Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
Br J Cancer ; 117(1): 65-77, 2017 Jun 27.
Article in English | MEDLINE | ID: mdl-28524160

ABSTRACT

BACKGROUND: The HMGA2 protein has experimentally been linked to EMT and cancer stemness. Recent studies imply that tumour-stroma interactions regulate these features and thereby contribute to tumour aggressiveness. METHODS: We analysed 253 cases of pancreatic ductal adenocarcinoma (PDAC) and 155 cases of ampullary adenocarcinoma (AAC) for HMGA2 expression by IHC. The data were correlated with stroma abundance and supplemented by experimental studies. RESULTS: HMGA2 acts as an independent prognostic marker associated with a significantly shorter overall survival in both tumour types. Overall, HMGA2-positivity was more frequent in patients with PDAC than with AAC. The HMGA2 status in tumour cells significantly correlated with the abundance of PDGFRß-defined stroma cells. In vivo co-injection of Panc-1 cancer cells with pancreatic stellate cells increased tumour growth in a manner associated with increased HMGA2 expression. Furthermore, in vitro treatment of Panc-1 with conditioned media from PDGF-BB-activated stellate cells increased their ability to form tumour spheroids. CONCLUSIONS: This study identifies HMGA2 expression in tumour cells as an independent prognostic marker in PDAC and AAC. Correlative data analysis gives novel tissue-based evidence for a heterotypic cross-talk with stroma cells as a possible mechanism for HMGA2 induction, which is further supported by experimental models.


Subject(s)
Adenocarcinoma/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Common Bile Duct Neoplasms/metabolism , HMGA2 Protein/metabolism , Pancreatic Neoplasms/metabolism , Adenocarcinoma/pathology , Aged , Ampulla of Vater , Animals , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Common Bile Duct Neoplasms/pathology , Female , Humans , Immunohistochemistry , Male , Mice , Mice, SCID , Middle Aged , Multivariate Analysis , Neoplasm Staging , Neoplasm Transplantation , Pancreatic Neoplasms/pathology , Pancreatic Stellate Cells/metabolism , Prognosis , Receptor, Platelet-Derived Growth Factor beta/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stromal Cells/metabolism , Survival Rate
2.
BMC Cancer ; 7: 224, 2007 Dec 12.
Article in English | MEDLINE | ID: mdl-18076756

ABSTRACT

BACKGROUND: Platelet-derived growth factor (PDGF)-BB and PDGF receptor (PDGFR)-beta are mainly expressed in the developing vasculature, where PDGF-BB is produced by endothelial cells and PDGFR-beta is expressed by mural cells, including pericytes. PDGF-BB is produced by most types of solid tumors, and PDGF receptor signaling participates in various processes, including autocrine stimulation of tumor cell growth, recruitment of tumor stroma fibroblasts, and stimulation of tumor angiogenesis. Furthermore, PDGF-BB-producing tumors are characterized by increased pericyte abundance and accelerated tumor growth. Thus, there is a growing interest in the development of tumor treatment strategies by blocking PDGF/PDGFR function. We have recently generated a mouse model carrying an activated PDGFR-beta by replacing the highly conserved aspartic acid residue (D) 849 in the activating loop with asparagine (N). This allowed us to investigate, in an orthotopic tumor model, the role of increased stromal PDGFR-beta signaling in tumor-stroma interactions. METHODS: B16 melanoma cells lacking PDGFR-beta expression and either mock-transfected or engineered to express PDGF-BB, were injected alone or in combination with matrigel into mice carrying the activated PDGFR-beta (D849N) and into wild type mice. The tumor growth rate was followed and the vessel status of tumors, i.e. total vessel area/tumor, average vessel surface and pericyte density of vessels, was analyzed after resection. RESULTS: Tumors grown in mice carrying an activated PDGFR-beta were established earlier than those in wild-type mice. In this early phase, the total vessel area and the average vessel surface were higher in tumors grown in mice carrying the activated PDGFR-beta (D849N) compared to wild-type mice, whereas we did not find a significant difference in the number of tumor vessels and the pericyte abundance around tumor vessels between wild type and mutant mice. At later phases of tumor progression, no significant difference in tumor growth rate was observed between wild type mice and mutant mice, although the pericyte coverage was higher around tumor vessels from mutant mice. CONCLUSION: Our findings suggest that the activated PDGFR-beta (D849N) in the host animal increased the total vessel area and the average vessel surface even in PDGF-negative tumors, resulting in a shorter lag phase during tumor establishment.


Subject(s)
Gene Expression Regulation, Neoplastic/genetics , Melanoma, Experimental/genetics , Mutation/genetics , Receptor, Platelet-Derived Growth Factor beta/genetics , Animals , Melanoma, Experimental/metabolism , Mice , Mice, Mutant Strains , Receptor, Platelet-Derived Growth Factor beta/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...