Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 102
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell Mol Life Sci ; 79(9): 475, 2022 Aug 09.
Article in English | MEDLINE | ID: mdl-35943635

ABSTRACT

In malignant melanoma, a highly aggressive form of skin cancer, many microRNAs are aberrantly expressed contributing to tumorigenesis and progression. Further, deregulation of microRNA processing enzymes, like the miRNA-binding protein Argonaute 2, significantly impacts microRNA function. This study characterizes a novel splice variant of Argonaut 2, AGO2-ex1/3. AGO2-ex1/3 is substantially expressed in different melanoma cell lines and patient-derived tissue samples. It is a mature mRNA, which is translated into an N-terminally truncated Argonaute 2 protein form. Molecular dynamics simulations show that the PAZ, MID, and PIWI domain largely retain their structure in AGO2-ex1/3 and that the truncation of the N-terminus leads to an increased interdomain flexibility. Expression of AGO2-ex1/3 provides a survival advantage for melanoma cells while the knockdown causes significantly reduced proliferation and increases apoptosis. RNA-sequencing revealed that in cells lacking AGO2-ex1/3 expression many miRNA target genes are deregulated, implicating a considerable role of AGO2-ex1/3 for miRNA function. This study inaugurates insights into an important role of a so far unknown splice variant of Argonaute 2 for the miRNA pathway as well as the mechanisms which drive growth and survival of melanoma cells. This knowledge provides the basis for potential new promising therapeutic targets focusing on small RNA-mediated gene regulation in melanoma.


Subject(s)
Melanoma , MicroRNAs , Skin Neoplasms , Apoptosis/genetics , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , Humans , Melanoma/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , RNA Interference , Skin Neoplasms/genetics
2.
Int J Mol Sci ; 23(2)2022 Jan 13.
Article in English | MEDLINE | ID: mdl-35055045

ABSTRACT

The neural crest transcription factor BRN3A is essential for the proliferation and survival of melanoma cells. It is frequently expressed in melanoma but not in normal melanocytes or benign nevi. The mechanisms underlying the aberrant expression of BRN3A are unknown. Here, we investigated the epigenetic regulation of BRN3A in melanocytes and melanoma cell lines treated with DNA methyltransferase (DNMT), histone acetyltransferase (HAT), and histone deacetylase (HDAC) inhibitors. DNMT and HAT inhibition did not significantly alter BRN3A expression levels, whereas panHDAC inhibition by trichostatin A led to increased expression. Treatment with the isoform-specific HDAC inhibitor mocetinostat, but not with PCI-34051, also increased BRN3A expression levels, suggesting that class I HDACs HDAC1, HDAC2, and HDAC3, and class IV HDAC11, were involved in the regulation of BRN3A expression. Transient silencing of HDACs 1, 2, 3, and 11 by siRNAs revealed that, specifically, HDAC2 inhibition was able to increase BRN3A expression. ChIP-Seq analysis uncovered that HDAC2 inhibition specifically increased H3K27ac levels at a distal enhancer region of the BRN3A gene. Altogether, our data suggest that HDAC2 is a key epigenetic regulator of BRN3A in melanocytes and melanoma cells. These results highlight the importance of epigenetic mechanisms in regulating melanoma oncogenes.


Subject(s)
Gene Expression Regulation , Histone Deacetylase 2/metabolism , Melanocytes/metabolism , Melanoma/etiology , Melanoma/metabolism , Transcription Factor Brn-3A/genetics , Cell Line , DNA Methylation , Epigenesis, Genetic , Gene Expression Regulation/drug effects , Gene Silencing , Histone Deacetylase 2/genetics , Histone Deacetylase Inhibitors/pharmacology , Humans , Melanocytes/pathology , Melanoma/pathology , Transcription Factor Brn-3A/metabolism
3.
Int J Mol Sci ; 22(17)2021 Aug 28.
Article in English | MEDLINE | ID: mdl-34502254

ABSTRACT

Molecular analyses of normal and diseased cells give insight into changes in gene expression and help in understanding the background of pathophysiological processes. Years after cDNA microarrays were established in research, RNA sequencing (RNA-seq) became a key method of quantitatively measuring the transcriptome. In this study, we compared the detection of genes by each of the transcriptome analysis methods: cDNA array, quantitative RT-PCR, and RNA-seq. As expected, we found differences in the gene expression profiles of the aforementioned techniques. Here, we present selected genes that exemplarily demonstrate the observed differences and calculations to reveal that a strong RNA secondary structure, as well as sample preparation, can affect RNA-seq. In summary, this study addresses an important issue with a strong impact on gene expression analysis in general. Therefore, we suggest that these findings need to be considered when dealing with data from transcriptome analyses.


Subject(s)
Gene Expression Profiling/methods , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, RNA , Adaptor Proteins, Signal Transducing/genetics , Cell Line, Tumor , Humans , Intracellular Signaling Peptides and Proteins/genetics , RNA/chemistry , SOX Transcription Factors/genetics , Transcription Factors/genetics , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Transcriptome , YAP-Signaling Proteins
4.
Am J Pathol ; 188(1): 72-83, 2018 01.
Article in English | MEDLINE | ID: mdl-29107073

ABSTRACT

Tumor immune microenvironment and tumor metabolism are major determinants of chemoradiotherapy response. The interdependency and prognostic significance of specific immune and metabolic phenotypes in head and neck squamous cell carcinoma (HNSCC) were assessed and changes in reactive oxygen species were evaluated as a mechanism of treatment response in tumor spheroid/immunocyte co-cultures. Pretreatment tumor biopsies were immunohistochemically characterized in 73 HNSCC patients treated by definitive chemoradiotherapy and correlated with survival. The prognostic significance of CD8A, GLUT1, and COX5B gene expression was analyzed within The Cancer Genome Atlas database. HNSCC spheroids were co-cultured in vitro with peripheral blood mononuclear cells (PBMCs) in the presence of the glycolysis inhibitor 2-deoxyglucose and radiation treatment followed by PBMC chemotaxis determination via fluorescence microscopy. In the chemoradiotherapy-treated HNSCC cohort, mitochondrial-rich (COX5B) metabolism correlated with increased and glucose-dependent (GLUT1) metabolism with decreased intratumoral CD8/CD4 ratios. High CD8/CD4, together with mitochondrial-rich or glucose-independent metabolism, was associated with improved short-term survival. The Cancer Genome Atlas analysis confirmed that patients with a favorable immune and metabolic gene signature (high CD8A, high COX5B, low GLUT1) had improved short- and long-term survival. In vitro, 2-deoxyglucose and radiation synergistically up-regulated reactive oxygen species-dependent PBMC chemotaxis to HNSCC spheroids. These results suggest that glucose-independent tumor metabolism is associated with CD8-dominant antitumor immune infiltrate, and together, these contribute to improved chemoradiotherapy response in HNSCC.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Squamous Cell/therapy , Chemoradiotherapy , Head and Neck Neoplasms/therapy , Aged , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , CD8 Antigens/genetics , CD8 Antigens/metabolism , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/mortality , Cell Line, Tumor , Cytochrome c Group/genetics , Cytochrome c Group/metabolism , Databases, Genetic , Electron Transport Complex IV , Female , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/mortality , Humans , Male , Middle Aged , Prognosis , Reactive Oxygen Species/metabolism , Survival Rate
5.
Photodermatol Photoimmunol Photomed ; 35(4): 255-260, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30815924

ABSTRACT

BACKGROUND: Phototherapy is a frequently used treatment modality for a variety of dermatologic diseases. UV radiation has different effects on the skin, for example increased production and release of cytokines and other proteins, and is involved in the initiation and progression of skin cancer. Objective of this clinical trial was to investigate potential systemic effects of UV phototherapy on cytokine profiles in blood. METHODS: In a prospective, mono-centric, one-armed study, the serum levels of the melanoma tumour marker "melanoma inhibitory activity" (MIA), Il-1α, Il-4, Il-6, Il-10, TNF-α and IFN-γ of 115 patients with different skin diseases were compared before and 24-48 hours as well as 2-4 weeks after the first phototherapy with PUVA (psoralen and ultraviolet A), UVA or UVB, or both. Data were analysed using linear mixed models. RESULTS: Estimated marginal means of MIA levels were 6.05 ng/mL (95%-CI: 5.37-6.72, range: 2.83-14.49) before the first treatment, which had significantly increased to 6.79 ng/mL 2-4 weeks after the first phototherapy (CI 95%: 6.12-7.47, range: 3.09-15.45; P = 0.0042). MIA levels 2-4 weeks after the first phototherapy were significantly higher than 24-48 hours after the first phototherapy (P = 0.0083). 2-4 weeks after the first treatment, TNF-α levels had decreased significantly (P = 0.033) more in patients with psoriasis who had responded well to phototherapy than in patients unresponsive to treatment. Serum levels of the other cytokines had not changed significantly. CONCLUSIONS: Short-term phototherapy significantly increased the serum levels of the melanoma tumour marker MIA. The potential clinical relevance of these findings (ie an increased risk of melanoma) is unclear and should be further investigated.


Subject(s)
Biomarkers, Tumor/blood , Cytokines/blood , Melanoma , PUVA Therapy , Skin Neoplasms , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Melanoma/blood , Melanoma/drug therapy , Melanoma/pathology , Middle Aged , Skin Neoplasms/blood , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology
6.
Biol Chem ; 400(1): 111-122, 2018 12 19.
Article in English | MEDLINE | ID: mdl-29908123

ABSTRACT

Plasma oncology is a relatively new field of research. Recent developments have indicated that cold atmospheric plasma (CAP) technology is an interesting new therapeutic approach to cancer treatment. In this study, p53 wildtype (LoVo) and human p53 mutated (HT29 and SW480) colorectal cancer cells were treated with the miniFlatPlaSter - a device particularly developed for the treatment of tumor cells - that uses the Surface Micro Discharge (SMD) technology for plasma production in air. The present study analyzed the effects of plasma on colorectal cancer cells in vitro and on normal colon tissue ex vivo. Plasma treatment had strong effects on colon cancer cells, such as inhibition of cell proliferation, induction of cell death and modulation of p21 expression. In contrast, CAP treatment of murine colon tissue ex vivo for up to 2 min did not show any toxic effect on normal colon cells compared to H2O2 positive control. In summary, these results suggest that the miniFlatPlaSter plasma device is able to kill colorectal cancer cells independent of their p53 mutation status. Thus, this device presents a promising new approach in colon cancer therapy.


Subject(s)
Atmospheric Pressure , Cell Proliferation/drug effects , Colorectal Neoplasms/pathology , Plasma Gases/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Colorectal Neoplasms/genetics , DNA Damage , Genes, p53 , Humans , Mice , Oncogene Protein p21(ras)/genetics , Oncogene Protein p21(ras)/metabolism , RNA, Messenger/metabolism
7.
J Cell Sci ; 126(Pt 13): 2890-902, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23606745

ABSTRACT

Sox9 is a key transcription factor in early chondrogenesis with distinct roles in differentiation processes and during embryonic development. Here, we report that Sox9 modulates cell survival and contributes to the commitment of mesenchymal stem cells (MSC) to adipogenic or osteogenic differentiation lineages. We found that the Sox9 activity level affects the expression of the key transcription factor in adipogenic differentiation, C/EBPß, and that cyclin D1 mediates the expression of the osteogenic marker osteocalcin in undifferentiated adult bone-marrow-derived rat MSC. Introducing a stable Sox9 knockdown into undifferentiated rat MSC resulted in a marked decrease in proliferation rate and an increase in apoptotic activity. This was linked to a profound upregulation of p21 and cyclin D1 gene and protein expression accompanied by an induction of caspase 3/7 activity and an inhibition of Bcl-2. We observed that Sox9 silencing provoked a delayed S-phase progression and an increased nuclear localization of p21. The protein stability of cyclin D1 was induced in the absence of Sox9 presumably as a function of altered p38 signalling. In addition, the major transcription factor for adipogenic differentiation, C/EBPß, was repressed after silencing Sox9. The nearly complete absence of C/EBPß protein as a result of increased destabilization of the C/EBPß mRNA and the impact on osteocalcin gene expression and protein synthesis, suggests that a delicate balance of Sox9 level is not only imperative for proper chondrogenic differentiation of progenitor cells, but also affects the adipogenic and probably osteogenic differentiation pathways of MSC. Our results identified Sox9 as an important link between differentiation, proliferation and apoptosis in undifferentiated adult rat mesenchymal stem cells, emphasizing the importance of the delicate balance of a precisely regulated Sox9 activity in MSC not only for proper skeletal development during embryogenesis but probably also for successful repair and regeneration of tissues and organs in adults.


Subject(s)
Adipogenesis/genetics , Bone Marrow Cells/metabolism , Gene Expression Regulation , Mesenchymal Stem Cells/metabolism , Osteogenesis/genetics , SOX9 Transcription Factor/genetics , Animals , Apoptosis , Bone Marrow Cells/cytology , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Differentiation , Cell Proliferation , Cell Survival/genetics , Cyclin D1/genetics , Cyclin D1/metabolism , Gene Knockdown Techniques , Mesenchymal Stem Cells/cytology , Osteocalcin/genetics , Osteocalcin/metabolism , Primary Cell Culture , Rats , SOX9 Transcription Factor/metabolism , Signal Transduction , p21-Activated Kinases/genetics , p21-Activated Kinases/metabolism
8.
FASEB J ; 28(2): 683-91, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24148886

ABSTRACT

Osteoarthritis (OA) is the most common form of arthritis. It is characterized by cartilage destruction and bone remodeling, mediated in part by synovial fibroblasts (SFs). Given the functional significance of cadherins in these cells, we aimed at determining the role of genetic variants of N-cadherin (CDH2) in OA of the knee and hip. Six single-nucleotide polymorphisms in the genomic region of the CDH2 gene were genotyped in 312 patients with OA and 259 healthy control subjects. Gene expression of CDH2 was analyzed by qRT-PCR. Liquid chromatography-mass spectrometry was used to identify a transcription factor isolated by DNA pulldown. Its potential for binding to gene variants was examined by electrophoretic mobility shift assay, enzyme-linked immunosorbent assay, and chromatin immunoprecipitation. Genetic analysis identified a polymorphism located in the CDH2 promoter region to be associated with risk of OA. The minor allele of rs11564299 had a protective effect against OA. Compared to carriers of the major allele, carriers of the minor allele of rs11564299 displayed increased N-cadherin levels in SFs. Based on in silico analysis, the minor allele was predicted to generate a novel transcription factor binding site, Direct-binding assays and mass spectrometric analysis identified hnRNP K as binding selectively to the minor allele. In summary, a CDH2 promoter polymorphism influences the risk of OA, and hnRNP K was found to be involved in the regulation of elevated N-cadherin expression in patients with OA carrying the minor allele of rs11564299.


Subject(s)
Cadherins/genetics , Osteoarthritis/genetics , Polymorphism, Genetic/genetics , Promoter Regions, Genetic/genetics , Adult , Aged , Aged, 80 and over , Binding Sites/genetics , Binding Sites/physiology , Cells, Cultured , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , Enzyme-Linked Immunosorbent Assay , Female , Genotype , Humans , Male , Middle Aged , Reverse Transcriptase Polymerase Chain Reaction , Young Adult
9.
Carcinogenesis ; 35(2): 461-8, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24104553

ABSTRACT

Posttranslational modification of different proteins via direct ubiquitin attachment is vital for mediating various cellular processes. Cylindromatosis (CYLD), a deubiquitination enzyme, is able to cleave the polyubiquitin chains from the substrate and to regulate different signaling pathways. Loss, or reduced expression, of CYLD is observed in different types of human cancer, such as hepatocellular carcinoma (HCC). However, the molecular mechanism by which CYLD affects cancerogenesis has to date not been unveiled. The aim of the present study was to examine how CYLD regulates cellular functions and signaling pathways during hepatocancerogenesis. We found that mice lacking CYLD were highly susceptible to chemically induced liver cancer. The mechanism behind proved to be an elevated proliferation rate of hepatocytes, owing to sustained c-Jun N-terminal kinase 1 (JNK1)-mediated signaling via ubiquitination of TNF receptor-associated factor 2 and expression of c-MYC. Overexpression of wild-type CYLD in HCC cell lines prevented cell proliferation, without affecting apoptosis, adhesion and migration. A combined immunohistochemical and tissue microarray analysis of 81 human HCC tissues revealed that CYLD expression is negatively correlated with expression of proliferation markers Ki-67 and c-MYC. To conclude, we found that downregulation of CYLD induces tumor cell proliferation, consequently contributing to the aggressive growth of HCC. Our findings suggest that CYLD holds potential to serve as a marker for HCC progression, and its link to c-MYC via JNK1 may provide the foundation for new therapeutic strategies for HCC patients.


Subject(s)
Carcinoma, Hepatocellular/pathology , Cysteine Endopeptidases/physiology , Liver Neoplasms/pathology , Mitogen-Activated Protein Kinase 8/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Tumor Suppressor Proteins/metabolism , Acute Lung Injury/metabolism , Acute Lung Injury/pathology , Animals , Blotting, Western , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Proliferation , Deubiquitinating Enzyme CYLD , Gene Expression Regulation, Neoplastic , Humans , Immunoenzyme Techniques , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Male , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 8/genetics , Proto-Oncogene Proteins c-myc/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , TNF Receptor-Associated Factor 2/genetics , TNF Receptor-Associated Factor 2/metabolism , Tissue Array Analysis , Tumor Suppressor Proteins/genetics
10.
Mol Carcinog ; 53(8): 635-47, 2014 Aug.
Article in English | MEDLINE | ID: mdl-23625515

ABSTRACT

An understanding of signaling pathways is a basic requirement for the treatment of melanoma. Currently, kinases are at the center of melanoma therapies. According to our research, additional alternative molecules are equally important for development of melanoma. In this regard, cancer progression is, among other factors, driven by an altered adhesion via cadherins. For instance, the de-regulated expression of the adhesion molecule T-cadherin is found in various cancer types, including melanoma, and influences migration and invasion. T-cadherin is thought to affect cellular function largely through its signaling and not its adhesion properties because the molecule is anchored into the cell membrane by a glycosylphosphatidylinositol (GPI) moiety. However, detailed knowledge about the consequences of the loss of T-cadherin in melanoma is currently lacking. For this reason, we were interested in assessing which signaling pathways are initiated by T-cadherin. The tumor growth of subcutaneously injected T-cadherin-positive melanoma cells was diminished compared with T-cadherin-negative cells in nude mice. The difference in tumor volume was not due to decreased proliferation but rather due to increased apoptosis. After the expression of T-cadherin was induced, we detected V-AKT murine thymoma viral oncogene homolog (AKT) and FoxO3a hypophosphorylation accompanied by the downregulation of the antiapoptotic molecules BCL-2, BCL-x and Clusterin. Furthermore, we detected a diminished transcriptional activity of CREB and AP-1. We demonstrated that T-cadherin functions as a pro-apoptotic tumor suppressor that antagonizes AKT/CREB/AP-1/FoxO3a signaling, whereas NFκB, TCF/LEF and mTOR are not part of the T-cadherin signaling pathway. Notably, we found that the restoration of T-cadherin in melanoma cells causes sensitization to apoptosis induced by CD95/Fas antibody CH-11.


Subject(s)
Apoptosis , Cadherins/metabolism , Cell Proliferation , Melanoma/pathology , Proto-Oncogene Proteins c-akt/metabolism , Animals , Blotting, Western , Cadherins/antagonists & inhibitors , Cadherins/genetics , Electrophoretic Mobility Shift Assay , Enzyme-Linked Immunosorbent Assay , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Humans , Immunoenzyme Techniques , Immunoprecipitation , Male , Melanoma/genetics , Melanoma/metabolism , Mice , Mice, Nude , Neovascularization, Pathologic , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
11.
Exp Eye Res ; 127: 59-68, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25051141

ABSTRACT

Although rare, uveal melanoma (UM) is the most common primary intraocular tumor in adults. About half of UM patients develop metastatic disease typically in the liver and die within a short period, due to ineffective systemic therapies. UM has unique and distinct genetic features predictive of metastasis. Animal models are required to improve our understanding of therapeutic options in disseminated UM. Since spontaneous murine UM models are lacking, our aim was to analyze the suitability of the established transgenic melanoma mouse model Tg(Grm1) as a new UM model system. We demonstrated that adult Grm1 transgenic mice develop choroidal thickening and uveal melanocytic neoplasia with expression of the melanocytic markers S100B and MelanA. Further, we showed that GRM1 is expressed in human UM, similar to skin melanoma. This study presents a new mouse model for spontaneous UM and suggests that the glutamate signaling pathway is a possible target for UM therapy.


Subject(s)
Choroid Neoplasms/pathology , Melanoma, Experimental/pathology , Animals , Biomarkers, Tumor/metabolism , Choroid/pathology , Choroid Neoplasms/genetics , Choroid Neoplasms/metabolism , Disease Models, Animal , Fluorescent Antibody Technique, Indirect , Ki-67 Antigen/metabolism , MART-1 Antigen/genetics , MART-1 Antigen/metabolism , Melanoma, Experimental/genetics , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, Metabotropic Glutamate/genetics , Receptors, Metabotropic Glutamate/metabolism , S100 Calcium Binding Protein beta Subunit/genetics , S100 Calcium Binding Protein beta Subunit/metabolism , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Tumor Cells, Cultured
12.
Mater Today Bio ; 26: 101071, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38736612

ABSTRACT

Although 2D cancer models have been the standard for drug development, they don't resemble in vivo properties adequately. 3D models can potentially overcome this. Bioprinting is a promising technique for more refined models to investigate central processes in tumor development such as proliferation, dormancy or metastasis. We aimed to analyze bioinks, which could mimic these different tumor stages in a cast vascularized arteriovenous loop melanoma model in vivo. It has the advantage to be a closed system with a defined microenvironment, supplied only with one vessel-ideal for metastasis research. Tested bioinks showed significant differences in composition, printability, stiffness and microscopic pore structure, which led to different tumor stages (Matrigel and Alg/HA/Gel for progression, Cellink Bioink for dormancy) and resulted in different primary tumor growth (Matrigel significantly higher than Cellink Bioink). Light-sheet fluorescence microscopy revealed differences in vascularization and hemorrhages with no additional vessels found in Cellink Bioink. Histologically, typical human melanoma with different stages was demonstrated. HMB-45-positive tumors in progression inks were infiltrated by macrophages (CD163), highly proliferative (Ki67) and metastatic (MITF/BRN2, ATX, MMP3). Stainings of lymph nodes revealed metastases even without significant primary tumor growth in Cellink Bioink. This model can be used to study tumor pathology and metastasis of different tumor stages and therapies.

13.
Sci Rep ; 14(1): 12945, 2024 06 05.
Article in English | MEDLINE | ID: mdl-38839791

ABSTRACT

Extrusion-based bioprinting is an established method in biofabrication. Suitable bioinks have fundamentally different compositions and characteristics, which should be examined, in order to find a perfect model system. Here, we investigate the effect of two alginate-based, yet unalike 3D-printed bioinks, pre-crosslinked alginate-dialdehyde gelatin (ADA-GEL) and a mixture of alginate, hyaluronic acid, and gelatin (Alg/HA/Gel), on the melanoma cell line Mel Im and vice versa in terms of stiffness, shrinkage, cellular behavior and colony formation over 15 days. Rheological stiffness measurements revealed two soft gels with similar storage moduli. The cells did not have a significant impact on the overall stiffness, whereas ADA-GEL (2.5/2.5%) was significantly stiffer than Alg/HA/Gel (0.5/0.1/3%). Regarding the shrinkage of printed constructs, cells had a significant influence, especially in ADA-GEL, which has covalent bonds between the oxidized alginate and gelatin. Multi-photon microscopy exhibited proliferation, cell spreading and migration in ADA-GEL with cell-cell and cell-matrix interaction, dissimilarly to Alg/HA/Gel, in which cells formed spherical, encapsulated colonies. Scanning electron microscopy and histology showed degradation and multi-layered growth on ADA-GEL and fewer examples of escaped cells on Alg/HA/Gel. Both gels serve as proliferation bioink for melanoma with more necrosis in deeper Alg/HA/Gel colonies and differences in spreading and matrix interaction. These findings show the importance of proper characterization of the bioinks for different applications.


Subject(s)
Alginates , Bioprinting , Cell Proliferation , Gelatin , Melanoma , Printing, Three-Dimensional , Alginates/chemistry , Melanoma/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Gelatin/chemistry , Bioprinting/methods , Humans , Ink , Hyaluronic Acid/chemistry , Rheology , Tissue Scaffolds/chemistry , Tissue Engineering/methods
14.
Exp Mol Pathol ; 95(2): 156-65, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23831429

ABSTRACT

Leukemia inhibitory factor (LIF) signaling regulates cellular processes to maintain the self-renewal and pluripotency of embryonic stem (ES) cells. Independent of these capabilities, LIF was also identified to be responsible for cancer development and progression. However, its detailed cellular function in cancer remains unclear thus far. We found LIF to be expressed in melanoma cell lines of primary and metastatic origin and in melanoma tissue. We further elucidated stimuli that are responsible for the high expression levels of LIF. Interestingly, hypoxia, specifically through HIF-1α, is involved in regulating LIF. Furthermore, our data showed that the signaling of LIF was not mediated by the classically described pathway via STAT3, but rather through BMP4 and BMP7. We hypothesize that the co-expression of LIF and BMP is necessary for a de-differentiated cancer phenotype. Ancillary to BMP4 and BMP7, classical stem cell proteins, e.g., SOX2, NANOG, OCT3/4 and GBX2, are regulated by LIF. We therefore speculate that LIF can induce a typical "cancer stem cell"-like behavior, as the appropriate genes are regulated by LIF. Particularly, the expression of these genes has been proposed as a driving force for tumorigenesis and the initiation of metastasis. Notably, LIF has an important role not only for ES cells but also for cancer development. Melanoblast-related cells (MBrcs), which resemble the neural crest precursor cells of melanocytes, expressed LIF in minor amounts compared to normal human melanocytes. These data, along with the data that LIF is upregulated in melanoma cell lines compared to melanocytes, strongly indicate that LIF is important for the stabilization of the melanoma phenotype. To elucidate the role of LIF in cellular melanoma behavior, we analyzed proliferation, attachment, migration and colony formation after silencing LIF by siRNA, and found all four characteristics restricted. In summary, we can show that LIF is an important factor in melanoma progression.


Subject(s)
Leukemia Inhibitory Factor/metabolism , Melanoma/metabolism , Signal Transduction/physiology , Blotting, Western , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/genetics , Gene Knockdown Techniques , Humans , Kruppel-Like Factor 4 , Leukemia Inhibitory Factor/genetics , Melanoma/genetics , Melanoma/pathology , RNA, Small Interfering , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transfection
15.
iScience ; 26(6): 106919, 2023 Jun 16.
Article in English | MEDLINE | ID: mdl-37283806

ABSTRACT

The bulge of hair follicles harbors Nestin+ (neural crest like) stem cells, which exhibit the potential to generate various cell types including melanocytes. In this study, we aimed to determine the role of Sox9, an important regulator during neural crest development, in melanocytic differentiation of those adult Nestin+ cells. Immunohistochemical analysis after conditional Sox9 deletion in Nestin+ cells of adult mice revealed that Sox9 is crucial for melanocytic differentiation of these cells and that Sox9 acts as a fate determinant between melanocytic and glial fate. A deeper understanding of factors that regulate fate decision, proliferation and differentiation of these stem cells provides new aspects to melanoma research as melanoma cells share many similarities with neural crest cells. In summary, we here show the important role of Sox9 in melanocytic versus glial fate decision of Nestin+ stem cells in the skin of adult mice.

16.
Sci Rep ; 13(1): 9561, 2023 06 12.
Article in English | MEDLINE | ID: mdl-37308689

ABSTRACT

Originally considered to act as a transcriptional co-factor, Pirin has recently been reported to play a role in tumorigenesis and the malignant progression of many tumors. Here, we have analyzed the diagnostic and prognostic value of Pirin expression in the early stages of melanoma, and its role in the biology of melanocytic cells. Pirin expression was analyzed in a total of 314 melanoma biopsies, correlating this feature with the patient's clinical course. Moreover, PIR downregulated primary melanocytes were analyzed by RNA sequencing, and the data obtained were validated in human melanoma cell lines overexpressing PIR by functional assays. The immunohistochemistry multivariate analysis revealed that early melanomas with stronger Pirin expression were more than twice as likely to develop metastases during the follow-up. Transcriptome analysis of PIR downregulated melanocytes showed a dampening of genes involved in the G1/S transition, cell proliferation, and cell migration. In addition, an in silico approach predicted that JARID1B as a potential transcriptional regulator that lies between PIR and its downstream modulated genes, which was corroborated by co-transfection experiments and functional analysis. Together, the data obtained indicated that Pirin could be a useful marker for the metastatic progression of melanoma and that it participates in the proliferation of melanoma cells by regulating the slow-cycling JARID1B gene.


Subject(s)
Melanoma , Humans , Prognosis , Melanocytes , Biopsy , Transcription Factors , Cell Proliferation , Nuclear Proteins , Repressor Proteins , Jumonji Domain-Containing Histone Demethylases
17.
Lab Invest ; 92(12): 1788-800, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23069940

ABSTRACT

T-cadherin (cadherin 13, H-cadherin, gene name CDH13) has been proposed to act as a tumor-suppressor gene as its expression is significantly diminished in several types of carcinomas, including melanomas. Allelic loss and promoter hypermethylation have been proposed as mechanisms for silencing of CDH13. However, they do not account for loss of T-cadherin expression in all carcinomas, and other genetic or epigenetic alterations can be presumed. The present study investigated transcriptional regulation of CDH13 in melanoma. Bioinformatical analysis pointed to the presence of known BRN2 (also known as POU3F2 and N-Oct-3)-binding motifs in the CDH13 promoter sequence. We found an inverse correlation between BRN2 and T-cadherin protein and transcript expression. Reporter gene analysis and electrophoretic mobility shift assays in melanoma cells demonstrated that CDH13 is a direct target of BRN2 and that BRN2 is a functional transcriptional repressor of CDH13 promoter activity. The regulatory binding element of BRN2 was located -219 bp of the CDH13 promoter proximal to the start codon and was identified as 5'-CATGCAAAA-3'. Ectopic expression of BRN2 in BRN2-negative/T-cadherin-positive melanoma cells resulted in suppression of CDH13 promoter activity, whereas BRN2 knockdown in BRN2-positive/T-cadherin-negative melanoma cells resulted in re-expression of T-cadherin transcripts and protein. Transcriptional repression of CDH13 by BRN2 may participate in malignant transformation of melanoma by increasing invasion and migration potentials of melanoma cells. The study has identified CDH13 as a novel direct BRN2 transcriptional target gene and has advanced knowledge of mechanisms underlying loss of T-cadherin expression in melanoma.


Subject(s)
Cadherins/genetics , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Melanoma/genetics , POU Domain Factors/genetics , Repressor Proteins/genetics , Base Sequence , Cadherins/metabolism , Cell Line, Tumor , Electrophoretic Mobility Shift Assay , Homeodomain Proteins/metabolism , Humans , Immunohistochemistry , Melanoma/metabolism , Molecular Sequence Data , POU Domain Factors/metabolism , Promoter Regions, Genetic , Real-Time Polymerase Chain Reaction , Repressor Proteins/metabolism
18.
Am J Pathol ; 178(3): 1145-52, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21356366

ABSTRACT

Recently, we have shown that down-regulation of methylthioadenosine phosphorylase (MTAP) in hepatocellular carcinoma (HCC) cells enhances the invasive potential and the resistance against cytokines. Here, we aimed at investigating the molecular mechanism underlying this tumor-promoting effect and expanded the analysis to a large series of human HCC tissues. Liquid chromatography tandem mass spectrometry revealed that reduced MTAP expression resulted in higher intra- and extracellular concentrations of 5'-deoxy-5'-methylthioadenosine (MTA) in cultivated HCC cells and, concordantly, higher levels of MTA in HCC tissue. MTA induced matrix metalloproteinase (MMP) and interleukin-8 transcription in HCC cells in vitro, accompanied by enhanced proliferation and activation of the transcription factor NFκB. In addition, MTA secreted by HCC cells induced expression of fibroblast growth factor-2 and MMP1 in stromal myofibroblasts. In human HCC tissues, MTAP mRNA correlated inversely with MTA levels, and immunohistochemical analysis of a tissue microarray of 140 human HCCs revealed that low MTAP protein expression correlated with advanced tumor stages. In conclusion, MTAP deficiency results in accumulation of MTA, which is associated with increased tumorigenicity. These data further indicate MTAP as a tumor suppressor in HCC, and MTA as a potential biomarker for HCC progression.


Subject(s)
Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/pathology , Deoxyadenosines/metabolism , Down-Regulation , Liver Neoplasms/enzymology , Liver Neoplasms/pathology , Purine-Nucleoside Phosphorylase/metabolism , Thionucleosides/metabolism , Aged , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Deoxyadenosines/pharmacology , Disease Progression , Down-Regulation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/enzymology , Humans , Liver Neoplasms/genetics , Male , Middle Aged , NF-kappa B/metabolism , Purine-Nucleoside Phosphorylase/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Thionucleosides/pharmacology
19.
Biosensors (Basel) ; 12(7)2022 Jul 01.
Article in English | MEDLINE | ID: mdl-35884286

ABSTRACT

Due to the occurrence of THz-excited vibrational modes in biomacromolecules, the THz frequency range has been identified as particularly suitable for developing and applying new bioanalytical methods. We present a scalable THz metamaterial-based biosensor being utilized for the multifrequency investigation of single- and double-stranded DNA (ssDNA and dsDNA) samples. It is demonstrated that the metamaterial resonance frequency shift by the DNA's presence depends on frequency. Our experiments with the scalable THz biosensors demonstrate a major change in the degree of the power function for dsDNA by 1.53 ± 0.06 and, in comparison, 0.34 ± 0.11 for ssDNA as a function of metamaterial resonance frequency. Thus, there is a significant advantage for dsDNA detection that can be used for increased sensitivity of biomolecular detection at higher frequencies. This work represents a first step for application-specific biosensors with potential advantages in sensitivity, specificity, and robustness.


Subject(s)
Biosensing Techniques , DNA , DNA, Single-Stranded
20.
Cells ; 11(13)2022 06 30.
Article in English | MEDLINE | ID: mdl-35805172

ABSTRACT

Pigmentation is an important process in skin physiology and skin diseases and presumably also plays a role in Parkinson's disease (PD). In PD, alpha-Synuclein (aSyn) has been shown to be involved in the pigmentation of neurons. The presynaptic protein is intensively investigated for its pathological role in PD, but its physiological function remains unknown. We hypothesized that aSyn is both involved in melanocytic differentiation and melanosome trafficking processes. We detected a strong expression of aSyn in human epidermal melanocytes (NHEMs) and observed its regulation in melanocytic differentiation via the microphthalmia-associated transcription factor (MITF), a central regulator of differentiation. Moreover, we investigated its role in pigmentation by performing siRNA experiments but found no effect on the total melanin content. We discovered a localization of aSyn to melanosomes, and further analysis of aSyn knockdown revealed an important role in melanocytic morphology and a reduction in melanosome release. Additionally, we found a reduction of transferred melanosomes in co-culture experiments of melanocytes and keratinocytes but no complete inhibition of melanosome transmission. In summary, this study highlights a novel physiological role of aSyn in melanocytic morphology and its so far unknown function in the pigment secretion in melanocytes.


Subject(s)
Melanocytes , alpha-Synuclein/metabolism , Humans , Keratinocytes/metabolism , Melanins/metabolism , Melanocytes/metabolism , Melanosomes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL