Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Angiogenesis ; 27(1): 37-49, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37493987

ABSTRACT

Modern drug development increasingly requires comprehensive models that can be utilized in the earliest stages of compound and target discovery. Here we report a phenotypic screening exercise in a high-throughput Organ-on-a-Chip setup. We assessed the inhibitory effect of 1537 protein kinase inhibitors in an angiogenesis assay. Over 4000 micro-vessels were grown under perfusion flow in microfluidic chips, exposed to a cocktail of pro-angiogenic factors and subsequently exposed to the respective kinase inhibitors. Efficacy of compounds was evaluated by reduced angiogenic sprouting, whereas reduced integrity of the main micro-vessel was taken as a measure for toxicity. The screen yielded 53 hits with high anti-angiogenicity and low toxicity, of which 44 were previously unassociated with angiogenic pathways. This study demonstrates that Organ-on-a-Chip models can be screened in high numbers to identify novel compounds and targets. This will ultimately reduce bias in early-stage drug development and increases probability to identify first in class compounds and targets for today's intractable diseases.


Subject(s)
Angiogenesis , Antineoplastic Agents , Humans , Microphysiological Systems , Antineoplastic Agents/therapeutic use , Neovascularization, Pathologic/drug therapy , Protein Kinase Inhibitors/pharmacology
2.
ALTEX ; 40(4): 649-664, 2023.
Article in English | MEDLINE | ID: mdl-37422924

ABSTRACT

Lung cancer is a leading cause of death worldwide, with only a fraction of patients responding to immunotherapy. The correlation between increased T-cell infiltration and positive patient outcomes has motivated the search for therapeutics promoting T-cell infiltration. While transwell and spheroid platforms have been employed, these models lack flow and endothelial barriers, and cannot faithfully model T-cell adhesion, extravasation, and migration through 3D tissue. Presented here is a 3D chemotaxis assay, in a lung tumor-on-chip model with 3D endothelium (LToC-Endo), to address this need. The described assay consists of a HUVEC-derived vascular tubule cultured under rocking flow, through which T-cells are added; a collagenous stromal barrier, through which T-cells migrate; and a chemoattractant/tumor (HCC0827 or NCI-H520) compartment. Here, activated T-cells extravasate and migrate in response to gradients of rhCXCL11 and rhCXCL12. Adopting a T-cell activation protocol with a rest period enables proliferative burst prior to introducing T-cells into chips and enhances assay sensitivity. In addition, incorporating this rest recovers endothelial activation in response to rhCXCL12. As a final control, we show that blocking ICAM-1 interferes with T-cell adhesion and chemotaxis. This microphysiological system, which mimics in vivo stromal and vascular barriers, can be used to evaluate potentiation of immune chemotaxis into tumors while probing for vascular responses to potential therapeutics. Finally, we propose translational strategies by which this assay could be linked to preclinical and clinical models to support human dose prediction, personalized medicine, and the reduction, refinement, and replacement of animal models.


Subject(s)
Lung Neoplasms , Microphysiological Systems , Animals , Humans , Cells, Cultured , Endothelium, Vascular , Lung Neoplasms/drug therapy , Cell Movement
3.
PLoS One ; 17(4): e0266812, 2022.
Article in English | MEDLINE | ID: mdl-35395060

ABSTRACT

Huntington's disease (HD) is caused by an expansion of the CAG trinucleotide repeat domain in the huntingtin gene that results in expression of a mutant huntingtin protein (mHTT) containing an expanded polyglutamine tract in the amino terminus. A number of therapeutic approaches that aim to reduce mHTT expression either locally in the CNS or systemically are in clinical development. We have previously described sensitive and selective assays that measure human HTT proteins either in a polyglutamine-independent (detecting both mutant expanded and non-expanded proteins) or in a polyglutamine length-dependent manner (detecting the disease-causing polyglutamine repeats) on the electrochemiluminescence Meso Scale Discovery detection platform. These original assays relied upon polyclonal antibodies. To ensure an accessible and sustainable resource for the HD field, we developed similar assays employing monoclonal antibodies. We demonstrate that these assays have equivalent sensitivity compared to our previous assays through the evaluation of cellular and animal model systems, as well as HD patient biosamples. We also demonstrate cross-site validation of these assays, allowing direct comparison of studies performed in geographically distinct laboratories.


Subject(s)
Huntington Disease , Animals , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Huntington Disease/genetics , Huntington Disease/metabolism , Peptides/genetics , Peptides/metabolism , Trinucleotide Repeat Expansion
4.
Int J Mol Sci ; 22(15)2021 Jul 30.
Article in English | MEDLINE | ID: mdl-34361000

ABSTRACT

The recruitment of T cells is a crucial component in the inflammatory cascade of the body. The process involves the transport of T cells through the vascular system and their stable arrest to vessel walls at the site of inflammation, followed by extravasation and subsequent infiltration into tissue. Here, we describe an assay to study 3D T cell dynamics under flow in real time using a high-throughput, artificial membrane-free microfluidic platform that allows unimpeded extravasation of T cells. We show that primary human T cells adhere to endothelial vessel walls upon perfusion of microvessels and can be stimulated to undergo transendothelial migration (TEM) by TNFα-mediated vascular inflammation and the presence of CXCL12 gradients or ECM-embedded melanoma cells. Notably, migratory behavior was found to differ depending on T cell activation states. The assay is unique in its comprehensiveness for modelling T cell trafficking, arrest, extravasation and migration, all in one system, combined with its throughput, quality of imaging and ease of use. We envision routine use of this assay to study immunological processes and expect it to spur research in the fields of immunological disorders, immuno-oncology and the development of novel immunotherapeutics.


Subject(s)
Microfluidics/methods , T-Lymphocytes/physiology , Transendothelial and Transepithelial Migration , Cell Adhesion , Cell Line, Tumor , Cells, Cultured , Chemokine CXCL12/metabolism , Endothelium, Vascular/physiology , Extracellular Matrix/metabolism , Humans , Melanoma/metabolism , Melanoma/pathology , T-Lymphocytes/metabolism , Tumor Necrosis Factor-alpha/metabolism
5.
Mol Cancer Res ; 15(12): 1714-1721, 2017 12.
Article in English | MEDLINE | ID: mdl-28860121

ABSTRACT

Nicotinamide phosphoribosyltransferase (NAMPT) and nicotinic acid phosphoribosyltransferase (NAPRT) are rate-limiting enzymes in the NAD+ synthesis pathway. Chondrosarcoma is a malignant cartilage forming bone tumor, in which mutations altering isocitrate dehydrogenase-1 and -2 (IDH1 and IDH2) activity have been identified as potential driver mutations. Vulnerability for NAD+ depletion has been reported for IDH1/2-mutant cells. Here, the potency of NAMPT inhibitors as a treatment of chondrosarcoma was explored. Eleven chondrosarcoma cell lines were treated with NAMPT inhibitors, in which the effect on cell viability, colony formation, and 3D collagen invasion was assessed. The expression level of NAMPT and NAPRT transcripts in chondrosarcoma cells was determined by qRT-PCR. Methylation of the NAPRT promoter was evaluated using a previously published dataset of genome-wide methylation. In addition, a methylation dataset was used to determine methylation of the NAPRT promoter in 20 IDH1/2-mutated cartilage tumors. Chondrosarcoma cells showed a dose-dependent decrease in cell viability, 3D collagen invasion, and colony formation upon treatment with NAMPT inhibitors, in which nearly half of the cell lines demonstrated absolute IC50s in the low nanomolar range. Increasing IC50s correlated to increasing NAPRT expression levels and decreasing NAPRT promoter methylation. No correlation between IDH1/2 mutation status and sensitivity for NAMPT inhibitors was observed. Strikingly, higher methylation of the NAPRT promoter was observed in high-grade versus low-grade chondrosarcomas. In conclusion, this study identified NAMPT as a potential target for treatment of chondrosarcoma.Implications: Chondrosarcoma patients, especially those of high histologic grade with lower expression and hypermethylation of NAPRT, may benefit from inhibition of the NAD synthesis pathway. Mol Cancer Res; 15(12); 1714-21. ©2017 AACR.


Subject(s)
Chondrosarcoma/genetics , Cytokines/genetics , Isocitrate Dehydrogenase/genetics , Neoplasms, Bone Tissue/genetics , Nicotinamide Phosphoribosyltransferase/genetics , Pentosyltransferases/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Chondrosarcoma/drug therapy , Chondrosarcoma/pathology , Cytokines/antagonists & inhibitors , Enzyme Inhibitors/administration & dosage , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mutation , NAD/antagonists & inhibitors , NAD/biosynthesis , NAD/genetics , Neoplasm Invasiveness/genetics , Neoplasms, Bone Tissue/drug therapy , Neoplasms, Bone Tissue/pathology , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Pentosyltransferases/antagonists & inhibitors , Promoter Regions, Genetic/drug effects , Signal Transduction/drug effects
6.
Clin Sarcoma Res ; 7: 8, 2017.
Article in English | MEDLINE | ID: mdl-28484589

ABSTRACT

BACKGROUND: Mutations in isocitrate dehydrogenase (IDH)1 or -2 are found in ~50% of conventional central chondrosarcomas and in up to 87% of their assumed benign precursors enchondromas. The mutant enzyme acquires the activity to convert α-ketoglutarate into the oncometabolite d-2-hydroxyglutarate (d-2-HG), which competitively inhibits α-ketoglutarate dependent enzymes such as histone- and DNA demethylases. METHODS: We therefore evaluated the effect of IDH1 or -2 mutations on histone modifications (H3K4me3, H3K9me3 and H3K27me3), chromatin remodeler ATRX expression, DNA modifications (5-hmC and 5-mC), and TET1 subcellular localization in a genotyped cohort (IDH, succinate dehydrogenase (SDH) and fumarate hydratase (FH)) of enchondromas and central chondrosarcomas (n = 101) using immunohistochemistry. RESULTS: IDH1 or -2 mutations were found in 60.8% of the central cartilaginous tumours, while mutations in FH and SDH were absent. The mutation status did not correlate with outcome. Chondrosarcomas are strongly positive for the histone modifications H3K4me3, H3K9me3 and H3K27me3, which was independent of the IDH1 or -2 mutation status. Two out of 36 chondrosarcomas (5.6%) show complete loss of ATRX. Levels of 5-hmC and 5-mC are highly variable in central cartilaginous tumours and are not associated with mutation status. In tumours with loss of 5-hmC, expression of TET1 was more prominent in the cytoplasm than the nucleus (p = 0.0001). CONCLUSIONS: In summary, in central chondrosarcoma IDH1 or -2 mutations do not affect immunohistochemical levels of 5-hmC, 5mC, trimethylation of H3K4, -K9 and K27 and outcome, as compared to wildtype.

7.
Lab Invest ; 96(10): 1128-37, 2016 10.
Article in English | MEDLINE | ID: mdl-27617402

ABSTRACT

Mesenchymal chondrosarcomas are rare and highly aggressive sarcomas occurring in bone and soft tissue, with poor overall survival. Bcl-2 expression was previously shown to be upregulated in mesenchymal chondrosarcomas. We here report on a newly derived mesenchymal chondrosarcoma cell line, MCS170, in which we investigated treatment with the BH3 mimetic ABT-737 alone or in combination with conventional chemotherapy as a possible new therapeutic strategy. The presence of the characteristic HEY1-NCOA2 fusion was confirmed in the MCS170 cell line using FISH, RT-PCR, and sequencing. The MCS170 cell line was treated with ABT-737 alone or in combination with doxorubicin or cisplatin. Cell viability and proliferation was determined using WST-1 viability assays and the xCELLigence system. Expression of Bcl-2 family members was studied using immunohistochemistry. Apoptosis was determined using the caspase-glo 3/7 assay and western blot for PARP cleavage. The MCS170 cell line was sensitive to doxorubicin treatment with an IC50 of 0.09 µM after 72 h, but more resistant to cisplatin treatment with an IC50 of 4.5 µM after 72 h. Cells showed little sensitivity toward ABT-737 with an IC50 of 1.8 µM after 72 h. Combination treatments demonstrated ABT-737 synergism with cisplatin as well as doxorubicin as shown by induction of apoptosis and reduction in cell proliferation. Restoration of the apoptotic machinery by inhibition of Bcl-2 family members sensitizes MCS170 mesenchymal chondrosarcoma cells to conventional chemotherapy. This indicates that combining the inhibition of Bcl-2 family members with conventional chemotherapy can be a possible therapeutic strategy for patients with mesenchymal chondrosarcoma.


Subject(s)
Cell Line, Tumor/drug effects , Chondrosarcoma, Mesenchymal , Drug Resistance, Neoplasm , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Adult , Antineoplastic Agents , Biphenyl Compounds , Cisplatin , Doxorubicin , Humans , Male , Nitrophenols , Piperazines , Sulfonamides
8.
Oncotarget ; 6(36): 38777-88, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-26472283

ABSTRACT

Succinate dehydrogenase (SDH) and fumarate hydratase (FH) are tricarboxylic acid (TCA) cycle enzymes and tumor suppressors. Loss-of-function mutations give rise to hereditary paragangliomas/pheochromocytomas and hereditary leiomyomatosis and renal cell carcinoma. Inactivation of SDH and FH results in an abnormal accumulation of their substrates succinate and fumarate, leading to inhibition of numerous α-ketoglutarate dependent dioxygenases, including histone demethylases and the ten-eleven-translocation (TET) family of 5-methylcytosine (5 mC) hydroxylases. To evaluate the distribution of DNA and histone methylation, we used immunohistochemistry to analyze the expression of 5 mC, 5-hydroxymethylcytosine (5 hmC), TET1, H3K4me3, H3K9me3, and H3K27me3 on tissue microarrays containing paragangliomas/pheochromocytomas (n = 134) and hereditary and sporadic smooth muscle tumors (n = 56) in comparison to their normal counterparts. Our results demonstrate distinct loss of 5 hmC in tumor cells in SDH- and FH-deficient tumors. Loss of 5 hmC in SDH-deficient tumors was associated with nuclear exclusion of TET1, a known regulator of 5 hmC levels. Moreover, increased methylation of H3K9me3 occurred predominantly in the chief cell component of SDH mutant tumors, while no changes were seen in H3K4me3 and H3K27me3, data supported by in vitro knockdown of SDH genes. We also show for the first time that FH-deficient smooth muscle tumors exhibit increased H3K9me3 methylation compared to wildtype tumors. Our findings reveal broadly similar patterns of epigenetic deregulation in both FH- and SDH-deficient tumors, suggesting that defects in genes of the TCA cycle result in common mechanisms of inhibition of histone and DNA demethylases.


Subject(s)
Adrenal Gland Neoplasms/genetics , Cytosine/analogs & derivatives , Fumarate Hydratase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Paraganglioma/genetics , Pheochromocytoma/genetics , Smooth Muscle Tumor/genetics , Succinate Dehydrogenase/genetics , 5-Methylcytosine/analogs & derivatives , Adrenal Gland Neoplasms/enzymology , Cell Nucleus/metabolism , Cytosine/metabolism , Fumarate Hydratase/deficiency , Fumarate Hydratase/metabolism , Gene Silencing , HEK293 Cells , Humans , Immunohistochemistry , Mixed Function Oxygenases/metabolism , Paraganglioma/enzymology , Paraganglioma/pathology , Pheochromocytoma/enzymology , Pheochromocytoma/pathology , Proto-Oncogene Proteins/metabolism , Smooth Muscle Tumor/enzymology , Succinate Dehydrogenase/deficiency , Succinate Dehydrogenase/metabolism
9.
Oncotarget ; 6(17): 14832-42, 2015 Jun 20.
Article in English | MEDLINE | ID: mdl-26046462

ABSTRACT

Mutations in isocitrate dehydrogenase 1 (IDH1) and IDH2 are found in a somatic mosaic fashion in patients with multiple enchondromas. Enchondromas are benign cartilaginous tumors arising in the medulla of bone. The mutant IDH1/2 causes elevated levels of D-2-hydroxyglutarate (D-2-HG). Mesenchymal stem cells (MSC) are the precursor of the osteoblastic, chondrogenic and adipocytic lineage and we hypothesized that increased levels of D-2-HG cause multiple enchondromas by affecting differentiation of MSCs. Bone marrow derived MSCs from different donors were differentiated towards osteoblastic, chondrogenic and adipocytic lineage in the presence or absence of 5 mM D-2-HG. Three of four MSCs showed near complete inhibition of calcification after 3 weeks under osteogenic differentiation conditions in the presence of D-2-HG, indicating a block in osteogenic differentiation. Two of four MSCs showed an increase in differentiation towards the chondrogenic lineage. To evaluate the effect of D-2-HG in vivo we monitored bone development in zebrafish, which revealed an impaired development of vertebrate rings in the presence of D-2-HG compared to control conditions (p-value < 0.0001). Our data indicate that increased levels of D-2-HG promote chondrogenic over osteogenic differentiation. Thus, mutations in IDH1/2 lead to a local block in osteogenic differentiation during skeletogenesis causing the development of benign cartilaginous tumors.


Subject(s)
Cell Differentiation/physiology , Glutarates/metabolism , Isocitrate Dehydrogenase/metabolism , Osteoblasts/metabolism , Animals , Bone Development/drug effects , Bone and Bones/drug effects , Bone and Bones/embryology , Cell Differentiation/drug effects , Cells, Cultured , Chondrogenesis/drug effects , Chondroma/genetics , Chondroma/metabolism , Glutarates/pharmacology , Humans , Immunohistochemistry , Isocitrate Dehydrogenase/genetics , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mutation , Osteoblasts/cytology , Osteoblasts/drug effects , Osteogenesis/drug effects , Zebrafish/embryology
10.
Oncotarget ; 6(14): 12505-19, 2015 May 20.
Article in English | MEDLINE | ID: mdl-25895133

ABSTRACT

Mutations in isocitrate dehydrogenase 1 (IDH1) and IDH2 are found in a subset of benign and malignant cartilage tumors, gliomas and leukaemias. The mutant enzyme causes the production of D-2-hydroxyglutarate (D-2-HG), affecting CpG island and histone methylation. While mutations in IDH1/2 are early events in benign cartilage tumors, we evaluated whether these mutations play a role in malignant chondrosarcomas. Compared to IDH1/2 wildtype cell lines, chondrosarcoma cell lines harboring an endogenous IDH1 (n=3) or IDH2 mutation (n=2) showed up to a 100-fold increase in intracellular and extracellular D-2-HG levels. Specific inhibition of mutant IDH1 using AGI-5198 decreased levels of D-2-HG in a dose dependent manner. After 72 hours of treatment one out of three mutant IDH1 cell lines showed a moderate decrease in viability , while D-2-HG levels decreased >90%. Likewise, prolonged treatment (up to 20 passages) did not affect proliferation and migration. Furthermore, global gene expression, CpG island methylation as well as histone H3K4, -9, and -27 trimethylation levels remained unchanged. Thus, while IDH1/2 mutations cause enchondroma, malignant progression towards central chondrosarcoma renders chondrosarcoma growth independent of these mutations. Thus, monotherapy based on inhibition of mutant IDH1 appears insufficient for treatment of inoperable or metastasized chondrosarcoma patients.


Subject(s)
Bone Neoplasms/genetics , Chondrosarcoma/genetics , Glutarates/metabolism , Isocitrate Dehydrogenase/genetics , Blotting, Western , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cell Line, Tumor , Chondrosarcoma/metabolism , Chondrosarcoma/pathology , Chromatography, Liquid , DNA Mutational Analysis , Humans , Isocitrate Dehydrogenase/metabolism , Mutation , Real-Time Polymerase Chain Reaction , Tandem Mass Spectrometry
11.
Biochem Biophys Res Commun ; 422(1): 169-73, 2012 May 25.
Article in English | MEDLINE | ID: mdl-22575449

ABSTRACT

The small viral protein apoptin is capable of inducing apoptosis selectively in human tumor cells. In normal cells apoptin localizes in the cytoplasm where it forms aggregates, becomes epitope-shielded and eventually degraded. By inhibiting the proteasome activity with the chemical inhibitors bortezomib and Ada-Ahx(3)L(3)VS apoptin levels can be stabilized in normal cells similar to the tumor suppressor p53 protein. In contrast, proteasome inhibition in tumor cells did not affect the apoptin stability while it still stabilized p53 levels. Apparently, apoptin is degraded by proteasomal activity in normal human cells, a process that no longer takes place in tumor cells. This loss of proteasomal susceptibility appears to be specific for apoptin.


Subject(s)
Capsid Proteins/metabolism , Neoplasms/metabolism , Proteasome Endopeptidase Complex/metabolism , Antineoplastic Agents/pharmacology , Boronic Acids/pharmacology , Bortezomib , Capsid Proteins/genetics , Capsid Proteins/pharmacology , Cell Line , Cell Line, Tumor , Fibroblasts/metabolism , Humans , Mutation , Phosphorylation , Protein Stability , Proteolysis , Pyrazines/pharmacology , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL