Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Cancer Discov ; 14(4): 683-689, 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38571435

ABSTRACT

Research on precancers, as defined as at-risk tissues and early lesions, is of high significance given the effectiveness of early intervention. We discuss the need for risk stratification to prevent overtreatment, an emphasis on the role of genetic and epigenetic aging when considering risk, and the importance of integrating macroenvironmental risk factors with molecules and cells in lesions and at-risk normal tissues for developing effective intervention and health policy strategies.


Subject(s)
Precancerous Conditions , Humans , Precancerous Conditions/genetics , Precancerous Conditions/pathology , Risk Factors
2.
bioRxiv ; 2023 Aug 15.
Article in English | MEDLINE | ID: mdl-37645752

ABSTRACT

Background: The development of diverse spatial profiling technologies has provided an unprecedented insight into molecular mechanisms driving cancer pathogenesis. Here, we conducted the first integrated cross-species assessment of spatial transcriptomics and spatial metabolomics alterations associated with progression of intraductal papillary mucinous neoplasms (IPMN), bona fide cystic precursors of pancreatic ductal adenocarcinoma (PDAC). Methods: Matrix Assisted Laster Desorption/Ionization (MALDI) mass spectrometry (MS)-based spatial imaging and Visium spatial transcriptomics (ST) (10X Genomics) was performed on human resected IPMN tissues (N= 23) as well as pancreata from a mutant Kras;Gnas mouse model of IPMN. Findings were further compared with lipidomic analyses of cystic fluid from 89 patients with histologically confirmed IPMNs, as well as single-cell and bulk transcriptomic data of PDAC and normal tissues. Results: MALDI-MS analyses of IPMN tissues revealed long-chain hydroxylated sulfatides, particularly the C24:0(OH) and C24:1(OH) species, to be selectively enriched in the IPMN and PDAC neoplastic epithelium. Integrated ST analyses confirmed that the cognate transcripts engaged in sulfatide biosynthesis, including UGT8, Gal3St1 , and FA2H , were co-localized with areas of sulfatide enrichment. Lipidomic analyses of cystic fluid identified several sulfatide species, including the C24:0(OH) and C24:1(OH) species, to be significantly elevated in patients with IPMN/PDAC compared to those with low-grade IPMN. Targeting of sulfatide metabolism via the selective galactosylceramide synthase inhibitor, UGT8-IN-1, resulted in ceramide-induced lethal mitophagy and subsequent cancer cell death in vitro , and attenuated tumor growth of mutant Kras;Gnas allografts. Transcript levels of UGT8 and FA2H were also selectively enriched in PDAC transcriptomic datasets compared to non-cancerous areas, and elevated tumoral UGT8 was prognostic for poor overall survival. Conclusion: Enhanced sulfatide metabolism is an early metabolic alteration in cystic pre-cancerous lesions of the pancreas that persists through invasive neoplasia. Targeting sulfatide biosynthesis might represent an actionable vulnerability for cancer interception.

3.
Cancer Discov ; 13(8): 1844-1861, 2023 08 04.
Article in English | MEDLINE | ID: mdl-37285225

ABSTRACT

Intraductal papillary mucinous neoplasms (IPMN) of the pancreas are bona fide precursor lesions of pancreatic ductal adenocarcinoma (PDAC). The most common subtype of IPMNs harbors a gastric foveolar-type epithelium, and these low-grade mucinous neoplasms are harbingers of IPMNs with high-grade dysplasia and cancer. The molecular underpinning of gastric differentiation in IPMNs is unknown, although identifying drivers of this indolent phenotype might enable opportunities for intercepting progression to high-grade IPMN and cancer. We conducted spatial transcriptomics on a cohort of IPMNs, followed by orthogonal and cross-species validation studies, which established the transcription factor NKX6-2 as a key determinant of gastric cell identity in low-grade IPMNs. Loss of NKX6-2 expression is a consistent feature of IPMN progression, while reexpression of Nkx6-2 in murine IPMN lines recapitulates the aforementioned gastric transcriptional program and glandular morphology. Our study identifies NKX6-2 as a previously unknown transcription factor driving indolent gastric differentiation in IPMN pathogenesis. SIGNIFICANCE: Identification of the molecular features driving IPMN development and differentiation is critical to prevent cancer progression and enhance risk stratification. We used spatial profiling to characterize the epithelium and microenvironment of IPMN, which revealed a previously unknown link between NKX6-2 and gastric differentiation, the latter associated with indolent biological potential. See related commentary by Ben-Shmuel and Scherz-Shouval, p. 1768. This article is highlighted in the In This Issue feature, p. 1749.


Subject(s)
Carcinoma, Pancreatic Ductal , Neoplasms, Cystic, Mucinous, and Serous , Pancreatic Intraductal Neoplasms , Pancreatic Neoplasms , Animals , Mice , Carcinoma, Pancreatic Ductal/pathology , Cell Differentiation/genetics , Pancreas/pathology , Pancreatic Intraductal Neoplasms/genetics , Pancreatic Neoplasms/pathology , Transcriptome , Tumor Microenvironment
4.
Gastroenterology ; 165(2): 374-390, 2023 08.
Article in English | MEDLINE | ID: mdl-37196797

ABSTRACT

BACKGROUND & AIMS: Elements of field cancerization, including atrophic gastritis, metaplasia, and dysplasia, promote gastric cancer development in association with chronic inflammation. However, it remains unclear how stroma changes during carcinogenesis and how the stroma contributes to progression of gastric preneoplasia. Here we investigated heterogeneity of fibroblasts, one of the most important elements in the stroma, and their roles in neoplastic transformation of metaplasia. METHODS: We used single-cell transcriptomics to evaluate the cellular heterogeneity of mucosal cells from patients with gastric cancer. Tissue sections from the same cohort and tissue microarrays were used to identify the geographical distribution of distinct fibroblast subsets. We further evaluated the role of fibroblasts from pathologic mucosa in dysplastic progression of metaplastic cells using patient-derived metaplastic gastroids and fibroblasts. RESULTS: We identified 4 subsets of fibroblasts within stromal cells defined by the differential expression of PDGFRA, FBLN2, ACTA2, or PDGFRB. Each subset was distributed distinctively throughout stomach tissues with different proportions at each pathologic stage. The PDGFRα+ subset expanded in metaplasia and cancer compared with normal, maintaining a close proximity with the epithelial compartment. Co-culture of metaplasia- or cancer-derived fibroblasts with gastroids showing the characteristics of spasmolytic polypeptide-expressing metaplasia-induced disordered growth, loss of metaplastic markers, and increases in markers of dysplasia. Culture of metaplastic gastroids with conditioned media from metaplasia- or cancer-derived fibroblasts also promoted dysplastic transition. CONCLUSIONS: These findings indicate that fibroblast associations with metaplastic epithelial cells can facilitate direct transition of metaplastic spasmolytic polypeptide-expressing metaplasia cell lineages into dysplastic lineages.


Subject(s)
Gastric Mucosa , Stomach Neoplasms , Humans , Gastric Mucosa/pathology , Stomach Neoplasms/pathology , Hyperplasia , Metaplasia/pathology , Fibroblasts/metabolism
5.
Gastroenterology ; 163(4): 875-890, 2022 10.
Article in English | MEDLINE | ID: mdl-35700772

ABSTRACT

BACKGROUND & AIMS: Dysplasia carries a high risk of cancer development; however, the cellular mechanisms for dysplasia evolution to cancer are obscure. We have previously identified 2 putative dysplastic stem cell (DSC) populations, CD44v6neg/CD133+/CD166+ (double positive [DP]) and CD44v6+/CD133+/CD166+ (triple positive [TP]), which may contribute to cellular heterogeneity of gastric dysplasia. Here, we investigated functional roles and cell plasticity of noncancerous Trop2+/CD133+/CD166+ DSCs initially developed in the transition from precancerous metaplasia to dysplasia in the stomach. METHODS: Dysplastic organoids established from active Kras-induced mouse stomachs were used for transcriptome analysis, in vitro differentiation, and in vivo tumorigenicity assessments of DSCs. Cell heterogeneity and genetic alterations during clonal evolution of DSCs were examined by next-generation sequencing. Tissue microarrays were used to identify DSCs in human dysplasia. We additionally evaluated the effect of casein kinase 1 alpha (CK1α) regulation on the DSC activities using both mouse and human dysplastic organoids. RESULTS: We identified a high similarity of molecular profiles between DP- and TP-DSCs, but more dynamic activities of DP-DSCs in differentiation and survival for maintaining dysplastic cell lineages through Wnt ligand-independent CK1α/ß-catenin signaling. Xenograft studies demonstrated that the DP-DSCs clonally evolve toward multiple types of gastric adenocarcinomas and promote cancer cell heterogeneity by acquiring additional genetic mutations and recruiting the tumor microenvironment. Last, growth and survival of both mouse and human dysplastic organoids were controlled by targeting CK1α. CONCLUSIONS: These findings indicate that the DSCs are de novo gastric cancer-initiating cells responsible for neoplastic transformation and a promising target for intervention in early induction of gastric cancer.


Subject(s)
Precancerous Conditions , Stomach Neoplasms , Animals , Casein Kinase I/metabolism , Cell Plasticity , Cell Transformation, Neoplastic/pathology , Gastric Mucosa/pathology , Humans , Hyperplasia/pathology , Ligands , Mice , Precancerous Conditions/pathology , Proto-Oncogene Proteins p21(ras)/metabolism , Stem Cells/metabolism , Stomach Neoplasms/pathology , Tumor Microenvironment , beta Catenin/metabolism
6.
Cell Stem Cell ; 29(5): 826-839.e9, 2022 05 05.
Article in English | MEDLINE | ID: mdl-35523142

ABSTRACT

Adult stem cells constantly react to local changes to ensure tissue homeostasis. In the main body of the stomach, chief cells produce digestive enzymes; however, upon injury, they undergo rapid proliferation for prompt tissue regeneration. Here, we identified p57Kip2 (p57) as a molecular switch for the reserve stem cell state of chief cells in mice. During homeostasis, p57 is constantly expressed in chief cells but rapidly diminishes after injury, followed by robust proliferation. Both single-cell RNA sequencing and dox-induced lineage tracing confirmed the sequential loss of p57 and activation of proliferation within the chief cell lineage. In corpus organoids, p57 overexpression induced a long-term reserve stem cell state, accompanied by altered niche requirements and a mature chief cell/secretory phenotype. Following the constitutive expression of p57 in vivo, chief cells showed an impaired injury response. Thus, p57 is a gatekeeper that imposes the reserve stem cell state of chief cells in homeostasis.


Subject(s)
Chief Cells, Gastric , Cyclin-Dependent Kinase Inhibitor p57/metabolism , Animals , Cell Lineage , Chief Cells, Gastric/metabolism , Mice , Organoids , Stem Cells , Stomach
7.
Ann Surg ; 275(4): 706-717, 2022 04 01.
Article in English | MEDLINE | ID: mdl-33086305

ABSTRACT

OBJECTIVE: To investigate the molecular characteristics of AGEJ compared with EAC and gastric adenocarcinoma. SUMMARY OF BACKGROUND DATA: Classification of AGEJ based on differential molecular characteristics between EAC and gastric adenocarcinoma has been long-standing controversy but rarely conducted due to anatomical ambiguity and epidemiologic difference. METHODS: The molecular classification model with Bayesian compound covariate predictor was developed based on differential mRNA expression of EAC (N = 78) and GCFB (N = 102) from the Cancer Genome Atlas (TCGA) cohort. AGEJ/cardia (N = 48) in TCGA cohort and AGEJ/upper third GC (N = 46 pairs) in Seoul National University cohort were classified into the EAC-like or GCFB-like groups whose genomic, transcriptomic, and proteomic characteristics were compared. RESULTS: AGEJ in both cohorts was similarly classified as EAC-like (31.2%) or GCFB-like (68.8%) based on the 400-gene classifier. The GCFB-like group showed significantly activated phosphoinositide 3-kinase-AKT signaling with decreased expression of ERBB2. The EAC-like group presented significantly different alternative splicing including the skipped exon of RPS24, a significantly higher copy number amplification including ERBB2 amplification, and increased protein expression of ERBB2 and EGFR compared with GCFB-like group. High-throughput 3D drug test using independent cell lines revealed that the EAC-like group showed a significantly better response to lapatinib than the GCFB-like group (P = 0.015). CONCLUSIONS: AGEJ was the combined entity of the EAC-like and GCFB-like groups with consistently different molecular characteristics in both Seoul National University and TCGA cohorts. The EAC-like group with a high Bayesian compound covariate predictor score could be effectively targeted by dual inhibition of ERBB2 and EGFR.


Subject(s)
Adenocarcinoma , Esophageal Neoplasms , Stomach Neoplasms , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Bayes Theorem , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Esophagogastric Junction/pathology , Humans , Phosphatidylinositol 3-Kinases , Proteomics , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology
8.
Cell Mol Gastroenterol Hepatol ; 13(1): 199-217, 2022.
Article in English | MEDLINE | ID: mdl-34455107

ABSTRACT

BACKGROUND & AIMS: Metaplasia in the stomach is highly associated with development of intestinal-type gastric cancer. Two types of metaplasias, spasmolytic polypeptide-expressing metaplasia (SPEM) and intestinal metaplasia (IM), are considered precancerous lesions. However, it remains unclear how SPEM and IM are related. Here we investigated a new lineage-specific marker for SPEM cells, aquaporin 5 (AQP5), to assist in the identification of these 2 metaplasias. METHODS: Drug- or Helicobacter felis (H felis) infection-induced mouse models were used to identify the expression pattern of AQP5 in acute or chronic SPEM. Gene-manipulated mice treated with or without drug were used to investigate how AQP5 expression is regulated in metaplastic lesions. Metaplastic samples from transgenic mice and human gastric cancer patients were evaluated for AQP5 expression. Immunostaining with lineage-specific markers was used to differentiate metaplastic gland characteristics. RESULTS: Our results revealed that AQP5 is a novel lineage-specific marker for SPEM cells that are localized at the base of metaplastic glands initially and expand to dominate glands after chronic H felis infection. In addition, AQP5 expression was up-regulated early in chief cell reprogramming and was promoted by interleukin 13. In humans, metaplastic corpus showed highly branched structures with AQP5-positive SPEM. Human SPEM cells strongly expressing AQP5 were present at the bases of incomplete IM glands marked by TROP2 but were absent from complete IM glands. CONCLUSIONS: AQP5-expressing SPEM cells are present in pyloric metaplasia and TROP2-positive incomplete IM and may be an important component of metaplasia that can predict a higher risk for gastric cancer development.


Subject(s)
Aquaporin 5 , Peptides , Animals , Aquaporin 5/genetics , Aquaporin 5/metabolism , Humans , Intercellular Signaling Peptides and Proteins , Metaplasia , Mice , Up-Regulation
9.
J Pathol ; 251(3): 336-347, 2020 07.
Article in English | MEDLINE | ID: mdl-32432338

ABSTRACT

Intestinal-type gastric adenocarcinoma arises in a field of pre-existing metaplasia. While biomarkers of cancer and metaplasia have been identified, the definition of dysplastic transition as a critical point in the evolution of cancer has remained obscure. We have evaluated Trop2 as a putative marker of the transition from metaplasia to dysplasia in the stomach in multiple mouse models of metaplasia induction and progression. In addition, TROP2 expression was evaluated in human samples by immunostaining tissue microarrays for metaplasia, dysplasia, and gastric cancer. Dysplastic mouse organoids were evaluated in vitro following shRNA knockdown of Trop2 expression. In mouse models, no Trop2 was observed in the normal corpus and Trop2 was not induced in acute models of metaplasia induction with either L635 or DMP-777. In Mist1-Kras mice, Trop2 expression was not observed in metaplasia at 1 month after Kras induction, but was observed in dysplastic glands at 3-4 months after Kras induction. In human tissues, no Trop2 was observed in normal corpus mucosa or SPEM, but Trop2 expression was observed in incomplete intestinal metaplasia, with significantly less expression in complete intestinal metaplasia. Trop2 expression was observed in all dysplastic and 84% of gastric cancer lesions, although expression levels were variable. Dysplastic mouse organoids from Mist1-Kras mice expressed Trop2 strongly. Knockdown of Trop2 with shRNA markedly reduced organoid growth and budding behavior, and induced the upregulation of apical villin expression. We conclude that Trop2 is upregulated in the transition to dysplasia in the stomach and promotes dysplastic cell behaviors. © 2020 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Antigens, Neoplasm/metabolism , Cell Adhesion Molecules/metabolism , Cell Transformation, Neoplastic/metabolism , Gastric Mucosa/metabolism , Precancerous Conditions/metabolism , Stomach Neoplasms/metabolism , Animals , Antigens, Neoplasm/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Adhesion Molecules/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Cells, Cultured , Disease Models, Animal , Gastric Mucosa/pathology , Genes, ras , Humans , Metaplasia , Mice, Transgenic , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Organoids , Precancerous Conditions/genetics , Precancerous Conditions/pathology , Signal Transduction , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Up-Regulation
10.
Gastric Cancer ; 23(4): 600-613, 2020 07.
Article in English | MEDLINE | ID: mdl-32112274

ABSTRACT

BACKGROUND: Spasmolytic polypeptide-expressing metaplasia (SPEM) is considered a precursor lesion of intestinal metaplasia and intestinal-type gastric cancer (GC), but little is known about microRNA alterations during metaplasia and GC developments. Here, we investigate miR-30a expression in gastric lesions and identify its novel target gene which is associated with the intestinal-type GC. METHODS: We conducted in situ hybridization and qRT-PCR to determine miR-30a expression in gastric tissues. miR-30a functions were determined through induction or inhibition of miR-30a in GC cell lines. A gene microarray was utilized to confirm miR-30a target genes in GC, and siRNA-mediated target gene suppression and immunostaining were performed. The Cancer Genome Atlas data were utilized to validate gene expressions. RESULTS: We found down-regulation of miR-30a during chief cell transdifferentiation into SPEM. MiR-30a level was also reduced in the early stage of GC, and its level was maintained in advanced GC. We identified a novel target gene of miR-30a and ITGA2, and our results showed that either ectopic expression of miR-30a or ITGA2 knockdown suppressed GC cell proliferation, migration, and tumorigenesis. Levels of ITGA2 inversely correlated with levels of miR-30a in human intestinal-type GC. CONCLUSION: We found down-regulation of miR-30a in preneoplastic lesions and its tumor-suppressive functions by targeting ITGA2 in GC. The level of ITGA2, which functions as an oncogene, was up-regulated in human GC. The results of this study suggest that coordination of the miR-30a-ITGA2 axis may serve as an important mechanism in the development of gastric precancerous lesions and intestinal-type GC.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinogenesis/pathology , Gene Expression Regulation, Neoplastic , Integrin alpha2/metabolism , Intestinal Neoplasms/pathology , MicroRNAs/genetics , Stomach Neoplasms/pathology , Animals , Apoptosis , Biomarkers, Tumor/genetics , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Movement , Cell Proliferation , Female , Humans , Integrin alpha2/genetics , Intestinal Neoplasms/genetics , Intestinal Neoplasms/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Prognosis , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
11.
Nat Commun ; 10(1): 5549, 2019 12 05.
Article in English | MEDLINE | ID: mdl-31804471

ABSTRACT

Dysplasia is considered a key transition state between pre-cancer and cancer in gastric carcinogenesis. However, the cellular or phenotypic heterogeneity and mechanisms of dysplasia progression have not been elucidated. We have established metaplastic and dysplastic organoid lines, derived from Mist1-Kras(G12D) mouse stomach corpus and studied distinct cellular behaviors and characteristics of metaplastic and dysplastic organoids. We also examined functional roles for Kras activation in dysplasia progression using Selumetinib, a MEK inhibitor, which is a downstream mediator of Kras signaling. Here, we report that dysplastic organoids die or show altered cellular behaviors and diminished aggressive behavior in response to MEK inhibition. However, the organoids surviving after MEK inhibition maintain cellular heterogeneity. Two dysplastic stem cell (DSC) populations are also identified in dysplastic cells, which exhibited different clonogenic potentials. Therefore, Kras activation controls cellular dynamics and progression to dysplasia, and DSCs might contribute to cellular heterogeneity in dysplastic cell lineages.


Subject(s)
Cell Lineage/genetics , Cell Transformation, Neoplastic/genetics , Gastric Mucosa/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Stomach Neoplasms/genetics , Animals , Benzimidazoles/pharmacology , Cell Lineage/drug effects , Cell Proliferation/genetics , Cell Transformation, Neoplastic/metabolism , Gastric Mucosa/pathology , Gene Expression Regulation, Neoplastic , Genetic Heterogeneity/drug effects , Humans , Kinetics , Mice, Inbred C57BL , Mice, Transgenic , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Stomach/pathology , Stomach Neoplasms/metabolism
12.
World J Surg ; 42(4): 1056-1064, 2018 04.
Article in English | MEDLINE | ID: mdl-28929278

ABSTRACT

BACKGROUND: To examine the effect and mechanism of Roux-en-Y gastric bypass (RYGB) on the improvement of diabetes according to the length of anastomosis and the gastric pouch volume in an animal model. METHODS: Glucose intolerance was induced with a high-fat diet for 3 months in Sprague-Dawley rats. The animals were subjected to conventional RYGB (cRYGB; 5% gastric pouch with 15-cm Roux limb, 40-cm biliopancreatic limb; n = 9), short-limb RYGB (sRYGB; 5%, 8, 4 cm; n = 9), fundus-sparing RYGB (fRYGB; 30%, 8, 4 cm; n = 9), or sham operation (n = 9). After 6 weeks, oral glucose tolerance tests (OGTTs) were performed, and gut hormones including insulin, total GLP-1, GIP, and ghrelin were analyzed. RESULTS: The cRYGB group showed significantly decreased food intake, body weight, and random glucose (p < 0.05). sRYGB resulted in a similar change of body weight loss to that of cRYGB, but with no improvement of hyperglycemia. The fRYGB group showed similar changes of body weight and random glucose to those of the sham group. In cRYGB and sRYGB, the level of insulin steeply increased until 30 min during OGTT. GLP-1 was higher at 30 min in cRYGB than in other groups, without significance. The fRYGB group showed a slowly increasing pattern in OGTT and GLP-1, and the lowest peak point in insulin and GIP. CONCLUSION: cRYGB with 95% gastric resection was needed to achieve not only weight loss but also diabetes improvement, which could be related to the increase in GLP-1.


Subject(s)
Diabetes Mellitus, Type 2/surgery , Gastrectomy/methods , Gastric Bypass/methods , Stomach Neoplasms/surgery , Animals , Diabetes Mellitus, Type 2/complications , Male , Rats , Rats, Sprague-Dawley , Stomach Neoplasms/complications , Treatment Outcome , Weight Loss
13.
Mol Cells ; 40(5): 346-354, 2017 May 31.
Article in English | MEDLINE | ID: mdl-28535662

ABSTRACT

Long interspersed nuclear element-1 (LINE-1) is a retrotransposon that contains a CpG island in its 5'-untranslated region. The CpG island of LINE-1 is often heavily methylated in normal somatic cells, which is associated with poor prognosis in various cancers. DNA methylation can differ between formalin-fixed paraffin-embedded (FFPE) and frozen tissues. Therefore, this study aimed to compare the LINE-1 methylation status between the two tissue-storage conditions in gastric cancer (GC) clinical samples and to evaluate whether LINE-1 can be used as an independent prognostic marker for each tissue-storage type. We analyzed four CpG sites of LINE-1 and examined the methylation levels at these sites in 25 FFPE and 41 frozen GC tissues by quantitative bisulfite pyrosequencing. The LINE-1 methylation status was significantly different between the FFPE and frozen GC tissues (p < 0.001). We further analyzed the clinicopathological features in the two groups separately. In the frozen GC tissues, LINE-1 was significantly hypomethylated in GC tissues compared to their corresponding normal gastric mucosa tissues (p < 0.001), and its methylation status was associated with gender, differentiation state, and lymphatic and venous invasion of GC. In the FFPE GC tissues, the methylation levels of LINE-1 differed according to tumor location and venous invasion of GC. In conclusion, LINE-1 can be used as a useful methylation marker for venous invasion in both FFPE and frozen tumor tissues of GC.


Subject(s)
Biomarkers, Tumor/genetics , DNA Methylation , Long Interspersed Nucleotide Elements , Stomach Neoplasms/pathology , Aged , Cryopreservation/methods , Female , Formaldehyde , Humans , Lymphatic Metastasis/genetics , Lymphatic Metastasis/pathology , Male , Middle Aged , Paraffin Embedding , Stomach Neoplasms/genetics , Tissue Fixation/methods , Veins/pathology
14.
Cancer Biomark ; 19(3): 231-239, 2017 Jul 04.
Article in English | MEDLINE | ID: mdl-28453457

ABSTRACT

BACKGROUND: Few reliable blood markers are available for detecting gastric cancer, mainly owing to the heterogeneity of the cancer. OBJECTIVE: To establish gastric cancer diagnostic markers, we evaluated the levels of plasma cadherin 17 (CDH17) and trefoil factor 3 (TFF3), which are secretory proteins and known markers for intestinal metaplasia (IM), in patients with gastric cancer. METHOD: The protein expression level was analyzed in blood plasma samples from 111 gastric cancer patients and 44 healthy individuals, using a sandwich ELISA kit, followed by statistical analyses. RESULT: Overall, the plasma levels of CDH17 and TFF3 were not significantly different between groups (p= 0.160 and p= 0.113, respectively). However, CDH17 expression was significantly elevated in patients with stage II and III gastric cancers compared to that in healthy controls (p= 0.023 and p= 0.037, respectively). In contrast, TFF3 levels were significantly elevated in patients with stage I (p= 0.001) and T1 gastric cancer (p= 0.013). The sensitivity and specificity of CDH17 were 66.7 and 61.4%, respectively (cutoff point: 0.189 ng/mL); for TFF3, these values were 62.2 and 56.8%, respectively (cutoff point: 5.215 ng/mL). CONCLUSIONS: These findings indicate that secretory protein markers for metaplastic lineages can be used as blood markers for gastric cancer.


Subject(s)
Biomarkers, Tumor/metabolism , Cadherins/metabolism , Intestines/growth & development , Stomach Neoplasms/genetics , Trefoil Factor-3/metabolism , Biomarkers, Tumor/analysis , Female , Humans , Male , Metaplasia , Middle Aged , Stomach Neoplasms/metabolism
15.
Biomarkers ; 22(1): 35-44, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27329150

ABSTRACT

CONTEXT: Gastric cancer (GC) is the fourth most common cause of cancer-related deaths worldwide. OBJECTIVE: To determine the mRNA-expression of the MAL, TMEM220, MMP28, IL-19 and HOPX genes and analyse the methylation statuses of MAL and TMEM220. MATERIALS AND METHODS: Gene-expression levels were analysed in 10 GC cell lines and 30 matched pairs of GC and normal mucosa (NM) gastric tissue specimens in real-time reverse-transcriptase polymerase chain reactions. Gene methylation was evaluated by bisulphite sequencing. Detailed gene-methylation patterns were confirmed by pyrosequencing analysis. RESULTS: MAL, TMEM220, MMP28 and IL-19 were significantly down-regulated in GC cell lines and GC tissues compared to NM tissues. MAL and TMEM220 were highly methylated in GC tissues, and methylation inversely correlated with expression. MAL and TMEM220 expression were restored by treatment with 5-aza-2'-deoxycytidine. MAL and TMEM220 were specifically methylated and were down-regulated in human GC. DISCUSSION AND CONCLUSION: These loci may serve as novel methylation markers for patients with GC.


Subject(s)
Biomarkers, Tumor/genetics , DNA Methylation , Gene Expression Regulation, Neoplastic , Membrane Proteins/genetics , Myelin and Lymphocyte-Associated Proteolipid Proteins/genetics , Stomach Neoplasms/genetics , Cell Line, Tumor , Down-Regulation , Gene Expression Profiling , Humans , RNA, Messenger/analysis
16.
Oncotarget ; 8(65): 108848-108858, 2017 Dec 12.
Article in English | MEDLINE | ID: mdl-29312573

ABSTRACT

Peritoneal carcinomatosis (PC) of gastric origin has a poor prognosis with short survival due to lack of effective therapeutic modalities. Here, we evaluated the therapeutic efficacy of an injectable thermosensitive poly (organophosphazene) (PPZ) hydrogel with docetaxel (DTX-gel) to develop an effective therapeutic agent for patient with PC. Three days after inoculation of highly metastatic 44As3Luc cells into peritoneal cavity, the mice were intravenously or intraperitoneally administered with docetaxel alone (DTX-sol IV or IP), and intraperitoneally injected with DTX-gel. The anti-tumor activity was monitored by bioluminescence live imaging system. Compared to DTX-sol IV or IP, the tumor growth was significantly reduced in the DTX-gel treated mice (p<0.0001, p=0.0001). Furthermore, the survival rate was significantly increased in the DTX-gel treated mice compared to DTX-sol IV or IP treated mice (p<0.0001, p=0.0068). Our results demonstrated that DTX-gel suppresses peritoneal metastasis by continuing release of chemotherapy agent, which leads to increase the survival rate in a PC model. Therefore, biodegradable thermosensitive hydrogel with docetaxel system can be a good anti-cancer agent for PC.

17.
Proc Natl Acad Sci U S A ; 112(40): 12492-7, 2015 Oct 06.
Article in English | MEDLINE | ID: mdl-26401016

ABSTRACT

Gastric cancer (GC) is the third leading cause of cancer-related deaths worldwide. Recent high-throughput analyses of genomic alterations revealed several driver genes and altered pathways in GC. However, therapeutic applications from genomic data are limited, largely as a result of the lack of druggable molecular targets and preclinical models for drug selection. To identify new therapeutic targets for GC, we performed array comparative genomic hybridization (aCGH) of DNA from 103 patients with GC for copy number alteration (CNA) analysis, and whole-exome sequencing from 55 GCs from the same patients for mutation profiling. Pathway analysis showed recurrent alterations in the Wnt signaling [APC, CTNNB1, and DLC1 (deleted in liver cancer 1)], ErbB signaling (ERBB2, PIK3CA, and KRAS), and p53 signaling/apoptosis [TP53 and BCL2L1 (BCL2-like 1)] pathways. In 18.4% of GC cases (19/103), amplification of the antiapoptotic gene BCL2L1 was observed, and subsequently a BCL2L1 inhibitor was shown to markedly decrease cell viability in BCL2L1-amplified cell lines and in similarly altered patient-derived GC xenografts, especially when combined with other chemotherapeutic agents. In 10.9% of cases (6/55), mutations in DLC1 were found and were also shown to confer a growth advantage for these cells via activation of Rho-ROCK signaling, rendering these cells more susceptible to a ROCK inhibitor. Taken together, our study implicates BCL2L1 and DLC1 as potential druggable targets for specific subsets of GC cases.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , GTPase-Activating Proteins/genetics , Genome-Wide Association Study/methods , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Tumor Suppressor Proteins/genetics , bcl-X Protein/genetics , Aged , Aged, 80 and over , Animals , Cell Line, Tumor , Cell Survival/genetics , Comparative Genomic Hybridization , DNA Copy Number Variations , Exome/genetics , Female , GTPase-Activating Proteins/metabolism , Humans , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , Male , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Middle Aged , Mutation , RNA Interference , Sequence Analysis, DNA/methods , Stomach Neoplasms/pathology , Tumor Suppressor Proteins/metabolism , Xenograft Model Antitumor Assays , bcl-X Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...