Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Cancer Lett ; 591: 216901, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38641311

RESUMEN

Diffuse-type gastric cancer (DGC) is a subtype of gastric cancer that is prone to peritoneal dissemination, with poor patient prognosis. Although intercellular adhesion loss between cancer cells is a major characteristic of DGCs, the mechanism underlying the alteration in cell-to-extracellular matrix (ECM) adhesion is unclear. We investigated how DGCs progress and cause peritoneal dissemination through interactions between DGC cells and the tumour microenvironment (TME). P53 knockout and KRASG12V-expressing (GAN-KP) cells and Cdh1-deleted GAN-KP (GAN-KPC) cells were orthotopically transplanted into the gastric wall to mimic peritoneal dissemination. The GAN-KPC tumour morphology was similar to that of human DGCs containing abundant stroma. RNA sequencing revealed that pathways related to Rho GTPases and integrin-ECM interactions were specifically increased in GAN-KPC cells compared with GAN-KP cells. Notably, we found that Rac Family Small GTPase 1 (RAC1) induces Integrin Subunit Alpha 6 (ITGA6) trafficking, leading to its enrichment on the GC cell membrane. Fibroblasts activate the FAK/AKT pathway in GC cells by mediating extracellular matrix (ECM)-Itga6 interactions, exacerbating the malignant phenotype. In turn, GC cells induce abnormal expression of fibroblast collagen and its transformation into cancer-associated fibroblasts (CAFs), resulting in DGC-like subtypes. These findings indicate that Cdh1 gene loss leads to abnormal expression and changes in the subcellular localization of ITGA6 through RAC1 signalling. The latter, through interactions with CAFs, allows for peritoneal dissemination.


Asunto(s)
Cadherinas , Neoplasias Peritoneales , Neoplasias Gástricas , Microambiente Tumoral , Proteína de Unión al GTP rac1 , Humanos , Neoplasias Gástricas/patología , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Cadherinas/metabolismo , Cadherinas/genética , Proteína de Unión al GTP rac1/metabolismo , Proteína de Unión al GTP rac1/genética , Línea Celular Tumoral , Neoplasias Peritoneales/secundario , Neoplasias Peritoneales/genética , Neoplasias Peritoneales/metabolismo , Neoplasias Peritoneales/patología , Animales , Antígenos CD/metabolismo , Antígenos CD/genética , Ratones , Transducción de Señal , Células del Estroma/metabolismo , Células del Estroma/patología , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Adhesión Celular , Regulación Neoplásica de la Expresión Génica
2.
Cell Rep ; 43(1): 113613, 2024 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-38232734

RESUMEN

Malignant ascites accompanied by peritoneal dissemination contain various factors and cell populations as well as cancer cells; however, how the tumor microenvironment is shaped in ascites remains unclear. Single-cell proteomic profiling and a comprehensive proteomic analysis are conducted to comprehensively characterize malignant ascites. Here, we find defects in immune effectors along with immunosuppressive cell accumulation in ascites of patients with gastric cancer (GC) and identify five distinct subpopulations of CD45(-)/EpCAM(-) cells. Mesothelial cells with mesenchymal features in CD45(-)/EpCAM(-) cells are the predominant source of chemokines involved in immunosuppressive myeloid cell (IMC) recruitment. Moreover, mesothelial-mesenchymal transition (MMT)-induced mesothelial cells strongly express extracellular matrix (ECM)-related genes, including tenascin-C (TNC), enhancing metastatic colonization. These findings highlight the definite roles of the mesenchymal cell population in the development of a protumorigenic microenvironment to promote peritoneal dissemination.


Asunto(s)
Ascitis , Neoplasias Peritoneales , Humanos , Ascitis/patología , Molécula de Adhesión Celular Epitelial , Proteómica , Peritoneo/patología , Neoplasias Peritoneales/patología , Línea Celular Tumoral , Microambiente Tumoral
3.
JHEP Rep ; 5(12): 100892, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37942226

RESUMEN

Background & Aims: Hepatocellular carcinoma (HCC) mainly develops from chronic hepatitis. Metabolic dysfunction-associated steatohepatitis (MASH) has gradually become the main pathogenic factor for HCC given the rising incidence of obesity and metabolic diseases. 15-Hydroxyprostaglandin dehydrogenase (15-PGDH) degrades prostaglandin 2 (PGE2), which is known to exacerbate inflammatory responses. However, the role of PGE2 accumulation caused by 15-PGDH downregulation in the development of MASH-HCC has not been determined. Methods: We utilised the steric animal model to establish a MASH-HCC model using wild-type and 15-Pgdh+/- mice to assess the significance of PGE2 accumulation in the development of MASH-HCC. Additionally, we analysed clinical samples obtained from patients with MASH-HCC. Results: PGE2 accumulation in the tumour microenvironment induced the production of reactive oxygen species in macrophages and the expression of cell growth-related genes and antiapoptotic genes. Conversely, the downregulation of fatty acid metabolism in the background liver promoted lipid accumulation in the tumour microenvironment, causing a decrease in mitochondrial membrane potential and CD8+ T-cell exhaustion, which led to enhanced development of MASH-HCC. Conclusions: 15-PGDH downregulation inactivates immune surveillance by promoting the proliferation of exhausted effector T cells, which enhances hepatocyte survival and proliferation and leads to the development of MASH-HCC. Impact and implications: The suppression of PGE2-related inflammation and subsequent lipid accumulation leads to a reduction in the severity of MASH and inhibition of subsequent progression toward MASH-HCC.

4.
Cancer Cell ; 41(12): 2019-2037.e8, 2023 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-37890493

RESUMEN

Intestinal metaplasia (IM) is a pre-malignant condition of the gastric mucosa associated with increased gastric cancer (GC) risk. Analyzing 1,256 gastric samples (1,152 IMs) across 692 subjects from a prospective 10-year study, we identify 26 IM driver genes in diverse pathways including chromatin regulation (ARID1A) and intestinal homeostasis (SOX9). Single-cell and spatial profiles highlight changes in tissue ecology and IM lineage heterogeneity, including an intestinal stem-cell dominant cellular compartment linked to early malignancy. Expanded transcriptome profiling reveals expression-based molecular subtypes of IM associated with incomplete histology, antral/intestinal cell types, ARID1A mutations, inflammation, and microbial communities normally associated with the healthy oral tract. We demonstrate that combined clinical-genomic models outperform clinical-only models in predicting IMs likely to transform to GC. By highlighting strategies for accurately identifying IM patients at high GC risk and a role for microbial dysbiosis in IM progression, our results raise opportunities for GC precision prevention and interception.


Asunto(s)
Lesiones Precancerosas , Neoplasias Gástricas , Humanos , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Estudios Prospectivos , Mucosa Gástrica/patología , Genómica , Metaplasia/genética , Lesiones Precancerosas/genética
5.
Cancer Res ; 83(5): 753-770, 2023 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-36543251

RESUMEN

Excess stroma and cancer-associated fibroblasts (CAF) enhance cancer progression and facilitate immune evasion. Insights into the mechanisms by which the stroma manipulates the immune microenvironment could help improve cancer treatment. Here, we aimed to elucidate potential approaches for stromal reprogramming and improved cancer immunotherapy. Platelet-derived growth factor C (PDGFC) and D expression were significantly associated with a poor prognosis in patients with gastric cancer, and PDGF receptor beta (PDGFRß) was predominantly expressed in diffuse-type gastric cancer stroma. CAFs stimulated with PDGFs exhibited markedly increased expression of CXCL1, CXCL3, CXCL5, and CXCL8, which are involved in polymorphonuclear myeloid-derived suppressor cell (PMN-MDSC) recruitment. Fibrotic gastric cancer xenograft tumors exhibited increased PMN-MDSC accumulation and decreased lymphocyte infiltration, as well as resistance to anti-PD-1. Single-cell RNA sequencing and spatial transcriptomics revealed that PDGFRα/ß blockade reversed the immunosuppressive microenvironment through stromal modification. Finally, combining PDGFRα/ß blockade and anti-PD-1 treatment synergistically suppressed the growth of fibrotic tumors. These findings highlight the impact of stromal reprogramming on immune reactivation and the potential for combined immunotherapy for patients with fibrotic cancer. SIGNIFICANCE: Stromal targeting with PDGFRα/ß dual blockade reverses the immunosuppressive microenvironment and enhances the efficacy of immune checkpoint inhibitors in fibrotic cancer. See related commentary by Tauriello, p. 655.


Asunto(s)
Receptor alfa de Factor de Crecimiento Derivado de Plaquetas , Neoplasias Gástricas , Humanos , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Fibrosis , Inmunoterapia , Microambiente Tumoral
6.
Gastric Cancer ; 26(2): 187-202, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36513910

RESUMEN

BACKGROUND: Remodeling the tumor microenvironment (TME) to benefit cancer cells is crucial for tumor progression. Although diffuse-type gastric cancer (DGC) preferentially interacts with the TME, the precise mechanism of the complicated network remains unknown. This study aimed to investigate the mutual activation mechanism underlying DGC progression. METHODS: Mass cytometry analysis of co-cultured macrophages, noncancerous fibroblasts (NFs), and DGC cells was performed. RNA sequencing was applied to examine gene expression in fibroblasts. DGC cells were treated with cytokines to examine their effect on characteristic changes. The TCGA and Kumamoto University cohorts were used to evaluate the clinical relevance of the in vitro findings. RESULTS: Cohort analysis revealed that DGC patients had a poor prognosis. The fibroblasts and macrophages interacted with DGC cells to form a cell cluster in the invasive front of DGC tissue. The original 3D triple co-culture system determined the promotional effects of nonmalignant cells on DGC invasive growth. We notably identified a mixed-polarized macrophage cell type with M1/M2 cell surface markers in a triple co-culture system. IL-1ß from mixed-polarized macrophages induced the conversion of NFs to cancer-associated fibroblast-like (CAF-like) cells, promoting the malignant phenotype of DGC cells by inducing the secretion of IL-6, IL-24, and leukemia inhibitory factor (LIF). Moreover, IL-6 and colony stimulating factor 2 (GM-CSF) cooperated to maintain the stable state of mixed-polarized macrophages. Finally, we found that mixed-polarized macrophages were frequently detected in DGC tissues. CONCLUSION: These findings demonstrated that mixed-polarized macrophages exist as a novel subtype through the reciprocal interaction between DGC cells and nonmalignant cells.


Asunto(s)
Interleucina-6 , Neoplasias Gástricas , Humanos , Interleucina-6/metabolismo , Interleucina-6/farmacología , Microambiente Tumoral , Neoplasias Gástricas/patología , Macrófagos/metabolismo , Fibroblastos
7.
FEBS J ; 290(10): 2604-2615, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36565059

RESUMEN

Fibroblast activation protein (FAP) generally shows low or undetectable expression in most normal tissues but is highly expressed in fibroblasts in almost all carcinomas. FAP is one of the potential molecules to detect activated fibroblasts and has multiple roles in tumour progression. We generated transgenic mice that specifically expressed tdTomato along with FAP promoter activity. Coculturing a mouse gastric cancer cell line and FAP-tdTomato transgenic mouse-derived fibroblasts showed that tdTomato expression was elevated in the cocultured fibroblasts. Moreover, stomach wall transplanted tumours in mice also showed FAP-tdTomato expression in fibroblasts of the stomach and each metastatic legion. These results indicated that FAP-tdTomato expression in fibroblasts was elevated by stimulation through the interaction with cancer cells. Functionally, collagen production was increased in FAP/tdTomato-positive fibroblasts cocultured with mouse cancer cells. These FAP-tdTomato transgenic mice have the potential to be used to investigate real-time FAP dynamics and the importance of FAP expression in tumour development.


Asunto(s)
Fibroblastos Asociados al Cáncer , Neoplasias Gástricas , Animales , Ratones , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Endopeptidasas/genética , Endopeptidasas/metabolismo , Ratones Transgénicos , Fibroblastos Asociados al Cáncer/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Fibroblastos/metabolismo , Neoplasias Gástricas/patología , Proteína Fluorescente Roja
8.
Cancer Sci ; 113(10): 3579-3592, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35848891

RESUMEN

The arachidonic acid cascade is a major inflammatory pathway that produces prostaglandin E2 (PGE2). Although inhibition of 15-hydroxyprostaglandin dehydrogenase (15-PGDH) is reported to lead to PGE2 accumulation, the role of 15-PGDH expression in the tumor microenvironment remains unclear. We utilized Panc02 murine pancreatic cancer cells for orthotopic transplantation into wild-type and 15-pgdh+/- mice and found that 15-pgdh depletion in the tumor microenvironment leads to enhanced tumorigenesis accompanied by an increase in cancer-associated fibroblasts (CAFs) and the promotion of fibrosis. The fibrotic tumor microenvironment is widely considered to be hypovascular; however, we found that the angiogenesis level is maintained in 15-pgdh+/- mice, and these changes were also observed in a genetically engineered PDAC mouse model. Further confirmation revealed that fibroblast growth factor 1 (FGF1) is secreted by pancreatic cancer cells after PGE2 stimulation, consequently promoting CAF proliferation and vascular endothelial growth factor A (VEGFA) expression in the tumor microenvironment. Finally, in 15-pgdh+/- Acta2-TK mice, depletion of fibroblasts inhibited angiogenesis and cancer cell viability in orthotopically transplanted tumors. These findings highlighted the role of 15-pgdh downregulation in enhancing PGE2 accumulation in the pancreatic tumor microenvironment and in subsequently maintaining the angiogenesis level in fibrotic tumors along with CAF expansion.


Asunto(s)
Neoplasias Pancreáticas , Factor A de Crecimiento Endotelial Vascular , Animales , Ácido Araquidónico , Línea Celular Tumoral , Dinoprostona/metabolismo , Dinoprostona/farmacología , Factor 1 de Crecimiento de Fibroblastos , Fibrosis , Hidroxiprostaglandina Deshidrogenasas/genética , Hidroxiprostaglandina Deshidrogenasas/metabolismo , Ratones , Neoplasias Pancreáticas/genética , Microambiente Tumoral , Factor A de Crecimiento Endotelial Vascular/genética , Neoplasias Pancreáticas
9.
In Vivo ; 36(4): 1982-1985, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35738623

RESUMEN

BACKGROUND: Appendicitis caused by a foreign body is extremely rare. We report a case of chronic appendicitis caused by a perforating fish bone. CASE REPORT: The patient was a 50-year-old Japanese man. He felt dull lower abdominal pain for 2 months and diagnosed as appendicitis caused by a perforating fish bone. He underwent emergency laparoscopic surgery. The fish bone had perforated through the appendix wall. The fish bone was initially removed followed by laparoscopic appendectomy. Pathological investigation revealed a transmural cut line of approximately 0.5 mm that was surrounded by fibrous tissue with inflammation. CONCLUSION: This is the first reported case of fish bone-induced chronic appendicitis that underwent laparoscopic appendectomy. For optimum outcome, a correct diagnosis based on a detailed consultation and imaging tests, and an operation performed after careful planning are needed.


Asunto(s)
Apendicitis , Laparoscopía , Dolor Abdominal/cirugía , Animales , Apendicectomía/efectos adversos , Apendicitis/diagnóstico , Apendicitis/etiología , Apendicitis/cirugía , Humanos , Laparoscopía/efectos adversos , Masculino , Alimentos Marinos
10.
Clin J Gastroenterol ; 15(3): 598-602, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35312955

RESUMEN

The accidental ingestion of foreign bodies is a common clinical issue. While most foreign bodies pass through the gastrointestinal (GI) tract without complications, a few cases unfortunately result in GI perforation. Fish bones are one of the most frequent foreign bodies found in the GI tract, and they are high-risk objects for GI perforation due to their hard and sharp-pointed ends. Here, we present a rare case of a 64-year-old man with perforation of a colorectal tumor by a fish bone. The patient received emergency Hartmann's operation with lymph node dissection. Although the patient experienced recurrence in the liver rather than peritoneal dissemination, systemic chemotherapy was considerably effective, and conversion therapy with hepatectomy was successfully performed; the patient achieved 5-year relapse-free survival after the operation. To our knowledge, this is the first report of the perforation of a GI tumor by a fish bone. This rare case suggests the significant clinical implication that proper preoperative diagnosis and prompt surgical treatment lead to better postoperative outcomes for patients with tumor perforation by a foreign body.


Asunto(s)
Neoplasias Colorrectales , Cuerpos Extraños , Perforación Intestinal , Animales , Neoplasias Colorrectales/complicaciones , Neoplasias Colorrectales/cirugía , Cuerpos Extraños/complicaciones , Cuerpos Extraños/cirugía , Humanos , Perforación Intestinal/etiología , Perforación Intestinal/cirugía , Recurrencia Local de Neoplasia
11.
Gastric Cancer ; 25(3): 542-557, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35166958

RESUMEN

BACKGROUND: Signet ring cell carcinoma (SRCC) is a particular histologic variant of gastric cancer (GC). However, the critical factor related to the aggressive characteristics of SRCC has not been determined. METHODS: We collected surgically resected tissues from 360 GC patients in the Kumamoto University cohort and generated survival curves via the Kaplan-Meier method. In vitro, we identified the specific transcript variant of MUC20 in SRCC cells by direct sequencing and investigated the role of MUC20 in GC progression using GC cells with MUC20 silencing and forced expression. In vivo, we examined chemoresistance using MUC20 variant 2 (MUC20v2)-overexpressing non-SRCC cells to construct a xenograft mouse model. RESULTS: We analyzed a comprehensive GC cell line database to identify the specifically expressed genes in gastric SRCC. We focused on MUC20 and investigated its role in GC progression. Survival analysis revealed that GC patients with high MUC20 expression exhibited a poor prognosis and that MUC20 expression was significantly correlated with SRCC histological type. Moreover, we found that gastric SRCC cells specifically expressed MUC20v2, which was dominantly expressed in the cytoplasm. Silencing MUC20v2 caused cell death with characteristic morphological changes in gastric SRCC cells. To further determine the types of cell death, we examined apoptosis, pyroptosis and ferroptosis by detecting cleaved PARP, gasdermin E-N-terminal (GSDME-N), and lipid reactive oxygen species (ROS) levels, respectively. We found that apoptosis and pyroptosis occurred in MUC20-silenced gastric SRCC cells. In addition, MUC20v2-overexpressing GC cells exhibited chemoresistance to cisplatin (CDDP) and paclitaxel (PTX). RNA sequencing revealed that the pathways involved in intracellular calcium regulation were significantly upregulated in MUC20v2-overexpressing GC cells. Notably, forced expression of MUC20v2 in the cytoplasm of GC cells led to the maintenance of mitochondrial calcium homeostasis and mitochondrial membrane potential (MMP), which promoted cell survival and chemoresistance by suppressing apoptosis and pyroptosis. Finally, we investigated the significance of MUC20v2 in a xenograft model treated with CDDP and showed that MUC20v2 overexpression caused chemoresistance by inhibiting cell death. CONCLUSION: These findings highlight the novel functions of MUC20v2, which may confer cell survival and drug resistance in GC cells. SIGNIFICANCE: MUC20v2 protects GC cells from apoptosis and pyroptosis by maintaining mitochondrial calcium levels and mitochondrial membrane potential and subsequently induces drug resistance.


Asunto(s)
Carcinoma de Células en Anillo de Sello , Neoplasias Gástricas , Animales , Calcio/uso terapéutico , Carcinoma de Células en Anillo de Sello/patología , Cisplatino , Resistencia a Medicamentos , Xenoinjertos , Homeostasis , Humanos , Ratones , Mucinas , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo
12.
Anticancer Res ; 42(1): 381-384, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34969748

RESUMEN

BACKGROUND: A desmoid tumor is a rare neoplasm that is derived from soft tissues. Although it shows benign characteristics pathologically, local recurrence can occur. CASE REPORT: We herein report the case of a patient with an intraabdominal desmoid tumor that developed 3 years after laparoscopic appendectomy for acute appendicitis. A 59-year-old male visited our emergency room with complaints of abdominal pain and fullness. Abdominal computed tomography revealed distention of the small intestine with a point of obstruction by an intraabdominal tumor-like region. Pathological findings showed that the tumor was compatible with desmoid fibromatosis. CONCLUSION: In cases with an intraabdominal tumor after laparoscopic surgery, it is important to consider the possibility of a desmoid tumor, since it is difficult to diagnose it accurately before surgery.


Asunto(s)
Dolor Abdominal/diagnóstico , Laparoscopía/efectos adversos , Recurrencia Local de Neoplasia/diagnóstico , Neoplasias de los Tejidos Blandos/diagnóstico , Dolor Abdominal/patología , Fibromatosis Abdominal/complicaciones , Fibromatosis Abdominal/patología , Fibromatosis Abdominal/cirugía , Fibromatosis Agresiva/patología , Humanos , Obstrucción Intestinal/complicaciones , Obstrucción Intestinal/patología , Obstrucción Intestinal/cirugía , Masculino , Mesenterio/patología , Persona de Mediana Edad , Recurrencia Local de Neoplasia/patología , Neoplasias de los Tejidos Blandos/patología
13.
Cancer Lett ; 523: 29-42, 2021 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-34508795

RESUMEN

Cancer cells craftily adapt their energy metabolism to their microenvironment. Nutrient deprivation due to hypovascularity and fibrosis is a major characteristic of pancreatic ductal adenocarcinoma (PDAC); thus, PDAC cells must produce energy intrinsically. However, the enhancement of energy production via activating Kras mutations is insufficient to explain the metabolic rewiring of PDAC cells. Here, we investigated the molecular mechanism underlying the metabolic shift in PDAC cells under serine starvation. Amino acid analysis revealed that the concentrations of all essential amino acids and most nonessential amino acids were decreased in the blood of PDAC patients. In addition, the plasma serine concentration was significantly higher in PDAC patients with PHGDH-high tumors than in those with PHGDH-low tumors. Although the growth and tumorigenesis of PK-59 cells with PHGDH promoter hypermethylation were significantly decreased by serine starvation, these activities were maintained in PDAC cell lines with PHGDH promoter hypomethylation by serine biosynthesis through PHGDH induction. In fact, DNA methylation analysis by pyrosequencing revealed that the methylation status of the PHGDH promoter was inversely correlated with the PHGDH expression level in human PDAC tissues. In addition to PHGDH induction by serine starvation, PDAC cells showed enhanced serine biosynthesis under serine starvation through 3-PG accumulation via PGAM1 knockdown, resulting in enhanced PDAC cell growth and tumor growth. However, PHGDH knockdown efficiently suppressed PDAC cell growth and tumor growth under serine starvation. These findings provide evidence that targeting the serine biosynthesis pathway by inhibiting PHGDH is a potent therapeutic approach to eliminate PDAC cells in nutrient-deprived microenvironments.


Asunto(s)
Carcinoma Ductal Pancreático/patología , Ácidos Glicéricos/metabolismo , Neoplasias Pancreáticas/patología , Fosfoglicerato-Deshidrogenasa/fisiología , Serina/biosíntesis , Animales , Línea Celular Tumoral , Islas de CpG , Metilación de ADN , Inducción Enzimática , Humanos , Ratones , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/metabolismo , Fosfoglicerato-Deshidrogenasa/antagonistas & inhibidores , Fosfoglicerato-Deshidrogenasa/genética , Fosfoglicerato Mutasa/fisiología
14.
Anticancer Res ; 41(6): 2895-2900, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-34083280

RESUMEN

BACKGROUND/AIM: Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignant tumor. Research using an innovative research approach is needed to identify effective biomarkers or therapeutic targets for PDAC. We aimed to identify proteins related to the peritoneal dissemination of PDAC. MATERIALS AND METHODS: We performed proteomic analysis using ascites samples from patients with advanced PDAC and peritoneal dissemination and patients with liver cirrhosis (LC). Proteins specific to PDAC were identified in comparison to the findings for ascites from patients with LC as a control group. RESULTS: In total, 336 proteins were identified in ascites from patients with PDAC. We identified 18 specific proteins in ascites from patients with advanced PDAC. Among these proteins, CD13, lymphatic vessel endothelial hyaluronan receptor 1, ficolin-3, and V-set and immunoglobulin domain containing 4 were the most frequently detected. In addition, these 18 proteins could be classified into four categories: extracellular matrix, immunity, metabolism, and others. CONCLUSION: The identified proteins could be informative for developing treatment strategies for patients with PDAC and peritoneal dissemination.


Asunto(s)
Ascitis/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Neoplasias Pancreáticas/metabolismo , Proteómica/métodos , Ascitis/patología , Biomarcadores de Tumor/metabolismo , Carcinoma Ductal Pancreático/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pancreáticas/patología
15.
Cell Rep ; 34(8): 108779, 2021 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-33626356

RESUMEN

In the tumor microenvironment, senescent non-malignant cells, including cancer-associated fibroblasts (CAFs), exhibit a secretory profile under stress conditions; this senescence-associated secretory phenotype (SASP) leads to cancer progression and chemoresistance. However, the role of senescent CAFs in metastatic lesions and the molecular mechanism of inflammation-related SASP induction are not well understood. We show that pro-inflammatory cytokine-driven EZH2 downregulation maintains the SASP by demethylating H3K27me3 marks in CAFs and enhances peritoneal tumor formation of gastric cancer (GC) through JAK/STAT3 signaling in a mouse model. A JAK/STAT3 inhibitor blocks the increase in GC cell viability induced by senescent CAFs and peritoneal tumor formation. Single-cell mass cytometry revealed that fibroblasts exist in the ascites of GC patients with peritoneal dissemination, and the fibroblast population shows p16 expression and SASP factors at high levels. These findings provide insights into the inflammation-related SASP maintenance by histone modification and the role of senescent CAFs in GC peritoneal dissemination.


Asunto(s)
Fibroblastos Asociados al Cáncer/enzimología , Citocinas/metabolismo , Mediadores de Inflamación/metabolismo , Neoplasias Peritoneales/metabolismo , Fenotipo Secretor Asociado a la Senescencia , Neoplasias Gástricas/metabolismo , Anciano , Animales , Antineoplásicos/farmacología , Fibroblastos Asociados al Cáncer/patología , Línea Celular Tumoral , Citocinas/genética , Proteína Potenciadora del Homólogo Zeste 2/genética , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Inhibidores de las Cinasas Janus/farmacología , Quinasas Janus/metabolismo , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Neoplasias Peritoneales/tratamiento farmacológico , Neoplasias Peritoneales/genética , Neoplasias Peritoneales/secundario , Piridinas/farmacología , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Microambiente Tumoral , Tirfostinos/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Cancer Sci ; 111(10): 3468-3477, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33044028

RESUMEN

The effectiveness of current chemotherapies for cancer is gradually progressing; however achieving a complete cure through chemotherapy is still difficult and has been the main goal in treatment of advanced cancer. Drug resistance is an issue in cancer therapy, therefore increasing numbers of investigations into drug resistance have focused on the characteristics of the cancer cells themselves. The interaction between the tumor microenvironment (TME) and cancer cells is also intimately involved in the development of drug resistance. Cancer-associated fibroblasts (CAFs) are a predominant component of the TME and affect tumor progression by secreting soluble factors. This review summarizes the most up-to-date knowledge of CAFs and drug resistance in cancer, with a focus on factors secreted from CAFs including proteins, cytokines, extracellular vesicles, and metabolites. A perspective on the potential role of anti-CAF therapies in overcoming CAF-induced drug resistance is also discussed.


Asunto(s)
Fibroblastos Asociados al Cáncer/metabolismo , Resistencia a Medicamentos , Neoplasias/tratamiento farmacológico , Humanos , Neoplasias/patología , Microambiente Tumoral
17.
Langenbecks Arch Surg ; 405(8): 1111-1118, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32860110

RESUMEN

PURPOSE: Advanced esophageal cancer often results in esophageal stenosis or tracheoesophageal fistula. Esophageal bypass surgery and esophageal stent insertion are palliative treatments for esophageal cancer. With improvements in metallic stents and the stent insertion technique, esophageal stent insertion appears to be performed more frequently than bypass surgery, worldwide. The aim of this study was to evaluate the outcomes of bypass surgery and stent insertion in our hospital and reevaluate which patients would benefit from bypass surgery. METHODS: A total of 70 esophageal cancer patients who could not tolerate oral feeding due to esophageal stenosis or tracheoesophageal fistula underwent palliative treatment [esophageal bypass surgery (N = 34) and esophageal stent insertion (N = 36)] at Kumamoto University. We retrospectively investigated the clinicopathological factors, postoperative outcomes, and complications. RESULTS: Both treatments could significantly improve the amount of food intake and the dietary form (P < 0.01). The length of hospital stay was shorter (P < 0.01) and complications associated with treatment were reduced in the stent group (P = 0.03). The overall survival did not differ significantly between the groups (log rank P = 0.22). Importantly, in the bypass surgery group, the patients who received postoperative treatment had a better prognosis than those who did not receive postoperative treatment (log rank P < 0.01). CONCLUSION: Both bypass surgery and stent insertion allowed oral intake in patients who could not tolerate oral feeding because of esophageal stenosis or tracheoesophageal fistula. Considering that patients who undergo stent insertion have a shorter hospital stay and fewer complications, stent insertion may be a better first choice for treatment than bypass surgery. However, bypass surgery may be an option for patients who can tolerate postoperative treatment.


Asunto(s)
Neoplasias Esofágicas , Estenosis Esofágica , Neoplasias Esofágicas/cirugía , Estenosis Esofágica/etiología , Estenosis Esofágica/cirugía , Humanos , Cuidados Paliativos , Estudios Retrospectivos , Stents , Resultado del Tratamiento
18.
Cancer Res ; 80(16): 3222-3235, 2020 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-32605995

RESUMEN

Extracellular vesicles (EV) from cancer-associated fibroblasts (CAF) are composed of diverse payloads. Although CAFs impact the aggressive characteristics of gastric cancer cells, the contribution of CAF-EV to gastric cancer progression has not been elucidated. Here, we investigated the molecular mechanism of the changes in gastric cancer characteristics induced by CAF-EV. CAF abundance in gastric cancer tissues was associated with poor prognosis of patients with gastric cancer receiving chemotherapy. Moreover, CAF-EV induced tubular network formation and drug resistance of gastric cancer cells in the extracellular matrix (ECM). Comprehensive proteomic analysis of CAF-EV identified that Annexin A6 plays a pivotal role in network formation and drug resistance of gastric cancer cells in the ECM via activation of ß1 integrin-focal adhesion kinase (FAK)-YAP. A peritoneal metastasis mouse model revealed that CAF-EV induced drug resistance in peritoneal tumors, and inhibition of FAK or YAP efficiently attenuated gastric cancer drug resistance in vitro and in vivo. These findings demonstrate that drug resistance is conferred by Annexin A6 in CAF-EV and provide a potential avenue for overcoming gastric cancer drug resistance through the inhibition of FAK-YAP signaling in combination with conventional chemotherapeutics. SIGNIFICANCE: This study elucidates a novel molecular mechanism through which Annexin A6 in CAF-EV activates FAK-YAP by stabilizing ß1 integrin at the cell surface of gastric cancer cells and subsequently induces drug resistance. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/16/3222/F1.large.jpg.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Anexina A6/metabolismo , Fibroblastos Asociados al Cáncer/metabolismo , Resistencia a Antineoplásicos , Vesículas Extracelulares/metabolismo , Quinasa 1 de Adhesión Focal/metabolismo , Integrina beta1/metabolismo , Neoplasias Gástricas/metabolismo , Factores de Transcripción/metabolismo , Animales , Secuencia de Bases , Línea Celular Tumoral , Progresión de la Enfermedad , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Humanos , Estimación de Kaplan-Meier , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Pronóstico , Reacción en Cadena en Tiempo Real de la Polimerasa , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/mortalidad , Neoplasias Gástricas/patología , Proteínas Señalizadoras YAP
19.
Biomedicines ; 8(1)2020 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-31935894

RESUMEN

Gastric cancer (GC) is a leading cause of cancer-related death worldwide. Cancer stem cells (CSCs) are known to be involved in chemotherapy resistance and the development of metastases. Although CSCs harbor self-renewal and tumorigenic abilities, the immune microenvironment surrounding CSCs provides various factors and supports the maintenance of CSC properties. The current review summarizes the accumulating findings regarding the relationship between the immune microenvironment and gastric CSCs (GCSCs), which will support the possibility of developing novel therapeutic strategies for targeting GCSCs.

20.
World J Surg ; 44(3): 831-837, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31701157

RESUMEN

BACKGROUND: Anatomical esophageal position may affect the short-term outcomes after minimally invasive esophagectomy (MIE). A previous single-institutional retrospective study suggested that the presence of a left-sided esophagus (LSE) made MIE more difficult and increased the incidence of postoperative complications. METHODS: The current study was a multicenter retrospective study of 303 patients with esophageal cancer who underwent MIE at six esophageal cancer high-volume centers in Kyushu, Japan, between April 2011 and August 2016. The patients were divided into the LSE (66 patients) and non-LSE groups (237 patients) based on the esophageal position on computed tomography images obtained with the patients in the supine position. RESULTS: Univariate analysis showed that patients with LSE were significantly older than those with non-LSE (69 ± 8 vs. 65 ± 9 years; P = 0.002), had a significantly greater incidence of cardiovascular comorbidity (65.2% vs. 47.7%; P = 0.013), and a significantly longer operating time (612 ± 112 vs. 579 ± 102 min; P = 0.025). Logistic regression analysis verified that LSE was an independent risk factor for the incidence of pneumonia (odds ratio 3.3, 95% confidence interval 1.254-8.695; P = 0.016). CONCLUSIONS: The presence of a LSE can increase the procedural difficulty of MIE and the incidence of morbidity after MIE. Thus, careful attention must be paid to anatomical esophageal position before performing MIE.


Asunto(s)
Neoplasias Esofágicas/cirugía , Esofagectomía/métodos , Procedimientos Quirúrgicos Mínimamente Invasivos/métodos , Anciano , Esofagectomía/efectos adversos , Femenino , Humanos , Incidencia , Masculino , Persona de Mediana Edad , Procedimientos Quirúrgicos Mínimamente Invasivos/efectos adversos , Complicaciones Posoperatorias/epidemiología , Estudios Retrospectivos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...