Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Cancers (Basel) ; 14(19)2022 Oct 08.
Article in English | MEDLINE | ID: mdl-36230845

ABSTRACT

Overexpression of the neurotensin receptor type 1 (NTS1R), a peptide receptor located at the plasma membrane, has been reported for a variety of malignant tumors. Thus, targeting the NTS1R with 18F- or 68Ga-labeled ligands is considered a straightforward approach towards in vivo imaging of NTS1R-expressing tumors via positron emission tomography (PET). The development of suitable peptidic NTS1R PET ligands derived from neurotensin is challenging due to proteolytic degradation. In this study, we prepared a series of NTS1R PET ligands based on the C-terminal fragment of neurotensin (NT(8-13), Arg8-Arg9-Pro10-Tyr11-Ile12-Leu13) by attachment of the chelator 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) via an Nω-carbamoylated arginine side chain. Insertion of Ga3+ in the DOTA chelator gave potential PET ligands that were evaluated concerning NTS1R affinity (range of Ki values: 1.2-21 nM) and plasma stability. Four candidates were labeled with 68Ga3+ and used for biodistribution studies in HT-29 tumor-bearing mice. [68Ga]UR-LS130 ([68Ga]56), containing an N-terminal methyl group and a ß,ß-dimethylated tyrosine instead of Tyr11, showed the highest in vivo stability and afforded a tumor-to-muscle ratio of 16 at 45 min p.i. Likewise, dynamic PET scans enabled a clear tumor visualization. The accumulation of [68Ga]56 in the tumor was NTS1R-mediated, as proven by blocking studies.

2.
Biomedicines ; 10(10)2022 Oct 18.
Article in English | MEDLINE | ID: mdl-36289872

ABSTRACT

Chemerin is a multifunctional adipokine that regulates adipogenesis, insulin signaling and blood pressure and has thus a central function in metabolism. Mounting evidence confirmed a function of chemerin in various cancers. In this study, we investigated the role of chemerin in rhabdomyosarcoma (RMS), an aggressive soft tissue cancer that affects mainly children and young adults. We found chemerin expression in 93.8% (90 of 96) of RMS cases, with a range of 86.7-96.7% for the four RMS subgroups. While chemerin is uniformly expressed in normal skeletal muscle, its expression in RMS is patchy with interspersed areas that are devoid of chemerin. This variable chemerin expression is reflected by RMS cell lines as two of them (Rh41 and Rd18) were found to secrete chemerin while the two other ones (JR1 and RD) were negative. Deletion of chemerin in Rh41 and Rd18 cells did not alter their growth rate or morphology. We investigated the potential influence of chemerin on immune surveillance by coculturing parental and chemerin-deficient RMS cells with resting- or lipopolysaccharide (LPS)-activated human peripheral monocytes. The absence of chemerin in the RMS cells led to increased expression levels of the coinhibitory molecules PD-L1 and PD-L2 while levels of the costimulatory molecule CD86 were not changed. Further, the absence of chemerin enhanced the secretion of cytokines (IL-1ß, IL-6, IL-10 and TNF) that have been shown to support RMS pathogenesis. These data indicate that the loss of chemerin expression by RMS cells repolarizes monocytes in the tumor microenvironment to supporting tumor progression.

3.
Otol Neurotol ; 40(5): e532-e541, 2019 06.
Article in English | MEDLINE | ID: mdl-31083091

ABSTRACT

HYPOTHESIS: The choice of the material for plugging a dehiscence of the superior semicircular canal is based on the ease of use and the success of the procedure to permanently relieve symptoms without adverse side effects. BACKGROUND: Dehiscence of the superior semicircular canal can lead to autophony, conductive hearing loss, and vertigo. Surgical treatment by plugging the canal is a highly effective treatment of the symptoms in many patients, although, the procedure can be associated with some degree of hearing loss in more than or equal to 25% of the patients. The available data indicate that adverse effects may be more frequently observed with bone wax as compared with other materials. METHODS: In the present study we compare the tissue reactions induced by plugging the superior semicircular canal with autologous bone pate/bone chips, muscle, fat, artificial bone wax, and teflon in the gerbil model in an attempt to identify the material leading to successful plugging with the least adverse tissue reactions. RESULTS: Our data show that successful plugging was achieved in 100% of the ears by bone pate/bone chips, teflon, and bone wax while the success rate was significantly lower (<50%) following muscle and fat. The proportion of adverse tissue reactions was significantly more pronounced using bone wax as compared with teflon and bone pate/bone chips. CONCLUSION: The use of teflon or autologous bone as a material for plugging a dehiscent superior semicircular canal should be favored over bone wax, muscle, and fat.


Subject(s)
Adhesives , Otologic Surgical Procedures/methods , Semicircular Canals/surgery , Surgical Wound Dehiscence/therapy , Adhesives/adverse effects , Adipose Tissue/transplantation , Animals , Bone Transplantation , Gerbillinae , Hearing Loss, Conductive/etiology , Male , Mastoid/growth & development , Muscles/transplantation , Polytetrafluoroethylene , Waxes
4.
Pigment Cell Melanoma Res ; 31(5): 614-629, 2018 09.
Article in English | MEDLINE | ID: mdl-29495114

ABSTRACT

The network of molecular players is similar when comparing neural crest-derived, actively migrating melanoblasts to melanoma cells. However, melanoblasts are sensitive to differentiation-initiating signals at their target site (epidermis), while melanoma cells maintain migratory and undifferentiated features. We aimed at identifying downregulated genes in melanoma that are particularly upregulated in melanoblasts. Loss of such genes could contribute to stabilization of a dedifferentiated, malignant phenotype in melanoma. We determined that microRNA-622 (miR-622) expression was strongly downregulated in melanoma cells and tissues compared to melanocytes and melanoblast-related cells. miR-622 expression correlated with survival of patients with melanoma. miR-622 re-expression inhibited clonogenicity, proliferation, and migration in melanoma. Inhibition of miR-622 in melanocytes induced enhanced migration. Kirsten rat sarcoma (KRAS) was identified as a major functional target of miR-622 in melanoma. We conclude that miR-622 is a novel tumor suppressor in melanoma and identify the miR-622-KRAS axis as potential therapeutic target.


Subject(s)
Biomarkers, Tumor/metabolism , Gene Expression Regulation, Neoplastic , Melanocytes/pathology , Melanoma/pathology , MicroRNAs/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Case-Control Studies , Cell Movement , Cell Proliferation , Humans , Male , Melanocytes/metabolism , Melanoma/genetics , Melanoma/metabolism , Mice , Mice, Nude , Prognosis , Proto-Oncogene Proteins p21(ras)/genetics , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
J Virol ; 91(1)2017 Jan 01.
Article in English | MEDLINE | ID: mdl-27795433

ABSTRACT

Previous observations that human amniotic fluid cells (AFC) can be transformed by human adenovirus type 5 (HAdV-5) E1A/E1B oncogenes prompted us to identify the target cells in the AFC population that are susceptible to transformation. Our results demonstrate that one cell type corresponding to mesenchymal stem/stroma cells (hMSCs) can be reproducibly transformed by HAdV-5 E1A/E1B oncogenes as efficiently as primary rodent cultures. HAdV-5 E1-transformed hMSCs exhibit all properties commonly associated with a high grade of oncogenic transformation, including enhanced cell proliferation, anchorage-independent growth, increased growth rate, and high telomerase activity as well as numerical and structural chromosomal aberrations. These data confirm previous work showing that HAdV preferentially transforms cells of mesenchymal origin in rodents. More importantly, they demonstrate for the first time that human cells with stem cell characteristics can be completely transformed by HAdV oncogenes in tissue culture with high efficiency. Our findings strongly support the hypothesis that undifferentiated progenitor cells or cells with stem cell-like properties are highly susceptible targets for HAdV-mediated cell transformation and suggest that virus-associated tumors in humans may originate, at least in part, from infections of these cell types. We expect that primary hMSCs will replace the primary rodent cultures in HAdV viral transformation studies and are confident that these investigations will continue to uncover general principles of viral oncogenesis that can be extended to human DNA tumor viruses as well. IMPORTANCE: It is generally believed that transformation of primary human cells with HAdV-5 E1 oncogenes is very inefficient. However, a few cell lines have been successfully transformed with HAdV-5 E1A and E1B, indicating that there is a certain cell type which is susceptible to HAdV-mediated transformation. Interestingly, all those cell lines have been derived from human embryonic tissue, albeit the exact cell type is not known yet. We show for the first time the successful transformation of primary human mesenchymal stromal cells (hMSCs) by HAdV-5 E1A and E1B. Further, we show upon HAdV-5 E1A and E1B expression that these primary progenitor cells exhibit features of tumor cells and can no longer be differentiated into the adipogenic, chondrogenic, or osteogenic lineage. Hence, primary hMSCs represent a robust and novel model system to elucidate the underlying molecular mechanisms of adenovirus-mediated transformation of multipotent human progenitor cells.


Subject(s)
Adenovirus E1A Proteins/genetics , Adenovirus E1B Proteins/genetics , Adenoviruses, Human/genetics , Cell Transformation, Viral , Gene Expression Regulation, Neoplastic , Gene Expression Regulation, Viral , Mesenchymal Stem Cells/virology , Adenovirus E1A Proteins/metabolism , Adenovirus E1B Proteins/metabolism , Adenoviruses, Human/growth & development , Adenoviruses, Human/metabolism , Animals , Cell Line, Transformed , Cell Proliferation , Chromosome Aberrations , Epithelial Cells/pathology , Epithelial Cells/virology , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Karyotype , Lentivirus/genetics , Lentivirus/metabolism , Mesenchymal Stem Cells/pathology , Oncogenes , Primary Cell Culture , Rats , Transfection
6.
Carcinogenesis ; 35(6): 1407-15, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24590895

ABSTRACT

In human cancers, giant cadherin FAT1 may function both, as an oncogene and a tumor suppressor. Here, we investigated the expression and function of FAT1 in hepatocellular carcinoma (HCC). FAT1 expression was increased in human HCC cell lines and tissues compared with primary human hepatocytes and non-tumorous liver tissue as assessed by quantitative PCR and western blot analysis. Combined immunohistochemical and tissue microarray analysis showed a significant correlation of FAT1 expression with tumor stage and proliferation. Suppression of FAT1 expression by short hairpin RNA impaired proliferation and migration as well as apoptosis resistance of HCC cells in vitro. In nude mice, tumors formed by FAT1-suppressed HCC cells showed a delayed onset and more apoptosis compared with tumors of control cells. Both hepatocyte growth factor and hypoxia-mediated hypoxia-inducible factor 1 alpha activation were identified as strong inducers of FAT1 in HCC. Moreover, demethylating agents induced FAT1 expression in HCC cells. Hypoxia lead to reduced levels of the methyl group donor S-adenosyl-L-methionine (SAM) and hypoxia-induced FAT1 expression was inhibited by SAM supplementation in HCC cells. Together, these findings indicate that FAT1 expression in HCC is regulated via promotor methylation. FAT1 appears as relevant mediator of hypoxia and growth receptor signaling to critical tumorigenic pathways in HCC. This knowledge may facilitate the rational design of novel therapeutics against this highly aggressive malignancy.


Subject(s)
Cadherins/metabolism , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Adult , Aged , Cadherins/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , DNA Methylation , Female , Gene Expression , Gene Expression Regulation, Neoplastic , Humans , Hypoxia/genetics , Hypoxia/metabolism , Immunohistochemistry , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Middle Aged , Neoplasm Grading , Neoplasm Staging , Tumor Burden
7.
PLoS One ; 7(5): e37941, 2012.
Article in English | MEDLINE | ID: mdl-22666418

ABSTRACT

Melanoma is the most aggressive form of skin cancer, with fast progression and early dissemination mediated by the melanoma inhibitory activity (MIA) protein. Here, we discovered that dimerization of MIA is required for functional activity through mutagenesis of MIA which showed the correlation between dimerization and functional activity. We subsequently identified the dodecapeptide AR71, which prevents MIA dimerization and thereby acts as a MIA inhibitor. Two-dimensional nuclear magnetic resonance (NMR) spectroscopy demonstrated the binding of AR71 to the MIA dimerization domain, in agreement with in vitro and in vivo data revealing reduced cell migration, reduced formation of metastases and increased immune response after AR71 treatment. We believe AR71 is a lead structure for MIA inhibitors. More generally, inhibiting MIA dimerization is a novel therapeutic concept in melanoma therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Extracellular Matrix Proteins/antagonists & inhibitors , Immune Tolerance/drug effects , Melanoma/immunology , Melanoma/pathology , Molecular Targeted Therapy/methods , Neoplasm Proteins/antagonists & inhibitors , Amino Acid Sequence , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Extracellular Matrix Proteins/chemistry , Extracellular Matrix Proteins/metabolism , Humans , Melanoma/drug therapy , Melanoma/metabolism , Melanoma, Experimental/drug therapy , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Models, Molecular , Molecular Sequence Data , Neoplasm Metastasis , Neoplasm Proteins/chemistry , Neoplasm Proteins/metabolism , Oligopeptides/pharmacology , Oligopeptides/therapeutic use , Protein Multimerization/drug effects , Protein Structure, Quaternary
8.
Mol Med ; 17(3-4): 221-8, 2011.
Article in English | MEDLINE | ID: mdl-21152698

ABSTRACT

Augmenter of liver regeneration (ALR), which is critically important in liver regeneration and hepatocyte proliferation, is highly expressed in cirrhotic livers and hepatocellular carcinomas (HCC). In the current study, the functional role of ALR in hepatocancerogenesis was analyzed in more detail. HepG2 cells, in which the cytosolic 15 kDa ALR isoform was reexpressed stably, (HepG2-ALR) were used in migration and invasion assays using modified Boyden chambers. Epithelial-mesenchymal transition (EMT) markers were determined in HepG2-ALR cells in vitro and in HepG2-ALR tumors grown in nude mice. ALR protein was quantified in HCC and nontumorous tissues by immunohistochemistry. HepG2-ALR, compared with HepG2 cells, demonstrated reduced cell motility and increased expression of the epithelial cell markers E-cadherin and Zona occludens-1 (ZO-1), whereas SNAIL, a negative regulator of E-cadherin, was diminished. Matrix metalloproteinase MMP1 and MMP3 mRNA expression and activity were reduced. HepG2-ALR cell-derived subcutaneously grown tumors displayed fewer necrotic areas, more epithelial-like cell growth and fewer polymorphisms and atypical mitotic figures than tumors derived from HepG2 cells. Analysis of tumor tissues of 53 patients with HCC demonstrated an inverse correlation of ALR protein with histological angioinvasion and grading. The 15 kDa ALR isoform was found mainly in HCC tissues without histological angioinvasion 0. In summary the present data indicate that cytosolic ALR reduces hepatoma cell migration, augments epithelial growth and, therefore, may act as an antimetastatic and EMT reversing protein.


Subject(s)
Carcinoma, Hepatocellular/physiopathology , Cytochrome Reductases/metabolism , Liver Neoplasms/physiopathology , Liver Regeneration , Animals , Blotting, Western , Cadherins/genetics , Cadherins/metabolism , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/pathology , Cell Movement , Cells, Cultured , Cytochrome Reductases/genetics , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Hep G2 Cells , Humans , Liver Neoplasms/enzymology , Liver Neoplasms/pathology , Male , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 3/genetics , Matrix Metalloproteinase 3/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred Strains , Mice, Nude , Neoplasm Metastasis , Neoplasms, Experimental/enzymology , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Oxidoreductases Acting on Sulfur Group Donors , Phosphoproteins/genetics , Phosphoproteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Transplantation, Heterologous , Zonula Occludens-1 Protein
9.
J Gen Virol ; 91(Pt 7): 1828-33, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20335490

ABSTRACT

Analysis of the molecular mechanisms of viral-mediated oncogenesis has contributed enormously to the understanding of the basic principles of normal/malignant cell growth. Transformation by human adenoviruses is a multi-step process involving the modulation of numerous cellular pathways, leading to inhibition of apoptosis and growth arrest. However, the molecular mechanism of how the adenovirus oncogenes facilitate transformation of rodent cells, while concurrently failing to do so for human cells, remains elusive. In this report, we demonstrate for the first time that the transformation capabilities of adenovirus type 5 oncogenes are not restricted to rodent cells, but include cells of the related mammalian order Lagomorpha, inducing considerable morphological alterations, enhanced cell growth and tumour induction in vivo. Furthermore, the established cell lines may represent a suitable tool for further development to generate E4-mutated adenoviruses, which has so far been difficult as mutations within the E4 region often prove to be lethal without a helper-cell system.


Subject(s)
Adenoviridae/metabolism , Adenovirus E1 Proteins/genetics , Adenovirus E4 Proteins/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Viral/genetics , Animals , Cell Line , Oncogenic Viruses , Rabbits
10.
Am J Pathol ; 176(3): 1433-42, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20093481

ABSTRACT

Fibroblast growth factor receptor 2 isoform b (FGFR2-IIIb) is highly expressed in hepatocytes and plays an important role in liver homeostasis and regeneration. Here, we analyzed the expression and function of FGFR2-IIIb in hepatocellular carcinoma (HCC). FGFR2-IIIb expression in HCC tissues and cell lines was lower than in primary human hepatocytes and nontumorous tissue. FGFR2-IIIb-negative HCCs showed a significantly higher Ki-67 labeling index, and loss of FGFR2-IIIb expression correlated significantly with vascular invasion and more advanced tumor stages. A decrease in FGFR-2IIIb expression in HCC cell lines was not related to promoter hypermethylation. However, PCR analysis indicated that chromosomal deletion at 10q accounted for the loss of FGFR2 expression in a subset of HCC cells. FGFR2-IIIb re-expression in stable transfected HCC cell lines induced a higher basal apoptosis rate and a significantly reduced proliferation and migratory potential in vitro. In nude mice, FGFR2-IIIb re-expressing HCC cells grew significantly slower, and terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling assay revealed higher apoptosis rates. The antitumorigenic effects of FGFR2-IIIb expression in HCC cells were not affected by keratinocyte growth factor or an inhibitor of FGFR-phosphorylation, indicating that they are independent of tyrosine kinase activation. In conclusion, our data indicate that FGFR2-IIIb inhibits tumorigenicity of HCC cells. Identification of the molecular mechanisms promoting regeneration in normal tissue while suppressing malignancy may lead to novel therapeutic targets of this highly aggressive tumor.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Receptor, Fibroblast Growth Factor, Type 2/genetics , Aged , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Methylation/drug effects , DNA Methylation/genetics , Down-Regulation/drug effects , Down-Regulation/genetics , Female , Fibroblast Growth Factor 7/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice , Middle Aged , Promoter Regions, Genetic/genetics , Protein Kinase Inhibitors/pharmacology , Pyrroles/pharmacology , Receptor, Fibroblast Growth Factor, Type 2/metabolism
11.
Cancer Sci ; 100(4): 646-53, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19175606

ABSTRACT

Liver cirrhosis is the main risk factor for the development of hepatocellular carcinoma (HCC). Activated hepatic stellate cells (HSC) are the effector cells of hepatic fibrosis and also infiltrate the HCC stroma where they might play a critical role in HCC progression. Here we aimed to analyze the effects of activated HSC on the proliferation and growth of HCC cell lines in vitro and in vivo. Conditioned media (CM) collected from HSC significantly induced proliferation and migration of HCC cells cultured in monolayers. In a 3-dimensional spheroid coculture system, HSC promoted HCC growth and diminished the extent of central necrosis. In accordance, in vivo simultaneous implantation of HSC and HCC cells into nude mice promoted tumor growth and invasiveness, and inhibited necrosis formation. As potential mechanism of the tumorigenic effects of HSC we identified activation of NFkappaB and extracellular-regulated kinase (ERK) in HCC cells, two signaling cascades that play a crucial role in HCC progression. In summary, our data indicate that stromal HSC promotes HCC progression and suggest the HSC-HCC interaction as an interesting tumor differentiation-independent target for therapy of this highly aggressive cancer.


Subject(s)
Carcinoma, Hepatocellular/pathology , Cell Movement/drug effects , Cell Proliferation/drug effects , Culture Media, Conditioned/pharmacology , Hepatic Stellate Cells/metabolism , Liver Neoplasms/pathology , Animals , Cell Line, Tumor , Coculture Techniques , Enzyme Activation , Humans , Mice , Mice, Nude , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , NF-kappa B/metabolism , Neurotrophin 3/genetics , Neurotrophin 3/metabolism , Organ Culture Techniques , RNA, Messenger/analysis , Xenograft Model Antitumor Assays
12.
J Exp Med ; 206(1): 221-32, 2009 Jan 16.
Article in English | MEDLINE | ID: mdl-19124656

ABSTRACT

High malignancy and early metastasis are hallmarks of melanoma. Here, we report that the transcription factor Snail1 inhibits expression of the tumor suppressor CYLD in melanoma. As a direct consequence of CYLD repression, the protooncogene BCL-3 translocates into the nucleus and activates Cyclin D1 and N-cadherin promoters, resulting in proliferation and invasion of melanoma cells. Rescue of CYLD expression in melanoma cells reduced proliferation and invasion in vitro and tumor growth and metastasis in vivo. Analysis of a tissue microarray with primary melanomas from patients revealed an inverse correlation of Snail1 induction and loss of CYLD expression. Importantly, tumor thickness and progression-free and overall survival inversely correlated with CYLD expression. Our data suggest that Snail1-mediated suppression of CYLD plays a key role in melanoma malignancy.


Subject(s)
Gene Expression Regulation, Neoplastic , Melanoma/pathology , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , Animals , B-Cell Lymphoma 3 Protein , Blotting, Western , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Cell Proliferation , Cyclin D1/genetics , Cyclin D1/metabolism , Deubiquitinating Enzyme CYLD , Down-Regulation , Female , Humans , Male , Melanoma/genetics , Melanoma/metabolism , Melanoma, Experimental/genetics , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred Strains , Mice, Nude , Neoplasm Invasiveness , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Snail Family Transcription Factors , Transcription Factors/metabolism , Transplantation, Heterologous , Tumor Suppressor Proteins/metabolism
13.
Tissue Eng Part A ; 14(2): 275-84, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18333780

ABSTRACT

One strategy of adipose tissue engineering is to transplant adipocytes or adipocyte precursor cells in combination with polymeric materials. However, a satisfying formation of fat tissue and its long-term survival still remain major problems. There is increasing evidence that treatment of the cells prior to implantation plays a critical role in the success of adipose tissue growth. In a previous study, we established a model system based on 3T3-L1 cells that allows for reproducible engineering of mature, coherent adipose tissues in vitro. We utilized this model system in the current study and systematically investigated the long-term in vivo development of cellular constructs with varying stages of adipogenic development at the time point of implantation. Blank polyglycolic acid fiber meshes, scaffolds seeded with uninduced 3T3-L1 preadipocytes, and cell-polymer constructs precultivated under adipogenic conditions for 2, 9, or 35 days were implanted subcutaneously into nude mice. Histological analysis revealed that no fat formation occurred in constructs without adipogenic precultivation. Implantation of mature fat pads (35 days) resulted in adiponecrosis within the constructs. In contrast, implants with an immature phenotype at the time point of implantation (2 and 9 days) gave rise to vascularized, mature adipose tissue in vivo. Further, these engineered adipose tissues showed long-term survival in vivo over the whole investigation period of 24 weeks. The results of this study can contribute to the development of future clinical approaches as they give clear evidence which precultivation strategy promotes successful development and long-term survival of engineered adipose tissue.


Subject(s)
Adipocytes/cytology , Adipose Tissue/cytology , Tissue Engineering/methods , 3T3-L1 Cells , Adipocytes/drug effects , Adipocytes/transplantation , Adipogenesis/drug effects , Adipose Tissue/drug effects , Adipose Tissue/surgery , Animals , Female , Mice , Mice, Nude , Polyglycolic Acid/chemistry , Polyglycolic Acid/pharmacology , Time Factors , Tissue Scaffolds/chemistry
14.
J Cancer Res Clin Oncol ; 134(5): 597-607, 2008 May.
Article in English | MEDLINE | ID: mdl-17932689

ABSTRACT

PURPOSE: Previously, we studied the effect of co-administration of paclitaxel with the second generation ABCB1 (p-gp) modulator valspodar on the intracerebral growth of human U118-MG glioblastoma in nude mice. Valspodar significantly increased the brain levels of paclitaxel by inhibition of p-gp expressed at the blood brain barrier. Thus, the tumour burden was reduced by 90%, which was considered as a proof of concept. However, the paclitaxel dose had to be reduced because of toxic side effects resulting from increased drug levels due to p-gp modulation in peripheral tissues. Therefore, in the present study we examined the co-application of paclitaxel with the third generation ABCB1 modulators elacridar and tariquidar, which were supposed to preferentially modulate p-gp in brain capillaries. METHODS: The inhibitory activity of the modulators was measured by a flow cytometric and a chemosensitivity assay in vitro. To determine the distribution of paclitaxel in vivo, nude mice received 50 mg/kg of valspodar, elacridar or tariquidar p.o. (control: vehicle) 4 h before i.v. injection of 8 mg/kg of paclitaxel. Brain, liver, kidney and plasma were collected and analyzed by RP-HPLC. RESULTS: Our in vitro experiments demonstrate that the new modulators are about 80 times more effective in comparison to valspodar. Co-administration of paclitaxel with elacridar and tariquidar led to a long lasting fivefold increase in the concentration of the cytostatic in the brain. Although the increase (2.5- to 7-fold) tended to be lower compared to that induced by co-administered valspodar (six- to eightfold), the brain/plasma ratios achieved with the new modulators were 2-15 times higher. CONCLUSIONS: Elacridar and tariquidar seem to modulate p-glycoprotein preferentially at the blood-brain barrier. Our results suggest that the systemic toxicity of cytostatics combined with elacridar or tariquidar should be lower than in combination with valspodar.


Subject(s)
ATP-Binding Cassette Transporters/drug effects , Acridines/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Paclitaxel/pharmacokinetics , Quinolines/pharmacokinetics , Tetrahydroisoquinolines/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1 , Animals , Brain/metabolism , Chromatography, High Pressure Liquid , Drug Interactions , Flow Cytometry , Mice , Mice, Nude
15.
J Neuroimmunol ; 187(1-2): 61-73, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17553571

ABSTRACT

We studied the role of the Transforming growth factor (TGF)-beta signaling antagonist Smad7 in autoimmune central nervous system (CNS) inflammation by using specific antisense oligonucleotides (Smad7-as). Elevated Smad7 protein expression was found in the spinal cord of SJL/J mice and DA rats with experimental autoimmune encephalomyelitis (EAE) and in effector T cells upon antigen stimulation. Smad7-as specifically decreased Smad7 mRNA and protein in cell lines and in ex-vivo-treated primary mouse lymph node cells (LNC). LNC exposed to Smad7-as during secondary activation showed reduced proliferation and encephalitogenicity. After systemic administration, Smad7-as ameliorated clinical signs of active and adoptively transferred EAE, diminished CNS inflammation, and reduced Smad7 protein levels in the brain. Smad7-as was found to be incorporated by peritoneal macrophages as well as by cells of the liver, kidneys, and peripheral lymph nodes. Importantly, Smad7-as treatment was not toxic and did not increase extracellular matrix formation. Smad7 inhibition thus represents a novel systemic treatment strategy for autoimmune CNS inflammation, targeting TGF-beta signaling without TGF-beta-associated toxicity.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/therapy , Signal Transduction/physiology , Smad7 Protein/metabolism , Transforming Growth Factor beta/physiology , Adoptive Transfer/methods , Animals , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Female , Gene Expression Regulation/drug effects , Lymphocytes/drug effects , Lymphocytes/metabolism , Mice , Mice, Inbred Strains , Oligonucleotides, Antisense/pharmacology , Oligonucleotides, Antisense/therapeutic use , RNA, Messenger/biosynthesis , Rats , Reverse Transcriptase Polymerase Chain Reaction/methods , Signal Transduction/drug effects , Smad7 Protein/chemistry , Thionucleotides/pharmacology , Thionucleotides/therapeutic use , Time Factors
16.
J Cancer Res Clin Oncol ; 133(3): 153-67, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17024493

ABSTRACT

[Meso-1,2-bis(2,6-dihalo-3/4-hydroxyphenyl)ethylenediamine]platinum(II) complexes (meso-1-PtLL': 2,6-F(2),3-OH; meso-2-PtLL': 2,6-F(2),4-OH; meso-3-PtLL': 2,6-Cl(2),3-OH; meso-4-PtLL': 2,6-Cl(2),4-OH; L = OH(2), L' = OSO(3) or L,L' = Cl(2)) were designed with the aim to get drugs comprising both cytotoxic and testosterone level lowering potencies. It is assumed that such compounds are more efficient than the established endocrine therapeutic measures and can affect the development of hormone refractory prostate cancer (PC). With exception of meso-3-PtLL' all Pt-complexes and the comparison compound cisplatin significantly reduced the testosterone level in experiments on male rats. However, in the test on the Dunning R3327 PC of the rat only cisplatin and meso-4-PtLL' showed a significant anti-tumor activity at well-tolerated dose ranges. Meso-4-PtLL' also significantly extended the time to disease progression in comparison with orchiectomy in this tumor model. Interestingly, the relapsed tumor, too, responded to meso-4-PtLL' as demonstrated in a long-term study on orchiectomized rats bearing Dunning R3327 PC grafts. This effect cannot be ascribed to cytotoxic effects of meso-4-PtLL' because of its inactivity on the human LNCaP/FGC PC cell line. Therefore, the contribution of an additional mechanism to the anti-prostate cancer activity of meso-4-PtLL', presumably owing to its estrogenic potency, must be considered. This assumption was supported by test results with diethylstilbestrol (DES) (non-steroidal estrogen) on the Dunning R3327 PC of the rat relapsed after orchiectomy. This tumor model was strongly inhibited by DES. The possible mode of action of meso-4-PtLL' is thoroughly discussed.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Neoplasm Recurrence, Local/prevention & control , Neoplasms, Hormone-Dependent/drug therapy , Prostatic Neoplasms/drug therapy , Testis/drug effects , Testis/metabolism , Testosterone/biosynthesis , Androgen Antagonists/pharmacology , Animals , Cell Line, Tumor , Cell Survival/drug effects , Diethylstilbestrol/pharmacology , Dose-Response Relationship, Drug , Furans/pharmacology , Humans , Lymphatic Metastasis , Male , Middle Aged , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/secondary , Organ Size/drug effects , Organoplatinum Compounds/pharmacology , Prostatic Neoplasms/metabolism , Pyrones/pharmacology , Rats , Rats, Sprague-Dawley , Testis/pathology , Testosterone/antagonists & inhibitors , Testosterone/blood
17.
FASEB J ; 21(2): 456-63, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17167064

ABSTRACT

The cytokine receptor CD137 is a member of the TNF receptor family and a potent T cell costimulatory molecule. Its ligand is expressed on antigen presenting cells as a transmembrane protein and it too can deliver signals into the cells it is expressed on (reverse signaling). In monocytes, immobilized CD137 protein induces activation, prolongation of survival and proliferation. Here we show that recombinant immobilized CD137 protein enhances migration of monocytes in vitro. Further, CD137 expression on spheroids leads to a significantly enhanced infiltration by monocytes. The migration-inducing activity of CD137 could be confirmed in vivo. Matrigel, which was coated with recombinant CD137 protein and was inserted into the flanks of mice attracted large numbers of monocytes and was heavily infiltrated by these cells. In vivo, expression of CD137 by blood vessel walls at sites of inflammation was detectable by immunohistochemistry. CD137 expression is inducible by proinflammatory cytokines in endothelial cells, suggesting that a physiological function of CD137 may be the facilitation of monocyte extravasation in inflammatory tissues.


Subject(s)
Cell Movement/physiology , Monocytes/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism , Vasculitis/metabolism , Animals , Blood Vessels/metabolism , Blood Vessels/pathology , COS Cells , Cells, Cultured , Chlorocebus aethiops , Female , Gene Expression , Humans , Immunohistochemistry , Inflammation/genetics , Inflammation/metabolism , Mice , Mice, Inbred Strains , Monocytes/cytology , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 9/physiology , Vasculitis/genetics
18.
Oncogene ; 24(1): 55-64, 2005 Jan 06.
Article in English | MEDLINE | ID: mdl-15480414

ABSTRACT

The 55-kDa gene product from subgroup C adenovirus type 5 (Ad5) early region 1 (E1B-55kDa) plays a central role in the oncogenic transformation of primary rodent cells primarily by inactivating transcriptional and presumably other functional properties of the tumor suppressor protein p53. We have previously shown that Ad5 E1B-55kDa possesses a leucine-rich nuclear export signal (NES), which confers rapid nucleocytoplasmic shuttling via the CRM1-dependent export pathway. In this study we report that an export-deficient mutant of the viral protein (E1B-NES) substantially enhances focus formation of primary baby rat kidney cells in combination with Ad E1A. Transformed rat cells stably expressing the E1B-NES protein exhibited increased tumorigenicity and accelerated tumor growth in nude mice compared to transformants containing the wild-type E1B product. This 'gain of function' correlated with enhanced inhibition of p53 transactivation in transient reporter assays and the accumulation of the mutant protein and p53 in several dot-like subnuclear aggregates. Interestingly, these structures also contained a large fraction of cellular promyelocytic leukemia protein (PML), a known regulator of p53. These data indicate that E1B-NES promotes oncogenic transformation by combinatorial mechanisms that involve modulation of p53 in the context of PML nuclear bodies. In sum, these results extend our previous observation that inhibition of PML activities by E1B-55kDa is required for efficient focus formation and provide further support for the view that blocking p53 transcriptional functions is the principal mechanism by which the Ad protein contributes to complete cell transformation in conjunction with Ad E1A.


Subject(s)
Adenoviridae/metabolism , Cell Nucleus/metabolism , Cell Transformation, Neoplastic/metabolism , Karyopherins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Viral Proteins/metabolism , Active Transport, Cell Nucleus/physiology , Animals , Epithelial Cells/metabolism , Mice , Mice, Nude , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Promyelocytic Leukemia Protein , Protein Sorting Signals/physiology , Rats , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins , Exportin 1 Protein
19.
Arch Pharm (Weinheim) ; 337(12): 672-81, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15597400

ABSTRACT

Although a few experimental approaches to isolated limb perfusion (ILP) are described in the literature, none of these animal models mimics the clinical perfusion techniques adequately to improve the technique of ILP on the basis of valid preclinical data. Therefore, we developed an ILP setup in rats allowing online monitoring of essential perfusion parameters such as temperature (in perfusate, various tissues, and rectum), pH (perfusate), perfusion pressure, and O(2) concentration (in perfusate, tissue), by a tailor-made data acquisition system. This setup permits close supervision of vital parameters during ILP. Various interdependencies, concerning the flow rate and the pressure of perfusate as well as tissue oxygenation were registered. For the measurement of pO(2) values in the perfusate and in different regions of the perfused hind limb, a novel type of microoptode based on quenching of a fluorescent dye was devised. Stable normothermic (37 degrees C) perfusion conditions were maintained at a constant perfusion pressure in the range of 40-60 mm Hg by administration of the spasmo lytic moxaverine (0.5 mg/mL of perfusate as initial dose) at a perfusate flow rate of 0.5 mL/min for 60 min. At the end of an ILP, there were no signs of tissue damage, neither concerning laboratory data (K(+), myoglobin, creatine kinase, lactic dehydrogenase) nor histopathological criteria. The reported ILP model is not only well suited to investigate the effects of hyperthermia but also to assess the efficacy of new antineoplastic approaches, when nude rats, bearing human tumours in the hind limbs, are used.


Subject(s)
Chemotherapy, Cancer, Regional Perfusion , Lower Extremity/blood supply , Microcomputers , Models, Animal , Animals , Chemotherapy, Cancer, Regional Perfusion/instrumentation , Chemotherapy, Cancer, Regional Perfusion/methods , Female , Hydrogen-Ion Concentration , Male , Muscle, Skeletal/blood supply , Muscle, Skeletal/metabolism , Oxygen/metabolism , Rats , Rats, Wistar , Regional Blood Flow , Temperature
20.
Exp Cell Res ; 300(1): 54-64, 2004 Oct 15.
Article in English | MEDLINE | ID: mdl-15383314

ABSTRACT

Tissue-inherent factors such as cell-cell and cell-extracellular matrix interactions are regarded to exert a potentially large impact on adipogenesis as well as on secretory functions of adipose tissue. However, an appropriate 3-D adipogenesis model useful for addressing such interactions is still lacking. In this study, using tissue-engineering techniques, we demonstrate for the first time the development of coherent fat pads consisting of unilocular signet-ring cells in vitro. The constructs were generated by differentiating 3T3-L1 preadipocytes on 3-D polymeric scaffolds for either 9, 21, or 35 days in vitro. Only long-term culture yielded uniform tissues histologically comparable to native fat. Light and scanning electron microscopy provided direct evidence of 3-D tissue coherence and cell-cell contact in a tissue context, which was in strong contrast to conventional 2-D monolayer culture. Further differences between the two culture systems included enhanced secretion of leptin in 3-D tissue culture and differences in laminin expression (mRNA and protein level). Increase of triglyceride content over culture time and mRNA expression of other adipocyte genes, such as PPARgamma and Glut-4, were found to be similar. Implantation of long-term differentiated tissue constructs in nude mice resulted in further development and maintenance of fat pads. The presented model system is suggested to contribute to a better understanding of adipose tissue development and function facilitating studies on tissue-inherent interactions in vitro and in vivo.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Cell Communication/physiology , Tissue Engineering/methods , 3T3-L1 Cells , Adipocytes/ultrastructure , Adipose Tissue/ultrastructure , Animals , Cell Adhesion/physiology , Cell Culture Techniques/methods , Extracellular Matrix/metabolism , Glucose Transporter Type 4 , Laminin/metabolism , Leptin/metabolism , Mice , Mice, Nude , Microscopy, Electron, Scanning , Monosaccharide Transport Proteins/genetics , Muscle Proteins/genetics , Organ Culture Techniques/methods , PPAR gamma/genetics , RNA, Messenger/metabolism , Time Factors , Triglycerides/metabolism , Up-Regulation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...