INVESTIGADORES
ALANIZ Laura Daniela
artículos
Título:
Hepatocellular Carcinoma Cells and Their Fibrotic Microenvironment Modulate Bone Marrow-Derived Mesenchymal Stromal Cell Migration in Vitro and in Vivo
Autor/es:
GARCIA, MARIANA G.; BAYO, JUAN; BOLONTRADE, MARCELA F.; SGANGA, LEONARDO; MALVICINI, MARIANA; ALANIZ, LAURA; AQUINO, JORGE B.; FIORE, ESTEBAN; RIZZO, MANGLIO M.; RODRIGUEZ, ANDRÉS; LORENTI, ALICIA; ANDRIANI, OSCAR; PODHAJCER, OSVALDO; MAZZOLINI, GUILLERMO
Revista:
MOLECULAR PHARMACEUTICS
Editorial:
AMER CHEMICAL SOC
Referencias:
Año: 2011 vol. 8 p. 1538 - 1548
ISSN:
1543-8384
Resumen:
Hepatocellular carcinoma (HCC) is the fifth most common cancer and the third cause of cancer-related death. Fibrogenesis is an active process characterized by the production of several proinflammatory cytokines, chemokines and growth factors. It involves the activation of hepatic stellate cells (HSCs) which accumulate at the site of injury and are the main source of the extracellular matrix deposits. There are no curative treatments for advanced HCC, thus, new therapies are urgently needed. Mesenchymal stromal cells (MSCs) have the ability to migrate to sites of injury or to remodeling tissues after in vivo administration; however, in several cancer models they demonstrated limited efficacy to eradicate experimental tumors partially due to poor engraftment. Thus, the aim of this work was to analyze the capacity of human MSCs (hMSCs) to migrate and anchor to HCC tumors. We observed that HCC and HSCs, but not nontumoral stroma, produce factors that induce hMSC migration in vitro. Conditioned media (CM) generated from established HCC cell lines were found to induce higher levels of hMSC migration than CM derived from fresh patient tumor samples. In addition, after exposure to CM from HCC cells or HSCs, hMSCs demonstrated adhesion and invasion capability to endothelial cells, type IV collagen and fibrinogen. Consistently, these cells were found to increase metalloproteinase-2 activity. In vivo studies with subcutaneous and orthotopic HCC models indicated that intravenously infused hMSCs migrated to lungs, spleen and liver. Seven days post-hMSC infusion cells were located also in the tumor in both models, but the signal intensity was significantly higher in orthotopic than in subcutaneous model. Interestingly, when orthotopic HCC tumors where established in noncirrhotic or cirrhotic livers, the amount of hMSCs localized in the liver was higher in comparison with healthy animals. A very low signal was found in lungs and spleens, indicating that liver tumors are able to recruit them at high efficiency. Taken together our results indicate that HCC and HSC cells produce factors that efficiently induce hMSC migration toward tumor microenvironment in vitro and in vivo and make MSCs candidates for cell-based therapeutic strategies to hepatocellular carcinoma associated with fibrosis