1
|
Businello G, Angerilli V, Parente P, Realdon S, Savarino E, Farinati F, Grillo F, Vanoli A, Galuppini F, Paccagnella S, Pennelli G, Mastracci L, Saragoni L, Fassan M. Molecular Landscapes of Gastric Pre-Neoplastic and Pre-Invasive Lesions. Int J Mol Sci 2021; 22:9950. [PMID: 34576114 PMCID: PMC8468646 DOI: 10.3390/ijms22189950] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/03/2021] [Accepted: 09/11/2021] [Indexed: 12/24/2022] Open
Abstract
Gastric carcinoma (GC) represents one of the most common and most lethal malignancies worldwide. The histopathological characterization of GC precursor lesions has provided great knowledge about gastric carcinogenesis, with the consequent introduction of effective strategies of primary and secondary prevention. In recent years, a large amount of data about the molecular events in GC development is emerging, flanking the histomorphological descriptions. In this review, we describe the landscape of molecular alterations in gastric pre-invasive lesions with a glance at their potential use in the diagnostic and therapeutic decision-making process.
Collapse
Affiliation(s)
- Gianluca Businello
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, 35121 Padua, Italy; (G.B.); (V.A.); (F.G.); (S.P.); (G.P.)
| | - Valentina Angerilli
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, 35121 Padua, Italy; (G.B.); (V.A.); (F.G.); (S.P.); (G.P.)
| | - Paola Parente
- Pathology Unit, Fondazione IRCCS Ospedale Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy;
| | - Stefano Realdon
- Veneto Institute of Oncology (IOV-IRCCS), 35128 Padua, Italy;
| | - Edoardo Savarino
- Division of Gastroenterology, Department of Surgery, Oncology and Gastroenterology, University of Padua, 35121 Padua, Italy; (E.S.); (F.F.)
| | - Fabio Farinati
- Division of Gastroenterology, Department of Surgery, Oncology and Gastroenterology, University of Padua, 35121 Padua, Italy; (E.S.); (F.F.)
| | - Federica Grillo
- Anatomic Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DICS), University of Genova, 16132 Genova, Italy; (F.G.); (L.M.)
- Ospedale Policlinico San Martino, IRCCS for Oncology and Neuroscience, 16132 Genova, Italy
| | - Alessandro Vanoli
- Anatomic Pathology Unit, Department of Molecular Medicine, University of Pavia and Fondazione IRCCS San Matteo Hospital, 27100 Pavia, Italy;
| | - Francesca Galuppini
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, 35121 Padua, Italy; (G.B.); (V.A.); (F.G.); (S.P.); (G.P.)
| | - Silvia Paccagnella
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, 35121 Padua, Italy; (G.B.); (V.A.); (F.G.); (S.P.); (G.P.)
| | - Gianmaria Pennelli
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, 35121 Padua, Italy; (G.B.); (V.A.); (F.G.); (S.P.); (G.P.)
| | - Luca Mastracci
- Anatomic Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DICS), University of Genova, 16132 Genova, Italy; (F.G.); (L.M.)
- Ospedale Policlinico San Martino, IRCCS for Oncology and Neuroscience, 16132 Genova, Italy
| | - Luca Saragoni
- UO Anatomia Patologica, Ospedale G.B. Morgagni-L. Pierantoni, 47121 Forlì, Italy;
| | - Matteo Fassan
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, 35121 Padua, Italy; (G.B.); (V.A.); (F.G.); (S.P.); (G.P.)
- Veneto Institute of Oncology (IOV-IRCCS), 35128 Padua, Italy;
| |
Collapse
|
2
|
Duong T, Vo DN, Nakayama T, Mukaisho KI, Bamba M, Nguyen T, Sugihara H. Rapidly and Slowly Growing Lineages in Chromosomal Instability-Type Gland-Forming Gastric Carcinomas as Revealed by Multisampling Analysis of DNA Copy-Number Profile. Pathobiology 2019; 86:118-127. [DOI: 10.1159/000494926] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Accepted: 10/28/2018] [Indexed: 11/19/2022] Open
|
3
|
Mizuguchi A, Takai A, Shimizu T, Matsumoto T, Kumagai K, Miyamoto S, Seno H, Marusawa H. Genetic features of multicentric/multifocal intramucosal gastric carcinoma. Int J Cancer 2018; 143:1923-1934. [DOI: 10.1002/ijc.31578] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 04/06/2018] [Accepted: 04/18/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Aya Mizuguchi
- Department of Gastroenterology and HepatologyGraduate School of Medicine, Kyoto UniversityKyoto Japan
| | - Atsushi Takai
- Department of Gastroenterology and HepatologyGraduate School of Medicine, Kyoto UniversityKyoto Japan
| | - Takahiro Shimizu
- Department of Gastroenterology and HepatologyGraduate School of Medicine, Kyoto UniversityKyoto Japan
| | - Tomonori Matsumoto
- Department of Gastroenterology and HepatologyGraduate School of Medicine, Kyoto UniversityKyoto Japan
| | - Ken Kumagai
- Department of Gastroenterology and HepatologyGraduate School of Medicine, Kyoto UniversityKyoto Japan
| | - Shin'ichi Miyamoto
- Department of Gastroenterology and HepatologyGraduate School of Medicine, Kyoto UniversityKyoto Japan
| | - Hiroshi Seno
- Department of Gastroenterology and HepatologyGraduate School of Medicine, Kyoto UniversityKyoto Japan
| | - Hiroyuki Marusawa
- Department of Gastroenterology and HepatologyGraduate School of Medicine, Kyoto UniversityKyoto Japan
| |
Collapse
|
4
|
Ichida K, Suzuki K, Fukui T, Takayama Y, Kakizawa N, Watanabe F, Ishikawa H, Muto Y, Kato T, Saito M, Futsuhara K, Miyakura Y, Noda H, Ohmori T, Konishi F, Rikiyama T. Overexpression of satellite alpha transcripts leads to chromosomal instability via segregation errors at specific chromosomes. Int J Oncol 2018; 52:1685-1693. [PMID: 29568894 DOI: 10.3892/ijo.2018.4321] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 02/22/2018] [Indexed: 11/05/2022] Open
Abstract
The impairment of the stability of the chromosomal structure facilitates the abnormal segregation of chromosomes, thus increasing the risk of carcinogenesis. Chromosomal stability during segregation is managed by appropriate methylation at the centromere of chromosomes. Insufficient methylation, or hypomethylation, results in chromosomal instability. The centromere consists of satellite alpha repetitive sequences, which are ideal targets for DNA hypomethylation, resulting in the overexpression of satellite alpha transcript (SAT). The overexpression of SAT has been reported to induce the abnormal segregation of chromosomes. In this study, we verified the oncogenic pathway via chromosomal instability involving DNA hypomethylation and the overexpression of SAT. For this purpose, we constructed lentiviral vectors expressing SAT and control viruses and then infected human mammary epithelial cells with these vectors. The copy number alterations and segregation errors of chromosomes were evaluated by microarray-based comparative genomic hybridization (array CGH) and immunocytochemistry, respectively. The levels of hypomethylation of satellite alpha sequences were determined by MethyLight polymerase chain reaction. Clinical specimens from 45 patients with breast cancer were recruited to verify the data in vitro. The results of immunocytochemistry revealed that the incidence of segregation errors was significantly higher in the cells overexpressing SAT than in the controls. An array CGH identified the specific chromosomes of 8q and 20q as frequent sites of copy number alterations in cells with SAT overexpression, although no such sites were noted in the controls, which was consistent with the data from clinical specimens. A regression analysis revealed that the expression of SAT was significantly associated with the levels of hypomethylation of satellite alpha sequences. On the whole, the overexpression of SAT led to chromosomal instability via segregation errors at specific chromosomes in connection with DNA hypomethylation, which was also recognized in clinical specimens of patients with breast cancer. Thus, this oncogenic pathway may be involved in the development of breast cancer.
Collapse
Affiliation(s)
- Kosuke Ichida
- Department of Surgery, Saitama Medical Center, Jichi Medical University, Saitama-shi, Saitama 330-8503, Japan
| | - Koichi Suzuki
- Department of Surgery, Saitama Medical Center, Jichi Medical University, Saitama-shi, Saitama 330-8503, Japan
| | - Taro Fukui
- Department of Surgery, Saitama Medical Center, Jichi Medical University, Saitama-shi, Saitama 330-8503, Japan
| | - Yuji Takayama
- Department of Surgery, Saitama Medical Center, Jichi Medical University, Saitama-shi, Saitama 330-8503, Japan
| | - Nao Kakizawa
- Department of Surgery, Saitama Medical Center, Jichi Medical University, Saitama-shi, Saitama 330-8503, Japan
| | - Fumiaki Watanabe
- Department of Surgery, Saitama Medical Center, Jichi Medical University, Saitama-shi, Saitama 330-8503, Japan
| | - Hideki Ishikawa
- Department of Surgery, Saitama Medical Center, Jichi Medical University, Saitama-shi, Saitama 330-8503, Japan
| | - Yuta Muto
- Department of Surgery, Saitama Medical Center, Jichi Medical University, Saitama-shi, Saitama 330-8503, Japan
| | - Takaharu Kato
- Department of Surgery, Saitama Medical Center, Jichi Medical University, Saitama-shi, Saitama 330-8503, Japan
| | - Masaaki Saito
- Department of Surgery, Saitama Medical Center, Jichi Medical University, Saitama-shi, Saitama 330-8503, Japan
| | - Kazushige Futsuhara
- Department of Surgery, Saitama Medical Center, Jichi Medical University, Saitama-shi, Saitama 330-8503, Japan
| | - Yasuyuki Miyakura
- Department of Surgery, Saitama Medical Center, Jichi Medical University, Saitama-shi, Saitama 330-8503, Japan
| | - Hiroshi Noda
- Department of Surgery, Saitama Medical Center, Jichi Medical University, Saitama-shi, Saitama 330-8503, Japan
| | - Tsukasa Ohmori
- Department of Biochemistry, Jichi Medical University, Shimotsuke-shi, Tochigi 329-0498, Japan
| | | | - Toshiki Rikiyama
- Department of Surgery, Saitama Medical Center, Jichi Medical University, Saitama-shi, Saitama 330-8503, Japan
| |
Collapse
|
5
|
Notch1 pathway-mediated microRNA-151-5p promotes gastric cancer progression. Oncotarget 2018; 7:38036-38051. [PMID: 27191259 PMCID: PMC5122370 DOI: 10.18632/oncotarget.9342] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 04/29/2016] [Indexed: 12/20/2022] Open
Abstract
Gastric carcinoma is the third leading cause of lethal cancer worldwide. Previous studies showed that Notch1 receptor intracellular domain (N1IC), the activated form of Notch1 receptor, promotes gastric cancer progression. It has been demonstrated that a significant cross-talk interplays between Notch pathways and microRNAs (miRNAs) in controlling tumorigenesis. This study identified an intronic microRNA-151 (miR-151), which consists of two mature miRNAs, miR-151-3p and miR-151-5p, as a Notch1 receptor-induced miRNA in gastric cancer cells. Activation of Notch1 pathway enhanced expressions of miR-151 and its host gene, focal adhesion kinase (FAK), in gastric cancer cells. The levels of miR-151 in gastric cancer samples were higher than those of adjacent non-tumor samples. Activated Notch1 pathway induced CBF1-dependent FAK promoter activity. The ectopic expression of miR-151 promoted growth and progression of SC-M1 gastric cancer cells including cell viability and colony formation, migration, and invasion abilities. Activated Notch1 pathway could augment progression of gastric cancer cells through miR-151-5p and FAK. The mRNA levels of pluripotency genes, Nanog and SOX-2, tumorsphere formation ability, tumor growth, and lung metastasis of SC-M1 cells were elevated by activated Notch1 pathway through miR-151-5p. Furthermore, miR-151-5p could target 3′-untranslated region (3′-UTR) of p53 mRNA and down-regulate p53 level in SC-M1 cells. Mechanistically, Notch1/miR-151-5p axis contributed to progression of SC-M1 cells through down-regulation of p53 which in turn repressed FAK promoter activity. Taken together, these results suggest that Notch1 pathway and miR-151-5p interplay with p53 in a reciprocal regulation loop in controlling gastric carcinogenesis.
Collapse
|
6
|
Birkness JE, Spada NG, Miller C, Luketich JD, Nason KS, Sun W, Davison JM. Extreme chromosome 17 copy number instability is a prognostic factor in patients with gastroesophageal adenocarcinoma: A retrospective cohort study. Genes Chromosomes Cancer 2018; 57:28-34. [PMID: 28913947 DOI: 10.1002/gcc.22504] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Revised: 09/09/2017] [Accepted: 09/11/2017] [Indexed: 12/30/2022] Open
Abstract
Gastric and esophageal cancers frequently show genomic instability and aneuploidy. Chromosomal copy number instability (CIN) is a form of genomic instability that exerts pleiotropic effects on cellular biology and is a source of genetic heterogeneity in a population of cells. CIN results in cell-to-cell variation in chromosome copy number which can be detected and quantified by fluorescence in situ hybridization (FISH). CIN is a biomarker associated with differential response to a number of chemotherapy compounds. We quantified chromosome 17 copy number instability (CIN-17) in 348 gastroesophageal adenocarcinomas by centromeric FISH in cases that were tested for HER2 amplification. We evaluated the association between CIN-17 and clinical outcome after surgical and nonsurgical treatment. CIN-17 was detected in 45.4% (158/348) and extreme CIN-17 in 28.4% (99/348). Extreme CIN-17 had no association with outcome in surgically treated patients. However, in patients treated with conventional radiation and/or chemotherapy, extreme CIN-17 was associated with 55% reduction in overall mortality (hazard ratio, 0.448; 95% confidence interval, 0.263-0.763) after adjusting for age and clinical stage at diagnosis. Extreme CIN-17 is detected in over a quarter of gastroesophageal adenocarcinomas and is a favorable prognostic marker in patients treated nonoperatively.
Collapse
Affiliation(s)
| | - Neal G Spada
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Caitlyn Miller
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - James D Luketich
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Katie S Nason
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Weijing Sun
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Jon M Davison
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
7
|
Abstract
Helicobacter pylori infection is the most important cause of human gastric cancer worldwide. Gastric cancer develops over a long time after H. pylori infection via stepwise accumulation of genetic alterations and positive selection of cells with growth advantages. H. pylori itself and the resultant chronic inflammation lead to the emergence of genetic alterations in gastric epithelial cells via increased susceptibility of these cells to DNA damage. Reactive oxygen species (ROS) and reactive nitrogen species (RNS) in inflammatory and gastric epithelial cells, as well as the expression of cytidine deaminase in gastric epithelial cells, may link H. pylori-related inflammation and DNA damage. Recent comprehensive analyses of gastric cancer genomes provide clues for the possible molecular mechanisms of gastric carcinogenesis. In this chapter, we describe how genetic alterations emerge during gastric carcinogenesis related to H. pylori infection.
Collapse
|
8
|
Min BH, Hwang J, Kim NKD, Park G, Kang SY, Ahn S, Ahn S, Ha SY, Lee YK, Kushima R, Van Vrancken M, Kim MJ, Park C, Park HY, Chae J, Jang SS, Kim SJ, Kim YH, Kim JI, Kim KM. Dysregulated Wnt signalling and recurrent mutations of the tumour suppressorRNF43in early gastric carcinogenesis. J Pathol 2016; 240:304-314. [DOI: 10.1002/path.4777] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 07/06/2016] [Accepted: 08/03/2016] [Indexed: 12/15/2022]
Affiliation(s)
- Byung-Hoon Min
- Department of Medicine, Samsung Medical Centre; Sungkyunkwan University School of Medicine; Seoul Korea
| | - Jinha Hwang
- Department of Biomedical Science; Seoul National University Graduate School; Seoul Korea
| | - Nayoung KD Kim
- Samsung Genome Institute; Samsung Medical Centre; Seoul Korea
| | - Gibeom Park
- Department of Biomedical Science; Seoul National University Graduate School; Seoul Korea
| | - So Young Kang
- Department of Pathology and Translational Genomics, Samsung Medical Centre; Sungkyunkwan University School of Medicine; Seoul Korea
| | - Sangjeong Ahn
- Department of Pathology and Translational Genomics, Samsung Medical Centre; Sungkyunkwan University School of Medicine; Seoul Korea
| | - Soomin Ahn
- Department of Pathology and Translational Genomics, Samsung Medical Centre; Sungkyunkwan University School of Medicine; Seoul Korea
| | - Sang Yun Ha
- Department of Pathology and Translational Genomics, Samsung Medical Centre; Sungkyunkwan University School of Medicine; Seoul Korea
| | - Yun Kyung Lee
- Department of Pathology and Translational Genomics, Samsung Medical Centre; Sungkyunkwan University School of Medicine; Seoul Korea
| | - Ryoji Kushima
- Department of Pathology, Undergraduate School of Medicine; Shiga University of Medical Science; Shiga Japan
| | - Michael Van Vrancken
- Department of Pathology and Laboratory Medicine; Tulane University School of Medicine; New Orleans LA USA
| | - Min Jung Kim
- Cancer Research Institute; Seoul National University College of Medicine; Seoul Korea
| | - Changho Park
- Department of Biomedical Science; Seoul National University Graduate School; Seoul Korea
| | - Ha Young Park
- Department of Biomedical Science; Seoul National University Graduate School; Seoul Korea
- Department of Pathology and Translational Genomics, Samsung Medical Centre; Sungkyunkwan University School of Medicine; Seoul Korea
| | - Jeesoo Chae
- Department of Biomedical Science; Seoul National University Graduate School; Seoul Korea
| | - Se Song Jang
- Department of Biomedical Science; Seoul National University Graduate School; Seoul Korea
| | - Sung Jin Kim
- Samsung Biomedical Research Institute; Samsung Medical Centre; Seoul Korea
| | - Young-Ho Kim
- Department of Medicine, Samsung Medical Centre; Sungkyunkwan University School of Medicine; Seoul Korea
| | - Jong-Il Kim
- Department of Biomedical Science; Seoul National University Graduate School; Seoul Korea
- Cancer Research Institute; Seoul National University College of Medicine; Seoul Korea
- Genomic Medicine Institute, Medical Research Centre; Seoul National University; Seoul Korea
| | - Kyoung-Mee Kim
- Department of Pathology and Translational Genomics, Samsung Medical Centre; Sungkyunkwan University School of Medicine; Seoul Korea
| |
Collapse
|
9
|
Recurrent amplification of MYC and TNFRSF11B in 8q24 is associated with poor survival in patients with gastric cancer. Gastric Cancer 2016; 19:116-27. [PMID: 25618371 DOI: 10.1007/s10120-015-0467-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 01/12/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND Gastric cancer (GC) is an aggressive malignancy whose mechanisms of development and progression are poorly understood. The identification of prognosis-related genomic loci and genes may suffer from the relatively small case numbers and a lack of systematic validation in previous studies. METHODS Array-based comparative genomic hybridization (aCGH) coupled with patient clinical information was applied to identify prognosis-related loci and genes with high-frequency recurrent gains in 129 GC patients. The candidate loci and genes were then validated using an independent cohort of 384 patients through branched DNA signal amplification analysis (QuantiGene assays). RESULTS In the 129 patients, a copy number gain of three chromosome regions-namely, 8q22 (including ESRP1 and CCNE2), 8q24 (including MYC and TNFRSF11B), and 20q11-q13 (including SRC, MMP9, and CSE1L)--conferred poor survival for patients. In addition, the correlation between the branched DNA signal amplification analysis results and the aCGH results was analyzed in 73 of these 129 patients, and MYC, TNFRSF11B, ESRP1, CSE1L, and MMP9 were found to be well correlated. Further validation using an independent cohort (n = 384) verified that only MYC and TNFRSF11B within 8q24 are related to survival. Patients with gains in both MYC and TNFRSF11B had poorer survival than those with no gains, particularly those with noncardia GC. Gains in both of these genes were also a significant independent prognostic indicator. CONCLUSIONS Our results revealed that copy number gains in MYC and TNFRSF11B located at 8q24 are associated with survival in GC, particularly noncardia GC.
Collapse
|
10
|
Abstract
Helicobacter pylori infection plays a crucial role in gastric carcinogenesis. H pylori exerts oncogenic effects on gastric mucosa through complex interaction between bacterial virulence factors and host inflammatory responses. On the other hand, gastric cancer develops via stepwise accumulation of genetic and epigenetic alterations in H pylori-infected gastric mucosa. Recent comprehensive analyses of gastric cancer genomes indicate a multistep process of genetic alterations as well as possible molecular mechanisms of gastric carcinogenesis. Both genetic processes of gastric cancer development and molecular oncogenic pathways related to H pylori infection are important to completely understand the pathogenesis of H pylori-related gastric cancer.
Collapse
|
11
|
Wei H, Li Z, Wang X, Wang J, Pang W, Yang G, Shen QW. microRNA-151-3p regulates slow muscle gene expression by targeting ATP2a2 in skeletal muscle cells. J Cell Physiol 2015; 230:1003-12. [PMID: 25200835 DOI: 10.1002/jcp.24793] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 09/05/2014] [Indexed: 12/21/2022]
Abstract
MicroRNAs (miRNAs) are a group of small noncoding RNAs that regulate the stability or translation of cognate mRNAs at the post-transcriptional level. Accumulating evidence indicates that miRNAs play important roles in many aspects of muscle function, including muscle growth and development, regeneration, contractility, and muscle fiber type plasticity. In the current study, we examined the function of miR-151-3p in myoblast proliferation and differentiation. Results show that overexpression of miR-151-3p not only upregulates myoblast proliferation, but also decreases slow muscle gene expression (such as MHC-β/slow and slow muscle troponin I) in both C2C12 myotubes and in primary cultures. Alternatively, inhibition of miR-151-3p by antisense RNA was found to upregulate MHC-β/slow expression, indicating that miR-151-3p plays a role in muscle fiber type determination. Further investigation into the underlying mechanisms revealed for the first time that miR-151-3p directly targets ATP2a2, a gene encoding for a slow skeletal and cardiac muscle specific Ca(2+) ATPase, SERCA2 thus downregulating slow muscle gene expression. Mechanisms by which the alteration in SERCA2 expression induces changes in other slow muscle gene expression levels needs to be defined in future research.
Collapse
Affiliation(s)
- Huan Wei
- Department of Animal Science, Northwest A&F University, Yangling, Shaanxi, China
| | | | | | | | | | | | | |
Collapse
|
12
|
Vo DTN, Nakayama T, Yamamoto H, Mukaisho KI, Hattori T, Sugihara H. Progression risk assessments of individual non-invasive gastric neoplasms by genomic copy-number profile and mucin phenotype. BMC Med Genomics 2015; 8:6. [PMID: 25881098 PMCID: PMC4346124 DOI: 10.1186/s12920-015-0080-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2014] [Accepted: 02/03/2015] [Indexed: 12/18/2022] Open
Abstract
Background Early detection and treatment of non-invasive neoplasms can effectively reduce the incidence of advanced gastric carcinoma (GC), but only when the lineage is continuous between non-invasive and advanced tumours. Although a fraction of non-invasive neoplasms progress to invasive GC, it is difficult to identify individual progression-prone non-invasive neoplasms. To classify non-invasive gland-forming gastric neoplasms into clusters of different levels of progression risk, we applied mucin phenotyping and genomic DNA microarray analyses to intramucosal gland-forming gastric neoplasms. Methods Formalin-fixed, paraffin-embedded tissues from 19 non-invasive and 24 invasive gland-forming neoplasms were obtained via endoscopic submucosal dissection or surgical excision. According to the Vienna classification, intramucosal neoplasms were classified as low-grade or high-grade non-invasive neoplasms (LGNs [category 3] and HGNs [category 4], respectively) or invasive carcinomas (intramucosal GCs and mucosal parts of submucosal or deeper GCs [category 5]). Neoplastic lesions were characterized by mucin phenotypes determined using monoclonal antibodies against MUC2, MUC5AC, MUC6, and CD10. Genomic DNA samples from mucosal neoplasms were subjected to array-based comparative genomic hybridization and subsequent unsupervised, hierarchical clustering with selected large-sized genes. Results There was no significant difference in mucin phenotype between HGNs/LGNs and invasive carcinomas. The clustering classified samples into stable, unstable, and intermediate. The histological tumour grade or mucin phenotype of non-invasive neoplasms did not correlate with the clustering results. Each cluster may represent an independent lineage of different outcome because the size distribution of non-invasive tumours among the 3 clusters almost overlapped. In contrast, the unstable cluster alone included invasive carcinomas. Conclusions These findings suggest that the outcome of individual tumours is not stochastically determined but can be predicted from the genomic copy-number profile even at the non-invasive stage. Non-invasive neoplasms of the unstable clusters, which accounted for 21% of non-invasive neoplasms, may progress to invasive carcinomas, whereas those of stable cluster may not. Electronic supplementary material The online version of this article (doi:10.1186/s12920-015-0080-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Diem Thi-Ngoc Vo
- Department of Pathology, Division of Molecular and Diagnostic Pathology, Shiga University of Medical Science, Otsu, 520-2192, Japan.
| | - Takahisa Nakayama
- Department of Pathology, Division of Molecular and Diagnostic Pathology, Shiga University of Medical Science, Otsu, 520-2192, Japan.
| | - Hiroto Yamamoto
- Department of Pathology, Division of Molecular and Diagnostic Pathology, Shiga University of Medical Science, Otsu, 520-2192, Japan.
| | - Ken-ichi Mukaisho
- Department of Pathology, Division of Molecular and Diagnostic Pathology, Shiga University of Medical Science, Otsu, 520-2192, Japan.
| | - Takanori Hattori
- Department of Pathology, Division of Molecular and Diagnostic Pathology, Shiga University of Medical Science, Otsu, 520-2192, Japan.
| | - Hiroyuki Sugihara
- Department of Pathology, Division of Molecular and Diagnostic Pathology, Shiga University of Medical Science, Otsu, 520-2192, Japan.
| |
Collapse
|
13
|
Xu X, Feng L, Liu Y, Zhou WX, Ma YC, Fei GJ, An N, Li Y, Wu X, Yao F, Cheng SJ, Lu XH. Differential gene expression profiling of gastric intraepithelial neoplasia and early-stage adenocarcinoma. World J Gastroenterol 2014; 20:17883-17893. [PMID: 25548486 PMCID: PMC4273138 DOI: 10.3748/wjg.v20.i47.17883] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Revised: 06/05/2014] [Accepted: 06/17/2014] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the differentiated whole genome expression profiling of gastric high- and low-grade intraepithelial neoplasia and early-stage adenocarcinoma.
METHODS: Gastric specimens from an upper magnifying chromoendoscopic targeted biopsy were collected from March 2010 to May 2013. Whole genome expression profiling was performed on 19 low-grade intraepithelial neoplasia (LGIN), 20 high-grade intraepithelial neoplasia (HGIN), 19 early-stage adenocarcinoma (EGC), and 19 chronic gastritis tissue samples using Agilent 4 × 44K Whole Human Genome microarrays. Differentially expressed genes between different types of lesions were identified using an unpaired t-test and corrected with the Benjamini and Hochberg false discovery rate algorithm. A gene ontology (GO) enrichment analysis was performed using the GeneSpring software GX 12.6. The differentially expressed gene was verified using a real-time TaqMan® PCR assay with independent tissue samples, including 26 LGIN, 15 HGIN, 14 EGC, and 20 chronic gastritis. The expression of G0S2 were further validated by immunohistochemical staining (IHC) in 24 LGIN, 40 HGIN, 30 EGC and 61 chronic gastritis specimens.
RESULTS: The gene expression patterns of LGIN and HGIN tissues were distinct. There were 2521 significantly differentially expressed transcripts in HGIN, with 951 upregulated and 1570 downregulated. A GO enrichment analysis demonstrated that the most striking overexpressed transcripts in HGIN compared with LGIN were in the category of metabolism, defense response, and nuclear factor κB (NF-κB) cascade. While the vast majority of transcripts had barely altered expression in HGIN and EGC tissues, only 38 transcripts were upregulated in EGC. A GO enrichment analysis revealed that the alterations of the immune response were most prominent in the progression from HGIN to EGC. It is worth noting that, compared with LGIN, 289 transcripts were expressed at higher levels both in HGIN and EGC. A characteristic gene, G0/G1 switch 2 (G0S2) was one of the 289 transcripts and related to metabolism, the immune response, and the NF-κB cascade, and its expression was validated in independent samples through real-time TaqMan® PCR and immunohistochemical staining. In real-time PCR analysis, the expression of G0S2 was elevated both in HGIN and EGC compared with that in LGIN (P < 0.01 and P < 0.001, respectively). In IHC analysis, G0S2 immunoreactivity was detected in the cytoplasmic of neoplastic cells, but was undetectable in chronic gastritis cells. The G0S2 expression in HGIN was higher than that of LGIN (P = 0.012, χ2 = 6.28) and EGC (P = 0.008, χ2 = 6.94).
CONCLUSION: A clear biological distinction between gastric high- and low-grade intraepithelial neoplasia was identified, and provides molecular evidence for clinical application.
Collapse
|
14
|
Tsai PC, Huang SW, Tsai HL, Ma CJ, Hou MF, Yang IP, Wang YS, Juo SHH, Wang JY. The association between DNA copy number aberrations at chromosome 5q22 and gastric cancer. PLoS One 2014; 9:e106624. [PMID: 25210923 PMCID: PMC4161348 DOI: 10.1371/journal.pone.0106624] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 07/30/2014] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Gastric cancer is common cancer. Discovering novel genetic biomarkers might help to identify high-risk individuals. Copy number variation (CNV) has recently been shown to influence risk for several cancers. The aim of the present study was sought to test the association between copy number at a variant region and GC. METHODS A total of 110 gastric cancer patients and 325 healthy volunteers were enrolled in this study. We searched for a CNV and found a CNV (Variation 7468) containing part of the APC gene, the SRP19 gene and the REEP5 gene. We chose four probes targeting at APC-intron8, APC-exon9, SRP19 and REEP5 to interrogate this CNV. Specific Taqman probes labeled by different reporter fluorophores were used in a real-time PCR platform to obtain copy number. Both the original non-integer data and transformed integer data on copy number were used for analyses. RESULTS Gastric caner patients had a lower non-integer copy number than controls for the APC-exon9 probe (Adjusted p = 0.026) and SRP19 probe (Adjusted p = 0.002). The analysis of integer copy number yielded a similar pattern although less significant (Adjusted p = 0.07 for APC-exon9 probe and Adjusted p = 0.02 for SRP19 probe). CONCLUSIONS Losses of a CNV at 5q22, especially in the DNA region surrounding APC-exon 9, may be associated with a higher risk of gastric cancer.
Collapse
Affiliation(s)
- Pei-Chien Tsai
- Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Szu-Wei Huang
- Department of Medical Genetics, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hsiang-Lin Tsai
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Graduate Institute of Clinical Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Cheng-Jen Ma
- Graduate Institute of Clinical Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Division of Gastrointestinal and General Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Ming-Feng Hou
- Graduate Institute of Clinical Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Division of Gastrointestinal and General Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - I-Ping Yang
- Department of Nursing, Shu-Zen College of Medicine and Management, Kaohsiung, Taiwan
| | - Yung-Song Wang
- Department of Medical Genetics, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Suh-Hang Hank Juo
- Department of Medical Genetics, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Graduate Institute of Clinical Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jaw-Yuan Wang
- Department of Medical Genetics, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Graduate Institute of Clinical Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Division of Gastrointestinal and General Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| |
Collapse
|
15
|
Jouni H, Shameer K, Asmann YW, Hazin R, de Andrade M, Kullo IJ. Clinical Correlates of Autosomal Chromosomal Abnormalities in an Electronic Medical Record-Linked Genome-Wide Association Study: A Case Series. J Investig Med High Impact Case Rep 2013; 1:2324709613508932. [PMID: 26425586 PMCID: PMC4528839 DOI: 10.1177/2324709613508932] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Although mosaic autosomal chromosomal abnormalities are being increasingly detected as part of high-density genotyping studies, the clinical correlates are unclear. From an electronic medical record (EMR)–based genome-wide association study (GWAS) of peripheral arterial disease, log-R-ratio and B-allele-frequency data were used to identify mosaic autosomal chromosomal abnormalities including copy number variation and loss of heterozygosity. The EMRs of patients with chromosomal abnormalities and those without chromosomal abnormalities were reviewed to compare clinical characteristics. Among 3336 study participants, 0.75% (n = 25, mean age = 74.8 ± 10.7 years, 64% men) had abnormal intensity plots indicative of autosomal chromosomal abnormalities. A hematologic malignancy was present in 8 patients (32%), of whom 4 also had a solid organ malignancy while 2 patients had a solid organ malignancy only. In 50 age- and sex-matched participants without chromosomal abnormalities, there was a lower rate of hematologic malignancies (2% vs 32%, P < .001) but not solid organ malignancies (20% vs 24%, P = .69). We also report the clinical characteristics of each patient with the observed chromosomal abnormalities. Interestingly, among 5 patients with 20q deletions, 4 had a myeloproliferative disorder while all 3 men in this group had prostate cancer. In summary, in a GWAS of 3336 adults, 0.75% had autosomal chromosomal abnormalities and nearly a third of them had hematologic malignancies. A potential novel association between 20q deletions, myeloproliferative disorders, and prostate cancer was also noted.
Collapse
Affiliation(s)
- Hayan Jouni
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN
| | - Khader Shameer
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN
| | - Yan W Asmann
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN
| | - Ribhi Hazin
- Department of Internal Medicine, Wayne State University, Detroit, MI
| | - Mariza de Andrade
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN
| | | |
Collapse
|
16
|
Abstract
MOTIVATION Data quality is a critical issue in the analyses of DNA copy number alterations obtained from microarrays. It is commonly assumed that copy number alteration data can be modeled as piecewise constant and the measurement errors of different probes are independent. However, these assumptions do not always hold in practice. In some published datasets, we find that measurement errors are highly correlated between probes that interrogate nearby genomic loci, and the piecewise-constant model does not fit the data well. The correlated errors cause problems in downstream analysis, leading to a large number of DNA segments falsely identified as having copy number gains and losses. METHOD We developed a simple tool, called autocorrelation scanning profile, to assess the dependence of measurement error between neighboring probes. RESULTS Autocorrelation scanning profile can be used to check data quality and refine the analysis of DNA copy number data, which we demonstrate in some typical datasets. CONTACT lzhangli@mdanderson.org. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Liangcai Zhang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77230, USA and Department of Biophysics, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | | |
Collapse
|
17
|
Abstract
The development of gastric adenocarcinoma is a complex multistep process involving multiple genetic alterations. Based on pathology, four different macroscopic types and at least two major histological types, intestinal and diffuse, have been described. Most gastric cancer (GC) show genetic instability, either microsatellite instability or chromosomal instability, which is considered an early event in gastric carcinogenesis. Molecular studies of alterations of single genes have provided evidence that intestinal and diffuse type GC evolve via different genetic pathways. Recent results from high-throughput whole-genome expression or copy number studies have demonstrated extensive genetic diversity between cases and within individual GC. Sets of commonly up- or downregulated microRNAs have been identified in GC and might be useful in the near future to identify pathways of GC progression. Results from detailed molecular and/or pathological GC studies, although promising, still have limited clinical utility in predicting survival and stratifying GC patients for appropriate treatment.
Collapse
Affiliation(s)
- Heike I Grabsch
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds LS9 7TF, UK.
| | | |
Collapse
|
18
|
Chiyomaru T, Yamamura S, Zaman MS, Majid S, Deng G, Shahryari V, Saini S, Hirata H, Ueno K, Chang I, Tanaka Y, Tabatabai ZL, Enokida H, Nakagawa M, Dahiya R. Genistein suppresses prostate cancer growth through inhibition of oncogenic microRNA-151. PLoS One 2012; 7:e43812. [PMID: 22928040 PMCID: PMC3426544 DOI: 10.1371/journal.pone.0043812] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Accepted: 07/26/2012] [Indexed: 01/12/2023] Open
Abstract
Genistein has been shown to suppress the growth of several cancers through modulation of various pathways. However, the effects of genistein on the regulation of oncogenic microRNA-151 (miR-151) have not been reported. In this study, we investigated whether genistein could alter the expression of oncogenic miR-151 and its target genes that are involved in the progression and metastasis of prostate cancer (PCa). Real-time RT-PCR showed that the expression of miR-151 was higher in PC3 and DU145 cells compared with RWPE-1 cells. Treatment of PC3 and DU145 cells with 25 µM genistein down-regulated the expression of miR-151 compared with vehicle control. Inhibition of miR-151 in PCa cells by genistein significantly inhibited cell migration and invasion. In-silico analysis showed that several genes (CASZ1, IL1RAPL1, SOX17, N4BP1 and ARHGDIA) suggested to have tumor suppressive functions were target genes of miR-151. Luciferase reporter assays indicated that miR-151 directly binds to specific sites on the 3′UTR of target genes. Quantitative real-time PCR analysis showed that the mRNA expression levels of the five target genes in PC3 and DU145 were markedly changed with miR-151 mimics and inhibitor. Kaplan-Meier curves and log-rank tests revealed that high expression levels of miR-151 had an adverse effect on survival rate. This study suggests that genistein mediated suppression of oncogenic miRNAs can be an important dietary therapeutic strategy for the treatment of PCa.
Collapse
Affiliation(s)
- Takeshi Chiyomaru
- Department of Urology, San Francisco Veterans Affairs Medical Center and University of California San Francisco, San Francisco, California, United States of America
| | - Soichiro Yamamura
- Department of Urology, San Francisco Veterans Affairs Medical Center and University of California San Francisco, San Francisco, California, United States of America
| | - Mohd Saif Zaman
- Department of Urology, San Francisco Veterans Affairs Medical Center and University of California San Francisco, San Francisco, California, United States of America
| | - Shahana Majid
- Department of Urology, San Francisco Veterans Affairs Medical Center and University of California San Francisco, San Francisco, California, United States of America
| | - Guoren Deng
- Department of Urology, San Francisco Veterans Affairs Medical Center and University of California San Francisco, San Francisco, California, United States of America
| | - Varahram Shahryari
- Department of Urology, San Francisco Veterans Affairs Medical Center and University of California San Francisco, San Francisco, California, United States of America
| | - Sharanjot Saini
- Department of Urology, San Francisco Veterans Affairs Medical Center and University of California San Francisco, San Francisco, California, United States of America
| | - Hiroshi Hirata
- Department of Urology, San Francisco Veterans Affairs Medical Center and University of California San Francisco, San Francisco, California, United States of America
| | - Koji Ueno
- Department of Urology, San Francisco Veterans Affairs Medical Center and University of California San Francisco, San Francisco, California, United States of America
| | - Inik Chang
- Department of Urology, San Francisco Veterans Affairs Medical Center and University of California San Francisco, San Francisco, California, United States of America
| | - Yuichiro Tanaka
- Department of Urology, San Francisco Veterans Affairs Medical Center and University of California San Francisco, San Francisco, California, United States of America
| | - Z. Laura Tabatabai
- Department of Pathology, San Francisco Veterans Affairs Medical Center and University of California San Francisco, San Francisco, California, United States of America
| | - Hideki Enokida
- Department of Urology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Masayuki Nakagawa
- Department of Urology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Rajvir Dahiya
- Department of Urology, San Francisco Veterans Affairs Medical Center and University of California San Francisco, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
19
|
Pérot G, Croce S, Ribeiro A, Lagarde P, Velasco V, Neuville A, Coindre JM, Stoeckle E, Floquet A, MacGrogan G, Chibon F. MED12 alterations in both human benign and malignant uterine soft tissue tumors. PLoS One 2012; 7:e40015. [PMID: 22768200 PMCID: PMC3386951 DOI: 10.1371/journal.pone.0040015] [Citation(s) in RCA: 109] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Accepted: 05/30/2012] [Indexed: 11/18/2022] Open
Abstract
The relationship between benign uterine leiomyomas and their malignant counterparts, i.e. leiomyosarcomas and smooth muscle tumors of uncertain malignant potential (STUMP), is still poorly understood. The idea that a leiomyosarcoma could derive from a leiomyoma is still controversial. Recently MED12 mutations have been reported in uterine leiomyomas. In this study we asked whether such mutations could also be involved in leiomyosarcomas and STUMP oncogenesis. For this purpose we examined 33 uterine mesenchymal tumors by sequencing the hot-spot mutation region of MED12. We determined that MED12 is altered in 66.6% of typical leiomyomas as previously reported but also in 11% of STUMP and 20% of leiomyosarcomas. The mutated allele is predominantly expressed in leiomyomas and STUMP. Interestingly all classical leiomyomas exhibit MED12 protein expression while 40% of atypical leiomyomas, 50% of STUMP and 80% of leiomyosarcomas (among them the two mutated ones) do not express MED12. All these tumors without protein expression exhibit complex genomic profiles. No mutations and no expression loss were identified in an additional series of 38 non-uterine leiomyosarcomas. MED12 mutations are not exclusive to leiomyomas but seem to be specific to uterine malignancies. A previous study has suggested that MED12 mutations in leiomyomas could lead to Wnt/β-catenin pathway activation however our immunohistochemistry results show that there is no association between MED12 status and β-catenin nuclear/cytoplasmic localization. Collectively, our results show that subgroups of benign and malignant tumors share a common genetics. We propose here that MED12 alterations could be implicated in the development of smooth muscle tumor and that its expression could be inhibited in malignant tumors.
Collapse
Affiliation(s)
- Gaëlle Pérot
- INSERM U916, Institut Bergonié Cancer Institute, Bordeaux, France
| | - Sabrina Croce
- Department of Pathology, Institut Bergonié Cancer Institute, Bordeaux, France
| | - Agnès Ribeiro
- Department of Pathology, Institut Bergonié Cancer Institute, Bordeaux, France
- Department of Molecular Pathology, Institut Bergonié Cancer Institute, Bordeaux, France
| | - Pauline Lagarde
- INSERM U916, Institut Bergonié Cancer Institute, Bordeaux, France
| | - Valérie Velasco
- Department of Pathology, Institut Bergonié Cancer Institute, Bordeaux, France
- Department of Molecular Pathology, Institut Bergonié Cancer Institute, Bordeaux, France
| | - Agnès Neuville
- Department of Pathology, Institut Bergonié Cancer Institute, Bordeaux, France
| | - Jean-Michel Coindre
- INSERM U916, Institut Bergonié Cancer Institute, Bordeaux, France
- Department of Pathology, Institut Bergonié Cancer Institute, Bordeaux, France
- University Victor Segalen, Bordeaux, France
| | - Eberhard Stoeckle
- Department of Surgery, Institut Bergonié Cancer Institute, Bordeaux, France
| | - Anne Floquet
- Department of Medical Oncology, Institut Bergonié Cancer Institute, Bordeaux, France
| | - Gaëtan MacGrogan
- INSERM U916, Institut Bergonié Cancer Institute, Bordeaux, France
- Department of Pathology, Institut Bergonié Cancer Institute, Bordeaux, France
| | - Frédéric Chibon
- INSERM U916, Institut Bergonié Cancer Institute, Bordeaux, France
- Department of Pathology, Institut Bergonié Cancer Institute, Bordeaux, France
- Department of Molecular Pathology, Institut Bergonié Cancer Institute, Bordeaux, France
- * E-mail:
| |
Collapse
|
20
|
Lo FY, Chang JW, Chang IS, Chen YJ, Hsu HS, Huang SFK, Tsai FY, Jiang SS, Kanteti R, Nandi S, Salgia R, Wang YC. The database of chromosome imbalance regions and genes resided in lung cancer from Asian and Caucasian identified by array-comparative genomic hybridization. BMC Cancer 2012; 12:235. [PMID: 22691236 PMCID: PMC3488578 DOI: 10.1186/1471-2407-12-235] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Accepted: 05/12/2012] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Cancer-related genes show racial differences. Therefore, identification and characterization of DNA copy number alteration regions in different racial groups helps to dissect the mechanism of tumorigenesis. METHODS Array-comparative genomic hybridization (array-CGH) was analyzed for DNA copy number profile in 40 Asian and 20 Caucasian lung cancer patients. Three methods including MetaCore analysis for disease and pathway correlations, concordance analysis between array-CGH database and the expression array database, and literature search for copy number variation genes were performed to select novel lung cancer candidate genes. Four candidate oncogenes were validated for DNA copy number and mRNA and protein expression by quantitative polymerase chain reaction (qPCR), chromogenic in situ hybridization (CISH), reverse transcriptase-qPCR (RT-qPCR), and immunohistochemistry (IHC) in more patients. RESULTS We identified 20 chromosomal imbalance regions harboring 459 genes for Caucasian and 17 regions containing 476 genes for Asian lung cancer patients. Seven common chromosomal imbalance regions harboring 117 genes, included gain on 3p13-14, 6p22.1, 9q21.13, 13q14.1, and 17p13.3; and loss on 3p22.2-22.3 and 13q13.3 were found both in Asian and Caucasian patients. Gene validation for four genes including ARHGAP19 (10q24.1) functioning in Rho activity control, FRAT2 (10q24.1) involved in Wnt signaling, PAFAH1B1 (17p13.3) functioning in motility control, and ZNF322A (6p22.1) involved in MAPK signaling was performed using qPCR and RT-qPCR. Mean gene dosage and mRNA expression level of the four candidate genes in tumor tissues were significantly higher than the corresponding normal tissues (P<0.001~P=0.06). In addition, CISH analysis of patients indicated that copy number amplification indeed occurred for ARHGAP19 and ZNF322A genes in lung cancer patients. IHC analysis of paraffin blocks from Asian Caucasian patients demonstrated that the frequency of PAFAH1B1 protein overexpression was 68% in Asian and 70% in Caucasian. CONCLUSIONS Our study provides an invaluable database revealing common and differential imbalance regions at specific chromosomes among Asian and Caucasian lung cancer patients. Four validation methods confirmed our database, which would help in further studies on the mechanism of lung tumorigenesis.
Collapse
Affiliation(s)
- Fang-Yi Lo
- Department of Pharmacology and Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, No,1, University Road, Tainan 701, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
A genome-wide study of cytogenetic changes in colorectal cancer using SNP microarrays: opportunities for future personalized treatment. PLoS One 2012; 7:e31968. [PMID: 22363777 PMCID: PMC3282791 DOI: 10.1371/journal.pone.0031968] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2011] [Accepted: 01/19/2012] [Indexed: 01/10/2023] Open
Abstract
In colorectal cancer (CRC), chromosomal instability (CIN) is typically studied using comparative-genomic hybridization (CGH) arrays. We studied paired (tumor and surrounding healthy) fresh frozen tissue from 86 CRC patients using Illumina's Infinium-based SNP array. This method allowed us to study CIN in CRC, with simultaneous analysis of copy number (CN) and B-allele frequency (BAF)--a representation of allelic composition. These data helped us to detect mono-allelic and bi-allelic amplifications/deletion, copy neutral loss of heterozygosity, and levels of mosaicism for mixed cell populations, some of which can not be assessed with other methods that do not measure BAF. We identified associations between CN abnormalities and different CRC phenotypes (histological diagnosis, location, tumor grade, stage, MSI and presence of lymph node metastasis). We showed commonalities between regions of CN change observed in CRC and the regions reported in previous studies of other solid cancers (e.g. amplifications of 20q, 13q, 8q, 5p and deletions of 18q, 17p and 8p). From Therapeutic Target Database, we identified relevant drugs, targeted to the genes located in these regions with CN changes, approved or in trials for other cancers and common diseases. These drugs may be considered for future therapeutic trials in CRC, based on personalized cytogenetic diagnosis. We also found many regions, harboring genes, which are not currently targeted by any relevant drugs that may be considered for future drug discovery studies. Our study shows the application of high density SNP arrays for cytogenetic study in CRC and its potential utility for personalized treatment.
Collapse
|
22
|
Inoue T, Matsuura K, Yoshimoto T, Nguyen LT, Tsukamoto Y, Nakada C, Hijiya N, Narimatsu T, Nomura T, Sato F, Nagashima Y, Kashima K, Hatakeyama S, Ohyama C, Numakura K, Habuchi T, Nakagawa M, Seto M, Mimata H, Moriyama M. Genomic profiling of renal cell carcinoma in patients with end-stage renal disease. Cancer Sci 2012; 103:569-76. [PMID: 22145865 DOI: 10.1111/j.1349-7006.2011.02176.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The purpose of the present study was to determine the genomic profile of renal cell carcinoma (RCC) in end-stage renal disease (ESRD) by analyzing genomic copy number aberrations. Seventy-nine tumor samples from 63 patients with RCC-ESRD were analyzed by array comparative genomic hybridization using the Agilent Whole Human Genome 4 × 44K Oligo Micro Array (Agilent Technologies Inc., Palo Alto, CA, USA). Unsupervised hierarchical clustering analysis revealed that the 63 cases could be divided into two groups, Clusters A and B. Cluster A was comprised mainly of clear cell RCC (CCRCC), whereas Cluster B was comprised mainly of papillary RCC (PRCC), acquired cystic disease (ACD)-associated RCC, and clear cell papillary RCC. Analysis of the averaged frequencies revealed that the genomic profiles of Clusters A and B resembled those of sporadic CCRCC and sporadic PRCC, respectively. Although it has been proposed on the basis of histopathology that ACD-associated RCC, clear cell papillary RCC and PRCC-ESRD are distinct subtypes, the present data reveal that the genomic profiles of these types, categorized as Cluster B, resemble one another. Furthermore, the genomic profiles of PRCC, ACD-associated RCC and clear cell papillary RCC admixed in one tissue tended to resemble one another. On the basis of genomic profiling of RCC-ESRD, we conclude that the molecular pathogenesis of CCRCC-ESRD resembles that of sporadic CCRCC. Although various histologic subtypes of non-clear cell RCC-ESRD have been proposed, their genomic profiles resemble those of sporadic PRCC, suggesting that the molecular pathogenesis of non-CCRCC-ESRD may be related to that of sporadic PRCC.
Collapse
Affiliation(s)
- Toru Inoue
- Department of Molecular Pathology, Faculty of Medicine, Oita University, Oita, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Yoon AR, Gao R, Kaul Z, Choi IK, Ryu J, Noble JR, Kato Y, Saito S, Hirano T, Ishii T, Reddel RR, Yun CO, Kaul SC, Wadhwa R. MicroRNA-296 is enriched in cancer cells and downregulates p21WAF1 mRNA expression via interaction with its 3' untranslated region. Nucleic Acids Res 2011; 39:8078-91. [PMID: 21724611 PMCID: PMC3185413 DOI: 10.1093/nar/gkr492] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2010] [Revised: 04/30/2011] [Accepted: 05/27/2011] [Indexed: 01/07/2023] Open
Abstract
MicroRNAs (miRNAs) are a class of noncoding small RNAs that act as negative regulators of gene expression. To identify miRNAs that may regulate human cell immortalization and carcinogenesis, we performed comparative miRNA array profiling of human normal and SV40-T antigen immortalized cells. We found that miR-296 was upregulated in immortalized cells that also had activation of telomerase. By an independent experiment on genomic analysis of cancer cells we found that chromosome region (20q13.32), where miR-296 is located, was amplified in 28/36 cell lines, and most of these showed enriched miR-296 expression. Overexpression of miR-296 in human cancer cells, with and without telomerase activity, had no effect on their telomerase function. Instead, it suppressed p53 function that is frequently downregulated during human cell immortalization and carcinogenesis. By monitoring the activity of a luciferase reporter connected to p53 and p21(WAF1) (p21) untranslated regions (UTRs), we demonstrate that miR-296 interacts with the p21-3'UTR, and the Hu binding site of p21-3'UTR was identified as a potential miR-296 target site. We demonstrate for the first time that miR-296 is frequently upregulated during immortalization of human cells and contributes to carcinogenesis by downregulation of p53-p21(WAF1) pathway.
Collapse
Affiliation(s)
- A-rum Yoon
- National Institute of Advanced Industrial Science & Technology (AIST), Central 4, 1-1-1 Higashi, Tsukuba, Ibaraki – 305 8562, Japan, Brain Korea 21 Project for Medical Science, Department of Biomedical Science, Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea, Majors of Medical Sciences, University of Tsukuba, Ibaraki – 305-8575, Japan and Children’s Medical Research Institute, 214 Hawkesbury Road, Westmead 2145, NSW and Sydney Medical School, University of Sydney 2006, NSW, Australia, Graduate Program for Nanomedical Science, Yonsei University, Seoul, South Korea, Department of Bioengineering, College of Engineering, Hanyang University, Seoul, South Korea, Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, South Korea
| | - Ran Gao
- National Institute of Advanced Industrial Science & Technology (AIST), Central 4, 1-1-1 Higashi, Tsukuba, Ibaraki – 305 8562, Japan, Brain Korea 21 Project for Medical Science, Department of Biomedical Science, Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea, Majors of Medical Sciences, University of Tsukuba, Ibaraki – 305-8575, Japan and Children’s Medical Research Institute, 214 Hawkesbury Road, Westmead 2145, NSW and Sydney Medical School, University of Sydney 2006, NSW, Australia, Graduate Program for Nanomedical Science, Yonsei University, Seoul, South Korea, Department of Bioengineering, College of Engineering, Hanyang University, Seoul, South Korea, Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, South Korea
| | - Zeenia Kaul
- National Institute of Advanced Industrial Science & Technology (AIST), Central 4, 1-1-1 Higashi, Tsukuba, Ibaraki – 305 8562, Japan, Brain Korea 21 Project for Medical Science, Department of Biomedical Science, Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea, Majors of Medical Sciences, University of Tsukuba, Ibaraki – 305-8575, Japan and Children’s Medical Research Institute, 214 Hawkesbury Road, Westmead 2145, NSW and Sydney Medical School, University of Sydney 2006, NSW, Australia, Graduate Program for Nanomedical Science, Yonsei University, Seoul, South Korea, Department of Bioengineering, College of Engineering, Hanyang University, Seoul, South Korea, Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, South Korea
| | - Il-Kyu Choi
- National Institute of Advanced Industrial Science & Technology (AIST), Central 4, 1-1-1 Higashi, Tsukuba, Ibaraki – 305 8562, Japan, Brain Korea 21 Project for Medical Science, Department of Biomedical Science, Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea, Majors of Medical Sciences, University of Tsukuba, Ibaraki – 305-8575, Japan and Children’s Medical Research Institute, 214 Hawkesbury Road, Westmead 2145, NSW and Sydney Medical School, University of Sydney 2006, NSW, Australia, Graduate Program for Nanomedical Science, Yonsei University, Seoul, South Korea, Department of Bioengineering, College of Engineering, Hanyang University, Seoul, South Korea, Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, South Korea
| | - Jihoon Ryu
- National Institute of Advanced Industrial Science & Technology (AIST), Central 4, 1-1-1 Higashi, Tsukuba, Ibaraki – 305 8562, Japan, Brain Korea 21 Project for Medical Science, Department of Biomedical Science, Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea, Majors of Medical Sciences, University of Tsukuba, Ibaraki – 305-8575, Japan and Children’s Medical Research Institute, 214 Hawkesbury Road, Westmead 2145, NSW and Sydney Medical School, University of Sydney 2006, NSW, Australia, Graduate Program for Nanomedical Science, Yonsei University, Seoul, South Korea, Department of Bioengineering, College of Engineering, Hanyang University, Seoul, South Korea, Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, South Korea
| | - Jane R. Noble
- National Institute of Advanced Industrial Science & Technology (AIST), Central 4, 1-1-1 Higashi, Tsukuba, Ibaraki – 305 8562, Japan, Brain Korea 21 Project for Medical Science, Department of Biomedical Science, Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea, Majors of Medical Sciences, University of Tsukuba, Ibaraki – 305-8575, Japan and Children’s Medical Research Institute, 214 Hawkesbury Road, Westmead 2145, NSW and Sydney Medical School, University of Sydney 2006, NSW, Australia, Graduate Program for Nanomedical Science, Yonsei University, Seoul, South Korea, Department of Bioengineering, College of Engineering, Hanyang University, Seoul, South Korea, Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, South Korea
| | - Yoshio Kato
- National Institute of Advanced Industrial Science & Technology (AIST), Central 4, 1-1-1 Higashi, Tsukuba, Ibaraki – 305 8562, Japan, Brain Korea 21 Project for Medical Science, Department of Biomedical Science, Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea, Majors of Medical Sciences, University of Tsukuba, Ibaraki – 305-8575, Japan and Children’s Medical Research Institute, 214 Hawkesbury Road, Westmead 2145, NSW and Sydney Medical School, University of Sydney 2006, NSW, Australia, Graduate Program for Nanomedical Science, Yonsei University, Seoul, South Korea, Department of Bioengineering, College of Engineering, Hanyang University, Seoul, South Korea, Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, South Korea
| | - Soichiro Saito
- National Institute of Advanced Industrial Science & Technology (AIST), Central 4, 1-1-1 Higashi, Tsukuba, Ibaraki – 305 8562, Japan, Brain Korea 21 Project for Medical Science, Department of Biomedical Science, Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea, Majors of Medical Sciences, University of Tsukuba, Ibaraki – 305-8575, Japan and Children’s Medical Research Institute, 214 Hawkesbury Road, Westmead 2145, NSW and Sydney Medical School, University of Sydney 2006, NSW, Australia, Graduate Program for Nanomedical Science, Yonsei University, Seoul, South Korea, Department of Bioengineering, College of Engineering, Hanyang University, Seoul, South Korea, Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, South Korea
| | - Takashi Hirano
- National Institute of Advanced Industrial Science & Technology (AIST), Central 4, 1-1-1 Higashi, Tsukuba, Ibaraki – 305 8562, Japan, Brain Korea 21 Project for Medical Science, Department of Biomedical Science, Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea, Majors of Medical Sciences, University of Tsukuba, Ibaraki – 305-8575, Japan and Children’s Medical Research Institute, 214 Hawkesbury Road, Westmead 2145, NSW and Sydney Medical School, University of Sydney 2006, NSW, Australia, Graduate Program for Nanomedical Science, Yonsei University, Seoul, South Korea, Department of Bioengineering, College of Engineering, Hanyang University, Seoul, South Korea, Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, South Korea
| | - Tetsuro Ishii
- National Institute of Advanced Industrial Science & Technology (AIST), Central 4, 1-1-1 Higashi, Tsukuba, Ibaraki – 305 8562, Japan, Brain Korea 21 Project for Medical Science, Department of Biomedical Science, Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea, Majors of Medical Sciences, University of Tsukuba, Ibaraki – 305-8575, Japan and Children’s Medical Research Institute, 214 Hawkesbury Road, Westmead 2145, NSW and Sydney Medical School, University of Sydney 2006, NSW, Australia, Graduate Program for Nanomedical Science, Yonsei University, Seoul, South Korea, Department of Bioengineering, College of Engineering, Hanyang University, Seoul, South Korea, Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, South Korea
| | - Roger R. Reddel
- National Institute of Advanced Industrial Science & Technology (AIST), Central 4, 1-1-1 Higashi, Tsukuba, Ibaraki – 305 8562, Japan, Brain Korea 21 Project for Medical Science, Department of Biomedical Science, Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea, Majors of Medical Sciences, University of Tsukuba, Ibaraki – 305-8575, Japan and Children’s Medical Research Institute, 214 Hawkesbury Road, Westmead 2145, NSW and Sydney Medical School, University of Sydney 2006, NSW, Australia, Graduate Program for Nanomedical Science, Yonsei University, Seoul, South Korea, Department of Bioengineering, College of Engineering, Hanyang University, Seoul, South Korea, Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, South Korea
| | - Chae-Ok Yun
- National Institute of Advanced Industrial Science & Technology (AIST), Central 4, 1-1-1 Higashi, Tsukuba, Ibaraki – 305 8562, Japan, Brain Korea 21 Project for Medical Science, Department of Biomedical Science, Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea, Majors of Medical Sciences, University of Tsukuba, Ibaraki – 305-8575, Japan and Children’s Medical Research Institute, 214 Hawkesbury Road, Westmead 2145, NSW and Sydney Medical School, University of Sydney 2006, NSW, Australia, Graduate Program for Nanomedical Science, Yonsei University, Seoul, South Korea, Department of Bioengineering, College of Engineering, Hanyang University, Seoul, South Korea, Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, South Korea
| | - Sunil C. Kaul
- National Institute of Advanced Industrial Science & Technology (AIST), Central 4, 1-1-1 Higashi, Tsukuba, Ibaraki – 305 8562, Japan, Brain Korea 21 Project for Medical Science, Department of Biomedical Science, Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea, Majors of Medical Sciences, University of Tsukuba, Ibaraki – 305-8575, Japan and Children’s Medical Research Institute, 214 Hawkesbury Road, Westmead 2145, NSW and Sydney Medical School, University of Sydney 2006, NSW, Australia, Graduate Program for Nanomedical Science, Yonsei University, Seoul, South Korea, Department of Bioengineering, College of Engineering, Hanyang University, Seoul, South Korea, Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, South Korea
| | - Renu Wadhwa
- National Institute of Advanced Industrial Science & Technology (AIST), Central 4, 1-1-1 Higashi, Tsukuba, Ibaraki – 305 8562, Japan, Brain Korea 21 Project for Medical Science, Department of Biomedical Science, Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea, Majors of Medical Sciences, University of Tsukuba, Ibaraki – 305-8575, Japan and Children’s Medical Research Institute, 214 Hawkesbury Road, Westmead 2145, NSW and Sydney Medical School, University of Sydney 2006, NSW, Australia, Graduate Program for Nanomedical Science, Yonsei University, Seoul, South Korea, Department of Bioengineering, College of Engineering, Hanyang University, Seoul, South Korea, Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
24
|
Kuroda A, Tsukamoto Y, Nguyen LT, Noguchi T, Takeuchi I, Uchida M, Uchida T, Hijiya N, Nakada C, Okimoto T, Kodama M, Murakami K, Matsuura K, Seto M, Ito H, Fujioka T, Moriyama M. Genomic profiling of submucosal-invasive gastric cancer by array-based comparative genomic hybridization. PLoS One 2011; 6:e22313. [PMID: 21811585 PMCID: PMC3141024 DOI: 10.1371/journal.pone.0022313] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2011] [Accepted: 06/19/2011] [Indexed: 12/28/2022] Open
Abstract
Genomic copy number aberrations (CNAs) in gastric cancer have already been extensively characterized by array comparative genomic hybridization (array CGH) analysis. However, involvement of genomic CNAs in the process of submucosal invasion and lymph node metastasis in early gastric cancer is still poorly understood. In this study, to address this issue, we collected a total of 59 tumor samples from 27 patients with submucosal-invasive gastric cancers (SMGC), analyzed their genomic profiles by array CGH, and compared them between paired samples of mucosal (MU) and submucosal (SM) invasion (23 pairs), and SM invasion and lymph node (LN) metastasis (9 pairs). Initially, we hypothesized that acquisition of specific CNA(s) is important for these processes. However, we observed no significant difference in the number of genomic CNAs between paired MU and SM, and between paired SM and LN. Furthermore, we were unable to find any CNAs specifically associated with SM invasion or LN metastasis. Among the 23 cases analyzed, 15 had some similar pattern of genomic profiling between SM and MU. Interestingly, 13 of the 15 cases also showed some differences in genomic profiles. These results suggest that the majority of SMGCs are composed of heterogeneous subpopulations derived from the same clonal origin. Comparison of genomic CNAs between SMGCs with and without LN metastasis revealed that gain of 11q13, 11q14, 11q22, 14q32 and amplification of 17q21 were more frequent in metastatic SMGCs, suggesting that these CNAs are related to LN metastasis of early gastric cancer. In conclusion, our data suggest that generation of genetically distinct subclones, rather than acquisition of specific CNA at MU, is integral to the process of submucosal invasion, and that subclones that acquire gain of 11q13, 11q14, 11q22, 14q32 or amplification of 17q21 are likely to become metastatic.
Collapse
Affiliation(s)
- Akiko Kuroda
- Department of Molecular Pathology, Faculty of Medicine, Oita University, Oita, Japan
- Department of General Medicine, Faculty of Medicine, Oita University, Oita, Japan
| | - Yoshiyuki Tsukamoto
- Department of Molecular Pathology, Faculty of Medicine, Oita University, Oita, Japan
- * E-mail:
| | - Lam Tung Nguyen
- Department of Molecular Pathology, Faculty of Medicine, Oita University, Oita, Japan
- Department of General Medicine, Faculty of Medicine, Oita University, Oita, Japan
| | - Tsuyoshi Noguchi
- Department of Gastrointestinal Surgery, Faculty of Medicine, Oita University, Oita, Japan
| | - Ichiro Takeuchi
- Department of Computer Science/Scientific and Engineering Simulation, Nagoya Institute of Technology, Nagoya, Japan
| | - Masahiro Uchida
- Department of Gastroenterology, Faculty of Medicine, Oita University, Oita, Japan
| | - Tomohisa Uchida
- Department of Molecular Pathology, Faculty of Medicine, Oita University, Oita, Japan
| | - Naoki Hijiya
- Department of Molecular Pathology, Faculty of Medicine, Oita University, Oita, Japan
| | - Chisato Nakada
- Department of Molecular Pathology, Faculty of Medicine, Oita University, Oita, Japan
| | - Tadayoshi Okimoto
- Department of General Medicine, Faculty of Medicine, Oita University, Oita, Japan
- Department of Gastroenterology, Faculty of Medicine, Oita University, Oita, Japan
| | - Masaaki Kodama
- Department of General Medicine, Faculty of Medicine, Oita University, Oita, Japan
- Department of Gastroenterology, Faculty of Medicine, Oita University, Oita, Japan
| | - Kazunari Murakami
- Department of General Medicine, Faculty of Medicine, Oita University, Oita, Japan
- Department of Gastroenterology, Faculty of Medicine, Oita University, Oita, Japan
| | - Keiko Matsuura
- Department of Molecular Pathology, Faculty of Medicine, Oita University, Oita, Japan
| | - Masao Seto
- Division of Molecular Medicine, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Hisao Ito
- Division of Organ Pathology, Department of Microbiology and Pathology, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Toshio Fujioka
- Department of General Medicine, Faculty of Medicine, Oita University, Oita, Japan
- Department of Gastroenterology, Faculty of Medicine, Oita University, Oita, Japan
| | - Masatsugu Moriyama
- Department of Molecular Pathology, Faculty of Medicine, Oita University, Oita, Japan
| |
Collapse
|