3 resultados para cancer cell

em ArchiMeD - Elektronische Publikationen der Universität Mainz - Alemanha


Relevância:

70.00% 70.00%

Publicador:

Resumo:

Mit der Zielsetzung der vorliegenden Arbeit wurde die detailierten Analyse von Migrationsdynamiken epithelilaler Monolayer anhand zweier neuartiger in vitro Biosensoren verfolgt, der elektrischen Zell-Substrat Impedanz Spektroskopie (electrical cell-substrate impedance sensing, ECIS) sowie der Quarz Kristall Mikrowaage (quartz crystal microbalance, QCM). Beide Methoden erwiesen sich als sensitiv gegenüber der Zellmotilität und der Nanozytotoxizität.rnInnerhalb des ersten Projektes wurde ein Fingerprinting von Krebszellen anhand ihrer Motilitätsdynamiken und der daraus generierten elektrischen oder akkustischen Fluktuationen auf ECIS oder QCM Basis vorgenommen; diese Echtzeitsensoren wurdene mit Hilfe klassicher in vitro Boyden-Kammer Migrations- und Invasions-assays validiert. Fluktuationssignaturen, also Langzeitkorrelationen oder fraktale Selbstähnlichkeit aufgrund der kollektiven Zellbewegung, wurden über Varianz-, Fourier- sowie trendbereinigende Fluktuationsanalyse quantifiziert. Stochastische Langzeitgedächtnisphänomene erwiesen sich als maßgebliche Beiträge zur Antwort adhärenter Zellen auf den QCM und ECIS-Sensoren. Des weiteren wurde der Einfluss niedermolekularer Toxine auf die Zytoslelettdynamiken verfolgt: die Auswirkungen von Cytochalasin D, Phalloidin und Blebbistatin sowie Taxol, Nocodazol und Colchicin wurden dabei über die QCM und ECIS Fluktuationsanalyse erfasst.rnIn einem zweiten Projektschwerpunkt wurden Adhäsionsprozesse sowie Zell-Zell und Zell-Substrat Degradationsprozesse bei Nanopartikelgabe charackterisiert, um ein Maß für Nanozytotoxizität in Abhangigkeit der Form, Funktionalisierung Stabilität oder Ladung der Partikel zu erhalten.rnAls Schlussfolgerung ist zu nennen, dass die neuartigen Echtzeit-Biosensoren QCM und ECIS eine hohe Zellspezifität besitzen, auf Zytoskelettdynamiken reagieren sowie als sensitive Detektoren für die Zellvitalität fungieren können.

Relevância:

70.00% 70.00%

Publicador:

Resumo:

Chemotherapy is a mainstay of cancer treatment. Due to increased drug resistance and the severe side effects of currently used therapeutics, new candidate compounds are required for improvement of therapy success. Shikonin, a natural naphthoquinone, was used in traditional Chinese medicine for the treatment of different inflammatory diseases and recent studies revealed the anticancer activities of shikonin. We found that shikonin has strong cytotoxic effects on 15 cancer cell lines, including multidrug-resistant cell lines. Transcriptome-wide mRNA expression studies showed that shikonin induced genetic pathways regulating cell cycle, mitochondrial function, levels of reactive oxygen species, and cytoskeletal formation. Taking advantage of the inherent fluorescence of shikonin, we analyzed its uptake and distribution in live cells with high spatial and temporal resolution using flow cytometry and confocal microscopy. Shikonin was specifically accumulated in the mitochondria, and this accumulation was associated with a shikonin-dependent deregulation of cellular Ca(2+) and ROS levels. This deregulation led to a breakdown of the mitochondrial membrane potential, dysfunction of microtubules, cell-cycle arrest, and ultimately induction of apoptosis. Seeing as both the metabolism and the structure of mitochondria show marked differences between cancer cells and normal cells, shikonin is a promising candidate for the next generation of chemotherapy.

Relevância:

70.00% 70.00%

Publicador:

Resumo:

Summary Antibody-based cancer therapies have been successfully introduced into the clinic and have emerged as the most promising therapeutics in oncology. The limiting factor regarding the development of therapeutical antibody vaccines is the identification of tumor-associated antigens. PLAC1, the placenta-specific protein 1, was categorized for the first time by the group of Prof. Sahin as such a tumor-specific antigen. Within this work PLAC1 was characterized using a variety of biochemical methods. The protein expression profile, the cellular localization, the conformational state and especially the interacting partners of PLAC1 and its functionality in cancer were analyzed. Analysis of the protein expression profile of PLAC1 in normal human tissue confirms the published RT-PCR data. Except for placenta no PLAC1 expression was detectable in any other normal human tissue. Beyond, an increased PLAC1 expression was detected in several cancer cell lines derived of trophoblastic, breast and pancreatic lineage emphasizing its properties as tumor-specific antigen. rnThe cellular localization of PLAC1 revealed that PLAC1 contains a functional signal peptide which conducts the propeptide to the endoplasmic reticulum (ER) and results in the secretion of PLAC1 by the secretory pathway. Although PLAC1 did not exhibit a distinct transmembrane domain, no unbound protein was detectable in the cell culture supernatant of overexpressing cells. But by selective isolation of different cellular compartments PLAC1 was clearly enriched within the membrane fraction. Using size exclusion chromatography PLAC1 was characterized as a highly aggregating protein that forms a network of high molecular multimers, consisting of a mixture of non-covalent as well as covalent interactions. Those interactions were formed by PLAC1 with itself and probably other cellular components and proteins. Consequently, PLAC1 localize outside the cell, where it is associated to the membrane forming a stable extracellular coat-like structure.rnThe first mechanistic hint how PLAC1 promote cancer cell proliferation was achieved identifying the fibroblast growth factor FGF7 as a specific interacting partner of PLAC1. Moreover, it was clearly shown that PLAC1 as well as FGF7 bind to heparin, a glycosaminoglycan of the ECM that is also involved in FGF-signaling. The participation of PLAC1 within this pathway was approved after co-localizing PLAC1, FGF7 and the FGF7 specific receptor (FGFR2IIIb) and identifying the formation of a trimeric complex (PLAC1, FGF7 and the specific receptor FGFR2IIIb). Especially this trimeric complex revealed the role of PLAC1. Binding of PLAC1 together with FGF7 leads to the activation of the intracellular tyrosine kinase of the FGFR2IIIb-receptor and mediate the direct phosphorylation of the AKT-kinase. In the absence of PLAC1, no FGF7 mediated phosphorylation of AKT was observed. Consequently the function of PLAC1 was clarified: PLAC1 acts as a co-factor by stimulating proliferation by of the FGF7-FGFR2 signaling pathway.rnAll together, these novel biochemical findings underline that the placenta specific protein PLAC1 could be a new target for cancer immunotherapy, especially considering its potential applicability for antibody therapy in tumor patients.