1
|
He R, Tang J, Lai H, Zhang T, Du L, Wei S, Zhao P, Tang G, Liu J, Luo X. Deciphering the role of sphingolipid metabolism in the immune microenvironment and prognosis of esophageal cancer via single-cell sequencing and bulk data analysis. Discov Oncol 2024; 15:505. [PMID: 39333432 PMCID: PMC11436545 DOI: 10.1007/s12672-024-01379-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 09/20/2024] [Indexed: 09/29/2024] Open
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) stands as a significant global health challenge, distinguished by its aggressive progression from the esophageal epithelium. Central to this malignancy is sphingolipid metabolism, a critical pathway that governs key cellular processes, including apoptosis and immune regulation, thereby influencing tumor behavior. The advent of single-cell and transcriptome sequencing technologies has catalyzed significant advancements in oncology research, offering unprecedented insights into the molecular underpinnings of cancer. METHODS We explored sphingolipid metabolism-related genes in ESCC using scRNA-seq data from GEO and transcriptome data from TCGA. We assessed 97 genes in epithelial cells with AUCell, UCell, and singscore algorithms, followed by bulk RNA-seq and differential analysis to identify prognosis-related genes. Immune infiltration and potential immunotherapeutic strategies were also investigated, and tumor gene mutations and drug treatment strategies were analyzed. RESULT Our study identified distinct gene expression patterns, highlighting ARSD, CTSA, DEGS1, and PPTQ's roles in later cellular stages. We identified seven independent prognostic genes and created a precise nomogram for prognosis. CONCLUSION This study integrates single-cell and transcriptomic data to provide a reliable prognostic model associated with sphingolipid metabolism and to inform immunotherapy and pharmacotherapy for ESCC at the genetic level. The findings have significant implications for precision therapy in esophageal cancer.
Collapse
Affiliation(s)
- Rongzhang He
- Gastroenterology Department, Guangyuan Central Hospital, Guangyuan, China
| | - Jing Tang
- Gastroenterology Department, Guangyuan Central Hospital, Guangyuan, China
| | - Haotian Lai
- School of Clinical Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Tianchi Zhang
- Department of General Surgery, Dazhou Central Hospital, Dazhou, China
| | - Linjuan Du
- Oncology department, Dazhou Central Hospital, Dazhou, China
| | - Siqi Wei
- Gastroenterology Department, Guangyuan Central Hospital, Guangyuan, China
| | - Ping Zhao
- Gastroenterology Department, Guangyuan Central Hospital, Guangyuan, China
| | - Guobin Tang
- Gastroenterology Department, Guangyuan Central Hospital, Guangyuan, China
| | - Jie Liu
- Department of General Surgery, Dazhou Central Hospital, Dazhou, China.
| | - Xiufang Luo
- Geriatric department, Dazhou Central Hospital, Dazhou, China.
| |
Collapse
|
2
|
Liu Y, Li J, Ding H, Ge D, Wang J, Xu C. Perfluorooctane sulfonate (PFOS) triggers migration and invasion of esophageal squamous cell carcinoma cells via regulation of Zeb1. Drug Chem Toxicol 2021; 45:2804-2813. [PMID: 34732098 DOI: 10.1080/01480545.2021.1991775] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Esophageal squamous cell carcinoma (ESCC) is one of the most prevalent and deadly cancers worldwide, especially in Eastern Asia. As a potential endocrine-disrupting chemical (EDC), perfluorooctane sulfonate (PFOS) can mimic estrogen, disturb the estrogen signals, and then cause various diseases. Although ESCC can be directly exposed to PFOS during food digestion, the effects and mechanisms of PFOS on the development of ESCC are still not well illustrated. This study showed that PFOS can promote the migration and invasion of ESCC cells. Further, PFOS treatment can increase the expression of matrix metalloproteinase-2 (MMP-2) and MMP-9, while decreasing the expression of E-Cadherin (E-Cad). Zeb1, an important transcription factor for cell motility, was essential for PFOS induced migration and invasion of ESCC cells. PFOS can increase the expression of Zeb1 via upregulation of its transcription and proteins stability. A-kinase interacting protein 1 (AKIP1) and ataxia-telangiectasia mutated (ATM) were responsible for PFOS induced transcription and proteins stability of Zeb1 in ESCC cells, respectively. Collectively, our data indicated that environmental exposure and body accumulation of PFOS might be an important risk factor for ESCC progression.
Collapse
Affiliation(s)
- Yaqing Liu
- Internal Medicine Department, The First People's Hospital of Shangqiu, Shangqiu, P. R. China.,Department of Gastroenterology, Shangqiu Clinical College, Xuzhou Medical University, Shangqiu, P. R. China
| | - Jian Li
- Department of Gastroenterology, Henan Provincial People's Hospital, Zhengzhou, P. R. China
| | - Hui Ding
- Department of Gastroenterology, Henan Provincial People's Hospital, Zhengzhou, P. R. China
| | - Dahe Ge
- Internal Medicine Department, The First People's Hospital of Shangqiu, Shangqiu, P. R. China.,Department of Gastroenterology, Shangqiu Clinical College, Xuzhou Medical University, Shangqiu, P. R. China
| | - Juntao Wang
- Internal Medicine Department, The First People's Hospital of Shangqiu, Shangqiu, P. R. China
| | - Chunjin Xu
- Internal Medicine Department, The First People's Hospital of Shangqiu, Shangqiu, P. R. China.,Department of Gastroenterology, Shangqiu Clinical College, Xuzhou Medical University, Shangqiu, P. R. China
| |
Collapse
|
3
|
de Oliveira Junior WE, Felix TF, Pires GDV, Lapa RML, Severino FE, Terra SA, Lourenção PLTDA, Dos Reis PP, Ortolan EVP. MicroRNA expression profiles in the esophagus of children with caustic stenosis: A pathway towards esophageal cancer? J Pediatr Surg 2020; 55:2144-2149. [PMID: 32111433 DOI: 10.1016/j.jpedsurg.2020.02.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 12/31/2019] [Accepted: 02/11/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Eighty percent of caustic ingestions occur in children and esophageal neoplasms may develop as a late complication of such injury. The identification of biomarkers is a promising strategy to improve early diagnosis of esophageal cancer or caustic lesions that are at an increased risk of progression. STUDY DESIGN/AIMS This study aimed at identifying global microRNA (miRNA) expression changes in esophageal mucosa from children with caustic stenosis. The study included 27 biopsy samples from 15 patients. Samples were divided into two groups, according to the time elapsed after injury (N = 15 in Group A, with less than five years of follow-up and N = 12 in Group B, with more than five years of follow-up). miRNA expression profiles were determined in each lesion, compared with normal esophageal tissues from control group. We used the TaqMan Human MicroRNA Arrays (Thermo Fisher) platform. Furthermore, bioinformatic algorithms were used to identify miRNA target genes and biological pathways including miRNAs and their target genes potentially associated with esophageal disease. RESULTS Thirteen miRNAs were significantly deregulated (9 over- and 4 underexpressed) in patients from Group A. In patients from Group B, two miRNAs were over- and two were underexpressed. Of note, miR-374 and miR-574 were deregulated in Group B patients and have been linked to esophageal tumorigenesis. We identified signal transduction and transcription factor networks with genes strongly related to development and progression of esophageal cancer. CONCLUSION miRNAs identified here contribute to a better understanding of pathways associated with malignant transformation from caustic stenosis to neoplastic lesions. This study may serve as a basis for validation of miRNAs, including miR-374 and miR-574, as potential biomarkers of early cancer detection.
Collapse
Affiliation(s)
- Wilson Elias de Oliveira Junior
- Division of Pediatric Surgery-Department of Surgery and Orthopedics, Faculty of Medicine, São Paulo State University (UNESP), Botucatu, SP, Brazil; Department of Pediatric Surgery, Barretos Children's Cancer Hospital from Barretos Cancer Center, Barretos, SP, Brazil.
| | - Tainara Francini Felix
- Department of Surgery and Orthopedics, Faculty of Medicine, São Paulo State University (UNESP), Botucatu, SP, Brazil; Experimental Research Unity (UNIPEX), Faculty of Medicine, São Paulo State University (UNESP), Botucatu, SP, Brazil
| | | | - Rainer Marco Lopez Lapa
- Experimental Research Unity (UNIPEX), Faculty of Medicine, São Paulo State University (UNESP), Botucatu, SP, Brazil; Department of Genetics, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Fábio Eduardo Severino
- Department of Surgery and Orthopedics, Faculty of Medicine, São Paulo State University (UNESP), Botucatu, SP, Brazil; Experimental Research Unity (UNIPEX), Faculty of Medicine, São Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Simone Antunes Terra
- Department of Pathology, Faculty of Medicine, São Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Pedro Luiz Toledo de Arruda Lourenção
- Division of Pediatric Surgery-Department of Surgery and Orthopedics, Faculty of Medicine, São Paulo State University (UNESP), Botucatu, SP, Brazil; Department of Surgery and Orthopedics, Faculty of Medicine, São Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Patricia Pintor Dos Reis
- Department of Surgery and Orthopedics, Faculty of Medicine, São Paulo State University (UNESP), Botucatu, SP, Brazil; Experimental Research Unity (UNIPEX), Faculty of Medicine, São Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Erika Veruska Paiva Ortolan
- Division of Pediatric Surgery-Department of Surgery and Orthopedics, Faculty of Medicine, São Paulo State University (UNESP), Botucatu, SP, Brazil; Department of Surgery and Orthopedics, Faculty of Medicine, São Paulo State University (UNESP), Botucatu, SP, Brazil
| |
Collapse
|
4
|
Esophageal Cancer Development: Crucial Clues Arising from the Extracellular Matrix. Cells 2020; 9:cells9020455. [PMID: 32079295 PMCID: PMC7072790 DOI: 10.3390/cells9020455] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 02/05/2020] [Accepted: 02/13/2020] [Indexed: 02/06/2023] Open
Abstract
In the last years, the extracellular matrix (ECM) has been reported as playing a relevant role in esophageal cancer (EC) development, with this compartment being related to several aspects of EC genesis and progression. This sounds very interesting due to the complexity of this highly incident and lethal tumor, which takes the sixth position in mortality among all tumor types worldwide. The well-established increase in ECM stiffness, which is able to trigger mechanotransduction signaling, is capable of regulating several malignant behaviors by converting alteration in ECM mechanics into cytoplasmatic biochemical signals. In this sense, it has been shown that some molecules play a key role in these events, particularly the different collagen isoforms, as well as enzymes related to its turnover, such as lysyl oxidase (LOX) and matrix metalloproteinases (MMPs). In fact, MMPs are not only involved in ECM stiffness, but also in other events related to ECM homeostasis, which includes ECM remodeling. Therefore, the crucial role of distinct MMPs isoform has already been reported, especially MMP-2, -3, -7, and -9, along EC development, thus strongly associating these proteins with the control of important cellular events during tumor progression, particularly in the process of invasion during metastasis establishment. In addition, by distinct mechanisms, a vast diversity of glycoproteins and proteoglycans, such as laminin, fibronectin, tenascin C, galectin, dermatan sulfate, and hyaluronic acid exert remarkable effects in esophageal malignant cells due to the activation of oncogenic signaling pathways mainly involved in cytoskeleton alterations during adhesion and migration processes. Finally, the wide spectrum of interactions potentially mediated by ECM may represent a singular intervention scenario in esophageal carcinogenesis natural history and, due to the scarce knowledge on the cellular and molecular mechanisms involved in EC development, the growing body of evidence on ECM’s role along esophageal carcinogenesis might provide a solid base to improve its management in the future.
Collapse
|
5
|
The Prominent Role of HMGA Proteins in the Early Management of Gastrointestinal Cancers. BIOMED RESEARCH INTERNATIONAL 2019; 2019:2059516. [PMID: 31737655 PMCID: PMC6815579 DOI: 10.1155/2019/2059516] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 09/23/2019] [Indexed: 12/24/2022]
Abstract
GI tumors represent a heterogeneous group of neoplasms concerning their natural history and molecular alterations harbored. Nevertheless, these tumors share very high incidence and mortality rates worldwide and patients' poor prognosis. Therefore, the identification of specific biomarkers could increase the development of personalized medicine, in order to improve GI cancer management. In this sense, HMGA family members (HMGA1 and HMGA2) comprise an important group of genes involved in the genesis and progression of malignant tumors. Additionally, it has also been reported that HMGA1 and HMGA2 display an important role in the detection and progression of GI tumors. In this way, HMGA family members could be used as reliable biomarkers able to efficiently track not only the tumor per se but also the main risk conditions related with their development of GI cancers in the future. Finally, it shall be a promising option to revert the current scenario, once HMGA genes and proteins could represent a convergence point in the complex landscape of GI tumors.
Collapse
|
6
|
Guan C, Liu Z, Lu C, Xiao M, Shi H, Ni R, Bian Z. Nucleolar spindle-associated protein 1 promotes tumorigenesis and predicts poor prognosis in human esophageal squamous cell carcinoma. J Cell Biochem 2019; 120:11726-11737. [PMID: 30793360 DOI: 10.1002/jcb.28452] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 01/23/2019] [Accepted: 01/24/2019] [Indexed: 01/24/2023]
Abstract
The microtubule binding protein, nucleolar spindle-associated protein 1 (NUSAP1), has a crucial function in mitosis and its expression is closely associated with carcinogenesis. Herein, we aimed to determine the function of NUSAP1 in the development of human esophageal squamous cell carcinoma (ESCC), and the association of NUSAP1 expression with ESCC. Immunohistochemical staining of ESCC tissue sections indicated that NUSAP1 was expressed to a higher degree in tumor tissues than in adjacent nontumor tissues. NUSAP1 levels were relevant closely to histological differentiation (P = 0.049). Overall survival was longer in patients with lower NUSAP1 levels ( P < 0.001). NUSAP1 expression ( P = 0.002), histological differentiation ( P < 0.001), tumor depth ( P = 0.045), lymph node metastases ( P < 0.001), and tumor-node-metastasis staging ( P = 0.008) were greatly associated with overall survival using univariate analysis. Multivariate analysis suggested that histological differentiation ( P = 0.014) and NUSAP1 expression ( P = 0.026) could be independent prognostic markers for ESCC. Additionally, the biological behavior of ESCC cells was investigated in vitro and in vivo. Suppression of NUSAP1 inhibited cellular proliferation and invasion, and induced cell cycle arrest and apoptosis in vitro. More importantly, knockdown of NUSAP1 led to inhibition of tumor formation in nude mice. These findings indicated that NUSAP1 is a potential prognostic biomarker in ESCC, and is an ESCC oncogene. Thus, NUSAP1 could represent a therapeutic target for ESCC.
Collapse
Affiliation(s)
- Chengqi Guan
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Zhaoxiu Liu
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Cuihua Lu
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Mingbing Xiao
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Hui Shi
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Runzhou Ni
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Zhaolian Bian
- Department of Gastroenterology and Hepatology, Nantong Institute of Liver Disease, Nantong Third People's Hospital Affiliated to Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
7
|
Huang H, Wei L, Qin T, Yang N, Li Z, Xu Z. Circular RNA ciRS-7 triggers the migration and invasion of esophageal squamous cell carcinoma via miR-7/KLF4 and NF-κB signals. Cancer Biol Ther 2018; 20:73-80. [PMID: 30207835 DOI: 10.1080/15384047.2018.1507254] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is one of the most prevalent and deadly cancers worldwide, especially in Eastern Asia. It has been indicated that circular RNAs (circRNA) are the key regulators in the development and progression of human cancers. We therefore evaluated the expression and regulation effects of ciRS-7 on the progression of ESCC, which is a recently identified circRNA and acts as a natural competing endogenous RNA. The expression of ciRS-7 was significantly increased in the ESCC tissues and cells as compared with their corresponding controls. In vitro study showed that ciRS-7 can promote the migration and invasion of ESCC cells. Over expression of miR-7, one of well-known targets of ciRS-7, can attenuate ciRS-7 induced invasion of ESCC cells and over expression of matrix metalloproteinase 2 (MMP2). The expression of stem cell marker Kruppel-like factor-4 (KLF-4), which has been reported as the target of miR7, increased significantly in ciRS-7 transfected ESCC cells. Knockdown of KLF-4 also attenuated over expression of ciRS-7 induced cell invasion. In addition, BAY 11-7082, the inhibitor of NF-κB, partially reversed ciRS-7 induced cell invasion. Mechanically studies indicated that ciRS-7 increased the expression of p65 via increasing the phosphorylation of IKK-α. Collectively, our present study revealed that ciRS-7 can trigger the migration and invasion of ESCC cells via miR-7/KLF4 and NF-κB signals. Targeted inhibition of ciRS-7 might be a potential approach for ESCC treatment.
Collapse
Affiliation(s)
- Hairong Huang
- a Department of Cardiothoracic Surgery, Changhai Hospital , Second Military Medical University , Shanghai , China
| | - Lei Wei
- a Department of Cardiothoracic Surgery, Changhai Hospital , Second Military Medical University , Shanghai , China
| | - Tao Qin
- b Department of Cardiothoracic Surgery, Jinling Hospital, Nanjing Clinical Medical College , Second Military Medical University , Nanjing , Jiangsu , China
| | - Nan Yang
- b Department of Cardiothoracic Surgery, Jinling Hospital, Nanjing Clinical Medical College , Second Military Medical University , Nanjing , Jiangsu , China
| | - Zhongdong Li
- b Department of Cardiothoracic Surgery, Jinling Hospital, Nanjing Clinical Medical College , Second Military Medical University , Nanjing , Jiangsu , China
| | - Zhiyun Xu
- b Department of Cardiothoracic Surgery, Jinling Hospital, Nanjing Clinical Medical College , Second Military Medical University , Nanjing , Jiangsu , China
| |
Collapse
|
8
|
UBE2C is overexpressed in ESCC tissues and its abrogation attenuates the malignant phenotype of ESCC cell lines. Oncotarget 2018; 7:65876-65887. [PMID: 27588470 PMCID: PMC5323199 DOI: 10.18632/oncotarget.11674] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Accepted: 08/13/2016] [Indexed: 11/25/2022] Open
Abstract
The esophageal squamous cell carcinoma (ESCC) is widely known as a highly lethal and poor understood cancer, then requiring the search for novel molecular markers to improve its management and patients survival. Recently, ubiquitin-conjugating enzyme E2C (UBE2C) has been figuring as a prominent tumor biomarker candidate, once it has been recognized as a key player in cell cycle progression. In this way, the aim of this study was to evaluate the expression profile of UBE2C gene and protein in ESCC samples, as well as its diagnostic and prognostic marker potential, and its contribution to ESSC genesis and/or progression by performing in vitro functional assays. The analysis of UBE2C gene expression in 52 paired ESCC samples (tumor and respective histologically normal surrounding tissue), by qRT-PCR, revealed that this gene is overexpressed in 73% of ESCC samples. Subsequently, immunohistochemical analysis confirmed that UBE2C protein expression was upregulated in all ESCC cases, but absent in the histologically normal tumor surrounding tissues. Moreover, we showed that UBE2C mRNA expression was able to accurately discriminate ESCC tissue from both healthy esophageal and histologically normal tumor surrounding tissues, pointing out its role as a diagnostic marker for this cancer. Finally, we report that UBE2C affects proliferation rates and cell cycle profile of ESCC cell lines, by directly interfering with cyclin B1 protein levels, suggesting its involvement in crucial steps of ESCC carcinogenesis.
Collapse
|
9
|
Palumbo A, Da Costa NM, Esposito F, De Martino M, D'Angelo D, de Sousa VPL, Martins I, Nasciutti LE, Fusco A, Ribeiro Pinto LF. HMGA2 overexpression plays a critical role in the progression of esophageal squamous carcinoma. Oncotarget 2017; 7:25872-84. [PMID: 27027341 PMCID: PMC5041951 DOI: 10.18632/oncotarget.8288] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 03/11/2016] [Indexed: 12/20/2022] Open
Abstract
Esophageal Squamous Cell Carcinoma (ESCC) is the most common esophageal tumor worldwide. However, there is still a lack of deeper knowledge about biological alterations involved in ESCC development. High Mobility Group A (HMGA) protein family has been related with poor outcome and malignant cell transformation in several tumor types. In this way, the aim of this study was to analyze the expression of HMGA1 and HMGA2 expression in ESCC and their role in crucial cellular features. We evaluated HMGA1 and HMGA2 mRNA expression in 52 paired ESCC and normal surrounding tissue samples by qRT-PCR. Here, we show that HMGA2, but not HMGA1, is overexpressed in ESCC samples. This result was further confirmed by the immunohistochemical analysis. Indeed, accordingly to mRNA expression data, HMGA2, but not HMGA1, was overexpressed in approximately 90% of ESCC samples, while it was barely expressed in the respective control. Conversely, HMGA1, but not HMGA2, was overexpressed in esophageal adenocarcinoma samples. Interestingly, HMGA2 abrogation attenuated the malignant phenotype of two ESCC cell lines, suggesting that HMGA2 overexpression is involved in ESCC progression.
Collapse
Affiliation(s)
- Antonio Palumbo
- Programa de Carcinogênese Molecular, Instituto Nacional de Câncer - INCA, Rio de Janeiro, RJ, Brazil.,Laboratório de Interações Celulares, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro Prédio de Ciências da Saúde - Cidade Universitária, Ilha do Fundão, Rio de Janeiro, RJ, Brazil
| | | | - Francesco Esposito
- Istituto di Endocrinologia e Oncologia Sperimentale - CNR c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli "Federico II", Naples, Italy
| | - Marco De Martino
- Istituto di Endocrinologia e Oncologia Sperimentale - CNR c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli "Federico II", Naples, Italy
| | - Daniela D'Angelo
- Istituto di Endocrinologia e Oncologia Sperimentale - CNR c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli "Federico II", Naples, Italy
| | | | - Ivanir Martins
- Divisão de Patologia, Instituto Nacional de Câncer - INCA, Rio de Janeiro, RJ, Brazil
| | - Luiz Eurico Nasciutti
- Laboratório de Interações Celulares, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro Prédio de Ciências da Saúde - Cidade Universitária, Ilha do Fundão, Rio de Janeiro, RJ, Brazil
| | - Alfredo Fusco
- Programa de Carcinogênese Molecular, Instituto Nacional de Câncer - INCA, Rio de Janeiro, RJ, Brazil.,Istituto di Endocrinologia e Oncologia Sperimentale - CNR c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli "Federico II", Naples, Italy
| | - Luis Felipe Ribeiro Pinto
- Programa de Carcinogênese Molecular, Instituto Nacional de Câncer - INCA, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
10
|
Sankalecha TH, Gupta SJ, Gaikwad NR, Shirole NU, Kothari HG. Yield of p53 expression in esophageal squamous cell cancer and its relationship with survival. Saudi J Gastroenterol 2017; 23:281-286. [PMID: 28937022 PMCID: PMC5625364 DOI: 10.4103/sjg.sjg_56_17] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND/AIMS Esophageal squamous cell carcinoma (ESCC) is the most aggressive type of cancer. Mutation of tumor suppressor gene p53 is observed in many gastrointestinal malignancies including ESCC. The immunohistochemical protein expression of mutant p53 has been proposed as a potential tool to evaluate the biological behavior of ESCC. Predictive value of p53 for survival is debatable, hence this study was formulated to know the survival of patients with p53 expression in ESCC. PATIENTS AND METHODS We prospectively included 91 consecutive patients of ESCC from August 2014 to August 2016. Biopsy specimens were treated immunohistochemically and expression of p53 gene was analyzed by Immunoreactive Score (IRS). These findings were then compared with clinicopathological parameters such as age, gender, histological grades, and TNM stages. All patients received treatment and were kept under regular follow-up. RESULTS M: F ratio was 2.03:1. p53 expression analyzed by IRS showed low expression (score ≤6) in 35 patients (38.46%) and high expression (>6) in 56 patients (61.54%). Level of p53 expression increased significantly with increasing histological grades of ESCC and TNM stage (P ≤ 0.001). Multivariate analysis shows p53 expression as independent predictor of survival. After 1 year of follow up, survival in the p53 high-expression group was 67.86% [standard error (SE) = 0.0473, confidence interval (CI) = 0.75-0.97) and in low p53 expression group was 91.43% (SE = 0.06, CI = 0.53-0.78) with statistically significant difference P = 0.0001 when analyzed with Kaplan-Meier method. CONCLUSION Expression of p53 correlates with the survival and is a simple, effective and reproducible modality to determine the prognosis and survival in ESCC.
Collapse
Affiliation(s)
- Tushar H. Sankalecha
- Department of Gastroenterology, Government Medical College and Super Speciality Hospital, Nagpur, Maharashtra, India
| | - Sudhir J. Gupta
- Department of Gastroenterology, Government Medical College and Super Speciality Hospital, Nagpur, Maharashtra, India
| | - Nitin R. Gaikwad
- Department of Gastroenterology, Government Medical College and Super Speciality Hospital, Nagpur, Maharashtra, India
| | - Nikhil U. Shirole
- Department of Gastroenterology, Government Medical College and Super Speciality Hospital, Nagpur, Maharashtra, India
| | - Harit G. Kothari
- Department of Gastroenterology, Government Medical College and Super Speciality Hospital, Nagpur, Maharashtra, India
| |
Collapse
|
11
|
Chen WX, Hong XB, Hong CQ, Liu M, Li L, Huang LS, Xu LY, Xu YW, Peng YH, Li EM. Tumor-associated autoantibodies against Fascin as a novel diagnostic biomarker for esophageal squamous cell carcinoma. Clin Res Hepatol Gastroenterol 2017; 41:327-332. [PMID: 27956255 DOI: 10.1016/j.clinre.2016.10.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2016] [Accepted: 10/29/2016] [Indexed: 02/05/2023]
Abstract
BACKGROUND AND OBJECTIVE Autoantibodies against tumor-associated antigens (TAAs) have been found in many kinds of cancers, and might serve as biomarkers for early cancer diagnosis. The present study was carried out to test if there is any relation between autoantibodies against Fascin and esophageal squamous cell carcinoma (ESCC). METHODS One hundred and forty-nine patients with ESCC and 98 control subjects were recruited in the study. The levels of circulating autoantibodies against Fascin were measured by enzyme-linked immunosorbent assay (ELISA). Receiver operating characteristic (ROC) was used to calculate diagnostic accuracy. RESULT The levels of autoantibodies against Fascin in patients with ESCC were significantly higher than in control subjects (P<0.001). Measurement of autoantibodies against Fascin provided an area under the curve (AUC) of 0.636, [95% confidence interval (CI): 0.568-0.704] 24.8% sensitivity (95% CI: 18.3%-37.2%) and 99.0% specificity (95% CI: 93.6%-99.9%). Moreover, serum level of autoantibodies against Fascin in early-stage ESCC was significantly higher than that of normal controls (P<0.05). The positive rates of autoantibodies against Fascin were correlated with age (P<0.05), but not with gender, tumor size, tumor site, histological grade, T stage, N stage or TNM stage (P>0.05). CONCLUSIONS Our results suggest that Fascin autoantibody may be a potential biomarker for the early detection of ESCC.
Collapse
Affiliation(s)
- Wen-Xia Chen
- Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Xin-Bin Hong
- Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Chao-Qun Hong
- Department of Oncological Research Lab, Cancer Hospital, Shantou University Medical College, Shantou 515041, China
| | - Ming Liu
- Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Lan Li
- Shantou University Medical College, Shantou 515041, Guangdong, China
| | - Li-Sheng Huang
- Department of Radiation Oncology, Cancer Hospital, Shantou University Medical College, Shantou 515041, China
| | - Li-Yan Xu
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, China
| | - Yi-Wei Xu
- Department of Clinical Laboratory, Cancer Hospital, Shantou University Medical College, Shantou 515041, China.
| | - Yu-Hui Peng
- Department of Clinical Laboratory, Cancer Hospital, Shantou University Medical College, Shantou 515041, China.
| | - En-Min Li
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China.
| |
Collapse
|
12
|
Palumbo Júnior A, Da Costa NM, Esposito F, Fusco A, Pinto LFR. High Mobility Group A proteins in esophageal carcinomas. Cell Cycle 2016; 15:2410-3. [PMID: 27484584 PMCID: PMC5026802 DOI: 10.1080/15384101.2016.1215388] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 07/15/2016] [Indexed: 10/21/2022] Open
Abstract
We have recently shown that HMGA2 is overexpressed in esophageal squamous cell carcinoma (ESCC) and its detection allows to discriminate between cancer and normal surrounding tissue proposing HMGA2 as a novel diagnostic marker. Interestingly, esophageal adenocarcinoma shows an opposite behavior with the overexpression of HMGA1 but not HMGA2. Moreover, we show that the suppression of HMGA2 in 2 ESCC cell lines reduces the malignant phenotype. Then, this paper highlights a differential induction of the HMGA proteins, depending on the cancer histological type, and reinforces the perspective of an innovative esophageal cancer therapy based on the suppression of the HMGA protein function and/or expression.
Collapse
Affiliation(s)
- Antonio Palumbo Júnior
- Programa de Carcinogênese Molecular, Instituto Nacional de Câncer - INCA, Rua André Cavalcanti, Rio de Janeiro, RJ, Brazil
- Laboratório de Interações Celulares, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro Prédio de Ciências da Saúde - Cidade Universitária, Ilha do Fundão, A. Carlos Chagas, Rio de Janeiro, RJ, Brasil
| | - Nathalia Meireles Da Costa
- Programa de Carcinogênese Molecular, Instituto Nacional de Câncer - INCA, Rua André Cavalcanti, Rio de Janeiro, RJ, Brazil
| | - Francesco Esposito
- Istituto di Endocrinologia e Oncologia Sperimentale - CNR c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II” - Naples, Italy
| | - Alfredo Fusco
- Istituto di Endocrinologia e Oncologia Sperimentale - CNR c/o Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II” - Naples, Italy
| | - Luis Felipe Ribeiro Pinto
- Programa de Carcinogênese Molecular, Instituto Nacional de Câncer - INCA, Rua André Cavalcanti, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
13
|
Alterations in glucose metabolism proteins responsible for the Warburg effect in esophageal squamous cell carcinoma. Exp Mol Pathol 2016; 101:66-73. [PMID: 27260309 DOI: 10.1016/j.yexmp.2016.05.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 05/14/2016] [Accepted: 05/28/2016] [Indexed: 12/14/2022]
Abstract
Esophageal squamous cell carcinoma (ESCC) is the most frequent esophageal tumor in the world. ESCC presents late diagnosis, highly aggressive behavior and poor survival. Changes in tumor cell energy metabolism appear to have a prominent role in malignant transformation. Tumor cells consume glucose avidly and produce lactic acid, even under normoxia. Among the factors that may contribute to the stimulation of glycolysis in tumor cells, there are changes in the glycolytic pathway enzymes such as: pyruvate kinase M1 and M2 (PKM2 and PKM1), hexokinase II (HKII), glucose transporter isoform 1 (GLUT-1), and transcription factor induced by hypoxia (HIF1α), responsible for the transcription of proteins cited. The objective of this study is to evaluate the alterations of these proteins and their association with clinicopathological data in ESCC. We performed immunohistochemistry to determine HIF-1α, GLUT-1, PKM1, PKM2, HK2 and Ki67-expression in ESCC patients and controls. Also, we used RT-qPCR to evaluated mRNA expression of GLUT-1 in esophageal mucosa of individuals without cancer, but are alcohol drinkers and tobacco smokers. Our results showed the exclusively expression of GLUT-1 in tumors cells and dysplastic samples. We also observed a compartmentalization of the expression of PKM1 and PKM2 in relation to tumor cells and stroma associated to tumor areas. All of the proteins evaluated, excepted GLUT-1, were frequently detected in normal mucosa. No correlations between clinicopathological features and protein expressions were observed. GLUT-1 expression appears in initial tumor lesions and is maintained through ESCC evolution. We reported for the first time PKM1 staining in normal esophagus and ESCC, being mostly present in more differentiated cells.
Collapse
|
14
|
Zeng D, Wu X, Zheng J, Zhuang Y, Chen J, Hong C, Zhang F, Wu M, Lin D. Loss of CADM1/TSLC1 Expression Is Associated with Poor Clinical Outcome in Patients with Esophageal Squamous Cell Carcinoma. Gastroenterol Res Pract 2016; 2016:6947623. [PMID: 26880895 PMCID: PMC4736003 DOI: 10.1155/2016/6947623] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 11/02/2015] [Accepted: 11/09/2015] [Indexed: 02/05/2023] Open
Abstract
Aims. We sought to determine the relationship between CADM1/TSLC1 expression and clinicopathological characteristics in patients with esophageal squamous cell carcinoma (ESCC) and the correlation with survival. Materials and Methods. Two hundred and ninety-three ESCC tissues and paired adjacent normal esophageal tissues were immunohistochemically assessed in this study. The association of CADM1/TSLC1 with clinicopathological parameters, as well as disease-free survival (DFS) and overall survival (OS), was determined based on the Kaplan-Meier method and Cox regression models. Results. CADM1/TSLC1 was detected in 236 (80.5%) tumor tissues and 19 (8.0%) paired adjacent normal esophageal tissues. Decreased CADM1/TSLC1 expression was correlated with more advanced histological grade. CADM1/TSLC1 negative tumors were more frequently observed in male cases than in female cases. DFS and OS in the CADM1/TSLC1 negative group were significantly shorter than those in the positive group, particularly in male patients with ESCC. Conclusion. Loss or reduction of CADM1/TSLC1 expression is associated with more advanced histological grade and predicts early recurrence and short survival duration. Thus, loss of CADM1/TSLC1 could be a prognostic factor that can be used to assess the risk of recurrence and survival.
Collapse
Affiliation(s)
- De Zeng
- Department of Medical Oncology, Cancer Hospital of Shantou University Medical College, No. 7 Raoping Road, Shantou 515031, China
| | - Xiao Wu
- Department of Clinical Pathology, Cancer Hospital of Shantou University Medical College, No. 7 Raoping Road, Shantou 515031, China
| | - Jin Zheng
- Department of Medical Oncology, Cancer Hospital of Shantou University Medical College, No. 7 Raoping Road, Shantou 515031, China
| | - Yixuan Zhuang
- Oncology Research Laboratory, Cancer Hospital of Shantou University Medical College, No. 7 Raoping Road, Shantou 515031, China
| | - Jiongyu Chen
- Oncology Research Laboratory, Cancer Hospital of Shantou University Medical College, No. 7 Raoping Road, Shantou 515031, China
| | - Chaoquan Hong
- Oncology Research Laboratory, Cancer Hospital of Shantou University Medical College, No. 7 Raoping Road, Shantou 515031, China
| | - Fan Zhang
- Oncology Research Laboratory, Cancer Hospital of Shantou University Medical College, No. 7 Raoping Road, Shantou 515031, China
| | - Mingyao Wu
- Department of Pathology, Shantou University Medical College, No. 22 Xinlin Road, Shantou 515041, China
| | - Danxia Lin
- Department of Medical Oncology, Cancer Hospital of Shantou University Medical College, No. 7 Raoping Road, Shantou 515031, China
- *Danxia Lin:
| |
Collapse
|