12 research outputs found

    The Role of the Tumor Microenvironment in the Development and Progression of Hepatocellular Carcinoma

    Get PDF
    There is a growing evidence that supports the role of the tumor microenvironment (TME) in the development and progression of cancer. TME is composed of cellular components, bioactive substances (e.g. growth factors) and extracellular matrix (ECM) comprising of proteins such as collagens, proteoglycans and the linear glycosaminoglycan hyaluronan, which is a key component of ECM. Hepatocellular carcinoma (HCC), generally arises from fibrotic or cirrhotic liver, characterized by excessive expression and alteration of ECM components which facilitates tumor development. On the other hand, non-tumoral cells, as such as the mesenchymal stem/stromal cells (MSCs) are typically recruited to the injured or hypoxic area within the tumor. Besides the secretion of immunoregulatory, growth factors and cytokines, MSCs and hepatic stellate cells (HSCs) can also synthesize hyaluronan, amongst other components, that affects several tumor processes. The TME also contains different types of immune cells. A key component in tumorigenesis in HCC are the macrophages, as tumor-associated macrophages (TAM). This chapter will describe specific data regarding the interaction of MSCs-hyaluronan-TAMs and tumor cells and how this interaction potentially contributes to the development and progression of HCC.Fil: Sevic, Ina. Consejo Nacional de Investigaciones Científicas y Técnicas. Centro de Investigaciones y Transferencia del Noroeste de la Provincia de Buenos Aires. Universidad Nacional del Noroeste de la Provincia de Buenos Aires. Centro de Investigaciones y Transferencia del Noroeste de la Provincia de Buenos Aires; ArgentinaFil: Spinelli, Fiorella Mercedes. Consejo Nacional de Investigaciones Científicas y Técnicas. Centro de Investigaciones y Transferencia del Noroeste de la Provincia de Buenos Aires. Universidad Nacional del Noroeste de la Provincia de Buenos Aires. Centro de Investigaciones y Transferencia del Noroeste de la Provincia de Buenos Aires; ArgentinaFil: Cantero, María José. Consejo Nacional de Investigaciones Científicas y Técnicas; Argentina. Universidad Austral. Facultad de Ciencias Biomédicas. Instituto de Investigaciones en Medicina Traslacional. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Parque Centenario. Instituto de Investigaciones en Medicina Traslacional; ArgentinaFil: Reszegi, Andrea. Semmelweis University; HungríaFil: Kovalszky, Ilona. Semmelweis University; HungríaFil: García, Mariana Gabriela. Universidad Austral. Facultad de Ciencias Biomédicas. Instituto de Investigaciones en Medicina Traslacional. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Parque Centenario. Instituto de Investigaciones en Medicina Traslacional; Argentina. Consejo Nacional de Investigaciones Científicas y Técnicas; ArgentinaFil: Alaniz, Laura Daniela. Consejo Nacional de Investigaciones Científicas y Técnicas. Centro de Investigaciones y Transferencia del Noroeste de la Provincia de Buenos Aires. Universidad Nacional del Noroeste de la Provincia de Buenos Aires. Centro de Investigaciones y Transferencia del Noroeste de la Provincia de Buenos Aires; Argentin

    SPOCK1 Promotes the Development of Hepatocellular Carcinoma

    Get PDF
    The extracellular matrix proteoglycan SPOCK1 is increasingly recognized as a contributor to the development and progression of cancers. Here, we study how SPOCK1, which is present in non-tumorous hepatocytes at low concentrations, promotes the development and progression of malignant hepatocellular tumors. Although SPOCK1 is an extracellular matrix proteoglycan, its concentration increases in the cytoplasm of hepatocytes starting with very low expression in the normal cells and then appearing in much higher quantities in cells of cirrhotic human liver and hepatocellular carcinoma. This observation is similar to that observed after diethylnitrosamine induction of mouse hepatocarcinogenesis. Furthermore, syndecan-1, the major proteoglycan of the liver, and SPOCK1 are in inverse correlation in the course of these events. In hepatoma cell lines, the cytoplasmic SPOCK1 colocalized with mitochondrial markers, such as MitoTracker and TOMM20, a characteristic protein of the outer membrane of the mitochondrion and could be detected in the cell nucleus. SPOCK1 downregulation of hepatoma cell lines by siRNA inhibited cell proliferation, upregulated p21 and p27, and interfered with pAkt and CDK4 expression. A tyrosine kinase array revealed that inhibition of SPOCK1 in the liver cancer cells altered MAPK signaling and downregulated several members of the Sarc family, all related to the aggressivity of the hepatoma cell lines. These studies support the idea that SPOCK1 enhancement in the liver is an active contributor to human and rodent hepatocarcinogenesis and cancer progression. However, its mitochondrial localization raises the possibility that it has a currently unidentified physiological function in normal hepatocytes

    Humán FXIII-B sushi domének expresszálása rovarsejtekben

    No full text
    Bevezetés: A XIII-as faktor egy protranszglutamináz, mely fontos szerepet tölt be a stabil véralvadék kialakulásában. A plazmában FXIII-A és FXIII-B alegységekből álló heterotetramer (FXIII-A2B2) formában kering. A FXIII-A alegység a katalitikusan aktív alegység, trombin és Ca2+ hatására aktiválódik, miközben a védő, szállító B alegység eltávozik. A FXIII-A homodimer formában megtalálható különböző sejtekben is. A FXIII-B alegységet, mely 10 sushi doménből épül fel, a májsejtek szekretálják, a két alegység kapcsolódása nagy valószínűséggel a plazmában történik. Amíg a FXIII-A struktúrája és funkciója intenzíven kutatott terület, viszonylag keveset tudunk a FXIII-B szerkezetéről, funkciójáról és a két alegység kapcsolódásában szerepet játszó struktúrákról. A korábbi irodalmi adatok alapján a két alegység kapcsolódásáért felelős szakasz a FXIII-B alegységben az első két sushi domén valamelyikében található. Célkitűzés: Munkánk során a FXIII-B 1-es sushi doménjéből és a szignál szekvenciából álló rekombináns FXIII-B fragmens (pp-s1), előállítását tűztük ki célul. Anyagok és módszerek: Az expressziót bakulovírus expressziós rendszerrel végeztük pFastBac vektor és sf9 sejtvonal alkalmazásával. Első lépésben a FXIII-B alegység első sushi doménjét az azt megelőző szignál (propeptid) szekvenciával együtt (pp-s1) PCR reakcióval felsokszoroztuk, majd beépítettük a vektorba. Az elkészített rekombináns plazmidot kompetens E. coli baktériumba transzformáltuk. A sikeresen transzformált baktériumokból plazmidokat izoláltunk, majd szekvenálással ellenőríztük, hogy tartalmazzák-e a pp-s1-et. A pp-s1-et tartalmazó plazmiddal sf9 rovarsejtvonalat transzfektáltunk. Eredmények és diszkusszió: A rovarsejtek mikroszkópban vizsgálva muttták a vírusfertőzés tipikus jeleit, azonban a sejtek felülúszójából ill. lizátumából még nem sikerült kimutatni a sushi domén jelenlétét Western blotting technikával. A FXIII-B specifikus poliklonális antitest nem adott reakciót, azonban a használt poliklonális antitest epitóp specificitása nem ismert, ezért csak a pozitív eredmény jelentene biztos eredményt. A továbbiakban más antitestek, ill. His-TAG ellenes antitest alkalmazásával kívánjuk igazolni a rekombináns fehérje jelenlétét. Nagyobb mennyiségű, tisztított sushi domén előállítása után lehetőség nyílik kötődési vizsgálatok (ELISA, felületi plazmon rezonancia) elvégzésére, az alegységek kapcsolódásáért felelős struktúra azonosítására.BSc/BAáltalános orvostudományimagyarOrvosi kutatólaboratóriumi analitikanappaliP.J

    SPOCK1 Overexpression Suggests Poor Prognosis of Ovarian Cancer

    No full text
    Purpose: Sparc/osteonectin, cwcv, and kazal-like domains proteoglycan 1 (SPOCK1) has been found in a variety of malignant tumors and is associated with a poor prognosis. We aimed to explore the role of SPOCK1 in ovarian cancer. Methods: Ovarian cancer cell lines SKOV3 and SW626 were transfected with SPOCK1 overexpressing or empty vector using electroporation. Cells were studied by immunostaining and an automated Western blotting system. BrdU uptake and wound healing assays assessed cell proliferation and migration. SPOCK1 expression in human ovarian cancer tissues and in blood samples were studied by immunostaining and ELISA. Survival of patients with tumors exhibiting low and high SPOCK1 expression was analyzed using online tools. Results: Both transfected cell lines synthesized different SPOCK1 variants; SKOV3 cells also secreted the proteoglycan. SPOCK1 overexpression stimulated DNA synthesis and cell migration involving p21CIP1. Ovarian cancer patients had increased SPOCK1 serum levels compared to healthy controls. Tumor cells of tissues also displayed abundant SPOCK1. Moreover, SPOCK1 levels were higher in untreated ovarian cancer serum and tissue samples and lower in recipients of chemotherapy. According to in silico analyses, high SPOCK1 expression was correlated with shorter survival. Conclusion: Our findings suggest SPOCK1 may be a viable anti-tumor therapeutic target and could be used for monitoring ovarian cancer

    Syndecan-1 inhibits early stages of liver fibrogenesis by interfering with TGFbeta1 action and upregulating MMP14

    No full text
    Increased expression of syndecan-1 is a characteristic feature of human liver cirrhosis. However, no data are available on the significance of this alteration. To address this question we designed a transgenic mouse strain that driven by albumin promoter, expresses human syndecan-1 in the hepatocytes. Liver cirrhosis was induced by thioacetamide in wild type and hSDC1(+/+) mice of the identical strain. The process of fibrogenesis, changes in signal transduction and proteoglycan expression were followed. In an in vitro experiment, the effect of syndecan-1 overexpression on the action of TGFbeta1 was determined. Human syndecan-1 and TGFbeta1 levels were measured by ELISA in the circulation. Without challenge, no morphological differences were observed between wild type and transgenic mice livers, although significant upregulation of phospho-Akt and FAK was observed in the latter. Fibrogenesis in the transgenic livers, characterized by picrosirius staining, collagen type I, and SMA levels, lagged behind that of control in the first and second months. Changes in signal transduction involved in the process of fibrogenesis, as SMAD, MAPK, Akt and GSK, pointed to the decreased effect of TGFbeta1, and this was corroborated by the decreased mRNA expression of TIEG and the growth factor itself. In vitro experiments exposing the LX2 hepatic stellate cell line to conditioned media of wild type and syndecan-1 transfected Hep3B cell lines proved that medium with high syndecan-1 content inhibits TGFbeta1-induced upregulation of SMA, TIEG, collagen type I and thrombospondin-1 expression. Detection of liver proteoglycans and heparan sulfate level revealed that their amounts are much higher in control transgenic liver, than that in the wild type. However, it decreases dramatically as a result of shedding after hepatic injury. Shedding is likely promoted by the upregulation of MMP14. As syndecan-1 can bind thrombospondin-1, and as our result demonstrated that the same is true for TGFbeta1, shed syndecan-1 can remove the growth factor and its activator together into the systemic circulation.Taking together, our results indicate that the effect of syndecan-1 is accomplished on two levels: a, the shedded syndecan can bind, inhibit and remove TGFbeta1; b, interferes with the activation of TGFbeta1 by downregulation and binding thrombospondin-1, the activator of the growth factor. However, by the end of the fourth month the protective effect was lost, which is explained by the considerable decrease of syndecan-1 and the almost complete loss of heparan sulfate from the surface of hepatocytes

    Two ways of epigenetic silencing of TFPI2 in cervical cancer.

    No full text
    ObjectiveComparison of human mRNA microarray results from tumor-associated and normal cervical fibroblasts revealed significant TFPI2 downregulation in tumor-associated fibroblasts isolated from cervical cancer, indicating that TFPI2 downregulation may play an important role in the pathogenesis of the disease. In the present work, we investigated the mechanism of TFPI2 downregulation in tumor-associated fibroblasts and tumor cells.MethodsIn vitro models of monocultures and co-cultures were established with tumor cells and fibroblasts to explore the changes of TFPI-2 expression and epigenetic modifications of the TFPI2 gene.ResultsThe TFPI2 gene was hypermethylated only in tumor cells. Reduction of TFPI-2 protein levels in tumor-associated fibroblasts, although the gene was not methylated, suggested alternative regulatory mechanisms of gene expression, such as inhibition by microRNAs. The expression pattern of miR-23a, a gene thought to inhibit TFPI2 translation, showed changes strongly correlated to detected TFPI-2 protein alterations. Transfections with miR-23a mimics resulted in a decrease of TFPI-2 protein expression whereas miR-23a inhibitors increased the TFPI-2 amount. Due to downregulation of miR-23a expression by HPV in cancer cells, TFPI2 was silenced by promoter methylation. In contrary, miR-23a was active in HPV-free fibroblasts and inactivated TFPI2.ConclusionThese results indicate dual epigenetic inhibition of TFPI2 on the transcription level by promoter methylation in cancer cells and on the translation level by miR-23a in tumor-associated fibroblasts. As a consequence, inactivation of the TFPI2 gene plays a strategic role in the progression of cervical cancer

    Dynamic Interplay in Tumor Ecosystems: Communication between Hepatoma Cells and Fibroblasts

    No full text
    Tumors are intricate ecosystems where cancer cells and non-malignant stromal cells, including cancer-associated fibroblasts (CAFs), engage in complex communication. In this study, we investigated the interaction between poorly (HLE) and well-differentiated (HuH7) hepatoma cells and LX2 fibroblasts. We explored various communication channels, including soluble factors, metabolites, extracellular vesicles (EVs), and miRNAs. Co-culture with HLE cells induced LX2 to produce higher levels of laminin β1, type IV collagen, and CD44, with pronounced syndecan-1 shedding. Conversely, in HuH7/LX2 co-culture, fibronectin, thrombospondin-1, type IV collagen, and cell surface syndecan-1 were dominant matrix components. Integrins α6β4 and α6β1 were upregulated in HLE, while α5β1 and αVβ1 were increased in HuH7. HLE-stimulated LX2 produced excess MMP-2 and 9, whereas HuH7-stimulated LX2 produced excess MMP-1. LX2 activated MAPK and Wnt signaling in hepatoma cells, and conversely, hepatoma-derived EVs upregulated MAPK and Wnt in LX2 cells. LX2-derived EVs induced over tenfold upregulation of SPOCK1/testican-1 in hepatoma EV cargo. We also identified liver cancer-specific miRNAs in hepatoma EVs, with potential implications for early diagnosis. In summary, our study reveals tumor type-dependent communication between hepatoma cells and fibroblasts, shedding light on potential implications for tumor progression. However, the clinical relevance of liver cancer-specific miRNAs requires further investigation

    Overexpression of Human Syndecan-1 Protects against the Diethylnitrosamine-Induced Hepatocarcinogenesis in Mice

    Get PDF
    Although syndecan-1 (SDC1) is known to be dysregulated in various cancer types, its implication in tumorigenesis is poorly understood. Its effect may be detrimental or protective depending on the type of cancer. Our previous data suggest that SDC1 is protective against hepatocarcinogenesis. To further verify this notion, human SDC1 transgenic (hSDC1+/+) mice were generated that expressed hSDC1 specifically in the liver under the control of the albumin promoter. Hepatocarcinogenesis was induced by a single dose of diethylnitrosamine (DEN) at an age of 15 days after birth, which resulted in tumors without cirrhosis in wild-type and hSDC1+/+ mice. At the experimental endpoint, livers were examined macroscopically and histologically, as well as by immunohistochemistry, Western blot, receptor tyrosine kinase array, phosphoprotein array, and proteomic analysis. Liver-specific overexpression of hSDC1 resulted in an approximately six month delay in tumor formation via the promotion of SDC1 shedding, downregulation of lipid metabolism, inhibition of the mTOR and the β-catenin pathways, and activation of the Foxo1 and p53 transcription factors that lead to the upregulation of the cell cycle inhibitors p21 and p27. Furthermore, both of them are implicated in the regulation of intermediary metabolism. Proteomic analysis showed enhanced lipid metabolism, activation of motor proteins, and loss of mitochondrial electron transport proteins as promoters of cancer in wild-type tumors, inhibited in the hSDC1+/+ livers. These complex mechanisms mimic the characteristics of nonalcoholic steatohepatitis (NASH) induced human liver cancer successfully delayed by syndecan-1
    corecore