Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(8)2024 Apr 20.
Article in English | MEDLINE | ID: mdl-38674102

ABSTRACT

Mesenchymal stem cells (MSCs) are an integral part of the tumor microenvironment (TME); however, their role is somewhat controversial: conflicting reports suggest that, depending on the stage of tumor development, MSCs can either support or suppress tumor growth and spread. Additionally, the influence of MSCs on drug resistance is also ambiguous. Previously, we showed that, despite MSCs proliferating significantly more slowly than cancer cells, there are chemotherapeutic drugs which proved to be similarly toxic to both cell types. Here we established 2D co-cultures and 3D co-culture spheroids from different ratios of GFP-expressing, adipose tissue-derived MSCs and A431 epidermoid carcinoma cells tagged with mCherry to investigate the effect of MSCs on cancer cell growth, survival, and drug sensitivity. We examined the cytokine secretion profile of mono- and co-cultures, explored the inner structure of the spheroids, applied MSC-(nutlin-3) and cancer cell-targeting (cisplatin) treatments separately, monitored the response with live-cell imaging and identified a new, double-fluorescent cell type emerging from these cultures. In 2D co-cultures, no effect on proliferation or drug sensitivity was observed, regardless of the changes in cytokine secretion induced by the co-culture. Conversely, 3D spheroids developed a unique internal structure consisting of MSCs, which significantly improved cancer cell survival and resilience to treatment, suggesting that physical proximity and cell-cell connections are required for MSCs to considerably affect nearby cancer cells. Our results shed light on MSC-cancer cell interactions and could help design new, better treatment options for tumors.


Subject(s)
Coculture Techniques , Mesenchymal Stem Cells , Spheroids, Cellular , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/cytology , Humans , Spheroids, Cellular/drug effects , Cell Line, Tumor , Tumor Microenvironment , Cell Proliferation/drug effects , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Cell Survival/drug effects , Cisplatin/pharmacology , Drug Tolerance , Cytokines/metabolism
2.
Cells ; 11(19)2022 09 20.
Article in English | MEDLINE | ID: mdl-36230904

ABSTRACT

Mesenchymal stem cells (MSCs) or fibroblasts are one of the most abundant cell types in the tumor microenvironment (TME) exerting various anti- and pro-apoptotic effects during tumorigenesis, invasion, and drug treatment. Despite the recently discovered importance of MSCs in tumor progression and therapy, the response of these cells to chemotherapeutics compared to cancer cells is rarely investigated. A widely accepted view is that these naive MSCs have higher drug tolerance than cancer cells due to a significantly lower proliferation rate. Here, we examine the differences and similarities in the sensitivity of MSCs and cancer cells to nine diverse chemotherapy agents and show that, although MSCs have a slower cell cycle, these cells are still sensitive to various drugs. Surprisingly, MSCs showed similar sensitivity to a panel of compounds, however, suffered fewer DNA double-stranded breaks, did not enter into a senescent state, and was virtually incapable of apoptosis. Our results suggest that MSCs and cancer cells have different cell fates after drug treatment, and this could influence therapy outcome. These findings could help design drug combinations targeting both MSCs and cancer cells in the TME.


Subject(s)
Antineoplastic Agents , Mesenchymal Stem Cells , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Apoptosis , Carcinogenesis/pathology , DNA/metabolism , Humans , Mesenchymal Stem Cells/metabolism , Tumor Microenvironment
3.
Molecules ; 25(3)2020 Feb 10.
Article in English | MEDLINE | ID: mdl-32050702

ABSTRACT

The resistance of tumors against anticancer drugs is a major impediment for chemotherapy. Tumors often develop multidrug resistance as a result of the cellular efflux of chemotherapeutic agents by ABC transporters such as P-glycoprotein (ABCB1/P-gp), Multidrug Resistance Protein 1 (ABCC1/MRP1), or Breast Cancer Resistance Protein (ABCG2/BCRP). By screening a chemolibrary comprising 140 compounds, we identified a set of naturally occurring aurones inducing higher cytotoxicity against P-gp-overexpressing multidrug-resistant (MDR) cells versus sensitive (parental, non-P-gp-overexpressing) cells. Follow-up studies conducted with the P-gp inhibitor tariquidar indicated that the MDR-selective toxicity of azaaurones is not mediated by P-gp. Azaaurone analogs possessing pronounced effects were then designed and synthesized. The knowledge gained from structure-activity relationships will pave the way for the design of a new class of anticancer drugs selectively targeting multidrug-resistant cancer cells.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Benzofurans/chemistry , Drug Resistance, Multiple , Drug Resistance, Neoplasm/drug effects , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Antineoplastic Agents/chemical synthesis , Cell Line, Tumor , Dogs , Drug Screening Assays, Antitumor , Humans , Madin Darby Canine Kidney Cells , Magnetic Resonance Spectroscopy , Structure-Activity Relationship
4.
Stem Cells Int ; 2016: 3595941, 2016.
Article in English | MEDLINE | ID: mdl-27999599

ABSTRACT

Bone tissue regeneration is a major, worldwide medical need, and several strategies have been developed to support the regeneration of extensive bone defects, including stem cell based bone grafts. In addition to the application of stem cells with high osteogenic potential, it is important to maintain proper blood flow in a bone graft to avoid inner graft necrosis. Mesenchymal stem cells (MSCs) may form both osteocytes and endothelial cells; therefore we examined the combined in vitro osteogenic and endothelial differentiation capacities of MSCs derived from adipose tissue, Wharton's jelly, and periodontal ligament. Based on a detailed characterization presented here, MSCs isolated from adipose tissue and periodontal ligament may be most appropriate for generating vascularized bone grafts.

5.
Stem Cells Dev ; 25(23): 1818-1832, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27596268

ABSTRACT

The role of extracellular vesicles (EVs) in mediating the immunosuppressory properties of mesenchymal stem cells (MSCs) has recently attracted remarkable scientific interest. The aim of this work was to analyze the transport mechanisms of membrane and cytoplasmic components between T lymphocytes and adipose tissue-derived MSCs (AD-MSCs), by focusing on the role of distinct populations of EVs, direct cell-cell contacts, and the soluble mediators per se in modulating T lymphocyte function. We found that neither murine thymocytes and human primary T cells nor Jurkat lymphoblastoid cells incorporated appreciable amounts of MSC-derived microvesicles (MVs) or exosomes (EXOs). Moreover, these particles had no effect on the proliferation and IFN-γ production of in vitro-stimulated primary T cells. In contrast, AD-MSCs incorporated large amounts of membrane components from T cells as an intensive uptake of EXOs and MVs could be observed. Interestingly, we found a bidirectional exchange of cytoplasmic components between human AD-MSCs and primary T lymphocytes, mediated by tunneling nanotubes (TNTs) derived exclusively from the T cells. In contrast, TNTs couldn't be observed between AD-MSCs and the Jurkat cells. Our results reveal a novel and efficient way of intercellular communication between MSCs and T cells, and may help a better understanding of the immunomodulatory function of MSCs.


Subject(s)
Extracellular Vesicles/metabolism , Mesenchymal Stem Cells/cytology , Nanotubes/chemistry , T-Lymphocytes/cytology , Adipose Tissue/cytology , Adult , Animals , Cell Membrane/metabolism , Child, Preschool , Coculture Techniques , Cytoplasm/metabolism , Exosomes/metabolism , Female , Flow Cytometry , Humans , Immunomodulation , Jurkat Cells , Lymphocyte Activation/immunology , Male , Mesenchymal Stem Cells/metabolism , Mice, Inbred C57BL , Microscopy, Confocal , T-Lymphocytes/metabolism
6.
Stem Cells Dev ; 24(2): 244-52, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25101689

ABSTRACT

Periodontal ligament stem cells (PDLSCs) provide an important source for tissue regeneration and may become especially useful in the formation of osteogenic seeds. PDLSCs can be cultured, expanded, and differentiated in vitro; thus, they may be applied in the long-term treatment of the defects in the dental regions. Here we studied numerous potential markers allowing the selection of human PDLSCs with a maximum differentiation potential. We followed the expression of the ATP-binding cassette subfamily G member 2 (ABCG2) membrane transporter protein and isolated ABCG2-expressing cells by using a monoclonal antibody, recognizing the transporter at the cell surface in intact cells. The expression of the ABCG2 protein, corresponding to the so-called side-population phenotype in various tissue-derived stem cells, was found to be a useful marker for the selection of PDLSCs with enhanced osteogenic, chondrogenic, and adipogenic differentiation. These findings may have important applications in achieving efficient dental tissue regeneration by using stem cells from extracted teeth.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Antigens, Differentiation/metabolism , Cell Differentiation/physiology , Neoplasm Proteins/metabolism , Periodontal Ligament/metabolism , Stem Cells/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Adolescent , Adult , Cells, Cultured , Female , Humans , Male , Periodontal Ligament/cytology , Stem Cells/cytology
7.
J Mater Sci Mater Med ; 24(2): 479-88, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23135412

ABSTRACT

Seeding of bone implants with mesenchymal stem cells (MSCs) may promote osseointegration and bone regeneration. However, implant material surfaces, such as titanium or bovine bone mineral, fail to support rapid and efficient attachment of MSCs, especially under serum-free conditions that may be desirable when human applications or tightly controlled experiments are envisioned. Here we demonstrate that a branched poly[Lys(Ser(i)-DL-Ala(m))] polymer functionalized with cyclic arginyl-glycyl-aspartate, when immobilized by simple adsorption to tissue culture plastic, surgical titanium alloy (Ti6Al4V), or Bio-Oss(®) bovine bone substitute, significantly accelerates serum-free adhesion and enhances seeding efficiency of human adipose tissue-derived MSCs. Moreover, when exposed to serum-containing osteogenic medium, MSCs survived and differentiated on the peptide-coated scaffolds. In summary, the presented novel polypeptide conjugate can be conveniently used for coating various surfaces, and may find applications whenever quick and efficient seeding of MSCs is required to various scaffolds in the absence of serum.


Subject(s)
Adipose Tissue/cytology , Bone Substitutes/metabolism , Bone Transplantation , Mesenchymal Stem Cells/drug effects , Peptides, Cyclic/pharmacology , Adipose Tissue/drug effects , Adult , Animals , Bone Substitutes/chemistry , Bone Substitutes/pharmacology , Bone Transplantation/methods , Cattle , Cell Adhesion/drug effects , Cells, Cultured , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Culture Media, Serum-Free/pharmacology , Humans , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Models, Biological , Osseointegration/drug effects , Osseointegration/physiology , Polymers/pharmacology , Surface Properties/drug effects
8.
Biochem Pharmacol ; 84(3): 260-7, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22548830

ABSTRACT

Human ABCG2 is a plasma membrane glycoprotein that provides physiological protection against xenobiotics. ABCG2 also significantly influences biodistribution of drugs through pharmacological tissue barriers and confers multidrug resistance to cancer cells. Moreover, ABCG2 is the molecular determinant of the side population that is characteristically enriched in normal and cancer stem cells. Numerous tumors depend on unregulated EGFR signaling, thus inhibition of this receptor by small molecular weight inhibitors such as gefitinib, and the novel second generation agents vandetanib, pelitinib and neratinib, is a promising therapeutic option. In the present study, we provide detailed biochemical characterization regarding the interaction of these EGFR inhibitors with ABCG2. We show that ABCG2 confers resistance to gefitinib and pelitinib, whereas the intracellular action of vandetanib and neratinib is unaltered by the presence of the transporter. At higher concentrations, however, all these EGFR inhibitors inhibit ABCG2 function, thereby promoting accumulation of ABCG2 substrate drugs. We also report enhanced expression of ABCG2 in gefitinib-resistant non-small cell lung cancer cells, suggesting potential clinical relevance of ABCG2 in acquired drug resistance. Since ABCG2 has important impact on both the pharmacological properties and anti-cancer efficiencies of drugs, our results regarding the novel EGFR inhibitors should provide useful information about their therapeutic applicability against ABCG2-expressing cancer cells depending on EGFR signaling. In addition, the finding that these EGFR inhibitors efficiently block ABCG2 function may help to design novel drug-combination therapeutic strategies.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Aminoquinolines/metabolism , Aniline Compounds/metabolism , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Neoplasm Proteins/metabolism , Piperidines/metabolism , Quinazolines/metabolism , Quinolines/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/physiology , Aminoquinolines/chemistry , Aminoquinolines/pharmacology , Aniline Compounds/chemistry , Aniline Compounds/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Cell Line , Cell Survival/drug effects , Cell Survival/physiology , Drug Resistance, Neoplasm/drug effects , Gefitinib , Humans , Neoplasm Proteins/physiology , Piperidines/chemistry , Piperidines/pharmacology , Protein Binding/drug effects , Quinazolines/chemistry , Quinazolines/pharmacology , Quinolines/chemistry , Quinolines/pharmacology
9.
Biochem Biophys Res Commun ; 422(1): 28-35, 2012 May 25.
Article in English | MEDLINE | ID: mdl-22554522

ABSTRACT

Adipose tissue-derived stromal cells (ASCs) are increasingly being studied for their usefulness in regenerative medicine. However, limited life span and donor-dependent variation of primary cells such as ASCs present major hurdles to controlled and reproducible experiments. We therefore aimed to establish immortalized ASC cell lines that provide steady supply of homogeneous cells for in vitro work while retain essential features of primary cells. To this end, combinations of human telomerase reverse transcriptase (hTERT), murine Bmi-1, and SV40 large T antigen (SV40T) were introduced by lentiviral transduction into ASCs. The resulting cell lines ASC(hTERT), ASC(Bmi-1), ASC(Bmi-1+hTERT) and ASC(SV40T+hTERT) were tested for transgene expression, telomerase activity, surface immunomarkers, proliferation, osteogenic and adipogenic differentiation, karyotype, tumorigenicity, and cellular senescence. All cell lines have maintained expression of characteristic surface immunomarkers, and none was tumorigenic. However, ASC(Bmi-1) had limited replicative potential, while the rapidly proliferating ASC(SV40T+hTERT) acquired chromosomal aberrations, departed from MSC phenotype, and lost differentiation capacity. ASC(hTERT) and ASC(hTERT+Bmi-1), on the other hand, preserved all essential MSC features and did not senesce after 100 population doublings. Notably, a subpopulation of ASC(hTERT) also acquired aberrant karyotype and showed signs of transformation after long-term culture. In conclusion, hTERT alone was sufficient to extend the life span of human ASC, but ASC(hTERT) are prone to transformation during extensive subculturing. The combination of Bmi-1 and hTERT successfully immortalized human ASCs without significantly perturbing their phenotype or biological behavior.


Subject(s)
Adipose Tissue/physiology , Cell Differentiation/genetics , Cell Transformation, Neoplastic/genetics , Cellular Senescence/physiology , Nuclear Proteins/genetics , Proto-Oncogene Proteins/genetics , Repressor Proteins/genetics , Telomerase/genetics , Adipose Tissue/cytology , Adipose Tissue/pathology , Animals , Cell Proliferation , Cellular Senescence/genetics , Gene Transfer Techniques , Humans , Karyotype , Lentivirus , Mice , Polycomb Repressive Complex 1 , Stromal Cells/cytology , Stromal Cells/pathology , Stromal Cells/physiology
10.
Biochem Biophys Res Commun ; 420(4): 869-74, 2012 Apr 20.
Article in English | MEDLINE | ID: mdl-22449574

ABSTRACT

The ATP-binding cassette (ABC) transporter ABCG2 plays an important role in tissue detoxification and confers multidrug resistance to cancer cells. Identification of expressional and functional cellular regulators of this multidrug transporter is therefore intensively pursued. The PI3-kinase/Akt signaling axis has been implicated as a key element in regulating various cellular functions, including the expression and plasma membrane localization of ABCG2. Here we demonstrate that besides inhibiting their respective target kinases, the pharmacological PI3-kinase inhibitor LY294002 and the downstream mTOR kinase inhibitor rapamycin also directly inhibit ABCG2 function. In contrast, wortmannin, another commonly used pharmacological inhibitor of PI3-kinase does not interact with the transporter. We suggest that direct functional modulation of ABCG2 should be taken into consideration when pharmacological agents are applied to dissect the specific role of PI3-kinase/Akt/mTOR signaling in cellular functions.


Subject(s)
ATP-Binding Cassette Transporters/antagonists & inhibitors , ATP-Binding Cassette Transporters/metabolism , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Androstadienes/pharmacology , Cell Line , Chromones/pharmacology , Humans , Morpholines/pharmacology , Sirolimus/pharmacology , Wortmannin
11.
Stem Cells Dev ; 21(5): 814-28, 2012 Mar 20.
Article in English | MEDLINE | ID: mdl-22149974

ABSTRACT

Although mesenchymal stem cells (MSCs) of distinct tissue origin have a large number of similarities and differences, it has not been determined so far whether tissue-resident MSCs are the progenies of one ancestor cell lineage or the results of parallel cell developmental events. Here we compared the expression levels of 177 genes in murine MSCs derived from adult and juvenile bone marrow and adult adipose tissue, as well as juvenile spleen, thymus, and aorta wall by quantitative real-time polymerase chain reaction and the results were partially validated at protein level. All MSC lines uniformly expressed a large set of genes including well-known mesenchymal markers, such as α-smooth muscle actin, collagen type I α-chain, GATA6, Mohawk, and vimentin. In contrast, pluripotency genes and the early mesodermal marker T-gene were not expressed. On the other hand, different MSC lines consistently expressed distinct patterns of Hox genes determining the positional identity of a given cell population. Moreover, MSCs of different origin expressed a few other transcription factors also reflecting their topological identity and so the body segment or organ to which they normally contributed in vivo: (1) thymus-derived cells specifically expressed Tbx5 and Pitx2; (2) spleen-derived MSCs were characterized with Tlx1 and Nkx2.5; (3) Pitx1 designated femoral bone marrow cells and (4) En2 appeared in aorta wall-derived MSCs. Thus, MSCs exhibited topographic identity and memory even after long-term cultivation in vitro. On the basis of these results, we suggest that postnatal MSCs isolated from different anatomical sites descend from precursor cells developing in the postsegmentation mesoderm.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation, Developmental , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mesoderm/cytology , Adipose Tissue/cytology , Adipose Tissue/growth & development , Animals , Aorta/cytology , Aorta/growth & development , Blotting, Western , Bone Marrow Cells/cytology , Cell Lineage/genetics , Cells, Cultured , Cluster Analysis , Flow Cytometry , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mesoderm/growth & development , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Spleen/cytology , Spleen/growth & development , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Thymus Gland/cytology , Thymus Gland/growth & development , Time Factors
12.
Biochem Biophys Res Commun ; 414(3): 474-80, 2011 Oct 28.
Article in English | MEDLINE | ID: mdl-21971558

ABSTRACT

Mesenchymal stem cell like (MSCl) cells were generated from human embryonic stem cells (hESC) through embryoid body formation, and isolated by adherence to plastic surface. MSCl cell lines could be propagated without changes in morphological or functional characteristics for more than 15 passages. These cells, as well as their fluorescent protein expressing stable derivatives, efficiently supported the growth of undifferentiated human embryonic stem cells as feeder cells. The MSCl cells did not express the embryonic (Oct4, Nanog, ABCG2, PODXL, or SSEA4), or hematopoietic (CD34, CD45, CD14, CD133, HLA-DR) stem cell markers, while were positive for the characteristic cell surface markers of MSCs (CD44, CD73, CD90, CD105). MSCl cells could be differentiated toward osteogenic, chondrogenic or adipogenic directions and exhibited significant inhibition of mitogen-activated lymphocyte proliferation, and thus presented immunosuppressive features. We suggest that cultured MSCl cells can properly model human MSCs and be applied as efficient feeders in hESC cultures.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Mesenchymal Stem Cells/cytology , Pluripotent Stem Cells/cytology , Adipogenesis , Biomarkers/metabolism , Biotechnology , Cell Adhesion , Cell Culture Techniques , Cell Line , Chondrogenesis , Humans , Immune Tolerance , Immunosuppression Therapy , Mesenchymal Stem Cells/immunology , Mesenchymal Stem Cells/metabolism , Osteogenesis
13.
BMC Cell Biol ; 10: 93, 2009 Dec 21.
Article in English | MEDLINE | ID: mdl-20025763

ABSTRACT

BACKGROUND: The ABCA1 protein plays a pivotal role in reverse cholesterol transport, by mediating the generation of HDL particles and removing cellular cholesterol. Both the proper expression of ABCA1 in the plasma membrane and the internalization along with apoA-I are required for function. Therefore, we developed a model system to investigate the effect of clinically relevant drugs on the cell surface appearance of ABCA1. RESULTS: By retroviral transduction system, we established stable mammalian cell lines expressing functional and non-functional ABCA1 variants, tagged with an extracellular hemagglutinin epitope. After characterization of the expression, proper localization and function of different ABCA1 variants, we followed quantitatively their cell surface expression by immunofluorescent staining, using flow cytometry. As expected, we found increased cell surface expression of ABCA1 after treatment with a calpain inhibitor, and observed a strong decrease in plasma membrane ABCA1 expression upon treatment with a trans-Golgi transport inhibitor, Brefeldin A. We tested cholesterol level lowering drugs and other potential inhibitors of ABCA1. Here we demonstrate that ezetimibe affects ABCA1 cell surface expression only in the case of a functional ABCA1. CONCLUSIONS: Our model system allows a quantitative detection of cell surface expression of ABCA1, screening of substrates or specific inhibitors, and investigating transport regulation.


Subject(s)
ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Cell Membrane/metabolism , Gene Expression , ATP Binding Cassette Transporter 1 , Animals , Apolipoprotein A-I/genetics , Apolipoprotein A-I/metabolism , Biological Transport , Cell Line , Cell Membrane/genetics , Dogs , Humans , Models, Biological
14.
Stem Cells ; 27(5): 1077-87, 2009 May.
Article in English | MEDLINE | ID: mdl-19415778

ABSTRACT

Human embryonic stem (HuES) cells represent a new potential tool for cell-therapy and gene-therapy applications. However, these approaches require the development of efficient, stable gene delivery, and proper progenitor cell and tissue separation methods. In HuES cell lines, we have generated stable, enhanced green fluorescent protein (EGFP)-expressing clones using a transposon-based (Sleeping Beauty) system. This method yielded high percentage of transgene integration and expression. Similarly to a lentiviral expression system, both the undifferentiated state and the differentiation pattern of the HuES cells were preserved. By using the CAG promoter, in contrast to several other constitutive promoter sequences (such as CMV, elongation factor 1alpha, or phosphoglycerate kinase), an exceptionally high EGFP expression was observed in differentiated cardiomyocytes. This phenomenon was independent of the transgene sequence, methods of gene delivery, copy number, and the integration sites. This "double-feature" promoter behavior, that is providing a selectable marker for transgene expressing undifferentiated stem cells, and also specifically labeling differentiated cardiomyocytes, was assessed by transcriptional profiling. We found a positive correlation between CAG promoter-driven EGFP transcription and expression of cardiomyocyte-specific genes. Our experiments indicate an efficient applicability of transposon-based gene delivery into HuES cells and provide a novel approach to identify differentiated tissues by exploiting a nontypical behavior of a constitutively active promoter, thereby avoiding invasive drug selection methods.


Subject(s)
Cell Differentiation , DNA Transposable Elements/genetics , Embryonic Stem Cells/cytology , Gene Transfer Techniques , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Promoter Regions, Genetic , Animals , Base Sequence , Biomarkers/metabolism , Cell Line , Clone Cells , Computational Biology , Gene Dosage , Gene Expression Profiling , Gene Expression Regulation , Genetic Vectors/genetics , Humans , Mice , Molecular Sequence Data , Mutagenesis, Insertional , Transcription, Genetic , Transgenes
15.
Biochim Biophys Acta ; 1778(12): 2700-9, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18793608

ABSTRACT

Expression of multidrug resistance ABC transporters has been suggested as a functional marker and chemoprotective element in early human progenitor cell types. In this study we examined the expression and function of the key multidrug-ABC transporters, ABCB1, ABCC1 and ABCG2 in two human embryonic stem (HuES) cell lines. We detected a high level ABCG2 expression in the undifferentiated HuES cells, while the expression of this protein significantly decreased during early cell differentiation. ABCG2 in HuES cells provided protection against mitoxantrone toxicity, with a drug-stimulated overexpression of the transporter. No significant expression of ABCB1/ABCC1 was found either in the undifferentiated or partially differentiated HuES cells. Examination of the ABCG2 mRNA in HuES cells indicated the use of selected promoter sites and a truncated 3' untranslated region, suggesting a functionally distinct regulation of this transporter in undifferentiated stem cells. The selective expression of the ABCG2 multidrug transporter indicates that ABCG2 can be applied as a marker for undifferentiated HuES cells. Moreover, protection of embryonic stem cells against xenobiotics and endobiotics may depend on ABCG2 expression and regulation.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Neoplasm Proteins/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , Antineoplastic Agents/metabolism , Biomarkers/analysis , Cell Differentiation , Cells, Cultured , Drug Resistance, Multiple/genetics , Fluorescent Antibody Technique, Direct , Humans , Mitoxantrone/metabolism , Neoplasm Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
16.
Cancer Lett ; 234(1): 62-72, 2006 Mar 08.
Article in English | MEDLINE | ID: mdl-16337740

ABSTRACT

The human multidrug resistance ABC transporters provide a protective function in our body against a large number of toxic compounds. These proteins, residing in the plasma membrane, perform an active, ATP-dependent extrusion of such xenobiotics. However, the same proteins are also used by the tumor cells to fight various anticancer agents. ABCG2 is an important member of the multidrug resistance proteins, an 'ABC half transporter', which functions as a homodimer in the cell membrane. In this review, we provide a basic overview of ABCG2 function in physiology and drug metabolism, but concentrate on the discussion of mutations and polymorphisms discovered in this protein. Interestingly, a single nucleotide mutation, changing amino acid 482 from arginine to threonine or glycine in ABCG2, results in a major increase in the catalytic activity and a wider drug recognition by this protein. Still, this mutation proved to be an in vitro artifact, produced only in heavily drug-selected cell lines. In contrast, at least two, but possibly more polymorphic variants of ABCG2 were found to be present in large human populations with different ethnic background. However, currently available experimental data regarding the cellular expression, localization and function of these ABCG2 variants are strongly contradictory. Since, the proteins produced by these variant alleles may differently modulate cancer treatment, general drug absorption and toxicity, may represent risk factors in fetal toxicity, or alter the differentiation of stem cells, their exact characterization is a major challenge in this field.


Subject(s)
ATP-Binding Cassette Transporters/physiology , Drug Resistance/physiology , Drug-Related Side Effects and Adverse Reactions , Neoplasm Proteins/physiology , Neoplasms/drug therapy , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Biological Transport , Humans , Pharmaceutical Preparations/metabolism
17.
Cancer Res ; 65(5): 1770-7, 2005 Mar 01.
Article in English | MEDLINE | ID: mdl-15753373

ABSTRACT

Iressa (ZD1839, Gefitinib), used in clinics to treat non-small cell lung cancer patients, is a tyrosine kinase receptor inhibitor that leads to specific decoupling of epidermal growth factor receptor (EGFR) signaling. Recent data indicate that Iressa is especially effective in tumors with certain EGFR mutations; however, a subset of these tumors does not respond to Iressa. In addition, certain populations have an elevated risk of side effects during Iressa treatment. The human ABCG2 (BCRP/MXR/ABCP) transporter causes cancer drug resistance by actively extruding a variety of cytotoxic drugs, and it functions physiologically to protect our tissues from xenobiotics. Importantly, ABCG2 modifies absorption, distribution, and toxicity of several pharmacologic agents. Previously, we showed that ABCG2 displays a high-affinity interaction with several tyrosine kinase receptor inhibitors, including Iressa. Here, we show that the expression of ABCG2, but not its nonfunctional mutant, protects the EGFR signaling-dependent A431 tumor cells from death on exposure to Iressa. This protection is reversed by the ABCG2-specific inhibitor, Ko143. These data, reinforced with cell biology and biochemical experiments, strongly suggest that ABCG2 can actively pump Iressa. Therefore, variable expression and polymorphisms of ABCG2 may significantly modify the antitumor effect as well as the absorption and tissue distribution of Iressa.


Subject(s)
ATP-Binding Cassette Transporters/antagonists & inhibitors , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Breast Neoplasms/drug therapy , ErbB Receptors/metabolism , Lung Neoplasms/drug therapy , Neoplasm Proteins/antagonists & inhibitors , Quinazolines/therapeutic use , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Adenosine Triphosphate/metabolism , Breast Neoplasms/metabolism , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , ErbB Receptors/antagonists & inhibitors , Gefitinib , Humans , Lung Neoplasms/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Phosphorylation , Tumor Cells, Cultured
18.
J Biol Chem ; 280(6): 4219-27, 2005 Feb 11.
Article in English | MEDLINE | ID: mdl-15557326

ABSTRACT

The human ABCG2 protein is an important primary active transporter for hydrophobic compounds in several cell types, and its overexpression causes multidrug resistance in tumors. A monoclonal antibody (5D3) recognizes this protein on the cell surface. In ABCG2-expressing cells 5D3 antibody showed a saturable labeling and inhibited ABCG2 transport and ATPase function. However, at low antibody concentrations 5D3 binding to intact cells depended on the actual conformation of the ABCG2 protein. ATP depletion or the addition of the ABCG2 inhibitor Ko143 significantly increased, whereas the vanadate-induced arrest of ABCG2 strongly decreased 5D3 binding. The binding of the 5D3 antibody to a non-functional ABCG2 catalytic center mutant (K86M) in intact cells was not affected by the addition of vanadate but still increased with the addition of Ko143. In isolated membrane fragments the ligand modulation of 5D3 binding to ABCG2 could be analyzed in detail. In this case 5D3 binding was maximum in the presence of ATP, ADP, or Ko143, whereas the non-hydrolysable ATP analog, adenosine 5'-(beta,gamma-imido)triphosphate (AMP-PNP), and nucleotide trapping by vanadate decreased antibody binding. In membranes expressing the ABCG2-K86M mutant, ATP, ADP, and AMP-PNP decreased, whereas Ko143 increased 5D3 binding. Based on these data we suggest that the 5D3 antibody can be used as a sensitive tool to reveal intramolecular changes, reflecting ATP binding, the formation of a catalytic intermediate, or substrate inhibition within the transport cycle of the ABCG2 protein.


Subject(s)
ATP-Binding Cassette Transporters/chemistry , ATP-Binding Cassette Transporters/physiology , Neoplasm Proteins/chemistry , Neoplasm Proteins/physiology , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Adenosine Diphosphate/chemistry , Adenosine Triphosphatases/chemistry , Adenosine Triphosphate/chemistry , Adenylyl Imidodiphosphate/chemistry , Animals , Antibodies, Monoclonal/chemistry , Benzimidazoles/pharmacology , Catalysis , Cell Line , Cell Membrane/metabolism , Cell Separation , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Flow Cytometry , Humans , Hydrolysis , Immunoblotting , Insecta , Ligands , Mitoxantrone/pharmacology , Mutation , Protein Binding , Protein Conformation , Substrate Specificity , Time Factors , Vanadates/chemistry , Vanadates/pharmacology
19.
Mol Pharmacol ; 65(6): 1485-95, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15155841

ABSTRACT

Tyrosine kinase inhibitors (TKIs) are promising new agents for specific inhibition of malignant cell growth and metastasis formation. Because most of the TKIs have to reach an intracellular target, specific membrane transporters may significantly modulate their effectiveness. In addition, the hydrophobic TKIs may interact with so-called multidrug transporters and thus alter the cellular distribution of unrelated pharmacological agents. In the present work, we show that certain TKIs, already in the clinical phase of drug development, directly interact with the ABCG2 multidrug transporter protein with a high affinity. We found that in several in vitro assay systems, STI-571 (Gleevec; imatinib mesylate), ZD1839 (Iressa; gefitinib), and N-[4-[(3-bromophenyl)amino]-6-quinazolinyl]-2-butynamide (EKI-785) interacted with ABCG2 at submicromolar concentrations, whereas other multidrug transporters, human multidrug resistance protein (P-glycoprotein, ABCB1) and human multidrug resistance protein 1 (ABCC1), showed much lower reactivity toward these agents. Low concentrations of the TKIs examined selectively modulated ABCG2-ATPase activity, inhibited ABCG2-dependent active drug extrusion, and significantly affected drug resistance patterns in cells expressing ABCG2. Our results indicate that multidrug resistance protein modulation by TKIs may be an important factor in the clinical treatment of cancer patients. These data also raise the possibility that an extrusion of TKIs by multidrug transporters, e.g., ABCG2, may be involved in tumor cell TKI resistance.


Subject(s)
Enzyme Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Adenosine Triphosphatases/metabolism , Animals , Cell Line , Cell Membrane/metabolism , Drug Resistance, Multiple/physiology , HL-60 Cells , Humans , Insecta/cytology
20.
FEBS Lett ; 567(1): 116-20, 2004 Jun 01.
Article in English | MEDLINE | ID: mdl-15165903

ABSTRACT

The human ABCG2 (ABCP/MXR/BCRP) protein is a recently recognized ABC half-transporter, which forms homodimers in the plasma membrane and actively extrudes a wide variety of chemically unrelated compounds from the cells. This protein protects our cells and tissues against various xenobiotics, with a crucial role in the intestine, liver, placenta, and the blood-brain barrier. Moreover, ABCG2 seems to have a key function in stem cell protection/regulation, and also in hypoxic defense mechanisms. Widely occurring single nucleotide polymorphisms in ABCG2 may affect absorption and distribution, altering the effectiveness and toxicity of drugs in large populations. At the clinics, overexpression of ABCG2 in tumor cells confers cancer multidrug resistance to a variety of newly developed anticancer agents. On the other hand, specific substrate mutants of ABCG2 are advocated for use as selectable markers in stem-cell based gene therapy.


Subject(s)
ATP-Binding Cassette Transporters/physiology , Neoplasm Proteins/physiology , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Biological Transport , Drug Resistance , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Genetic Therapy , Humans , Hypoxia , Models, Biological , Models, Genetic , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Polymorphism, Genetic , Protein Structure, Tertiary , Stem Cells/cytology , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...