Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Am J Pathol ; 194(5): 828-848, 2024 May.
Article in English | MEDLINE | ID: mdl-38320632

ABSTRACT

Cancer-associated fibroblasts (CAFs) in the tumor microenvironment are involved in the progression of various cancers, including esophageal squamous cell carcinoma (ESCC). CAF-like cells were generated through direct co-culture of human bone marrow-derived mesenchymal stem cells, one of CAF origins, with ESCC cells. Periostin (POSTN) was found to be highly expressed in CAF-like cells. After direct co-culture, ESCC cells showed increased malignant phenotypes, such as survival, growth, and migration, as well as increased phosphorylation of Akt and extracellular signal-regulated kinase (Erk). Recombinant human POSTN activated Akt and Erk signaling pathways in ESCC cells, enhancing survival and migration. The suppression of POSTN in CAF-like cells by siRNA during direct co-culture also suppressed enhanced survival and migration in ESCC cells. In ESCC cells, knockdown of POSTN receptor integrin ß4 inhibited Akt and Erk phosphorylation, and survival and migration increased by POSTN. POSTN also enhanced mesenchymal stem cell and macrophage migration and endowed macrophages with tumor-associated macrophage-like properties. Immunohistochemistry showed that high POSTN expression in the cancer stroma was significantly associated with tumor invasion depth, lymphatic and blood vessel invasion, higher pathologic stage, CAF marker expression, and infiltrating tumor-associated macrophage numbers. Moreover, patients with ESCC with high POSTN expression exhibited poor postoperative outcomes. Thus, CAF-secreted POSTN contributed to tumor microenvironment development. These results indicate that POSTN may be a novel therapeutic target for ESCC.


Subject(s)
Cancer-Associated Fibroblasts , Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Humans , Cancer-Associated Fibroblasts/metabolism , Cell Line, Tumor , Cell Movement , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Periostin , Proto-Oncogene Proteins c-akt/metabolism , Tumor Microenvironment
2.
Cells ; 12(22)2023 11 10.
Article in English | MEDLINE | ID: mdl-37998338

ABSTRACT

Tumor-associated macrophages (TAMs), one of the major components of the tumor microenvironment, contribute to the progression of esophageal squamous cell carcinoma (ESCC). We previously established a direct co-culture system of human ESCC cells and macrophages and reported the promotion of malignant phenotypes, such as survival, growth, and migration, in ESCC cells. These findings suggested that direct interactions between cancer cells and macrophages contribute to the malignancy of ESCC, but its underlying mechanisms remain unclear. In this study, we compared the expression levels of the interferon-induced genes between mono- and co-cultured ESCC cells using a cDNA microarray and found that interferon-inducible protein 16 (IFI16) was most significantly upregulated in co-cultured ESCC cells. IFI16 knockdown suppressed malignant phenotypes and also decreased the secretion of interleukin-1α (IL-1α) from ESCC cells. Additionally, recombinant IL-1α enhanced malignant phenotypes of ESCC cells through the Erk and NF-κB signaling. Immunohistochemistry revealed that high IFI16 expression in human ESCC tissues tended to be associated with disease-free survival and was significantly associated with tumor depth, lymph node metastasis, and macrophage infiltration. The results of this study reveal that IFI16 is involved in ESCC progression via IL-1α and imply the potential of IFI16 as a novel prognostic factor for ESCC.


Subject(s)
Carcinoma, Squamous Cell , Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Humans , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/pathology , Interferons/metabolism , Interleukin-1alpha/metabolism , Macrophages/metabolism , Neoplastic Processes , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Tumor Microenvironment
3.
J Pathol ; 261(1): 55-70, 2023 09.
Article in English | MEDLINE | ID: mdl-37436683

ABSTRACT

M2 macrophages contribute to the progression of oesophageal squamous cell carcinoma (ESCC); however, the roles of M2 macrophages in early ESCC remain unclear. To clarify the biological mechanisms underlying the interaction between M2 macrophages and oesophageal epithelial cells in early-stage ESCC, in vitro co-culture assays between the immortalised oesophageal epithelial cell line Het-1A and cytokine-defined M2 macrophages were established. Co-culture with M2 macrophages promoted the proliferation and migration of Het-1A cells via the mTOR-p70S6K signalling pathway activated by YKL-40, also known as chitinase 3-like 1, and osteopontin (OPN) that were hypersecreted in the co-culture supernatants. YKL-40 and OPN promoted the above phenotypes of Het-1A by making a complex with integrin ß4 (ß4). Furthermore, YKL-40 and OPN promoted M2 polarisation, proliferation, and migration of macrophages. To validate the pathological and clinical significances of in vitro experimental results, immunohistochemistry of human early ESCC tissues obtained by endoscopic submucosal dissection (ESD) was performed, confirming the activation of the YKL-40/OPN-ß4-p70S6K axis in the tumour area. Moreover, epithelial expression of ß4 and the number of epithelial and stromal infiltrating YKL-40- and OPN-positive cells correlated with the Lugol-voiding lesions (LVLs), a well-known predictor of the incidence of metachronous ESCC. Furthermore, the combination of high expression of ß4 and LVLs or high numbers of epithelial and stromal infiltrating YKL-40- and OPN-positive immune cells could more clearly detect the incidence of metachronous ESCC than each of the parameters alone. Our results demonstrated that the YKL-40/OPN-ß4-p70S6K axis played important roles in early-stage ESCC, and the high expression levels of ß4 and high numbers of infiltrating YKL-40- and OPN-positive immune cells could be useful predictive parameters for the incidence of metachronous ESCC after ESD. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Humans , Esophageal Squamous Cell Carcinoma/metabolism , Integrin beta4/metabolism , Osteopontin/genetics , Osteopontin/metabolism , Esophageal Neoplasms/pathology , Chitinase-3-Like Protein 1/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Clinical Relevance , Macrophages/pathology , Cell Line, Tumor
4.
Cancers (Basel) ; 15(11)2023 May 30.
Article in English | MEDLINE | ID: mdl-37296952

ABSTRACT

Tumor-associated macrophages (TAMs) contribute to disease progression in various cancers, including esophageal squamous cell carcinoma (ESCC). We have previously used an indirect co-culture system between ESCC cell lines and macrophages to analyze their interactions. Recently, we established a direct co-culture system to closely simulate actual ESCC cell-TAM contact. We found that matrix metalloproteinase 9 (MMP9) was induced in ESCC cells by direct co-culture with TAMs, not by indirect co-culture. MMP9 was associated with ESCC cell migration and invasion, and its expression was controlled by the Stat3 signaling pathway in vitro. Immunohistochemical analyses revealed that MMP9 expression in cancer cells at the invasive front ("cancer cell MMP9") was related to high infiltration of CD204 positive M2-like TAMs (p < 0.001) and was associated with worse overall and disease-free survival of patients (p = 0.036 and p = 0.038, respectively). Furthermore, cancer cell MMP9 was an independent prognostic factor for disease-free survival. Notably, MMP9 expression in cancer stroma was not associated with any clinicopathological factors or patient prognoses. Our results suggest that close interaction with TAMs infiltrating in cancer stroma or cancer nests induces MMP9 expression in ESCC cells, equipping them with more malignant features.

5.
Cancers (Basel) ; 15(2)2023 Jan 06.
Article in English | MEDLINE | ID: mdl-36672342

ABSTRACT

High infiltration of tumor-associated macrophages (TAMs), which contribute to the progression of several cancer types, is correlated with poor prognosis of esophageal squamous cell carcinoma (ESCC). In addition to the previously reported increase in migration and invasion, ESCC cells co-cultured directly with macrophages exhibited enhanced survival and growth. Furthermore, interleukin-related molecules are associated with ESCC; however, the precise mechanism underlying this association is unclear. Therefore, we explored the role of interleukin-related molecules in ESCC progression. A cDNA microarray analysis of monocultured and co-cultured ESCC cells revealed that the interleukin 7 receptor (IL-7R) was upregulated in ESCC cells co-cultured with macrophages. Overexpression of IL-7R promoted the survival and growth of ESCC cells by activating the Akt and Erk1/2 signaling pathways. The IL-7/IL-7R axis also contributed to the promotion of ESCC cell migration via the Akt and Erk1/2 signaling pathways. Furthermore, immunohistochemistry showed that ESCC patients with high IL-7R expression in cancer nests exhibited a trend toward poor prognosis in terms of disease-free survival, and showed significant correlation with increased numbers of infiltrating macrophages and cancer-associated fibroblasts. Therefore, IL-7R, which is upregulated when directly co-cultured with macrophages, may contribute to ESCC progression by promoting the development of various malignant phenotypes in cancer cells.

6.
Biomedicines ; 11(1)2023 Jan 10.
Article in English | MEDLINE | ID: mdl-36672681

ABSTRACT

Although a therapeutic response to neoadjuvant chemoradiotherapy (NACRT) is important to improve oncological outcomes after surgery in patients with locally advanced rectal cancer, there is no reliable predictor for this. The Wnt/ß-catenin signal is known to be crucial for the tumorigenesis of colorectal cancer. This study aimed to investigate the association of Wnt/ß-catenin signal activation with a pathological response to NACRT. The immunohistochemical expression of nuclear and membranous ß-catenin was analyzed in biopsy samples obtained from 60 patients with locally advanced rectal cancer who received curative surgery following NACRT. The association of Wnt/ß-catenin signal activation with their clinical outcomes was investigated. Notably, the body mass index of these patients was significantly higher in the low nuclear ß-catenin expression group. Moreover, patients in the high nuclear ß-catenin expression group tended to have more advanced disease and a higher rate of positive vascular invasion than those in the low expression group. Furthermore, the rate of good histological responses was significantly higher in the low nuclear ß-catenin expression group (72% vs. 37.1%, p < 0.01). Overall, relapse-free survival tended to be better in patients with low nuclear/high membranous ß-catenin expression (n = 9) than in other individuals (n = 51) (p = 0.093 and p = 0.214, respectively). Activation of the Wnt/ß-catenin signal pathway represented by nuclear ß-catenin accumulation was significantly associated with a poor response to NACRT in patients with rectal cancer. Analysis of nuclear ß-catenin accumulation before starting treatment might help predict the therapeutic response to NACRT.

7.
Gan To Kagaku Ryoho ; 50(13): 1370-1373, 2023 Dec.
Article in Japanese | MEDLINE | ID: mdl-38303278

ABSTRACT

Although a therapeutic response to neoadjuvant chemoradiotherapy(NACRT)is important to improve oncological outcomes after surgery in patients with locally advanced rectal cancer, there is no reliable predictor for this. The Wnt/ß-catenin signal is known to be crucial for the tumorigenesis of colorectal cancer. This study aimed to investigate the association between Wnt/ß-catenin signal activation and the response to NACRT in patients with locally advanced rectal cancer. We analyzed the expression of nuclear and membranous ß-catenin in biopsy samples obtained from 60 patients who underwent curative surgery following NACRT. We found that patients with low nuclear ß-catenin expression had a higher rate of good histological responses compared to those with high nuclear ß-catenin expression. Additionally, patients with low nuclear/high membranous ß-catenin expression tended to have better relapse-free survival. The activation of the Wnt/ß- catenin signal pathway, represented by nuclear ß-catenin accumulation, was significantly associated with a poor response to NACRT in patients with rectal cancer. The analysis of nuclear ß-catenin accumulation before starting treatment could potentially predict the therapeutic response to NACRT.


Subject(s)
Neoadjuvant Therapy , Rectal Neoplasms , Humans , beta Catenin/metabolism , Neoplasm Recurrence, Local/drug therapy , Rectal Neoplasms/drug therapy , Retrospective Studies , Treatment Outcome
8.
Gan To Kagaku Ryoho ; 46(4): 787-789, 2019 Apr.
Article in Japanese | MEDLINE | ID: mdl-31164536

ABSTRACT

A 61-year-old man was examined for cervical pain and CT showed a 9 cm tumor to the third part of the duodenum and proximal jejunum. CT /MRI showed that the tumor was separated from the pancreas body. We scheduled a laparoscopic partial resection of the intestine with a suspected diagnosis of GIST of the intestine. The tumor was adhered to both the proximal jejunum and uncinate process of the pancreas. Therefore, we converted to an open surgery and resected part of the pancreas, duodenum, and proximal jejunum including the tumor. Histopathological examination showed the tumor capsule included the tissue of the pancreas and that the border between the intestine and the tumor was clear, suggesting that the origin of the tumor was the pancreas. We diagnosed the patient as having a grade 2 pancreatic neuroendocrine tumor based on the tumor growth pattern and immunohistochemistry findings. We examined the preoperative CT images retrospectively and found that the tumor had adhered to the uncinate process of the pancreas, which extends over the left side of the superior mesenteric artery. When GIST close to the proximal jejunum is suspected, the possibility of pancreatic neuroendocrine tumor should be considered.


Subject(s)
Gastrointestinal Stromal Tumors , Jejunal Neoplasms , Pancreatic Neoplasms , Diagnosis, Differential , Gastrointestinal Stromal Tumors/diagnosis , Gastrointestinal Stromal Tumors/surgery , Humans , Jejunal Neoplasms/diagnosis , Jejunum , Male , Middle Aged , Pancreas , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/surgery , Retrospective Studies
9.
Gan To Kagaku Ryoho ; 45(13): 2390-2392, 2018 Dec.
Article in Japanese | MEDLINE | ID: mdl-30692474

ABSTRACT

A 73-year-old man visited our hospital with the chief complaints of anorexia and weight loss. Computed tomography showed a 5 cm tumor(diameter)in the sigmoid mesocolon. We performed laparoscopic Hartmann operation; however, because of sacral invasion of tumor, curative surgery was difficult. Therefore, the tumor was excised together with the sigmoid colon. Histopathological examination of the tumor confirmed the diagnosis of epithelioid sarcoma. Postoperatively, the patient received heavy-particle radiotherapy at another facility as treatment for the residual tumor. The size of the residual tumor decreased 6 months after the surgery. Up until 1 year after surgery, the patient's condition has remained stable without any disease progression. Epithelioid sarcoma is a rare soft-tissue tumor and often leads to a poor prognosis. We present a case of epithelioid sarcoma occurring in the sigmoid mesocolon.


Subject(s)
Mesocolon , Sarcoma , Soft Tissue Neoplasms , Aged , Colon, Sigmoid , Humans , Male , Mesocolon/diagnostic imaging , Mesocolon/pathology , Sarcoma/diagnostic imaging , Sarcoma/surgery , Soft Tissue Neoplasms/diagnostic imaging , Soft Tissue Neoplasms/surgery , Tomography, X-Ray Computed
SELECTION OF CITATIONS
SEARCH DETAIL
...