and the lowest (0.269) in intestinal type. Null expression and a p53 IHC cutoff at >10 %, but not mutTP53 per se, provided the optimal prediction of survival outcome (p = 0.043), particularly in diffuse type (p = 0.044). Multivariate analysis showed that aberrant p53 IHC expression was not an independent prognostic factor. CONCLUSIONS P53 IHC patterns are predictive biomarkers for mutTP53 and gastric cancer outcomes, where a prerequisite involves a nuanced approach considering cutoff values and molecular-histologic subtyping.
Collapse
Affiliation(s)
- Shih-Chiang Huang
- Department of Anatomic Pathology, Chang Gung Memorial Hospital at Linko, Chang Gung University, Taiwan
| | - Ian Yi-Feng Chang
- Molecular Medicine Research Center, Chang Gung University, Taiwan; Department of Neurosurgery, Chang Gung Memorial Hospital at Linko, Chang Gung University, Taiwan
| | - Tse-Ching Chen
- Department of Anatomic Pathology, Chang Gung Memorial Hospital at Linko, Chang Gung University, Taiwan
| | - Hsiao-Ching Lin
- Department of Anatomic Pathology, Chang Gung Memorial Hospital at Linko, Chang Gung University, Taiwan
| | - Chun-Yi Tsai
- Department of Surgery, Chang Gung Memorial Hospital at Linko, Chang Gung University, Taiwan
| | - Jun-Te Hsu
- Department of Surgery, Chang Gung Memorial Hospital at Linko, Chang Gung University, Taiwan
| | - Chun-Nan Yeh
- Department of Surgery, Chang Gung Memorial Hospital at Linko, Chang Gung University, Taiwan
| | - Shih-Cheng Chang
- Department of Laboratory Medicine, Chang Gung Memorial Hospital at Linko, Chang Gung University, Taiwan
| | - Ta-Sen Yeh
- Department of Surgery, Chang Gung Memorial Hospital at Linko, Chang Gung University, Taiwan.
| |
Collapse
|
2
|
Li F, Wang Y, Ping X, Yin JC, Wang F, Zhang X, Li X, Zhai J, Shen L. Molecular evolution of intestinal-type early gastric cancer according to Correa cascade. J Biomed Res 2024; 38:1-16. [PMID: 39314047 DOI: 10.7555/jbr.38.20240118] [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: 09/25/2024] Open
Abstract
Early screening is crucial for the prevention of intestinal-type gastric cancer. The objective of the current study was to ascertain molecular evolution of intestinal-type gastric cancer according to the Correa cascade for the precise gastric cancer screening. We collected sequential lesions of the Correa cascade in the formalin-fixed and paraffin-embedded endoscopic submucosal dissection-resected specimens from 14 Chinese patients by microdissection, and subsequently determined the profiles of somatic aberrations during gastric carcinogenesis using the whole exome sequencing, identifying multiple variants at different Correa stages. The results showed that TP53, PCLO, and PRKDC were the most frequently mutated genes in the early gastric cancer (EGC). A high frequency of TP53 alterations was found in low-grade intraepithelial neoplasia (LGIN), which further increased in high-grade intraepithelial neoplasia (HGIN) and EGC. Intestinal metaplasia (IM) had no significant correlation with EGC in terms of mutational spectra, whereas both LGIN and HGIN showed higher genomic similarities to EGC, compared with IM. Based on Jaccard similarity coefficients, three evolutionary models were further constructed, and most patients showed linear progression from LGIN to HGIN, ultimately resulting in EGC. The ECM-receptor interaction pathway was revealed to be involved in the linear evolution. Additionally, the retrospective validation study of 39 patients diagnosed with LGIN indicated that PRKDC mutations, in addition to TP53 mutations, may drive LGIN progression to HGIN or EGC. In conclusion, the current study unveils the genomic evolution across the Correa cascade of intestinal-type gastric cancer, elucidates the underlying molecular mechanisms of gastric carcinogenesis, and provides some evidence for potential personalized gastric cancer surveillance.
Collapse
Affiliation(s)
- Fangyuan Li
- Digestive Endoscopy Center, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, China
| | - Yaohui Wang
- Department of Pathology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, China
| | - Xiaochun Ping
- Department of General Surgery, the First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Jiani C Yin
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu 210061, China
| | - Fufeng Wang
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu 210061, China
| | - Xian Zhang
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu 210061, China
| | - Xiang Li
- Department of Surgical Oncology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, China
| | - Jing Zhai
- Department of Surgical Oncology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, China
| | - Lizong Shen
- Department of General Surgery, the First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Department of Surgical Oncology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, China
| |
Collapse
|
3
|
Liu P, Ding P, Guo H, Yang J, Wu H, Wu J, Yang P, Zhao Q. Clinical calculator based on CT and clinicopathologic characteristics predicts short-term prognosis following resection of microsatellite-stabilized diffuse gastric cancer. Abdom Radiol (NY) 2024; 49:2165-2176. [PMID: 38727742 DOI: 10.1007/s00261-024-04350-4] [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: 03/27/2024] [Revised: 04/15/2024] [Accepted: 04/17/2024] [Indexed: 07/30/2024]
Abstract
PURPOSE Although microsatellite stability/Epithelial-mesenchymal transition (MSS/EMT) subtypes have been reported in multiple cancer prognosis studies, strong confounding factors between MSS/EMT (usually with Lauren's diffuse phenotype) and diffuse gastric cancer (GC) may obscure the independent prognostic value of diffuse GC. Additionally, recent studies suggest a strong correlation between mural stratification based on CT and diffuse GC. This study aims to investigate potential prognostic factors of MSS diffuse GC using mural stratification and to develop a risk assessment model. METHODS This retrospective study included 131 patients with MSS diffuse GC who underwent radical surgery. Univariate and multivariate Cox proportional hazards regression analysis was used to identify model predictors and construct a nomogram for overall survival (OS) and recurrence-free survival (RFS) risks. The model's performance was evaluated using ROC, accuracy, and C-index. Internal validation of the model was conducted using the bootstrap resampling method. RESULTS Among 131 cases, 60 cases (45.8%) exhibited grade 2 mural stratification, which correlated with a poorer tumor prognosis and a more invasive phenotype. Furthermore, a nomogram for predicting OS and RFS prognosis was established based on multivariate results (age, extranodal invasion, mural stratification, and/or P53). The nomogram demonstrated excellent performance, with an AUC of 0.859 (95% CI 0.794-0.924) for OS and 0.859 (95% CI 0.789-0.929) for RFS. Internal validation using 1000 bootstrap samples yielded AUC values of 0.845 and 0.846 for OS and RFS, respectively. CONCLUSION Grade 2 mural stratification based on CT imaging revealed a more aggressive invasive phenotype, characterized by increased LN metastasis, higher rates of peritoneal metastasis, and a poorer short-term prognosis. Furthermore, the CT phenotype-based nomogram demonstrates favorable discrimination and calibration, enabling convenient individual short-term prognostic evaluation following resection of MSS diffuse GC.
Collapse
Affiliation(s)
- Pengpeng Liu
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China
| | - Ping'an Ding
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China
| | - Honghai Guo
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China
| | - Jiaxuan Yang
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China
| | - Haotian Wu
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China
| | - Jiaxiang Wu
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China
| | - Peigang Yang
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China
| | - Qun Zhao
- The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, Hebei, China.
- Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang, 050011, China.
| |
Collapse
|
4
|
Cao F, Sun H, Yang Z, Bai Y, Hu X, Hou Y, Bian X, Liu Y. Multiple approaches revealed MGc80-3 as a somatic hybrid with HeLa cells rather than a gastric cancer cell line. Int J Cancer 2024; 154:155-168. [PMID: 37543987 DOI: 10.1002/ijc.34677] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 08/08/2023]
Abstract
The short-tandem-repeats (STR) profiles of MGc80-3 and HeLa partially overlap, raising suspicion of contamination in the MGc80-3 cell line. However, there has not been any relevant study demonstrating whether MGc80-3 was fully replaced by HeLa cells, just mixed with HeLa cells (co-existing), or was a somatic hybrid with HeLa cells. In addition to STR profiling, various approaches, including single nucleotide polymorphisms genotyping, polymerase chain reaction, screening for human papillomaviruses type 18 (HPV-18) fragment, chromosome karyotyping, pathological examination of xenografts, tissue-specific-90-gene expression signature and high-throughput RNA sequencing were used to determine the nature of MGc80-3. Our study found that the abnormal STR profile, partially overlapping with that of HeLa cells (64.62% to 71.64%), could not verify MGc80-3 as a HeLa cell line. However, the STR 13.3 repeat allele in the D13S317 locus that seemed to be unique to HeLa cells was detected in MGc80-3. Almost all the MGc80-3 cells exhibited HPV-18 fragments in the genome as well as certain HeLa marker chromosomes, such as M7 and M12. The molecular assay of the 90-gene expression signature still considered MGc80-3 as a stomach cancer using an algorithmic analysis. The expression pattern of multiple genes in MGc80-3 was quite different from that in HeLa cells, which showed that certain characteristics belonged to gastric cancer cell lines. High throughput RNA sequencing showed the distinct patterns of gene expression in MGc80-3. In conclusion, MGc80-3 cell line is a somatic hybrid with HeLa cells rather than a pure gastric cancer cell line.
Collapse
Affiliation(s)
- Fang Cao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital &Institute, Beijing, China
| | - Hao Sun
- Department of Pathology, Cell Resource Center, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Zhenli Yang
- Department of Pathology, Cell Resource Center, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Yanhua Bai
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital &Institute, Beijing, China
| | - Xiao Hu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital &Institute, Beijing, China
| | - Yuhong Hou
- Department of Pathology, Cell Resource Center, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Xiaocui Bian
- Department of Pathology, Cell Resource Center, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Yuqin Liu
- Department of Pathology, Cell Resource Center, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & School of Basic Medicine, Peking Union Medical College, Beijing, China
| |
Collapse
|
5
|
Chen W, Guo Q, Zhang H, Du Y, Zhou Y, Huang Z, Zhang M, Qin S. Differentially expressed microRNA in prognosis of gastric cancer with Lauren classification. Cancer Biomark 2024; 41:41-54. [PMID: 39177588 PMCID: PMC11492042 DOI: 10.3233/cbm-230303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 07/19/2024] [Indexed: 08/24/2024]
Abstract
BACKGROUND Gastric cancer (GC) is one of the most common tumors. There were several classifications of GC recently. The value of Lauren classification in evaluating the prognosis after radical gastrectomy was still unclear and the prognosis of gastric cancer remained relatively poor in the absence of prognostic biomarkers. This study aimed to explore microRNA (miRNA) in the prognosis of GC with different Lauren classification. METHODS A retrospective study of 1144 patients was performed in this study. Quantificational reverse transcription-PCR (qRT-PCR) was used to examine the expression of miRNAs. Univariate and multivariate analysis were performed to evaluate prognosis value of Lauren classification. RESULTS Total 1144 GC patients were recruited in this cohort, including 302 diffuse type (26.4%), 436 intestinal type (38.1%) and 406 mixed type (35.5%) GC. Multivariate analysis showed that Lauren classification, patients' age, tumor size, tumor infiltrating depth, vascular nerve infiltrating and metastatic lymph nodes ration were significantly correlated with GC patients' OS and DFS. The miR-141-3p, miR-200b-3p and miR-133a-5p were significantly down-regulated in diffuse type compared to intestinal type GC tissues, the miR-105-5p had significant lower expression in diffuse type compared with intestinal type and mixed type GC tissues. As a consequence of univariate analysis, low miR-141-3p in diffuse type GC showed significant worse OS and DFS than high miR-141-3p. CONCLUSIONS Lauren classification was an independent prognostic factor in GC. MiR-141-3p was an independent prognostic factor and a promising prognostic biomarker in Lauren classification GC.
Collapse
Affiliation(s)
- Wenjiao Chen
- Department of Radiation Oncology, First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Oncology, Affiliated Yixing Hospital of Jiangsu University, Wuxi, China
| | - Qin Guo
- Department of Radiation Oncology, First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Clinical Laboratory, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Huo Zhang
- Department of Radiation Oncology, First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Medical Oncology, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Yiping Du
- Department of Oncology, First People’s Hospital of Kunshan Affiliated with Jiangsu University, Suzhou, China
| | - Yan Zhou
- Department of Oncology, Affiliated Yixing Hospital of Jiangsu University, Wuxi, China
| | - Zebo Huang
- Department of Oncology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Meiling Zhang
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Songbing Qin
- Department of Radiation Oncology, First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
6
|
Matsuoka T, Yashiro M. Molecular Insight into Gastric Cancer Invasion-Current Status and Future Directions. Cancers (Basel) 2023; 16:54. [PMID: 38201481 PMCID: PMC10778111 DOI: 10.3390/cancers16010054] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/15/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Gastric cancer (GC) is one of the most common malignancies worldwide. There has been no efficient therapy for stage IV GC patients due to this disease's heterogeneity and dissemination ability. Despite the rapid advancement of molecular targeted therapies, such as HER2 and immune checkpoint inhibitors, survival of GC patients is still unsatisfactory because the understanding of the mechanism of GC progression is still incomplete. Invasion is the most important feature of GC metastasis, which causes poor mortality in patients. Recently, genomic research has critically deepened our knowledge of which gene products are dysregulated in invasive GC. Furthermore, the study of the interaction of GC cells with the tumor microenvironment has emerged as a principal subject in driving invasion and metastasis. These results are expected to provide a profound knowledge of how biological molecules are implicated in GC development. This review summarizes the advances in our current understanding of the molecular mechanism of GC invasion. We also highlight the future directions of the invasion therapeutics of GC. Compared to conventional therapy using protease or molecular inhibitors alone, multi-therapy targeting invasion plasticity may seem to be an assuring direction for the progression of novel strategies.
Collapse
Affiliation(s)
| | - Masakazu Yashiro
- Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, Osaka 5458585, Japan;
| |
Collapse
|
7
|
Boldrin E, Piano MA, Bernaudo F, Alfieri R, Biasin MR, Montagner IM, Volpato A, Mattara G, Lamacchia F, Magni G, Rosato A, Scapinello A, Pilati P, Curtarello M. p53/ TP53 Status Assessment in Gastroesophageal Adenocarcinoma. Cancers (Basel) 2023; 15:2783. [PMID: 37345120 DOI: 10.3390/cancers15102783] [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: 03/31/2023] [Revised: 05/05/2023] [Accepted: 05/11/2023] [Indexed: 06/23/2023] Open
Abstract
Chromosomal instability (CIN) is very frequent in gastroesophageal adenocarcinoma (GEA) and it is characterized by TP53 deletions/mutations resulting in p53 nuclear accumulation, as revealed by immunohistochemistry (IHC), which considers the cases with "high" staining levels to be positive. Aiming to improve aberrant TP53 detection, droplet digital PCR (ddPCR) was used to evaluate TP53 deletion in formalin-fixed, paraffin-embedded DNA (FFPE-DNA) and cell-free DNA (cfDNA). To further investigate the mutational TP53 profile, next-generation sequencing (NGS) was performed in a subset of FFPE samples. After combining "low" and "high" IHC staining level groups, the proportion of deletion events was significantly higher compared to the "intermediate" group (72.9% vs. 47.5%, p-value = 0.002). The ddPCR TP53 deletion assay was feasible for cfDNA but only had good agreement (72.7%, Cohen's kappa = 0.48) with the assay performed with FFPE-DNA of the "low-level" group. NGS analysis confirmed that, in the "low-level" group, a high percentage (66.7%) of cases were aberrant, with disruptive mutations that probably led to p53 loss. Data suggested that p53 IHC alone underestimates the CIN phenotype in GEA and that molecular analysis in both solid and liquid biopsies could be integrated with it; in particular, in cases of completely negative staining.
Collapse
Affiliation(s)
- Elisa Boldrin
- Immunology and Molecular Oncology Diagnostics Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy
| | - Maria Assunta Piano
- Immunology and Molecular Oncology Diagnostics Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy
| | - Francesco Bernaudo
- Immunology and Molecular Oncology Diagnostics Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy
| | - Rita Alfieri
- Surgical Oncology of Digestive Tract Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy
| | - Maria Raffaella Biasin
- Anatomy and Pathological Histology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy
| | - Isabella Monia Montagner
- Anatomy and Pathological Histology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy
| | - Alice Volpato
- Anatomy and Pathological Histology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy
| | - Genny Mattara
- Surgical Oncology of Digestive Tract Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy
| | - Francesco Lamacchia
- Surgical Oncology of Digestive Tract Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy
- Department of Surgery Oncology and Gastroenterology, University of Padova, 35122 Padova, Italy
| | - Giovanna Magni
- Clinical Research Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy
| | - Antonio Rosato
- Immunology and Molecular Oncology Diagnostics Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy
- Department of Surgery Oncology and Gastroenterology, University of Padova, 35122 Padova, Italy
| | - Antonio Scapinello
- Anatomy and Pathological Histology Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy
| | - Pierluigi Pilati
- Surgical Oncology of Digestive Tract Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy
| | - Matteo Curtarello
- Immunology and Molecular Oncology Diagnostics Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padova, Italy
| |
Collapse
|
8
|
Zhou Z, Huang D, Dai Y, Yin Y, Jiang N, Zhou Z. Clinicopathological analysis of a type of "low grade" poorly cohesive gastric adenocarcinoma not otherwise specified with a good prognosis. Ann Diagn Pathol 2023; 63:152105. [PMID: 36621077 DOI: 10.1016/j.anndiagpath.2023.152105] [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: 11/10/2022] [Revised: 01/01/2023] [Accepted: 01/01/2023] [Indexed: 01/05/2023]
Abstract
Poorly cohesive carcinoma not otherwise specified (PCGCA-NOS) is regarded in the most recent WHO classification as a high-grade malignancy; however, some cases may be associated with a relatively good prognosis. We have studied a series of 115 cases of PCGCA-NOS and were able to identify low-grade features in 14 cases based on three morphological manifestations. Immunohistochemical staining, EBER in situ hybridization, Feulgen staining and flow cytometry were employed. Among the 115 cases of PCGAC-NOS, 14 cases met the criteria of "low grade", accounting for 12.2 %. The "low grade" cases exhibited more shallow invasion and less lymph node metastasis (both P < 0.05); showed less frequent expression of MUC5AC, E-cadherin and p53 (all P < 0.05). Moreover, "low grade" PCGAC-NOS had a lower proliferative index(P < 0.001). We also found that the DNA content was lower in the "low grade" group, and aneuploidy was not detected in the "low grade" group, which was sharply different from the control group (50 %). Last, "low grade" PCGAC-NOS had a more favorable prognosis. A small subset of PCGAC-NOS cases have a low grade nature, and the clinicopathological features, immunophenotypes, and cytogenetics of these "low grade" cases differ from those of traditional PCGAC-NOS.
Collapse
Affiliation(s)
- Zhiyi Zhou
- Department of Pathology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi 214023, China
| | - Dandan Huang
- Center of Digestive Endoscopy, the Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi 214023, China
| | - Youai Dai
- Laboratory of the Institute of Organ Transplantation, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi 214023, China
| | - Yin Yin
- Department of Laboratory Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi 214023, China
| | - Nanxing Jiang
- Department of Pathology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi 214023, China
| | - Zhihua Zhou
- Department of Pathology, The 904 Hospital of Joint Logistic Support Force of People's Liberation Army, Wuxi 214044, China.
| |
Collapse
|
9
|
A novel metabolism-related prognostic gene development and validation in gastric cancer. CLINICAL & TRANSLATIONAL ONCOLOGY : OFFICIAL PUBLICATION OF THE FEDERATION OF SPANISH ONCOLOGY SOCIETIES AND OF THE NATIONAL CANCER INSTITUTE OF MEXICO 2023; 25:447-459. [PMID: 36168087 DOI: 10.1007/s12094-022-02958-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 09/15/2022] [Indexed: 01/29/2023]
Abstract
BACKGROUND The importance of metabolism-related alterations in the development of gastric cancer (GC) is increasingly recognized. The present study aimed to identify metabolism-related genes to facilitate prognosis of GC patients. METHODS Gene expression datasets and clinical information of GC patients were downloaded from TCGA and GEO databases. We scored the enrichment of human metabolism-related pathways (n = 86) in GC samples by GSV, constructed prognostic risk models using LASSO algorithm and multivariate Cox regression analysis, combined with clinical information to construct a nomogram, and finally cis score algorithm to analyze the abundance of immune-related cells in different subtypes. We used Weka software to screen for prognosis-related marker genes and finally validated the expression of the selected genes in clinical cancer patient tissues. RESULTS We identified that two GC metabolism-related signatures were strongly associated with OS and the levels of immune cell infiltration. Moreover, a survival prediction model for GC was established based on six GC metabolism-related genes. Time-dependent ROC analysis showed good stability of the risk prediction scoring model. The model was successfully validated in an independent ACRG cohort, and the expression trends of key genes were also verified in the GC tissues of patients. DLX1, LTBP2, FGFR1 and MMP2 were highly expressed in the cluster with poorer prognosis while SLC13A2 and SLCO1B3 were highly expressed in the cluster with better prognosis. CONCLUSIONS We identified a risk predictive score model based on six metabolism-related genes related to survival, which may serve as prognostic indicators and potential therapeutic targets for GC.
Collapse
|
10
|
Li F, Li S, Wang X, Liu C, Li X, Li Y, Liu Y. To investigate the prognostic factors of stage Ⅰ-Ⅱ gastric cancer based on P53 mutation and tumor budding. Pathol Res Pract 2022; 240:154195. [PMID: 36356333 DOI: 10.1016/j.prp.2022.154195] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/27/2022] [Accepted: 10/29/2022] [Indexed: 11/05/2022]
Abstract
BACKGROUND P53 is a tumor suppressor and genome guardian factor, and tumor budding is a key link in tumor metastasis. The purpose of this study was to investigate P53 mutation and tumor budding in stage Ⅰ-Ⅱ gastric cancer, to explore the correlation with clinicopathological features, and to reveal the independent prognostic factors of gastric cancer. METHODS The data of 588 patients with stage Ⅰ-Ⅱ gastric cancer who underwent radical surgical resection from April 2015 to December 2016 in the Fourth Hospital of Hebei Medical University were retrospectively analyzed and followed up. Immunohistochemistry Envision method was used to conduct P53 staining for paraffin-embedded gastric cancer tissues, and ITBCC recommended tumor budding evaluation method was used to count tumor budding in gastric cancer tissues. The factors affecting the prognosis of gastric cancer were analyzed. RESULTS There were 209 cases (35.54%) of P53 wild-type and 379 cases (64.46 %) of P53 mutant in 588 patients with stage Ⅰ-Ⅱ gastric cancer. P53 mutation rate were closely correlated with Lauren classification (χ2 =8.152, p = 0.017), degree of differentiation (χ2 =10.495, p = 0.004), number of lymph node metastasis (χ2 =25.550, p < 0.001), and clinical stage (χ2 =7.617, p = 0.016). Tumor budding were closely correlated with Lauren classification (χ2 =194.533, p < 0.001), degree of tissue differentiation (χ2 =22.719, p < 0.001), depth of tumor invasion (χ2 =19.204, p = 0.004), number of lymph node metastasis (χ2 =22.555, p = 0.001), clinical stage (χ2 =10.769, p = 0.005), and vascular tumors bolt (χ2 =12.478, p = 0.002). The higher the tumor budding grade was, the higher the P53 mutation rate was (χ2 =12.933, p = 0.002). Lauren classification (p < 0.001), degree of tissue differentiation (p = 0.005), vascular tumors bolt (p < 0.001) and P53 mutation (p = 0.006) were independent influencing factors for 5-year survival of patients with stage Ⅰ-Ⅱ gastric cancer. CONCLUSION P53 mutation status is an independent prognostic factor for gastric cancer patients and a promising cancer treatment target. Tumor budding is a very reliable independent prognostic parameter with important clinical value and should be routinely reported as a biomarker.
Collapse
Affiliation(s)
- Fang Li
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Shi Li
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xinran Wang
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Chang Liu
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiaoya Li
- Department of Scientific Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yong Li
- Department of General Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yueping Liu
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China.
| |
Collapse
|
11
|
Choi S, Kim N, Park JH, Nam RH, Song CH, Lee HS. Effect of Helicobacter pylori infection and its eradication on the expression of tight junction proteins in the gastric epithelium in relation to gastric carcinogenesis. Helicobacter 2022; 27:e12929. [PMID: 36063450 DOI: 10.1111/hel.12929] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 08/10/2022] [Accepted: 08/15/2022] [Indexed: 12/09/2022]
Abstract
BACKGROUND Tight junction proteins (TJPs) play a role in epithelial defense mechanisms. However, the effect of Helicobacter pylori (Hp) on TJPs remains unclear. This study aimed to evaluate the expression of TJPs in relation to Hp infection and eradication in gastric carcinogenesis. METHODS In total, 510 subjects (284 controls and 226 gastric cancer [GC] patients) were prospectively enrolled in the study. The expression of claudin-1 and -2 (CLDN-1, -2), occludin (OCLN), and tight junction protein 1 (TJP1) was measured based on their Hp infection status in normal corpus mucosa and evaluated following Hp eradication using quantitative real-time polymerase chain reaction (qPCR) and immunohistochemistry (IHC). RESULTS The expression of TJP1 in Hp+ controls was significantly lower than that in Hp- controls (p = 0.006), whereas it was higher in Hp+ than in Hp- GC patients (p = 0.001). Moreover, the increased expression of TJP1 in Hp+ GC patients was reduced to levels in Hp- within a year after Hp eradication and was maintained for more than 5 years. Furthermore, IHC results for TJP1 were similar to qPCR results. In particular, the higher IHC staining intensity of TJP1 in the cytosol of GC patients (p = 0.019) decreased after Hp eradication (p = 0.040). CONCLUSION Hp infection affects TJP expression. The high expression of TJP1 in Hp+ GC patients was restored to control levels after Hp eradication, suggesting that TJP1 plays a role in gastric carcinogenesis.
Collapse
Affiliation(s)
- SooIn Choi
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Nayoung Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea.,Department of Internal Medicine and Liver Research Institute, Seoul National University, Seoul, South Korea
| | - Ji Hyun Park
- Department of Internal Medicine and Liver Research Institute, Seoul National University, Seoul, South Korea
| | - Ryoung Hee Nam
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Chin-Hee Song
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Hye Seung Lee
- Department of Pathology, Seoul National University, Seoul, South Korea
| |
Collapse
|
12
|
Li C, Peng X, Peng Z, Yan B. circBGN accelerates gastric cancer cell proliferation and invasion via activating IL6/STAT3 signaling pathway. FASEB J 2022; 36:e22604. [PMID: 36250950 DOI: 10.1096/fj.202200957rr] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 09/20/2022] [Accepted: 09/28/2022] [Indexed: 01/17/2023]
Abstract
Circular RNAs participate in the pathogenesis of various tumors, including gastric cancer (GC). In this study, we investigated the role of circBGN in regulating proliferation and invasion of GC cells and elucidated the mechanism. The expression of circBGN was assessed by quantitative reverse-transcription PCR and in situ hybridization. In addition, loss- and gain-of-function investigations in vitro and in vivo were performed to determine the biological functions of circBGN. Luciferase reporter assays and rescue experiments were applied to investigate the interaction between circBGN and miR-149-5p as well as the relationship between miR-149-5p and IL6. Our results showed that circBGN expression was significantly elevated in GC tissues and cells. Knockdown of circBGN dramatically suppressed GC cell proliferation and invasion in vitro. Xenograft experiments revealed that knockdown of circBGN delayed tumor growth in vivo. Furthermore, circBGN can directly bind to miR-149-5p, thereby preventing miR-149-5p from binding to its target mRNA [IL6 mRNA], thus activating IL6/STAT3 signaling pathway. Rescue assays indicated that circBGN regulates GC cell proliferation and invasion by upregulating miR-149-5p/IL6 axis output. Taken together, our investigation indicates that circBGN supports GC progression by activating IL6/STAT3 signaling pathway, thus pointing to a new possible therapeutic target in GC.
Collapse
Affiliation(s)
- Chenghui Li
- Department of General Surgery, Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai, China
| | - Xiang Peng
- Department of General Surgery, Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai, China
| | - Zhiyong Peng
- Department of General Surgery, Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai, China
| | - Bin Yan
- Department of General Surgery, Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai, China
| |
Collapse
|
13
|
Choi Y, Kim N, Kim KW, Jo HH, Park J, Yoon H, Shin CM, Park YS, Lee DH. Gastric cancer in older adult patients: a retrospective study and literature review. Ann Geriatr Med Res 2022; 26:33-41. [PMID: 35298880 PMCID: PMC8984166 DOI: 10.4235/agmr.21.0144] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 03/11/2022] [Indexed: 11/13/2022] Open
Abstract
Background With increasing life expectancy, the incidence of gastric cancer (GC) in older adults is increasing. This study analyzed differences in GC characteristics according to age and sex among patients who underwent surgical treatment for GC. Methods A total of 2,983 patients diagnosed with gastric adenocarcinoma who underwent surgical treatment at Seoul National University Bundang Hospital between 2003 and 2017 were classified into three groups: I (<65 years, n=1,680), II (60–74 years, n=919), and III (≥75 years, n=384). We compared the baseline clinical characteristics, pathological characteristics of the tumor, overall and GC-specific survival rates, and associated risk factors between the groups. Results Cancer of the distal third of the stomach (p<0.001), with intestinal-type histology (p<0.001), and with p53 overexpression (p=0.004) were more common in groups II and III than in group I, and the proportion of intestinal-type GC increased with age. The cancer type, lymph node metastasis, and cancer stage did not differ significantly. In terms of overall survival, survival decreased with increasing age (p<0.001), but this difference decreased significantly for GC-specific survival. Cox multivariate analyses revealed age, histologic type (diffuse or mixed type), and advanced cancer stage (p=0.002, 0.001, and <0.001, respectively) as risk factors for GC-related mortality. Conclusion Age itself was found to be one of the most important prognostic factors for overall and disease-specific survival in elderly GC patients, along with cancer stage.
Collapse
Affiliation(s)
- Yonghoon Choi
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Nayoung Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea.,Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Ki Wook Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Hyeong Ho Jo
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Jaehyung Park
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Hyuk Yoon
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Cheol Min Shin
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Young Soo Park
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Dong Ho Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea.,Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
14
|
Choi Y, Kim N, Kim KW, Jo HH, Park J, Yoon H, Shin CM, Park YS, Lee DH, Oh HJ, Lee HS, Park YS, Ahn SH, Suh YS, Park DJ, Kim HH, Kim JW, Kim JW, Lee KW, Chang W, Park JH, Lee YJ, Lee KH, Kim YH. Sex-based differences in histology, staging, and prognosis among 2983 gastric cancer surgery patients. World J Gastroenterol 2022; 28:933-947. [PMID: 35317055 PMCID: PMC8908285 DOI: 10.3748/wjg.v28.i9.933] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 11/08/2021] [Accepted: 01/26/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Few studies have been conducted on sex differences in the incidence, pathophysiology, and prognosis of gastric cancer (GC).
AIM To analyze the differences in GC characteristics according to sex in patients who underwent surgical treatment for GC.
METHODS A total of 2983 patients diagnosed with gastric adenocarcinoma who received surgical treatment at the Seoul National University Bundang Hospital between 2003 and 2017 were included. Baseline clinicopathological characteristics, histologic type of GC, overall and GC-specific survival rates, and associated risk factors were analyzed.
RESULTS Among the 2983 patients, 2005 (67.2%) and 978 (32.8%) were males and females, respectively. The average age of the female group (59.36 years) was significantly younger than that of the male group (61.66 years; P < 0.001). Cancer of the gastric body (P < 0.001) and diffuse-type histology (P < 0.001) were more common in females than in males. This trend was more prominent in females younger than 60 years of age, with a significantly higher proportion of diffuse-type cancer than in the male group. Regardless of sex, diffuse-type GC was more common in younger patients, and the proportion of intestinal-type GC increased with age. The overall survival rate was significantly higher in females (P < 0.001). However, this difference disappeared for GC-specific survival (P = 0.168), except for the poor GC-specific survival rate in advanced-stage cancer (stage III or above) in females (P = 0.045). The risk factors for GC-related mortality were older age, upper location of GC, and diffuse- or mixed-type histology. In terms of comorbidities, more males died from diseases other than GC, including other malignancies such as lung cancer, hepatocellular carcinoma, and pancreatic cancer, and respiratory diseases such as interstitial lung disease and chronic obstructive pulmonary disease, while there were relatively more cardiovascular or cerebrovascular deaths in females.
CONCLUSION Sex-based differences in GC were observed in clinicopathological features, including age at diagnosis, tumor location, histologic type, survival rate, and comorbidities.
Collapse
Affiliation(s)
- Yonghoon Choi
- Department ofInternal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, South Korea
| | - Nayoung Kim
- Department ofInternal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, South Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, South Korea
| | - Ki Wook Kim
- Department ofInternal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, South Korea
| | - Hyeong Ho Jo
- Department ofInternal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, South Korea
| | - Jaehyung Park
- Department ofInternal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, South Korea
| | - Hyuk Yoon
- Department ofInternal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, South Korea
| | - Cheol Min Shin
- Department ofInternal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, South Korea
| | - Young Soo Park
- Department ofInternal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, South Korea
| | - Dong Ho Lee
- Department ofInternal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, South Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, South Korea
| | - Hyeon Jeong Oh
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam 13620, South Korea
| | - Hye Seung Lee
- Department of Pathology, Seoul National University College of Medicine, Seoul 03080, South Korea
| | - Young Suk Park
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam 13620, South Korea
| | - Sang-Hoon Ahn
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam 13620, South Korea
| | - Yun-Suhk Suh
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam 13620, South Korea
- Department of Surgery, Seoul National University College of Medicine, Seoul 03080, South Korea
| | - Do Joong Park
- Department of Surgery, Seoul National University College of Medicine, Seoul 03080, South Korea
| | - Hyung-Ho Kim
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam 13620, South Korea
- Department of Surgery, Seoul National University College of Medicine, Seoul 03080, South Korea
| | - Ji-Won Kim
- Department ofInternal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, South Korea
| | - Jin Won Kim
- Department ofInternal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, South Korea
| | - Keun-Wook Lee
- Department ofInternal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, South Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, South Korea
| | - Won Chang
- Department of Radiology, Seoul National University Bundang Hospital, Seongnam 13620, South Korea
| | - Ji Hoon Park
- Department of Radiology, Seoul National University Bundang Hospital, Seongnam 13620, South Korea
| | - Yoon Jin Lee
- Department of Radiology, Seoul National University Bundang Hospital, Seongnam 13620, South Korea
| | - Kyoung Ho Lee
- Department of Radiology, Seoul National University Bundang Hospital, Seongnam 13620, South Korea
- Department of Radiology, Seoul National University College of Medicine, Seoul 03080, South Korea
| | - Young Hoon Kim
- Department of Radiology, Seoul National University Bundang Hospital, Seongnam 13620, South Korea
- Department of Radiology, Seoul National University College of Medicine, Seoul 03080, South Korea
| |
Collapse
|
15
|
Chen J, Yang S, Li P, Wu A, Nepovimova E, Long M, Wu W, Kuca K. MicroRNA regulates the toxicological mechanism of four mycotoxins in vivo and in vitro. J Anim Sci Biotechnol 2022; 13:37. [PMID: 35197116 PMCID: PMC8867758 DOI: 10.1186/s40104-021-00653-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 11/21/2021] [Indexed: 11/30/2022] Open
Abstract
Mycotoxins can cause body poisoning and induce carcinogenesis, often with a high mortality rate. Therefore, it is of great significance to seek new targets that indicate mycotoxin activity and to diagnose and intervene in mycotoxin-induced diseases in their early stages. MicroRNAs (miRNAs) are physiological regulators whose dysregulation is closely related to the development of diseases. They are thus important markers for the occurrence and development of diseases. In this review, consideration is given to the toxicological mechanisms associated with four major mycotoxins (ochratoxin A, aflatoxin B1, deoxynivalenol, and zearalenone). The roles that miRNAs play in these mechanisms and the interactions between them and their target genes are explained, and summarize the important role of histone modifications in their toxicity. As a result, the ways that miRNAs are regulated in the pathogenicity signaling pathways are revealed which highlights the roles played by miRNAs in preventing and controlling the harmful effects of the mycotoxins. It is hoped that this review will provide a theoretical basis for the prevention and control of the damage caused by these mycotoxins.
Collapse
Affiliation(s)
- Jia Chen
- College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China
| | - Shuhua Yang
- College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China
| | - Peng Li
- College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China
| | - Aibo Wu
- SIBS-UGENT-SJTU Joint Laboratory of Mycotoxin Research, CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, 50003, Czech Republic
| | - Miao Long
- College of Animal Science & Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China.
| | - Wenda Wu
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, 50003, Czech Republic. .,MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, 50003, Czech Republic. .,Biomedical Research Center, University Hospital Hradec Kralove, Hradec Kralove, 50003, Czech Republic.
| |
Collapse
|
16
|
Jung YJ, Kim HJ, Park CH, Park SJ, Kim N. Effects of Reproductive Factors on Lauren Intestinal-Type Gastric Cancers in Females: A Multicenter Retrospective Study in South Korea. Gut Liver 2022; 16:706-715. [PMID: 35000933 PMCID: PMC9474480 DOI: 10.5009/gnl210293] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 10/05/2021] [Accepted: 10/21/2021] [Indexed: 12/13/2022] Open
Abstract
Background/Aims Gastric cancers (GCs), particularly the Lauren intestinal type, show a male predominance. The aim of this study was to investigate the effects of reproductive factors on GCs in females, according to Lauren classification. Methods Medical records of 1,849 males and 424 females who underwent radical gastrectomy or endoscopic resection for GCs between 2010 and 2018 were reviewed. The incidences of intestinal-type GCs were compared between males and groups of females stratified according to postmenopausal period. Associations between reproductive factors in females and intestinaltype GCs were analyzed using multivariate models. Results The proportions of intestinal-type GCs were significantly lower in premenopausal (19%), less than 10 years postmenopausal (30.4%), and 10 to 19 years postmenopausal females (44.1%) than in males (61.0%) (p<0.05 for all). Females ≥20 years postmenopause had a proportion of intestinal-type GCs similar to that in males (60.6% vs 61.0%; p=0.948). Multivariate analysis revealed that age (odds ratio [OR], 1.075; 95% confidence interval [CI], 1.039 to 1.113; p<0.001) and parity ≥3 (OR, 1.775; 95% CI, 1.012 to 3.114; p=0.045) were positively associated with an increased risk of intestinal-type GCs in postmenopausal females, while long fertility duration (OR, 1.147; 95% CI, 1.043 to 1.261; p=0.005) was positively associated with an increased risk of intestinal-type GCs in premenopausal females. Conclusions There were no significant differences in the proportions of intestinal-type GCs between males and females ≥20 years postmenopause, suggesting that female reproductive factors play a role in the prevention of intestinal-type GC.
Collapse
Affiliation(s)
- Yoon Ju Jung
- Division of Gastrointestinal Surgery, Department of Surgery, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Incheon, Korea
| | - Hee Jin Kim
- Department of Internal Medicine, Gyeongsang National University Changwon Hospital, Gyeongsang National University College of Medicine, Changwon, Korea
| | - Cho Hyun Park
- Division of Gastrointestinal Surgery, Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Seun Ja Park
- Department of Internal Medicine, Kosin University College of Medicine, Busan, Korea
| | - Nayoung Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea.,Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
17
|
Kim HY, Chang W, Lee YJ, Park JH, Cho J, Na HY, Ahn H, Hwang SI, Lee HJ, Kim YH, Lee KH. Adrenal Nodules Detected at Staging CT in Patients with Resectable Gastric Cancers Have a Low Incidence of Malignancy. Radiology 2021; 302:129-137. [PMID: 34665031 DOI: 10.1148/radiol.2021211210] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background Guidelines recommending additional imaging for adrenal nodules lack relevant epidemiologic evidence. Purpose To measure the prevalence of adrenal nodules detected at staging CT in patients with potentially resectable gastric cancer and the proportion of patients with malignant nodules among them. Materials and Methods This retrospective study included 10 250 consecutive patients (median age, 63 years; interquartile range, 53-71 years; 6884 men) who underwent staging CT and had potentially resectable gastric cancer in a tertiary center (May 2003 to December 2018). All 10 250 CT studies were retrospectively reviewed, and patients with adrenal nodules (or thickening ≥10 mm) were identified to measure the prevalence of adrenal nodules. Among patients with adrenal nodules, the per-patient proportions of malignant nodules, adrenal metastasis from gastric cancer, and additional adrenal examinations were measured. A secondary analysis was performed by using data from the original CT reports. The same metrics that were used in the retrospective review were assessed. Results The prevalence of adrenal nodules was 4.5% (95% CI: 4.1, 4.9; 462 of 10 250). The proportions of malignant nodules and adrenal metastasis from gastric cancer were 0.4% ( 95% CI: 0.1, 1.6; two of 462) and 0% (95% CI: 0.0, 0.8; 0 of 462), respectively. A total of 27% of the patients (95% CI: 23, 31; 123 of 462) underwent additional adrenal examination. According to original CT reports, the prevalence of adrenal nodules and the proportions of malignant nodules, adrenal metastases from gastric cancer, and additional adrenal examination were 2.7% (95% CI: 2.4, 3.0; 272 of 10 250), 0.7% (95% CI: 0.1, 2.6; two of 272), 0% (95% CI: 0.0, 1.4; 0 of 272), and 42.6% (95% CI: 36.7, 48.8; 116 of 272), respectively. Conclusion Although adrenal nodules were detected frequently on staging CT images of patients with otherwise resectable gastric cancer, these nodules were rarely malignant. ©RSNA, 2021 Online supplemental material is available for this article. See also the editorial by Baumgarten in this issue.
Collapse
Affiliation(s)
- Hae Young Kim
- From the Departments of Radiology (H.Y.K., W.C., Y.J.L., J.H.P., J.C., H.A., S.I.H., H.J.L., Y.H.K., K.H.L.) and Pathology (H.Y.N.), Seoul National University Bundang Hospital, 82 Gumi-ro-173-beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 13620, Korea; Department of Pathology, Seoul National University College of Medicine, Seoul, Korea (H.Y.N.); Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea (K.H.L.); Department of Radiology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Gyeonggi-do, Korea (H.J.L., Y.H.K., K.H.L.); and Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, Korea (K.H.L.)
| | - Won Chang
- From the Departments of Radiology (H.Y.K., W.C., Y.J.L., J.H.P., J.C., H.A., S.I.H., H.J.L., Y.H.K., K.H.L.) and Pathology (H.Y.N.), Seoul National University Bundang Hospital, 82 Gumi-ro-173-beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 13620, Korea; Department of Pathology, Seoul National University College of Medicine, Seoul, Korea (H.Y.N.); Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea (K.H.L.); Department of Radiology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Gyeonggi-do, Korea (H.J.L., Y.H.K., K.H.L.); and Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, Korea (K.H.L.)
| | - Yoon Jin Lee
- From the Departments of Radiology (H.Y.K., W.C., Y.J.L., J.H.P., J.C., H.A., S.I.H., H.J.L., Y.H.K., K.H.L.) and Pathology (H.Y.N.), Seoul National University Bundang Hospital, 82 Gumi-ro-173-beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 13620, Korea; Department of Pathology, Seoul National University College of Medicine, Seoul, Korea (H.Y.N.); Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea (K.H.L.); Department of Radiology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Gyeonggi-do, Korea (H.J.L., Y.H.K., K.H.L.); and Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, Korea (K.H.L.)
| | - Ji Hoon Park
- From the Departments of Radiology (H.Y.K., W.C., Y.J.L., J.H.P., J.C., H.A., S.I.H., H.J.L., Y.H.K., K.H.L.) and Pathology (H.Y.N.), Seoul National University Bundang Hospital, 82 Gumi-ro-173-beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 13620, Korea; Department of Pathology, Seoul National University College of Medicine, Seoul, Korea (H.Y.N.); Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea (K.H.L.); Department of Radiology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Gyeonggi-do, Korea (H.J.L., Y.H.K., K.H.L.); and Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, Korea (K.H.L.)
| | - Jungheum Cho
- From the Departments of Radiology (H.Y.K., W.C., Y.J.L., J.H.P., J.C., H.A., S.I.H., H.J.L., Y.H.K., K.H.L.) and Pathology (H.Y.N.), Seoul National University Bundang Hospital, 82 Gumi-ro-173-beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 13620, Korea; Department of Pathology, Seoul National University College of Medicine, Seoul, Korea (H.Y.N.); Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea (K.H.L.); Department of Radiology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Gyeonggi-do, Korea (H.J.L., Y.H.K., K.H.L.); and Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, Korea (K.H.L.)
| | - Hee Young Na
- From the Departments of Radiology (H.Y.K., W.C., Y.J.L., J.H.P., J.C., H.A., S.I.H., H.J.L., Y.H.K., K.H.L.) and Pathology (H.Y.N.), Seoul National University Bundang Hospital, 82 Gumi-ro-173-beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 13620, Korea; Department of Pathology, Seoul National University College of Medicine, Seoul, Korea (H.Y.N.); Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea (K.H.L.); Department of Radiology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Gyeonggi-do, Korea (H.J.L., Y.H.K., K.H.L.); and Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, Korea (K.H.L.)
| | - Hyungwoo Ahn
- From the Departments of Radiology (H.Y.K., W.C., Y.J.L., J.H.P., J.C., H.A., S.I.H., H.J.L., Y.H.K., K.H.L.) and Pathology (H.Y.N.), Seoul National University Bundang Hospital, 82 Gumi-ro-173-beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 13620, Korea; Department of Pathology, Seoul National University College of Medicine, Seoul, Korea (H.Y.N.); Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea (K.H.L.); Department of Radiology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Gyeonggi-do, Korea (H.J.L., Y.H.K., K.H.L.); and Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, Korea (K.H.L.)
| | - Sung Il Hwang
- From the Departments of Radiology (H.Y.K., W.C., Y.J.L., J.H.P., J.C., H.A., S.I.H., H.J.L., Y.H.K., K.H.L.) and Pathology (H.Y.N.), Seoul National University Bundang Hospital, 82 Gumi-ro-173-beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 13620, Korea; Department of Pathology, Seoul National University College of Medicine, Seoul, Korea (H.Y.N.); Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea (K.H.L.); Department of Radiology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Gyeonggi-do, Korea (H.J.L., Y.H.K., K.H.L.); and Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, Korea (K.H.L.)
| | - Hak Jong Lee
- From the Departments of Radiology (H.Y.K., W.C., Y.J.L., J.H.P., J.C., H.A., S.I.H., H.J.L., Y.H.K., K.H.L.) and Pathology (H.Y.N.), Seoul National University Bundang Hospital, 82 Gumi-ro-173-beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 13620, Korea; Department of Pathology, Seoul National University College of Medicine, Seoul, Korea (H.Y.N.); Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea (K.H.L.); Department of Radiology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Gyeonggi-do, Korea (H.J.L., Y.H.K., K.H.L.); and Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, Korea (K.H.L.)
| | - Young Hoon Kim
- From the Departments of Radiology (H.Y.K., W.C., Y.J.L., J.H.P., J.C., H.A., S.I.H., H.J.L., Y.H.K., K.H.L.) and Pathology (H.Y.N.), Seoul National University Bundang Hospital, 82 Gumi-ro-173-beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 13620, Korea; Department of Pathology, Seoul National University College of Medicine, Seoul, Korea (H.Y.N.); Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea (K.H.L.); Department of Radiology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Gyeonggi-do, Korea (H.J.L., Y.H.K., K.H.L.); and Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, Korea (K.H.L.)
| | - Kyoung Ho Lee
- From the Departments of Radiology (H.Y.K., W.C., Y.J.L., J.H.P., J.C., H.A., S.I.H., H.J.L., Y.H.K., K.H.L.) and Pathology (H.Y.N.), Seoul National University Bundang Hospital, 82 Gumi-ro-173-beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do 13620, Korea; Department of Pathology, Seoul National University College of Medicine, Seoul, Korea (H.Y.N.); Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Korea (K.H.L.); Department of Radiology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Gyeonggi-do, Korea (H.J.L., Y.H.K., K.H.L.); and Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, Korea (K.H.L.)
| |
Collapse
|
18
|
Cimbalo A, Alonso-Garrido M, Font G, Frangiamone M, Manyes L. Transcriptional Changes after Enniatins A, A1, B and B1 Ingestion in Rat Stomach, Liver, Kidney and Lower Intestine. Foods 2021; 10:foods10071630. [PMID: 34359500 PMCID: PMC8303686 DOI: 10.3390/foods10071630] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/18/2021] [Accepted: 06/26/2021] [Indexed: 12/12/2022] Open
Abstract
Enniatins (ENs) are depsipeptide mycotoxins produced by Fusarium fungi. They are known for their capacity to modulate cell membrane permeability and disruption of ionic gradients, affecting cell homeostasis and initiating oxidative stress mechanisms. The effect of the acute toxicity of ENs A, A1, B and B1 at two different concentrations after 8 h of exposure was analysed in Wistar rats by a transcriptional approach. The following key mitochondrial and nuclear codified genes related to the electron transport chain were considered for gene expression analysis in stomach, liver, kidney and lower intestine by quantitative Real-Time PCR: mitochondrially encoded NADH dehydrogenase 1 (MT-ND1), mitochondrially encoded cytochrome c oxidase 1 (MT-COX1), succinate dehydrogenase flavoprotein subunit A and ATP synthase F1 subunit alpha, respectively. Moreover, the expression of markers involved in oxidative stresssuperoxide dismutase 1 (SOD1), glutathione peroxidase 1 (Gpx1), heme oxygenase 1, apoptosis B-cell lymphoma 2, Bcl2 Associated protein X (Bax), tumor suppressor protein (p53), inhibition of apoptosis nuclear factor kappa of activated B cells, immune system interleukin 1β and intestinal tight junction Occludin merely in lower intestine tissues have been investigated. For mitochondrial genes, the main differences were observed for MT-ND1 and MT-COX1, showing its deficiency in all selected organs. With regard to the intestinal barrier’s cellular response to oxidative stress, the activity of the antioxidant gene SOD1 was decreased in a dose-dependent manner. Similarly, the catalytic enzyme GPx1 was also downregulated though merely at medium dose employed. On the contrary, the pro-apoptotic Bax and p53 regulators were activated after ENs exposure, reporting a significant increase in their expression. Furthermore, the alteration of intestinal permeability was assessed by the abnormal activity of the tight junction protein occludin. In summary, ENs may generate mitochondrial disorders and induce oxidative stress in intestinal barrier function.
Collapse
|