Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Signal ; 8(398): ra102, 2015 Oct 13.
Article in English | MEDLINE | ID: mdl-26462735

ABSTRACT

The activation of T cells is the fundamental on switch for the adaptive immune system. Ca(2+) signaling is essential for T cell activation and starts as initial, short-lived, localized Ca(2+) signals. The second messenger nicotinic acid adenine dinucleotide phosphate (NAADP) forms rapidly upon T cell activation and stimulates early Ca(2+) signaling. We developed a high-resolution imaging technique using multiple fluorescent Ca(2+) indicator dyes to characterize these early signaling events and investigate the channels involved in NAADP-dependent Ca(2+) signals. In the first seconds of activation of either primary murine T cells or human Jurkat cells with beads coated with an antibody against CD3, we detected Ca(2+) signals with diameters close to the limit of detection and that were close to the activation site at the plasma membrane. In Jurkat cells in which the ryanodine receptor (RyR) was knocked down or in primary T cells from RyR1(-/-) mice, either these early Ca(2+) signals were not detected or the number of signals was markedly reduced. Local Ca(2+) signals observed within 20 ms upon microinjection of Jurkat cells with NAADP were also sensitive to RyR knockdown. In contrast, TRPM2 (transient receptor potential channel, subtype melastatin 2), a potential NAADP target channel, was not required for the formation of initial Ca(2+) signals in primary T cells. Thus, through our high-resolution imaging method, we characterized early Ca(2+) release events in T cells and obtained evidence for the involvement of RyR and NAADP in such signals.


Subject(s)
Calcium Signaling , Calcium/metabolism , NADP/analogs & derivatives , Ryanodine Receptor Calcium Release Channel/metabolism , T-Lymphocytes/metabolism , Aniline Compounds/chemistry , Animals , Benzofurans/chemistry , Cells, Cultured , Fluorometry/methods , Humans , Imidazoles/chemistry , Jurkat Cells , Lymphocyte Activation/drug effects , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Muromonab-CD3/pharmacology , NADP/metabolism , Ryanodine Receptor Calcium Release Channel/genetics , T-Lymphocytes/drug effects , TRPM Cation Channels/metabolism , Time Factors , Xanthenes/chemistry
2.
Dis Colon Rectum ; 50(10): 1618-26; discussion 1626-7, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17762966

ABSTRACT

PURPOSE: To investigate a potential mode of noninvasive screening for colorectal cancer, we evaluated the hypermethylation of the secreted frizzled-related protein-1 gene promoter in human stool DNA. METHODS: In stool samples from 36 patients with colorectal neoplasia (7 adenoma, 29 colorectal cancer) and 17 healthy control subjects, isolated DNA was treated with sodium bisulfite and analyzed by methylation-specific polymerase chain reaction with primers specific for methylated or unmethylated promoter sequences of the secreted frizzled-related protein-1 gene. RESULTS: Hypermethylation of the secreted frizzled-related protein-1 promoter was present in the stool DNA of patients with adenoma and colorectal cancer. A sensitivity of 89 percent and specificity of 86 percent were achieved in the detection of colorectal neoplasia. The difference in hypermethylation status of the secreted frizzled-related protein-1 promoter between the patients with colorectal neoplasia and the control group was statistically highly significant (P < 0.001). Adenoma and early tumor Stage I (International Union Against Cancer) displayed both unmethylated and methylated secreted frizzled-related protein-1 promoter sequences, whereas advanced tumor stages showed only methylated secreted frizzled-related protein-1 (P = 0.05). CONCLUSIONS: The results indicate that this DNA stool test of hypermethylation of the secreted frizzled-related protein-1 promoter is a sensitive and specific method. It has the potential of a clinically useful test for the early detection of colorectal cancer.


Subject(s)
Adenoma/metabolism , Carcinoma/metabolism , Colorectal Neoplasms/metabolism , DNA Methylation , Feces , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Adenoma/diagnosis , Adenoma/genetics , Adult , Aged , Aged, 80 and over , Carcinoma/diagnosis , Carcinoma/genetics , Case-Control Studies , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/genetics , Feasibility Studies , Female , Humans , Intercellular Signaling Peptides and Proteins/genetics , Male , Membrane Proteins/genetics , Middle Aged , Polymerase Chain Reaction , Promoter Regions, Genetic/physiology
3.
Cancer Res ; 65(10): 4218-27, 2005 May 15.
Article in English | MEDLINE | ID: mdl-15899813

ABSTRACT

In many cases, silencing of gene expression by CpG methylation is causally involved in carcinogenesis. Furthermore, cancer-specific CpG methylation may serve as a tumor marker. In order to identify candidate genes for inactivation by CpG methylation in prostate cancer, the prostate cancer cell lines LNCaP, PC3, and Du-145 were treated with 5-aza-2' deoxycytidine and trichostatin A, which leads to reversion of epigenetic silencing. By microarray analysis of 18,400 individual transcripts, several hundred genes were found to be induced when compared with cells treated with trichostatin A. Fifty re-expressed genes were selected for further analysis based on their known function, which implied a possible involvement in tumor suppression. Twelve of these genes showed a significant degree of CpG methylation in their promoters. Six genes were silenced by CpG methylation in the majority of five analyzed prostate cancer cell lines, although they displayed robust mRNA expression in normal prostate epithelial cells obtained from four different donors. In primary prostate cancer samples derived from 41 patients, the frequencies of CpG methylation detected in the promoter regions of these genes were: GPX3, 93%; SFRP1, 83%; COX2, 78%; DKK3, 68%; GSTM1, 58%; and KIP2/p57, 56%. Ectopic expression of SFRP1 or DKK3 resulted in decreased proliferation. The expression of DKK3 was accompanied by attenuation of the mitogen-activated protein kinase pathway. The high frequency of CpG methylation detected in the promoters of the identified genes suggests a potential causal involvement in prostate cancer and may prove useful for diagnostic purposes.


Subject(s)
Gene Silencing , Prostatic Neoplasms/genetics , Adaptor Proteins, Signal Transducing , Cell Line, Tumor , Chemokines , CpG Islands , DNA Methylation , Gene Expression Regulation, Neoplastic , Humans , Intercellular Signaling Peptides and Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/genetics , Male , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Oligonucleotide Array Sequence Analysis , Prostatic Neoplasms/metabolism , Proteins/genetics , Proteins/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics
4.
Mod Pathol ; 18(3): 340-8, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15257317

ABSTRACT

The 14-3-3sigma inhibitor of cell cycle progression has been shown to be target of epigenetic deregulation in many forms of human cancers; however, its role in urological and gynecological cancers has not been studied. Here, we have analyzed the expression of 14-3-3sigma, wild-type p53 and mutated p53 in over 300 cases of the most common cancers occurring in the urological and gynecological tracts and its normal counterpart tissue by immunohistochemistry using the multiple tumor tissue microarrays. 14-3-3sigma expression was detected in normal epithelia from most organs with sporadic expression in renal tubules and absence in the testis. In contrast to normal tissue, 14-3-3sigma expression was lost in 40-60% of adenocarcinomas of the breast, ovary, endometrium and prostate. There was no association between 14-3-3sigma and wild-type/mutated p53 expression. By performing methylation-specific PCR, we showed a close association of 14-3-3sigma CpG island methylation and low protein expression levels of 14-3-3sigma. In addition, a direct link of 14-3-3sigma mRNA expression levels to CpG island methylation is demonstrated in two human cancer cell lines. Loss of 14-3-3sigma expression due to promoter hypermethylation may represent the most frequent molecular aberration in ovarian, endometrial and prostate adenocarcinomas.


Subject(s)
Biomarkers, Tumor/genetics , CpG Islands/genetics , DNA Methylation , Exonucleases/genetics , Neoplasm Proteins/genetics , Neoplasms/genetics , 14-3-3 Proteins , Azacitidine/pharmacology , Biomarkers, Tumor/biosynthesis , Cell Line, Tumor , Down-Regulation , Endometrial Neoplasms/genetics , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Exonucleases/biosynthesis , Exoribonucleases , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunohistochemistry , Male , Mutation , Neoplasm Proteins/biosynthesis , Neoplasms/metabolism , Neoplasms/pathology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tissue Array Analysis , Tumor Suppressor Protein p53/genetics
5.
Oncogene ; 23(56): 9034-41, 2004 Dec 02.
Article in English | MEDLINE | ID: mdl-15489902

ABSTRACT

In order to identify tumor suppressive genes silenced by CpG methylation in prostate carcinoma (PCa), we determined genome-wide expression changes after pharmacological reversal of CpG methylation-mediated transcriptional repression in three PCa cell lines using microarray analysis. Thereby, epigenetic silencing of the 14-3-3sigma gene was detected in the cell line LNCaP. 14-3-3sigma encodes a p53-regulated inhibitor of cell cycle progression. Laser microdissection was used to isolate different cell types present in diseased prostatic tissue. Subsequent methylation-specific PCR analysis showed CpG methylation of 14-3-3sigma in all 41 primary PCa samples analysed, which was accompanied by a decrease or loss of 14-3-3sigma protein expression. In contrast, normal prostate epithelial and benign prostate hyperplasia cells showed high levels of 14-3-3sigma expression. PCa-precursor lesions (prostatic intraepithelial neoplasia) also displayed decreased levels of 14-3-3sigma expression in luminal cells, which are known to contain shortened telomeres. RNA interference-mediated inactivation of 14-3-3sigma compromised a DNA damage-induced G(2)/M arrest in the PCa cell line PC3. The generality of CpG methylation and downregulation of 14-3-3sigma expression in PCa suggests that it significantly contributes to the formation of PCa, potentially by allowing the escape from a DNA damage-induced arrest elicited by telomere shortening.


Subject(s)
Biomarkers, Tumor/genetics , Epigenesis, Genetic , Exonucleases/genetics , Gene Silencing , Neoplasm Proteins/genetics , Prostatic Neoplasms/genetics , 14-3-3 Proteins , Base Sequence , Cell Line, Tumor , CpG Islands , DNA Methylation , DNA Primers , Down-Regulation , Exoribonucleases , Humans , Male , Oligonucleotide Array Sequence Analysis , Prostatic Neoplasms/pathology , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction
6.
Oncogene ; 22(35): 5519-24, 2003 Aug 21.
Article in English | MEDLINE | ID: mdl-12934112

ABSTRACT

The p53-regulated 14-3-3sigma gene encodes an inhibitor of cell cycle progression essential for senescence and clonal evolution of keratinocytes in vitro. Here we analysed the in vivo expression of 14-3-3sigma protein in several skin diseases, which are characterized by hyperproliferative keratinocytes. Unexpectedly, the 14-3-3sigma protein was expressed at high levels in psoriasis (11 of 11 patients), condylomata acuminata (11/11), actinic keratoses (11/11) and squamous cell carcinomas (SCC) (11/11). However, keratinocytes that had undergone transformation to basal cell carcinoma (BCC) showed partial (10 of 41; 24.4%) or complete (19 of 41; 46.3%) loss of 14-3-3sigma protein expression. BCC (5/5), SCC (6/6) and actinic keratoses (7/7) concomitantly expressed the p53-homolog p63 and 14-3-3sigma at high levels, ruling out potential inhibitory effects of p63 isoforms on 14-3-3sigma transcription as the basis for loss of 14-3-3sigma expression. Of 41 BCC samples isolated by laser-capture microdissection, 28 (68.3%) showed CpG-hypermethylation of the 14-3-3sigma promoter combined with reduced or absent 14-3-3sigma protein levels in 22 cases (78.6%). Since it has been reported that BCC retain wild-type p16(INK4A) and here BCC with CpG-methylation of 14-3-3sigma did not show CpG-methylation of p16(INK4A) (0/17), silencing of 14-3-3sigma may contribute to evasion of senescence in BCC. As experimental removal of 14-3-3sigma sensitizes to DNA damage, silencing of 14-3-3sigma may explain the high efficacy of radiation therapy in the treatment of BCC.


Subject(s)
Biomarkers, Tumor , Carcinoma, Basal Cell/genetics , DNA Methylation , Exonucleases/genetics , Neoplasm Proteins , 14-3-3 Proteins , Carcinoma, Basal Cell/metabolism , Exonucleases/biosynthesis , Exoribonucleases , Humans , Skin Diseases/pathology
7.
J Clin Invest ; 110(11): 1717-27, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12464677

ABSTRACT

Using microarray analysis, we have detected downregulation of several components of the cGMP signaling pathway during replicative senescence of primary human diploid fibroblasts (HDFs). Therefore, the effect of pharmacological inhibition of cGMP synthesis was analyzed in HDFs. Treatment with 6-anilino-5,8-quinolinequinone (LY83583, referred to as LY hereafter), a previously described inhibitor of guanylate cyclase, induced cellular senescence. Microarray analysis revealed that LY treatment induced the Cdk inhibitor p21(WAF1/SDI/CIP1). In colorectal cancer cells, transcription of p21 was induced by LY in a p53-independent manner. Furthermore, p21, but not p53, was required for inhibition of proliferation by LY. The lack of p53 involvement suggests that LY does not induce DNA damage. Growth inhibition was also observed in malignant melanoma and breast cancer cell lines. Functional inactivation of the retinoblastoma tumor-suppressor protein, an effector of p21-mediated cell-cycle inhibition, converted LY-induced growth arrest to apoptosis. These results suggest that LY, or derivatives, may be useful therapeutic agents for the treatment of tumors.


Subject(s)
Aminoquinolines/pharmacology , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclins/genetics , Enzyme Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Cell Division/drug effects , Cellular Senescence/drug effects , Cellular Senescence/genetics , Cyclic GMP/physiology , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/drug effects , Fibroblasts/cytology , Fibroblasts/drug effects , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Humans , Infant, Newborn , Kinetics , Signal Transduction/drug effects , Signal Transduction/physiology , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...