Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Plast Reconstr Surg ; 2023 Oct 13.
Article in English | MEDLINE | ID: mdl-37832143

ABSTRACT

BACKGROUND: The driver of secondary lymphedema (SL) progression is chronic inflammation, which promotes fibrosis. Despite advances in preclinical research, a specific effector cell subpopulation as a biomarker for therapy response or stage progression is still missing for SL. METHODS: Whole skin samples of 35 murine subjects of a microsurgical-induced SL model and 12 patients with SL were collected and their fibroblasts were isolated. These lymphedema-derived fibroblasts (LAF) were cultured in a collagen I-poly-D-Lysine 3D hydrogel to mimic skin conditions. Fibroblasts from non-lymphedema skin were used as negative control and TGF-ß-stimulated fibroblasts were used to recreate profibrotic myofibroblasts. Quantitative immunocytofluorescence confocal microscopy analysis and invasion functional assays were performed in all subpopulations and statistically compared. RESULTS: In contrast to normal skin fibroblasts, LAF exhibit α-SMA-positive stress fibers and a reduced number of tight junctions in 3D hydrogel conditions. The switch from normal E-cadherin high phenotype to an N-cadherin high-E-cadherin low morphology suggests epithelial to mesenchymal transition for expansion and proliferation. This pathological behavior of LAF was confirmed by live cell imaging analysis of invasion assays. The significant activation of markers of the TGFBR2-Smad pathway and collagen synthesis (HSP-47) in LAF supports EMT phenotypic changes and previous findings relating to TGF-ß1 and fibrosis with lymphedema. CONCLUSIONS: A characteristic SL myofibroblast subpopulation was identified and translationally related to fibrosis and TGF-ß1-associated stage progression. This SL-related subpopulation was termed lymphedema-associated fibroblasts. A comprehensive stage-related characterization is required to validate LAF as a reliable biomarker for SL disease progression.

2.
Transfus Med Hemother ; 48(1): 48-59, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33708052

ABSTRACT

BACKGROUND/AIMS: Extracellular vesicles (EVs), including microvesicles and exosomes, deliver bioactive cargo mediating intercellular communication in physiological and pathological conditions. EVs are increasingly investigated as therapeutic agents and targets, but also as disease biomarkers. However, a definite consensus regarding EV isolation methods is lacking, which makes it intricate to standardize research practices and eventually reach a desirable level of data comparability. In our study, we performed an inter-laboratory comparison of EV isolation based on a differential ultracentrifugation protocol carried out in 4 laboratories in 2 independent rounds of isolation. METHODS: Conditioned medium of colorectal cancer cells was prepared and pooled by 1 person and distributed to each of the participating laboratories for isolation according to a pre-defined protocol. After EV isolation in each laboratory, quantification and characterization of isolated EVs was collectively done by 1 person having the highest expertise in the respective test method: Western blot, flow cytometry (fluorescence-activated cell sorting [FACS], nanoparticle tracking analysis (NTA), and transmission electron microscopy (TEM). RESULTS: EVs were visualized with TEM, presenting similar cup-shaped and spherical morphology and sizes ranging from 30 to 150 nm. NTA results showed similar size ranges of particles in both isolation rounds. EV preparations showed high purity by the expression of EV marker proteins CD9, CD63, CD81, Alix, and TSG101, and the lack of calnexin. FACS analysis of EVs revealed intense staining for CD63 and CD81 but lower levels for CD9 and TSG101. Preparations from 1 laboratory presented significantly lower particle numbers (p < 0.0001), most probably related to increased processing time. However, even when standardizing processing time, particle yields still differed significantly between groups, indicating inter-laboratory differences in the efficiency of EV isolation. Importantly, no relation was observed between centrifugation speed/k-factor and EV yield. CONCLUSIONS: Our findings demonstrate that quantitative differences in EV yield might be due to equipment- and operator-dependent technical variability in ultracentrifugation-based EV isolation. Furthermore, our study emphasizes the need to standardize technical parameters such as the exact run speed and k-factor in order to transfer protocols between different laboratories. This hints at substantial inter-laboratory biases that should be assessed in multi-centric studies.

3.
Cancers (Basel) ; 12(1)2019 Dec 21.
Article in English | MEDLINE | ID: mdl-31877768

ABSTRACT

Extracellular vesicles (EVs) are shed by many different cell types. Their nucleic acids content offers new opportunities for biomarker research in different solid tumors. The role of EV RNA in prostate cancer (PCa) is still largely unknown. EVs were isolated from different benign and malignant prostate cell lines and blood plasma from patients with PCa (n = 18) and controls with benign prostatic hyperplasia (BPH) (n = 7). Nanoparticle tracking analysis (NTA), Western blot, electron microscopy, and flow cytometry analysis were used for the characterization of EVs. Non-coding RNA expression profiling of PC3 metastatic PCa cells and their EVs was performed by next generation sequencing (NGS). miRNAs differentially expressed in PC3 EVs were validated with qRT-PCR in EVs derived from additional cell lines and patient plasma and from matched tissue samples. 92 miRNAs were enriched and 48 miRNAs were depleted in PC3 EVs compared to PC3 cells, which could be confirmed by qRT-PCR. miR-99b-5p was significantly higher expressed in malignant compared to benign EVs. Furthermore, expression profiling showed miR-10a-5p (p = 0.018) and miR-29b-3p (p = 0.002), but not miR-99b-5p, to be overexpressed in plasma-derived EVs from patients with PCa compared with controls. In the corresponding tissue samples, no significant differences in the miRNA expression could be observed. We thus propose that EV-associated miR-10a-5p and miR-29b-3p could serve as potential new PCa detection markers.

4.
Int J Mol Sci ; 20(17)2019 Aug 26.
Article in English | MEDLINE | ID: mdl-31454892

ABSTRACT

Microsatellite unstable (MSI) colorectal cancers (CRCs) are characterized by mutational inactivation of Transforming Growth Factor Beta Receptor Type 2 (TGFBR2). TGFBR2-deficient CRCs present altered target gene and protein expression. Such cellular alterations modulate the content of CRC-derived extracellular vesicles (EVs). EVs function as couriers of proteins, nucleic acids, and lipids in intercellular communication. At a qualitative level, we have previously shown that TGFBR2 deficiency causes overall alterations in the EV protein content. To deepen the basic understanding of altered protein dynamics, this work aimed to determine TGFBR2-dependent EV protein signatures in a quantitative manner. Using a stable isotope labeling with amino acids in cell culture (SILAC) approach for mass spectrometry-based quantification, 48 TGFBR2-regulated proteins were identified in MSI CRC-derived EVs. Overall, TGFBR2 deficiency caused upregulation of several EV proteins related to the extracellular matrix and nucleosome as well as downregulation of proteasome-associated proteins. The present study emphasizes the general overlap of proteins between EVs and their parental CRC cells but also highlights the impact of TGFBR2 deficiency on EV protein composition. From a clinical perspective, TGFBR2-regulated quantitative differences of protein expression in EVs might nominate novel biomarkers for liquid biopsy-based MSI typing in the future.


Subject(s)
Biological Assay , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Extracellular Vesicles/metabolism , Microsatellite Instability , Receptor, Transforming Growth Factor-beta Type II/metabolism , Amino Acids/metabolism , Biological Assay/methods , Cell Line, Tumor , Colorectal Neoplasms/pathology , Extracellular Vesicles/ultrastructure , Gene Expression Regulation, Neoplastic , Humans , Isotope Labeling , Protein Biosynthesis , Protein Interaction Mapping , Protein Interaction Maps , Reproducibility of Results
5.
Int J Oncol ; 55(4): 925-937, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31432155

ABSTRACT

In colorectal cancer (CRC) with microsatellite instability (MSI), >90% of cases are affected by inactivating frameshift mutations of transforming growth factor ß receptor type 2 (TGFBR2). TGFBR2 deficiency is considered to drive MSI tumor progression by abrogating downstream TGF­ß signaling. This pathway can alter the expression of coding and non­coding RNAs, including microRNAs (miRNAs), which are also present in extracellular vesicles (EVs) as post­transcriptional modulators of gene expression. In our previous study, it was shown that TGFBR2 deficiency alters the protein composition and function of EVs in MSI tumors. To investigate whether mutant TGFBR2 may also affect the miRNA cargo of EVs, the present study characterized miRNAs in EVs and their parental MSI tumor cells that differed only in TGFBR2 expression status. The HCT116­TGFBR2 MSI cell line model enables the doxycycline (dox)­inducible reconstituted expression of TGFBR2 in an isogenic background (­dox, TGFBR2 deficient; +dox, TGFBR2 proficient). Small RNA sequencing of cellular and EV miRNAs showed that the majority of the miRNAs (263/471; 56%) were shared between MSI tumor cells and their EVs. Exploratory data analysis revealed the TGBFR2­dependent cluster separation of miRNA profiles in EVs and MSI tumor cells. This segregation appeared to result from two subsets of miRNAs, the expression of which were regulated in a TGFBR2­dependent manner (EVs: n=10; MSI cells: n=15). In the EV subset, 7/10 miRNAs were downregulated and 3/10 were upregulated by TGFBR2 deficiency. In the cellular subset, 13/15 miRNAs were downregulated and 2/15 miRNAs were upregulated in the TGFBR2­deficient cells. The present study emphasizes the general overlap of miRNA profiles in MSI tumor cells and their EVs, but also highlights the impact of a single tumor driver mutation on the expression of individual miRNAs, as exemplified by the downregulation of miR­381­3p in TGFBR2­deficient MSI tumor cells and their secreted EVs.


Subject(s)
Colorectal Neoplasms/genetics , Extracellular Vesicles/genetics , MicroRNAs/genetics , Receptor, Transforming Growth Factor-beta Type II/genetics , Colorectal Neoplasms/metabolism , Doxycycline/pharmacology , Extracellular Vesicles/metabolism , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Microsatellite Instability , Mutation , Receptor, Transforming Growth Factor-beta Type II/metabolism
6.
Cell Commun Signal ; 15(1): 14, 2017 04 04.
Article in English | MEDLINE | ID: mdl-28376875

ABSTRACT

BACKGROUND: Colorectal cancers (CRCs) that lack DNA mismatch repair function exhibit the microsatellite unstable (MSI) phenotype and are characterized by the accumulation of frameshift mutations at short repetitive DNA sequences (microsatellites). These tumors recurrently show inactivating frameshift mutations in the tumor suppressor Transforming Growth Factor Beta Receptor Type 2 (TGFBR2) thereby abrogating downstream signaling. How altered TGFBR2 signaling affects exosome-mediated communication between MSI tumor cells and their environment has not been resolved. Here, we report on molecular alterations of exosomes shed by MSI cells and the biological response evoked in recipient cells. METHODS: Exosomes were isolated and characterized by electron microscopy, nanoparticle tracking, and western blot analysis. TGFBR2-dependent effects on the cargo and functions of exosomes were studied in a MSI CRC model cell line enabling reconstituted and inducible TGFBR2 expression and signaling. Microsatellite frameshift mutations in exosomal and cellular DNA were examined by PCR-based DNA fragment analysis and exosomal protein profiles were identified by mass spectrometry. Uptake of fluorescent-labeled exosomes by hepatoma recipient cells was monitored by confocal microscopy. TGFBR2-dependent exosomal effects on secreted cytokine levels of recipient cells were analyzed by Luminex technology and ELISA. RESULTS: Frameshift mutation patterns in microsatellite stretches of TGFBR2 and other MSI target genes were found to be reflected in the cargo of MSI CRC-derived exosomes. At the proteome level, reconstituted TGFBR2 expression and signaling uncovered two protein subsets exclusively occurring in exosomes derived from TGFBR2-deficient (14 proteins) or TGFBR2-proficient (five proteins) MSI donor cells. Uptake of these exosomes by recipient cells caused increased secretion (2-6 fold) of specific cytokines (Interleukin-4, Stem Cell Factor, Platelet-derived Growth Factor-B), depending on the TGFBR2 expression status of the tumor cell. CONCLUSION: Our results indicate that the coding MSI phenotype of DNA mismatch repair-deficient CRC cells is maintained in their exosomal DNA. Moreover, we uncovered that a recurrent MSI tumor driver mutation like TGFBR2 can reprogram the protein content of MSI cell-derived exosomes and in turn modulate the cytokine secretion profile of recipient cells. Apart from its diagnostic potential, these TGFBR2-dependent exosomal molecular and proteomic signatures might help to understand the signaling routes used by MSI tumors. Fricke et al. uncovered coding microsatellite instability-associated mutations of colorectal tumor driver genes like TGFBR2 in MSI tumor cellderived exosomes. Depending on the TGFBR2 expression status of their donor cells, shed exosomes show distinct proteomic signatures and promote altered cytokine secretion profiles in recipient cells.


Subject(s)
Colorectal Neoplasms/metabolism , DNA Mismatch Repair , Exosomes/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Chemokines/metabolism , Enzyme-Linked Immunosorbent Assay , Exosomes/ultrastructure , Frameshift Mutation/genetics , HCT116 Cells , Hep G2 Cells , Humans , Microsatellite Instability , Platelet-Derived Growth Factor/metabolism , Proteome/metabolism , Receptor, Transforming Growth Factor-beta Type II , Reproducibility of Results
7.
Neurol Res ; 38(5): 397-405, 2016 May.
Article in English | MEDLINE | ID: mdl-27095151

ABSTRACT

OBJECTIVES: Surgical interventions can cause systemic postoperative inflammation, which in turn can induce neuroinflammation. A close link between immune reaction and cholinergic metabolism has been postulated. Pharmacological enhancement of cholinergic activity by administering physostigmine is known to induce protective effects. It is not known, however, whether physostigmine has an impact on postoperative inflammation and acetylcholine metabolism after a partial liver resection (PLR) surgery. METHODS: Rats (n = 100) underwent a PLR or sham surgery. Rats were investigated before the intervention and 120 min and 24 h postoperatively. The control group only received sevoflurane anaesthesia. Half of each treatment group received a single intraoperative dose of physostigmine, whereas the others were given placebo. Acetylcholine (ACHE) and butyrylcholinesterase (BuCHE) activity and IL1ß, IL6 and corticosterone levels were measured in rat plasma and brain. Acetylcholine (ACH) concentrations were determined additionally in cerebral tissue. RESULTS: Surgical interventions induced a peripheral stress reaction, which was characterized by an increase (p < 0.05) in pro-inflammatory cytokines, cholinergic esterases and corticosterone at 120 min postoperatively in rat blood and in cerebral tissue. At 24 h postoperatively, all measured cerebral parameters reached control values. In blood, IL1ß and BuCHE were still increased, suggesting they are peripheral markers of a stress reaction. The reduced cerebral acetylcholine is increased after physostigmine administration. Furthermore, physostigmine reduced IL1ß (p < 0.05). CONCLUSION: We show in this observational study that a single intraoperative dose of physostigmine produced a sustained anti-inflammatory effect in rat blood and brain up to 120 min postoperatively, which was especially pronounced under the condition of PLR surgery.


Subject(s)
Anesthesia/methods , Cholinesterase Inhibitors/therapeutic use , Encephalitis , Physostigmine/therapeutic use , Postoperative Complications/prevention & control , Stress, Psychological , Acetylcholine/blood , Animals , Cholinesterases/blood , Corticosterone/blood , Cytokines/blood , Disease Models, Animal , Encephalitis/blood , Encephalitis/etiology , Encephalitis/prevention & control , Liver/surgery , Male , Rats , Rats, Wistar , Statistics, Nonparametric , Stress, Psychological/blood , Stress, Psychological/etiology , Stress, Psychological/prevention & control
8.
PLoS One ; 10(6): e0131506, 2015.
Article in English | MEDLINE | ID: mdl-26114631

ABSTRACT

Although inactivating frameshift mutations in the Transforming growth factor beta receptor type 2 (TGFBR2) gene are considered as drivers of microsatellite unstable (MSI) colorectal tumorigenesis, consequential alterations of the downstream target proteome are not resolved completely. Applying a click-it chemistry protein labeling approach combined with mass spectrometry in a MSI colorectal cancer model cell line, we identified 21 de novo synthesized proteins differentially expressed upon reconstituted TGFBR2 expression. One candidate gene, the TGF-ß family member Growth differentiation factor-15 (GDF-15), exhibited TGFBR2-dependent transcriptional upregulation causing increased intracellular and extracellular protein levels. As a new TGFBR2 target gene it may provide a link between the TGF-ß branch and the BMP/GDF branch of SMAD-mediated signaling.


Subject(s)
Colorectal Neoplasms/genetics , Growth Differentiation Factor 15/genetics , Protein Serine-Threonine Kinases/physiology , Receptors, Transforming Growth Factor beta/physiology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic/drug effects , Growth Differentiation Factor 15/metabolism , Growth Differentiation Factor 15/pharmacology , HCT116 Cells , Humans , Protein Serine-Threonine Kinases/genetics , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Smad Proteins/metabolism , Transcriptional Activation/genetics , Up-Regulation/drug effects , Up-Regulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...