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










Publication year range
1.
Neuropsychopharmacology ; 44(2): 344-355, 2019 01.
Article in English | MEDLINE | ID: mdl-30054584

ABSTRACT

Astrocytes are ubiquitous CNS cells that support tissue homeostasis through ion buffering, neurotransmitter recycling, and regulation of CNS vasculature. Yet, despite the essential functional roles they fill, very little is known about the physiology of astrocytes in the ventral midbrain, a region that houses dopamine-releasing neurons and is critical for reward learning and motivated behaviors. Here, using a combination of whole-transcriptome sequencing, histology, slice electrophysiology, and calcium imaging, we performed the first functional and molecular profiling of ventral midbrain astrocytes and observed numerous differences between these cells and their telencephalic counterparts, both in their gene expression profile and in their physiological properties. Ventral midbrain astrocytes have very low membrane resistance and inward-rectifying potassium channel-mediated current, and are extensively coupled to surrounding oligodendrocytes through gap junctions. They exhibit calcium responses to glutamate but are relatively insensitive to norepinephrine. In addition, their calcium activity can be dynamically modulated by dopamine D2 receptor signaling. Taken together, these data indicate that ventral midbrain astrocytes are physiologically distinct from astrocytes in cortex and hippocampus. This work provides new insights into the extent of functional astrocyte heterogeneity within the adult brain and establishes the foundation for examining the impact of regional astrocyte differences on dopamine neuron function and susceptibility to degeneration.


Subject(s)
Astrocytes/physiology , Cerebral Cortex/metabolism , Mesencephalon/metabolism , Receptors, Dopamine D2/metabolism , Animals , Astrocytes/cytology , Astrocytes/drug effects , Calcium/metabolism , Cell Shape/physiology , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Female , Gap Junctions/metabolism , Glutamic Acid/pharmacology , Male , Mesencephalon/cytology , Mesencephalon/drug effects , Mice , Norepinephrine/pharmacology
2.
Mol Cancer Ther ; 5(2): 467-75, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16505122

ABSTRACT

Inhibitors of DNA methyltransferases (DNMT) and histone deacetylases can reactivate epigenetically silenced tumor suppressor genes and thereby decrease tumor cell growth. Little, however, is known on the effects of these compounds in endothelial cell biology and tumor angiogenesis. Here, we show that the DNMT inhibitors 5-aza-2'-deoxycytidine and zebularine markedly decrease vessel formation in different tumor models. We show that DNMT inhibitors are antiproliferative for tumor-conditioned endothelial cells, without affecting endothelial cell apoptosis and migration. Furthermore, these compounds inhibit angiogenesis in vitro and in vivo as shown by inhibition of endothelial cells sprouting in a three-dimensional gel and inhibition of microvessel formation in the chorioallantoic membrane, respectively. 5-Aza-2'-deoxycytidine, as well as the histone deacetylase inhibitor trichostatin A, reactivates the growth-inhibiting genes TSP1, JUNB, and IGFBP3, which are suppressed in tumor-conditioned endothelial cells. Despite enhanced DNMT activity and increased overall genomic methylation levels in tumor-conditioned endothelial cells, silencing of these genes seemed not to be regulated by direct promoter hypermethylation. For IGFBP3, gene expression in endothelial cells correlated with histone H3 acetylation patterns. In conclusion, our data show that DNMT inhibitors have angiostatic activity in addition to their inhibitory effects on tumor cells. This dual action of these compounds makes them promising anticancer therapeutics.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Azacitidine/pharmacology , Cytidine/analogs & derivatives , DNA Modification Methylases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Neovascularization, Pathologic/enzymology , Neovascularization, Physiologic/drug effects , Acetylation , Angiogenesis Inhibitors/therapeutic use , Animals , Azacitidine/therapeutic use , Cell Movement/drug effects , Cytidine/pharmacology , Cytidine/therapeutic use , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Enzyme Inhibitors/therapeutic use , Gene Expression/drug effects , Histones/antagonists & inhibitors , Histones/metabolism , Humans , Hydroxamic Acids/pharmacology , Insulin-Like Growth Factor Binding Protein 3/genetics , Melanoma, Experimental/drug therapy , Melanoma, Experimental/enzymology , Melanoma, Experimental/genetics , Mice , Mice, Mutant Strains , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/genetics , Neovascularization, Physiologic/genetics , Thrombospondin 1/genetics
3.
Cancer Res ; 66(2): 682-92, 2006 Jan 15.
Article in English | MEDLINE | ID: mdl-16423997

ABSTRACT

A major obstacle toward understanding how patterns of abnormal mammalian cytosine DNA methylation are established is the difficulty in quantitating the de novo methylation activities of DNA methyltransferases (DNMT) thought to catalyze these reactions. Here, we describe a novel method, using native human CpG island substrates from genes that frequently become hypermethylated in cancer, which generates robust activity for measuring de novo CpG methylation. We then survey colon cancer cells with genetically engineered deficiencies in different DNMTs and find that the major activity against these substrates in extracts of these cells is DNMT1, with minor contribution from DNMT 3b and none from DNMT3a, the only known bona fide de novo methyltransferases. The activity of DNMT1 against unmethylated CpG rich DNA was further tested by introducing CpG island substrates and DNMT1 into Drosophila melanogaster cells. The exogenous DNMT1 methylates the integrated mammalian CpG islands but not the Drosophila DNA. Additionally, in human cancer cells lacking DNMT1 and DNMT3b and having nearly absent genomic methylation, gene-specific de novo methylation can be initiated by reintroduction of DNMT1. Our studies provide a new assay for de novo activity of DNMTs and data suggesting a potential role for DNMT1 in the initiation of promoter CpG island hypermethylation in human cancer cells.


Subject(s)
Colonic Neoplasms/genetics , CpG Islands , DNA (Cytosine-5-)-Methyltransferases/physiology , DNA Methylation , Animals , Colonic Neoplasms/pathology , DNA (Cytosine-5-)-Methyltransferase 1 , Drosophila/genetics , Genetic Engineering , Humans , Tumor Cells, Cultured
4.
Cancer Res ; 66(2): 729-35, 2006 Jan 15.
Article in English | MEDLINE | ID: mdl-16424002

ABSTRACT

Previous work has shown that DNA hypermethylation of tumor suppressor genes in colorectal cancer cells may be maintained in the absence of the major mammalian methyltransferase, DNA methyltransferase 1 (DNMT1). In an effort to dissect the dependency on DNMT1 to maintain such hypermethylation in different cancer types, we performed a systematic analysis of depletion of DNMT1 in colorectal (SW48), bladder (T24), and breast (T47D) cancer cells by DNMT1-specific small hairpin RNA (shRNA) targeting. We show that although DNMT1-deficient SW48 and T24 cells exhibited no observable growth defects and were able to maintain promoter hypermethylation, DNMT1-deficient T47D breast cells failed to form comparable numbers of colonies when stably selected for the incorporation of the DNMT1-specific shRNA expression vector, suggesting a growth defect with reduced levels of DNMT1. Further treatment of T47D cells with transient transfection of small interfering RNA targeting DNMT1 revealed that severely DNMT1-deficient T47D cells could not fully maintain promoter hypermethylation, and gene silencing was partially reversed at two of the three assayed loci. These observations suggest that human cancer cells may differ in their reliance on DNMT1 for maintaining DNA methylation.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , DNA (Cytosine-5-)-Methyltransferase 1 , Female , Gene Deletion , Humans , Promoter Regions, Genetic , RNA, Small Interfering , Transfection , Tumor Cells, Cultured , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology
5.
Cell Cycle ; 3(8): 1024-6, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15280656

ABSTRACT

The role of DNA methyltransferase 1, DNMT1, in human cancer cells has recently been contested. In this setting, DNMT1's function as the sole maintenance methyltransferase was based on the assumption that its biological activity is identical to the mouse homologue. However, the application of recent technological advances, including gene targeting and siRNA mediated ablation studies, has cast doubt on this presumed role. Here, we attempt to shed light on these new data within the context of previous experiments.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/physiology , Animals , DNA (Cytosine-5-)-Methyltransferase 1 , Humans , Mammals , Neoplasms/enzymology
6.
Nat Genet ; 36(6): 582-4, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15156141

ABSTRACT

The role of the primary mammalian DNA methyltransferase, DNMT1, in maintaining CpG island methylation in human colon cancer cells has recently been questioned. This controversy has arisen from discrepancies between genetic knockout and RNA interference-mediated knockdown studies. Here, we re-examined the RNA interference-based approach and found that hypermethylation of single-copy genes is maintained in cells transiently and stably depleted of DNMT1.


Subject(s)
Colorectal Neoplasms/enzymology , Colorectal Neoplasms/genetics , CpG Islands , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , Base Sequence , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , Gene Silencing , Genes, Tumor Suppressor , HCT116 Cells , Humans , Promoter Regions, Genetic , RNA Interference , RNA, Neoplasm/genetics , RNA, Small Interfering/genetics
7.
Nat Genet ; 36(4): 417-22, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15034581

ABSTRACT

Aberrant WNT pathway signaling is an early progression event in 90% of colorectal cancers. It occurs through mutations mainly of APC and less often of CTNNB1 (encoding beta-catenin) or AXIN2 (encoding axin-2, also known as conductin). These mutations allow ligand-independent WNT signaling that culminates in abnormal accumulation of free beta-catenin in the nucleus. We previously identified frequent promoter hypermethylation and gene silencing of the genes encoding secreted frizzled-related proteins (SFRPs) in colorectal cancer. SFRPs possess a domain similar to one in the WNT-receptor frizzled proteins and can inhibit WNT receptor binding to downregulate pathway signaling during development. Here we show that restoration of SFRP function in colorectal cancer cells attenuates WNT signaling even in the presence of downstream mutations. We also show that the epigenetic loss of SFRP function occurs early in colorectal cancer progression and may thus provide constitutive WNT signaling that is required to complement downstream mutations in the evolution of colorectal cancer.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Gene Silencing , Glycoproteins/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction , Cell Line, Tumor , Colorectal Neoplasms/pathology , Humans , Reverse Transcriptase Polymerase Chain Reaction , Wnt Proteins
8.
Mol Cell Biol ; 23(23): 8429-39, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14612389

ABSTRACT

The GATA family of transcription factors participates in gastrointestinal (GI) development. Increases in GATA-4 and -5 expression occur in differentiation and GATA-6 expression in proliferation in embryonic and adult settings. We now show that in colorectal cancer (CRC) and gastric cancer promoter hypermethylation and transcriptional silencing are frequent for GATA-4 and -5 but are never seen for GATA-6. Potential antitumor target genes upregulated by GATA-4 and -5, the trefoil factors, inhibinalpha, and disabled-2 (Dab2) are also silenced, in GI cancers, with associated methylation of the promoters. Drug or genetically induced demethylation simultaneously leads to expression, in CRC cells, of all of the GATA-4, -5, and downstream genes. Expression of exogenous GATA-5 overrides methylation at the downstream promoters to activate the target genes. Selection for silencing of both upstream transcription factors and their target genes in GI cancers could indicate that epigenetic silencing of the involved genes provides a summated contribution to tumor progression.


Subject(s)
Colorectal Neoplasms/genetics , DNA-Binding Proteins/genetics , Gene Silencing , Stomach Neoplasms/genetics , Transcription Factors/genetics , Cell Line, Tumor , Colorectal Neoplasms/etiology , CpG Islands , DNA Methylation , DNA, Neoplasm/genetics , Epigenesis, Genetic , GATA4 Transcription Factor , GATA5 Transcription Factor , GATA6 Transcription Factor , Humans , Oncogenes , Promoter Regions, Genetic , Stomach Neoplasms/etiology
9.
Proc Natl Acad Sci U S A ; 100(13): 7818-23, 2003 Jun 24.
Article in English | MEDLINE | ID: mdl-12810945

ABSTRACT

Cell-cycle checkpoints controlling the orderly progression through mitosis are frequently disrupted in human cancers. One such checkpoint, entry into metaphase, is regulated by the CHFR gene encoding a protein possessing forkhead-associated and RING finger domains as well as ubiquitin-ligase activity. Although defects in this checkpoint have been described, the molecular basis and prevalence of CHFR inactivation in human tumors are still not fully understood. To address this question, we analyzed the pattern of CHFR expression in a number of human cancer cell lines and primary tumors. We found CpG methylation-dependent silencing of CHFR expression in 45% of cancer cell lines, 40% of primary colorectal cancers, 53% of colorectal adenomas, and 30% of primary head and neck cancers. Expression of CHFR was precisely correlated with both CpG methylation and deacetylation of histones H3 and H4 in the CpG-rich regulatory region. Moreover, CpG methylation and thus silencing of CHFR depended on the activities of two DNA methyltransferases, DNMT1 and DNMT3b, as their genetic inactivation restored CHFR expression. Finally, cells with CHFR methylation had an intrinsically high mitotic index when treated with microtubule inhibitor. This means that cells in which CHFR was epigenetically inactivated constitute loss-of-function alleles for mitotic checkpoint control. Taken together, these findings shed light on a pathway by which mitotic checkpoint is bypassed in cancer cells and suggest that inactivation of checkpoint genes is much more widespread than previously suspected.


Subject(s)
Cell Cycle Proteins/genetics , Gene Expression Regulation, Neoplastic , Models, Genetic , Neoplasm Proteins , Neoplasms/genetics , Chromatin/metabolism , Colony Count, Microbial , CpG Islands , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methylation , Histone Deacetylases/metabolism , Histones/metabolism , Humans , Mitosis , Poly-ADP-Ribose Binding Proteins , Precipitin Tests , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Ubiquitin-Protein Ligases , DNA Methyltransferase 3B
10.
Nature ; 416(6880): 552-6, 2002 Apr 04.
Article in English | MEDLINE | ID: mdl-11932749

ABSTRACT

Inactivation of tumour suppressor genes is central to the development of all common forms of human cancer. This inactivation often results from epigenetic silencing associated with hypermethylation rather than intragenic mutations. In human cells, the mechanisms underlying locus-specific or global methylation patterns remain unclear. The prototypic DNA methyltransferase, Dnmt1, accounts for most methylation in mouse cells, but human cancer cells lacking DNMT1 retain significant genomic methylation and associated gene silencing. We disrupted the human DNMT3b gene in a colorectal cancer cell line. This deletion reduced global DNA methylation by less than 3%. Surprisingly, however, genetic disruption of both DNMT1 and DNMT3b nearly eliminated methyltransferase activity, and reduced genomic DNA methylation by greater than 95%. These marked changes resulted in demethylation of repeated sequences, loss of insulin-like growth factor II (IGF2) imprinting, abrogation of silencing of the tumour suppressor gene p16INK4a, and growth suppression. Here we demonstrate that two enzymes cooperatively maintain DNA methylation and gene silencing in human cancer cells, and provide compelling evidence that such methylation is essential for optimal neoplastic proliferation.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/physiology , Gene Expression Regulation, Neoplastic , Gene Silencing , Genes, Tumor Suppressor , Neoplasms/genetics , Amino Acid Sequence , Cyclin-Dependent Kinase Inhibitor p16/genetics , DNA (Cytosine-5-)-Methyltransferase 1 , DNA Methylation , DNA, Neoplasm/metabolism , Gene Targeting , Humans , Insulin-Like Growth Factor II/genetics , Molecular Sequence Data , Neoplasms/enzymology , Promoter Regions, Genetic , Tissue Inhibitor of Metalloproteinase-3/genetics , Tumor Cells, Cultured , DNA Methyltransferase 3B
11.
Genome Res ; 12(1): 153-7, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11779840

ABSTRACT

We have developed a novel quantitative method for rapidly assessing the CpG methylation density of a DNA region in mammalian cells. After bisulfite modification of genomic DNA, the region of interest is PCR amplified with primers containing two dam sites (GATC). The purified PCR products are then incubated with 14C-labeled S-adenosyl-L-methionine (SAM) and dam methyltransferase as an internal control to standardize DNA quantity. This is followed by an incubation with 3H-labeled SAM and SssI methyltransferase for methylation quantification. By use of standard mixtures of cell line DNA with a defined methylation status in every assay, the ratio (3H/14C signal) of each sample can be converted into percentage values to assess the methylation density of the amplified sequence. This methylation-sensitive technique, termed ERMA (Enzymatic Regional Methylation Assay) provides several advantages over existing methods used for methylation analysis as it determines an exact measurement of the methylation density of the region studied. We demonstrate a use of this technique in determining the methylation density of the promoter region of the tumor suppressor gene p15INK4B and changes that occur after treatment with demethylating agents.


Subject(s)
CpG Islands , DNA Methylation , DNA, Neoplasm/chemistry , Tumor Suppressor Proteins , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , CpG Islands/genetics , Cyclin-Dependent Kinase Inhibitor p15 , Cyclin-Dependent Kinase Inhibitor p16/chemistry , Cyclin-Dependent Kinase Inhibitor p16/genetics , DNA, Neoplasm/genetics , DNA-Cytosine Methylases/metabolism , Genes, Tumor Suppressor , HL-60 Cells/chemistry , HL-60 Cells/metabolism , Humans , Promoter Regions, Genetic/genetics , S-Adenosylmethionine/metabolism , Site-Specific DNA-Methyltransferase (Adenine-Specific)/metabolism , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...