Our Promise to You

Guaranteed product quality, expert customer support

Our Promise to You

Gastric Tumor Cells

  • Product List
  • Background
  • Applications
  • Scientific Data
  • FAQ

Background

Gastric tumor cells are the basis of gastric cancer (GC), which is one of the five most common malignancies and the third leading cause of cancer mortality worldwide. Despite the significant advancements in treatment, the prognosis for GC patients remains poor, primarily due to frequent relapse, metastasis, and drug resistance.

These cells are abnormal cells that grow uncontrollably in the stomach, forming a tumor. They can originate from different parts of the stomach, leading to different types of GC. The exact cause of their formation is not fully known, but it is believed to involve a combination of genetic factors, environmental factors, and infection with certain types of bacteria, such as Helicobacter pylori.

Gastric Tumor Cell Characteristics

  • Cellular morphology and phenotypic features. Gastric tumor cells often exhibit altered cell shapes, sizes, and organization compared to normal gastric epithelial cells. They may display an increased nuclear-to-cytoplasmic ratio, irregular nuclear shapes, and prominent nucleoli. Gastric tumor cells can lose their polarized epithelial morphology and adopt a more dedifferentiated, mesenchymal-like phenotype. Aberrant expression of cell surface markers, such as adhesion molecules and growth factor receptors, is commonly observed in gastric tumor cells.
  • Genetic and molecular alterations. Gastric tumor cells harbor various genetic and epigenetic changes that contribute to their malignant transformation. Commonly reported genetic alterations include mutations in tumor suppressor genes (e.g., TP53, ARID1A) and oncogenes (e.g., KRAS, HER2/ERBB2). Dysregulation of signaling pathways, such as the PI3K/AKT, RAS/MAPK, and Wnt/β-catenin pathways, is frequently observed in gastric tumor cells. Epigenetic modifications, including aberrant DNA methylation and histone modifications, can lead to the silencing of tumor suppressor genes and activation of oncogenes in gastric tumor cells.
  • Tumor heterogeneity. Gastric tumors exhibit substantial heterogeneity both between different GC subtypes and within individual tumors. This heterogeneity is reflected in the diverse morphological, genetic, and phenotypic characteristics of gastric tumor cells.

In Vitro Cell Line Models

One of the primary applications of gastric tumor cells lies in the development and utilization of in vitro cell line models. These models, established from the tumor cells of GC patients, serve as invaluable tools for researchers.

By cultivating and maintaining these cell lines, scientists can closely study the behavior and properties of gastric tumor cells under controlled laboratory conditions. This allows for a deeper exploration of the underlying mechanisms that drive their proliferation, invasion, and metastasis, as well as the evaluation of the efficacy and specificity of potential anti-cancer compounds. The availability of these well-characterized gastric tumor cell lines has been instrumental in accelerating the drug discovery and development process, as they provide a reliable platform for high-throughput screening and preclinical testing of novel therapeutic interventions.

Biomarker Identification

Gastric tumor cells also play a crucial role in the identification and validation of biomarkers for GC. By analyzing the genetic, molecular, and phenotypic profiles of these cells, researchers have been able to uncover a range of potential diagnostic and prognostic markers. For instance, detecting specific genetic alterations, such as mutations in TP53 or HER2 amplification, in gastric tumor cells can serve as valuable indicators for disease diagnosis and risk stratification. Furthermore, the expression patterns of cell surface markers and secreted factors by these cells can provide insights into the progression and metastatic potential of gastric tumors.

Advancing Cancer Research

Beyond the clinical applications, gastric tumor cells have also been instrumental in advancing the fundamental understanding of cancer biology. Investigations into the signaling pathways, epigenetic modifications, and the tumor microenvironment factors that influence gastric tumor cell behavior have led to groundbreaking discoveries. These insights have not only expanded our understanding of GC but have also informed research on other types of malignancies, fostering a more comprehensive approach to cancer research and treatment development.

Cell State-Specific Transcriptional Patterns Associated With GC Uncovered by scRNA-seq

To examine tumor-associated expression programs at the single-cell level, primary gastric tumor samples were compared to normal tissues (n = 26 tumor; n = 10 normal). After random downsampling to ensure statistical equivalency (Fig. 1A), the proportion of epithelial cells in tumors was lower compared with normal tissues (P = 0.002), whereas myeloid cells in tumors were higher (P = 0.05). The proportions of lymphoid, plasma, and stromal cells were not statistically different between tumor and normal samples.

Compared with their cognate cell states in normal tissues, tumor-associated epithelial cells exhibited higher expression of multiple oncogenic gene signatures related to epithelial-mesenchymal transition (EMT), cell motility, and cancer signatures derived from previous GC single-cell RNA sequencing (scRNA-seq) studies (Fig. 1B; P < 0.0001). When the expression status of individual GC oncogenes was assessed (e.g., CEACAM6, EGFR, MET, CCND1, and KRAS) among the tumor epithelial clusters, the EpiInt1 tumor epithelial cluster exhibited the largest increase (Fig. 1C). Comparisons in a cell state-specific manner revealed that "chief cells" and "intestinal-type cells" showed the greatest differences in transcriptomic profiles between GC and their cognate cell state in normal gastric tissues (Fig. 1E; "chief cells": 1,232 genes/EpiC3 cluster; "intestinal-type cells": 775 genes/EpiInt2 cluster). Pathway analysis indicated that each tumor epithelial cluster shared both common upregulated modules relevant to cancer, but also cluster-specific tumor-associated modules. Among downregulated genes, the classic chief cell markers LIPF and PGA3 were among the top downregulated genes in tumor epithelial cells (Fig. 1F; P < 0.0001). Using spatial DSP, the loss of LIPF was confirmed in tumor epithelial cells (Pan-CK+) compared with normal samples (n = 13, P = 0.0035; Fig. 1G).

A, Density plot of UMAP representation comparing normal and gastric tumor samples after random downsampling to approximately 30,000 cells each to allow statistical equivalence. B, Split violin plot of EMT oncogenic gene signature score in normal and tumor cells, showing a significantly higher score in tumor cells. C, Bubble plot depicting the expression of GC oncogenes in tumor epithelial cell clusters. D, Box plot depicting CNV scores for epithelial cells (green) and macrophage cells (blue) in normal and tumor samples. E, Bar graph depicting differences in transcriptomic profiles between tumor and normal tissue by the number of upregulated and downregulated genes in epithelial cell clusters. F, Heat map of LIPF and PGA3 gene expression (classic chief cell marker genes) in tumor versus normal samples. G, Gene expression of LIPF by DSP analysis, in tumor epithelial cells (Pan-CK+) compared with normal samples (n = 13).Fig. 1 scRNA-seq deconvolutes gastric tumor programs associated with distinct cell states. (Kumar V, et al, 2022)

Neutrophils Promote Migration and Invasiveness and EMT of GC Cells

To study the effect of neutrophils on GC cells, a Transwell migration/invasion assay was used to explore the effect of neutrophils on the migration and invasion ability of GC cells. The results showed that neutrophils promote GC cell (MKN45 and MKN74) migration (P < 0.001 and P < 0.001) and invasion (P < 0.001 and P < 0.001) (Fig. 2a and b), when an IL-17a neutralizing antibody was added to the Transwell co-culture chamber, GC cell (MKN45 and MKN74) migration (P < 0.001 and P < 0.001) and invasion was decreased (P < 0.001 and P < 0.001) (Fig. 2a and b). Therefore, neutrophil promotes the migration and invasiveness of GC cells through IL-17a.

Western blot results showed that neutrophils promote the expression of EMT-related markers in GC cells (MKN45 and MKN74). E-cadherin expression was significantly decreased (P < 0.05 and P < 0.05 in MKN45 and MKN74 cells, respectively), and Vimentin (P < 0.05 and P < 0.05 in MKN45 and MKN74 cells, respectively) and ZEB1 (P < 0.05 and P < 0.05 in MKN45 and MKN74 cells, respectively) expression was significantly upregulated (Fig. 2c) when an IL-17a neutralizing antibody was added to the Transwell co-culture system, The expression of E-cadherin protein in the epithelium of GC cells was upregulated (P < 0.05 and P < 0.05 in MKN45 and MKN74 cells, respectively), while the expression of Vimentin protein in the interstitial space decreased significantly (P < 0.05 and P < 0.05 in MKN45 and MKN74 cells, respectively). The expression of the transcription factor ZEB1 was also significantly downregulated (P < 0.05 and P < 0.05 in MKN45 and MKN74 cells, respectively) (Fig. 2c). Therefore, neutrophils promote EMT in GC cells, and further promote the migration and invasiveness of GC cells, which may be mediated by IL-17a.

(a, b) The effect of neutrophils on the migration and invasion ability of GC cells (MKN45 and MKN74) was determined 24 h when IL-17a neutralizing antibody or IgG isotype control antibody was added to Transwell co-culture chamber. c Protein expression of E-cadherin, Vimentin, and ZEB1 in GC cells (MKN45 and MKN74) co-cultured with neutrophils were analyzed by western blot when IL-17a neutralizing antibody or IgG isotype control antibody was added to the Transwell co-culture system.Fig. 2 Neutrophils enhance the migration, invasiveness, and EMT of GC cells through IL-17a. (Li S, et al., 2019)

How are gastric tumor cell lines developed and utilized?

Gastric tumor cell lines are established by isolating and culturing cells directly from the tumors of GC patients. These cell lines are extensively characterized to ensure they faithfully represent the properties of the original tumors.

How do gastric tumor cells differ from normal gastric epithelial cells?

These include uncontrolled proliferation and disregard for normal growth signals; increased invasive and metastatic behavior; altered gene expression profiles, such as mutations in key oncogenes and tumor suppressor genes; metabolic reprogramming to support rapid growth and survival; increased expression of cell surface markers associated with cancer stem cells.

What are the key signaling pathways that are dysregulated in the transformation of normal gastric cells into tumor cells?

Some of the critical signaling pathways involved in this process include PI3K/Akt/mTOR, Ras/MAPK, Wnt/β-catenin, Notch signaling, and TGF-β/Smad pathways.

Species:
Tissue:
Keywords:

Description: Species: human female 55 years old;
Tissue: colon;
Tumor: adenocarcinoma

Cat#: CSC-6251W INQUIRY

Description: Species: human, Caucasian female 82 years old;
Tissue: colon;
Tumor: adenocarcinoma, grade IV

Cat#: CSC-6316W INQUIRY

Description: Established from the primary tumor of a 72-year-old man with gastric adenocarcinoma in 1987

Cat#: CSC-C0324 INQUIRY

Description: Established from the poorly differentiated adenocarcinoma of the stomach (medullary...

Cat#: CSC-C0453 INQUIRY

Description: Established in 1989 from the primary tumor of a 72-year-old Hispanic man with a poorly...

Cat#: CSC-C0682 INQUIRY

Description: Species: human female 69 years;
Tissue: colon;
Tumor: carcinoma;
Derived from: pelvic wall metastasis

Cat#: CSC-C2204 INQUIRY

Description: Human cell line derived from stomach cancer (gastric cancer; highly differentiated tubular adenocarcinoma).

Cat#: CSC-C6250J INQUIRY

Description: Rectal carcinoma. Culture cell line of SCC.

Cat#: CSC-C6252J INQUIRY
GSS

Description: Human gastric cancer cell line derived from metastasis at liver.

Cat#: CSC-C6316J INQUIRY
GSU

Description: Human cell line derived from gastric cancer. Established from the cells in ascites.

Cat#: CSC-C6317J INQUIRY

Description: Human cell line derived from stomach cancer.

Cat#: CSC-C6321J INQUIRY

Description: Human cell line derived from signet ring cell carcinoma of stomach.

Cat#: CSC-C6343J INQUIRY

Description: Human cell line derived from intraperitoneal metastasis of gastric cancer

Cat#: CSC-C6375J INQUIRY

Description: Human cell line derived from gastric cancer

Cat#: CSC-C6378J INQUIRY

Description: Stomach cancer, but producing hybrid type of alkaline phosphatase.

Cat#: CSC-C6407J INQUIRY

Description: Stomach cancer, but producing Nagao-type alkaline phosphatase.

Cat#: CSC-C6408J INQUIRY

Description: Japanese gastric cancer, lymph node metastated.

Cat#: CSC-C6413J INQUIRY

Description: Human cell line derived from lymph node metastasis of gastric cancer.

Cat#: CSC-C6416J INQUIRY

For research use only. Not for any other purpose.