Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Neoplasma ; 71(2): 99-116, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38766858

ABSTRACT

Cancer is one of the leading causes of death worldwide. We still do not understand all the details of carcinogenesis, and effective treatment is lacking for many oncological diseases. Animal models provide an irreplaceable tool to observe the growth and spreading of neoplastic cells in an environment of living organisms, to test the efficacy of cancer treatment, side effects, and toxicity, and to study the tumor microenvironment. Mice are the most often used model organisms because of their easy handling, short reproductive period, multiple strains, and complete DNA sequencing. An ideal model should accurately recapitulate each step of tumor development. Recent techniques have established models that enable the study of different aspects of cancer, but choosing a particular model depends on the application of output data. This article aimed to review induced, transplantable, and engineered mice and highlight their significance for recent and future cancer research.


Subject(s)
Disease Models, Animal , Neoplasms , Animals , Mice , Neoplasms/pathology , Neoplasms/therapy , Humans , Tumor Microenvironment
2.
Front Pharmacol ; 13: 991751, 2022.
Article in English | MEDLINE | ID: mdl-36278182

ABSTRACT

Decitabine (DAC), a DNA methyltransferase (DNMT) inhibitor, is tested in combination with conventional anticancer drugs as a treatment option for various solid tumors. Although epigenome modulation provides a promising avenue in treating resistant cancer types, more studies are required to evaluate its safety and ability to normalize the aberrant transcriptional profiles. As deoxycytidine kinase (DCK)-mediated phosphorylation is a rate-limiting step in DAC metabolic activation, we hypothesized that its intracellular overexpression could potentiate DAC's effect on cell methylome and thus increase its therapeutic efficacy. Therefore, two breast cancer cell lines, JIMT-1 and T-47D, differing in their molecular characteristics, were transfected with a DCK expression vector and exposed to low-dose DAC (approximately IC20). Although transfection resulted in a significant DCK expression increase, further enhanced by DAC exposure, no transfection-induced changes were found at the global DNA methylation level or in cell viability. In parallel, an integrative approach was applied to decipher DAC-induced, methylation-mediated, transcriptomic reprogramming. Besides large-scale hypomethylation, accompanied by up-regulation of gene expression across the entire genome, DAC also induced hypermethylation and down-regulation of numerous genes in both cell lines. Interestingly, TET1 and TET2 expression halved in JIMT-1 cells after DAC exposure, while DNMTs' changes were not significant. The protein digestion and absorption pathway, containing numerous collagen and solute carrier genes, ranking second among membrane transport proteins, was the top enriched pathway in both cell lines when hypomethylated and up-regulated genes were considered. Moreover, the calcium signaling pathway, playing a significant role in drug resistance, was among the top enriched in JIMT-1 cells. Although low-dose DAC demonstrated its ability to normalize the expression of tumor suppressors, several oncogenes were also up-regulated, a finding, that supports previously raised concerns regarding its broad reprogramming potential. Importantly, our research provides evidence about the involvement of active demethylation in DAC-mediated transcriptional reprogramming.

3.
BMC Cancer ; 21(1): 507, 2021 May 06.
Article in English | MEDLINE | ID: mdl-33957885

ABSTRACT

BACKGROUND: Tumor necrosis factor alpha (TNFα) is a pleiotropic cytokine with both anti-tumorigenic and pro-tumorigenic activity, affecting tumor cell biology, the balance between cell survival and death. The final effect of TNFα is dependent on the type of malignant cells, with the potential to arrest cancer progression. METHODS: In order to explain the diverse cellular response to TNFα, we engineered melanoma and colorectal carcinoma cell lines stably overexpressing this cytokine. RESULTS: Under the TNFα overexpression, significant upregulation of two genes was observed: proinflammatory cytokine IL6 gene in melanoma cells A375 and gene for pro-apoptotic ligand TRAIL in colorectal carcinoma cells HT29, both mediated by TNFα/TNFR1 signaling. Malignant melanoma line A375 displayed also increased autophagy on day 3, followed by premature senescence on day 6. Both processes seem to be interconnected, following earlier apoptosis induction and deregulation of mitochondrial functions. We documented altered mitochondrial status, lowered ATP production, lowered mitochondrial mass, and changes in mitochondrial morphology (shortened and condensed mitochondria) both in melanoma and colorectal carcinoma cells. Overexpression of TNFα was not linked with significant affection of the subpopulation of cancer stem-like cells in vitro. However, we could demonstrate a decrease in aldehyde dehydrogenase (ALDH) activity up to 50%, which is associated with to the stemness phenotype. CONCLUSIONS: Our in vitro study of direct TNFα influence demonstrates two distinct outcomes in tumor cells of different origin, in non-epithelial malignant melanoma cells of neural crest origin, and in colorectal carcinoma cells derived from the epithelium.


Subject(s)
Autophagy/physiology , Melanoma/pathology , Mitochondria/physiology , Tumor Necrosis Factor-alpha/physiology , Aldehyde Dehydrogenase/metabolism , Cell Line, Tumor , Cellular Senescence , Colorectal Neoplasms/pathology , Humans , Interleukin-6/genetics , Receptors, Tumor Necrosis Factor, Type I/physiology , Receptors, Tumor Necrosis Factor, Type II/physiology , TNF-Related Apoptosis-Inducing Ligand/genetics
4.
J Cell Mol Med ; 23(5): 3641-3655, 2019 05.
Article in English | MEDLINE | ID: mdl-30916466

ABSTRACT

We report results showing that the silencing of carbonic anhydrase I (siCA1) in prostatic (PC3) tumour cells has a significant impact on exosome formation. An increased diameter, concentration and diversity of the produced exosomes were noticed as a consequence of this knock-down. The protein composition of the exosomes' cargo was also altered. Liquid chromatography and mass spectrometry analyses identified 42 proteins significantly altered in PC3 siCA1 exosomes compared with controls. The affected proteins are mainly involved in metabolic processes, biogenesis, cell component organization and defense/immunity. Interestingly, almost all of them have been described as 'enhancers' of tumour development through the promotion of cell proliferation, migration and invasion. Thus, our results indicate that the reduced expression of the CA1 protein enhances the malignant potential of PC3 cells.


Subject(s)
Carbonic Anhydrase I/genetics , Exosomes/genetics , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Prostatic Neoplasms/genetics , RNA Interference , Carbonic Anhydrase I/metabolism , Cell Movement/genetics , Cell Proliferation/genetics , Energy Metabolism/genetics , Exosomes/metabolism , Humans , Male , PC-3 Cells , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology
5.
Cancer Gene Ther ; 25(11-12): 285-299, 2018 12.
Article in English | MEDLINE | ID: mdl-29925898

ABSTRACT

Gene-directed enzyme/prodrug therapy (GDEPT) mediated by mesenchymal stromal cells (MSC) was already approved for clinical study on a progressive disease refractory to standard therapy. In this work, we examined the effect of several GDEPT approaches on chemoresistant cells. First, we derived 5-fluorouracil (5-FU)-resistant variant of human colorectal adenocarcinoma cells HT-29 designated HT-29/EGFP/FUR. Our data show that the upregulation of thymidylate synthase (TS) and downregulation of thymidine phosphorylase (TP), orotate phosphoribosyl transferase (OPRT) and dihydropyrimidine dehydrogenase (DPD) contributed to the 5-FU resistance in cancer cells. Next, we combined the MSC expressing either yeast cytosine deaminase (CD-MSC) or fusion yeast CD::uracil phosphoribosyl transferase (CD::UPRT-MSC) and prodrug 5-fluorocytosine (5-FC) in a cell-mediated GDEPT approach. Bystander cytotoxic effect in the direct co-cultures of the tumor and therapeutic cells mixed in a 5:1 ratio resulted in 55% and 70% inhibition of proliferation, respectively. However, the acquired chemoresistance to 5-FU can be overcome by introducing the prodrug-converting transgene into the tumor cells. When the transgene CD::UPRT was expressed in the chemoresistant cells (CD::UPRT-FUR), substantial suicide effect and a 90% decrease in viability was observed using non-toxic concentration of 62.5 µg/ml 5-FC. In summary, we demonstrate here that the transgene introduction circumvented 5-FU resistance in the tumor cells.


Subject(s)
Fluorouracil/therapeutic use , Genetic Therapy/methods , Prodrugs/therapeutic use , Animals , Cell Line, Tumor , Fluorouracil/pharmacology , Humans , Mice , Mice, SCID , Prodrugs/pharmacology
6.
J Cell Mol Med ; 22(1): 695-699, 2018 01.
Article in English | MEDLINE | ID: mdl-28782909

ABSTRACT

We report the silencing of CA1 mRNA in PC3 and MDA cells. The levels of mRNA coding CA1 protein in the knock-down mRNA (CA1 siRNA) cells have been measured by RT-PCR and were approximately 5% (PC3) and 20% (MDA-MB-231), respectively, of the level of control (Mock siRNA) used during silencing. In PC3 and MDA-MB-231 cells, the mRNAs for COL1A1 and COL4A4 were up-regulated. The mRNAs for CTHRC1, LAMC2, and WNT7B were not changed when compared to the control. The morphology of the cells during the treatments remained the same. On the Western blots, the lysate from the silenced cells showed lower levels of CA I as well.


Subject(s)
Carbonic Anhydrases/genetics , Extracellular Matrix Proteins/genetics , Gene Expression Regulation, Neoplastic , Gene Silencing , Neoplasms/enzymology , Neoplasms/genetics , Carbonic Anhydrases/metabolism , Cell Line, Tumor , Extracellular Matrix Proteins/metabolism , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism
7.
J Cell Mol Med ; 21(3): 543-551, 2017 03.
Article in English | MEDLINE | ID: mdl-27704726

ABSTRACT

Spontaneous tumour regression after high-dose therapy and autologous stem cell transplantation is associated with the aplastic anaemia-like syndrome and the presence of polyclonal autoantibodies against carbonic anhydrase I (CA I). When tumour cells were grown in vitro in the presence of patients' sera positive for anti-CA I autoantibodies, their morphological pattern was altered. These changes were accompanied by modifications in the gene expression profile. We observed downregulation of genes of the basal lamina assembly (collagen type IV alpha 4, the laminin subunit gamma 2), the extracellular matrix (collagen type I alpha 1), the cytoskeleton (keratin 14 type I), the collagen triple helix repeat containing 1 and the proto-oncogene WNT7B. On the other hand, the expression of the CA 1 gene was increased in the tumour cells. It was also noticed that the presence of anti-CA I autoantibodies did not impair tumour cell proliferation and cell viability in vitro. These findings were observed only in the presence of patients' sera positive for anti-CA I autoantibodies.


Subject(s)
Autoantibodies/blood , Autoantibodies/immunology , Carbonic Anhydrase I/immunology , Extracellular Matrix Proteins/metabolism , Extracellular Matrix/metabolism , Neoplasms/blood , Neoplasms/immunology , Adult , Aged , Antibody Specificity/immunology , Basement Membrane/immunology , Basement Membrane/metabolism , Carbonic Anhydrase I/metabolism , Cell Proliferation/physiology , Cell Survival/immunology , Collagen/immunology , Collagen/metabolism , Collagen Type I, alpha 1 Chain , Cytoskeleton/immunology , Cytoskeleton/metabolism , Down-Regulation/immunology , Extracellular Matrix/immunology , Extracellular Matrix Proteins/immunology , Female , Gene Expression/genetics , Gene Expression/immunology , Humans , Male , Middle Aged , Neoplasms/metabolism , Proto-Oncogene Mas
9.
Cell Commun Signal ; 14: 4, 2016 Jan 12.
Article in English | MEDLINE | ID: mdl-26759169

ABSTRACT

BACKGROUND: Cells of the tumor microenvironment are recognized as important determinants of the tumor biology. The adjacent non-malignant cells can regulate drug responses of the cancer cells by secreted paracrine factors and direct interactions with tumor cells. RESULTS: Human mesenchymal stromal cells (MSC) actively contribute to tumor microenvironment. Here we focused on their response to chemotherapy as during the treatment these cells become affected. We have shown that the secretory phenotype and behavior of mesenchymal stromal cells influenced by cisplatin differs from the naïve MSC. MSC were more resistant to the concentrations of cisplatin, which was cytotoxic for tumor cells. They did not undergo apoptosis, but a part of MSC population underwent senescence. However, MSC pretreatment with cisplatin led to changes in phosphorylation profiles of many kinases and also increased secretion of IL-6 and IL-8 cytokines. These changes in cytokine and phosphorylation profile of MSC led to increased chemoresistance and stemness of breast cancer cells. CONCLUSION: Taken together here we suggest that the exposure of the chemoresistant cells in the tumor microenvironment leads to substantial alterations and might lead to promotion of acquired microenvironment-mediated chemoresistance and stemness.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Cisplatin/pharmacology , Mesenchymal Stem Cells/drug effects , Tumor Microenvironment/drug effects , Apoptosis/drug effects , Breast/drug effects , Breast/immunology , Breast Neoplasms/immunology , Cell Line, Tumor , Cellular Senescence/drug effects , Drug Resistance, Neoplasm , Female , Humans , Interleukin-6/immunology , Interleukin-8/immunology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/immunology
10.
J Exp Clin Cancer Res ; 34: 33, 2015 Apr 09.
Article in English | MEDLINE | ID: mdl-25884597

ABSTRACT

BACKGROUND: Metastatic spread of tumor cells remains a serious problem in cancer treatment. Gene-directed enzyme/prodrug therapy mediated by tumor-homing genetically engineered mesenchymal stromal cells (MSC) represents a promising therapeutic modality for elimination of disseminated cells. Efficacy of gene-directed enzyme/prodrug therapy can be improved by combination of individual systems. We aimed to define the combination effect of two systems of gene therapy mediated by MSC, and evaluate the ability of systemically administered genetically engineered mesenchymal stromal cells to inhibit the growth of experimental metastases derived from human breast adenocarcinoma cells MDA-MB-231/EGFP. METHODS: Human adipose tissue-derived mesenchymal stromal cells (AT-MSC) were retrovirally transduced with fusion yeast cytosine deaminase::uracil phosphoribosyltransferase (CD::UPRT) or with Herpes simplex virus thymidine kinase (HSVtk). Engineered MSC were cocultured with tumor cells in the presence of prodrugs 5-fluorocytosin (5-FC) and ganciclovir (GCV). Combination effect of these enzyme/prodrug approaches was calculated. SCID/bg mice bearing experimental lung metastases were treated with CD::UPRT-MSC, HSVtk-MSC or both in combination in the presence of respective prodrug(s). Treatment efficiency was evaluated by EGFP-positive cell detection by flow cytometry combined with real-time PCR quantification of human cells in mouse organs. Results were confirmed by histological and immunohistochemical examination. RESULTS: We demonstrated various extent of synergy depending on tested cell line and experimental setup. The strongest synergism was observed on breast cancer-derived cell line MDA-MB-231/EGFP. Systemic administration of CD::UPRT-MSC and HSVtk-MSC in combination with 5-FC and GCV inhibited growth of MDA-MB-231 induced lung metastases. CONCLUSIONS: Combined gene-directed enzyme/prodrug therapy mediated by MSC exerted synergic cytotoxic effect and resulted in high therapeutic efficacy in vivo.


Subject(s)
Ganciclovir/pharmacology , Genetic Therapy , Lung Neoplasms/pathology , Mesenchymal Stem Cells/metabolism , Thymidine Kinase/genetics , Transduction, Genetic , Animals , Cell Line, Tumor , Disease Models, Animal , Humans , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Mesenchymal Stem Cell Transplantation , Mice , Moloney murine leukemia virus/genetics , Xenograft Model Antitumor Assays
11.
J Gene Med ; 17(1-2): 54-67, 2015.
Article in English | MEDLINE | ID: mdl-25677845

ABSTRACT

BACKGROUND: Mesenchymal stromal cells (MSC) are a promising tool for targeted cancer therapy due to their tumour-homing ability. Intrinsic resistance enables the MSC to longer tolerate therapeutic factors, such as prodrug converting enzymes, cytokines and pro-apoptotic proteins. Tumour necrosis factor alpha (TNFα) is known to be cytotoxic to a variety of cancer cells and exert a tumour-destructive capacity. METHODS: MSC were retrovirally transduced to stable express an exogenous gene encoding the desired therapeutic agent hTNFα. The effect of a TNFα-producing adipose tissue-derived MSC (AT-MSC/hTNFα) was tested on the tumour cell lines of different origins: melanoma (A375), breast carcinoma (SKBR3, MDA-MB-231), colon carcinoma (HT29), ovarian carcinoma (SKOV3) and glioblastoma (U87-MG) cells. The tumour suppressing effect of AT-MSC/hTNFα on A375 melanoma xenografts was monitored in an immunodeficient mouse model in vivo. RESULTS: Engineered AT-MSC are able to constitutively secrete human TNFα protein, induce apoptosis of tumour cell lines via caspase 3/7 activation and inhibit the tumour cell proliferation in vitro. Melanoma A375 and breast carcinoma SKBR3 cells were the most sensitive, and their proliferation in vitro was reduced by conditioned media produced by AT-MSC/hTNFα to 60% and 40%, respectively. The previously reported tumour supportive effect of AT-MSC on subcutaneous A375 melanoma xenograft growth was neutralised and suppressed by engineered AT-MSC stably producing hTNFα. When AT-MSC/hTNFα were coinjected with A375 melanoma cells, the tumour mass inhibition was up to 97.5%. CONCLUSIONS: The results of the present study demonstrate that tumour cells respond to hTNFα-based treatment mediated by genetically engineered AT-MSC/hTNFα both in vitro and in vivo.


Subject(s)
Genetic Engineering , Melanoma/genetics , Melanoma/pathology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Tumor Necrosis Factor-alpha/genetics , Animals , Cell Line , Cell Line, Tumor , Disease Models, Animal , Gene Expression , Genetic Therapy , Genetic Vectors/genetics , Humans , Melanoma/metabolism , Melanoma/therapy , Mice , Retroviridae/genetics , Transduction, Genetic , Tumor Burden/genetics , Tumor Necrosis Factor-alpha/biosynthesis , Xenograft Model Antitumor Assays
12.
Stem Cell Res ; 8(2): 247-58, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22265744

ABSTRACT

Mesenchymal stromal cells (MSCs) are considered to be suitable vehicles for cellular therapy in various conditions. The expression of reporter and/or effector protein(s) enabled both the identification of MSCs within the organism and the exploitation in targeted tumor therapies. The aim of this study was to evaluate cellular changes induced by retrovirus-mediated transgene expression in MSCs in vitro. Human Adipose Tissue-derived MSCs (AT-MSCs) were transduced to express (i) the enhanced green fluorescent protein (EGFP) reporter transgene, (ii) the fusion yeast cytosine deaminase::uracil phosphoribosyltransferase (CDy::UPRT) enzyme along with the expression of dominant positive selection gene NeoR or (iii) the selection marker NeoR alone (MOCK). CDy::UPRT expression resulted in increased proliferation of CDy::UPRT-MSCs versus naïve AT-MSCs, MOCK-MSCs or EGFP-MSCs. Furthermore, CDy::UPRT-MSCs were significantly more sensitive to 5-fluorouracil (5FU), cisplatin, cyclophosphamide and cytosine arabinoside as determined by increased Caspase 3/7 activation and/or decreased relative proliferation. CDy::UPRT-MSCs in direct cocultures with breast cancer cells MDA-MB-231 increased tumor cell killing induced by low concentrations of 5FU. Our data demonstrated the changes in proliferation and chemoresistance in engineered MSCs expressing transgene with enzymatic function and suggested the possibilities for further augmentation of targeted MSC-mediated antitumor therapy.


Subject(s)
Cytosine Deaminase/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Adipose Tissue/cytology , Apoptosis/drug effects , Azacitidine/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cytarabine/pharmacology , Drug Screening Assays, Antitumor , Female , Fluorouracil/pharmacology , Genetic Engineering , Green Fluorescent Proteins/metabolism , Humans , Mesenchymal Stem Cells/drug effects , Retroviridae/drug effects , Retroviridae/genetics , Saccharomyces cerevisiae/drug effects , Transduction, Genetic , Transgenes/genetics
13.
J Gene Med ; 10(10): 1071-82, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18671316

ABSTRACT

BACKGROUND: Previously, we validated capability of human adipose tissue-derived mesenchymal stem cells (AT-MSC) to serve as cellular vehicles for gene-directed enzyme prodrug molecular chemotherapy. Yeast fusion cytosine deaminase : uracil phosphoribosyltransferase expressing AT-MSC (CD y-AT-MSC) combined with systemic 5-fluorocytosine (5FC) significantly inhibited growth of human colon cancer xenografts. We aimed to determine the cytotoxic efficiency to other tumour cells both in vitro and in vivo. METHODS: CD y-AT-MSC/5FC-mediated proliferation inhibition against a panel of human tumour cells lines was evaluated in direct and indirect cocultures in vitro. Antitumour effect was tested on immunodeficient mouse model in vivo. RESULTS: Although culture expansion of CD y-AT-MSC sensitized these cells to 5FC mediated suicide effect, expanded CD y-AT-MSC/5FC still exhibited strong bystander cytotoxic effect towards human melanoma, glioblastoma, colon, breast and bladder carcinoma in vitro. Most efficient inhibition (91%) was observed in melanoma A375 cell line when directly cocultured with 2% of therapeutic cells CD y-AT-MSC/5FC. The therapeutic paradigm of the CD y -AT-MSC/5FC system was further evaluated on melanoma A375 xenografts on nude mice in vivo. Complete regression in 89% of tumours was achieved when 20% CD y-AT-MSC/5FC were co-injected along with tumour cells. More importantly, systemic CD y-AT-MSC administration resulted in therapeutic cell homing into subcutaneous melanoma and mediated tumour growth inhibition. CONCLUSIONS: CD y-AT-MSC capability of targeting subcutaneous melanoma offers a possibility to selectively produce cytotoxic agent in situ. Our data further demonstrate beneficial biological properties of AT-MSC as a cellular vehicle for enzyme/prodrug therapy approach to molecular chemotherapy.


Subject(s)
Cytosine Deaminase/genetics , Melanoma, Experimental/drug therapy , Mesenchymal Stem Cells/enzymology , Adipose Tissue/metabolism , Adult , Animals , Apoptosis , Cancer Vaccines/genetics , Cell Line, Tumor , Cytosine Deaminase/metabolism , Flucytosine/metabolism , Flucytosine/pharmacology , Genetic Therapy , Genetic Vectors/administration & dosage , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Nude , Neoplasm Transplantation , Pentosyltransferases/genetics , Pentosyltransferases/metabolism , Prodrugs/pharmacology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transduction, Genetic
14.
Cancer Res ; 67(13): 6304-13, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17616689

ABSTRACT

Human adipose tissue-derived mesenchymal stem cells (AT-MSC) are considered to be a promising source of autologous stem cells in personalized cell-based therapies. Tumor tracking properties of MSC provide an attractive opportunity for targeted transgene delivery into the sites of tumor formation. In the present study, we addressed whether the suicide gene introduction into human AT-MSC could produce a tumor-specific prodrug converting cellular vehicle for targeted chemotherapy. We prepared yeast fusion cytosine deaminase::uracil phosphoribosyltransferase gene-expressing cells [cytosine deaminase (CD)-expressing AT-MSC (CD-AT-MSC)] by retrovirus transduction. We explored their therapeutic potential on a model of human colon cancer in the presence of prodrug 5-fluorocytosine (5-FC). Gene manipulation of human AT-MSC did not sensitize CD-AT-MSC to 5-FC, thus overcoming the inherent disadvantage of suicide effect on cellular vehicle. CD-AT-MSC in combination with 5-FC augmented the bystander effect and selective cytotoxicity on target tumor cells HT-29 in direct coculture in vitro. We confirmed directed migration ability of AT-MSC and CD-AT-MSC toward tumor cells HT-29 in vitro. Moreover, we achieved significant inhibition of s.c. tumor xenograft growth by s.c. or i.v. administered CD-AT-MSC in immunocompromised mice treated with 5-FC. We confirmed the ability of CD-AT-MSC to deliver the CD transgene to the site of tumor formation and mediate strong antitumor effect in vivo. Taken together, these data characterize MSC derived from adipose tissue as suitable delivery vehicles for prodrug converting gene and show their utility for a personalized cell-based targeted cancer gene therapy.


Subject(s)
Adipose Tissue/metabolism , Antineoplastic Agents/pharmacology , Flucytosine/pharmacology , Gene Expression Regulation , Genetic Therapy/methods , Mesenchymal Stem Cells/cytology , Neoplasms/drug therapy , Prodrugs/pharmacology , Animals , Cell Line, Tumor , Cytosine Deaminase/genetics , Cytosine Deaminase/metabolism , Female , Humans , Mice , Mice, Inbred BALB C , Neoplasm Transplantation
15.
FEMS Yeast Res ; 5(1): 19-27, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15381119

ABSTRACT

The phosphatidylglycerolphosphate synthase (CDP-diacylglycerol:sn-glycerol-3-phosphate 3-phosphatidyltransferase, EC 2.7.8.5) is an essential enzyme in biosynthesis of cardiolipin. In this work we report the isolation, heterological cloning, molecular characterization and physical mapping of the Saccharomyces cerevisiae PEL1/PGS1 homologue from Kluyveromyces lactis. The pel1 mutant strain of S. cerevisiae was used to isolate this homologue by screening a K. lactis genomic library. The novel cloned gene was named KlPGS1. Its coding region was found to consist of 1623 bp. The corresponding protein exhibits 55% amino acid identity to its S. cerevisiae counterpart. The presence of the mitochondrial presequence indicates its mitochondrial localization. Sporulation and ascus dissection of diploids heterozygous for single-copy disruption of KlPGS1 revealed that the KlPGS1 gene, is essential in K. lactis. Using a DIG-dUTP-labeled DNA probe-originated from the KlPGS1 gene and Southern hybridization of contour-clamped homogeneous electric field (CHEF)-separated K. lactis chromosomal DNA, the KlPGS1 gene was assigned to chromosome I. The nucleotide sequence data reported in this paper were submitted to GenBank and assigned the Accession No. AY176328.


Subject(s)
CDPdiacylglycerol-Serine O-Phosphatidyltransferase/genetics , Chromosomes, Fungal/genetics , Genes, Fungal/genetics , Kluyveromyces/enzymology , Kluyveromyces/genetics , Amino Acid Sequence , Base Sequence , Blotting, Southern , Chromosome Mapping , Cloning, Molecular , DNA, Fungal/chemistry , DNA, Fungal/genetics , Electrophoresis, Gel, Pulsed-Field , Molecular Sequence Data , Mutagenesis, Insertional , Polymerase Chain Reaction , Sequence Alignment
SELECTION OF CITATIONS
SEARCH DETAIL
...