Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
PLoS One ; 10(9): e0136505, 2015.
Article in English | MEDLINE | ID: mdl-26352266

ABSTRACT

The MYD88 L265P is a recurrent somatic mutation in neoplastic cells from patients with Waldenström Macroglobulinemia (WM). We identified the MYD88 L265P mutation in three individuals from unrelated families, but its presence did not explain the disease segregation within these WM pedigrees. We observed the mutation in these three individuals at high allele fractions in DNA extracted from EBV-immortalized Lymphoblastoid cell lines established from peripheral blood (LCL), but at much lower allele fractions in DNA extracted directly from peripheral blood, suggesting that this mutation is present in a clonal cell subpopulation rather than of germ-line origin. Furthermore, we observed that the MYD88 L265P mutation is enriched in WM families, detected in 40.5% of patients with familial WM or MGUS (10/22 WM, 5/15 MGUS), compared to 3.5% of patients with familial MM or MGUS (0/72 MM, 4/41 MGUS) (p = 10-7). The mutant allele frequency increased with passages in vitro after immortalization with Epstein-Barr virus (EBV) consistent with the MYD88 L265P described gain-of-function proposed for this mutation. The MYD88 L265P mutation appears to be frequently present in circulating cells in patients with WM, and MGUS, and these cells are amenable to immortalization by EBV.


Subject(s)
Cell Transformation, Viral , Clone Cells/pathology , Lymphocytes/pathology , Mutation, Missense , Myeloid Differentiation Factor 88/genetics , Point Mutation , Waldenstrom Macroglobulinemia/pathology , Adult , Aged , Aged, 80 and over , Alleles , Cell Division , Cells, Cultured , Clone Cells/metabolism , Clone Cells/virology , Female , Genetic Predisposition to Disease , Herpesvirus 4, Human/physiology , Humans , Immunoglobulin M/genetics , Lymphocytes/metabolism , Lymphocytes/virology , Male , Middle Aged , Monoclonal Gammopathy of Undetermined Significance/blood , Monoclonal Gammopathy of Undetermined Significance/genetics , Monoclonal Gammopathy of Undetermined Significance/pathology , Waldenstrom Macroglobulinemia/blood , Waldenstrom Macroglobulinemia/genetics
2.
Development ; 141(19): 3683-96, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25249460

ABSTRACT

The molecular signals driving tendon development are not fully identified. We have undertaken a transcriptome analysis of mouse limb tendon cells that were isolated at different stages of development based on scleraxis (Scx) expression. Microarray comparisons allowed us to establish a list of genes regulated in tendon cells during mouse limb development. Bioinformatics analysis of the tendon transcriptome showed that the two most strongly modified signalling pathways were TGF-ß and MAPK. TGF-ß/SMAD2/3 gain- and loss-of-function experiments in mouse limb explants and mesenchymal stem cells showed that TGF-ß signalling was sufficient and required via SMAD2/3 to drive mouse mesodermal stem cells towards the tendon lineage ex vivo and in vitro. TGF-ß was also sufficient for tendon gene expression in late limb explants during tendon differentiation. FGF does not have a tenogenic effect and the inhibition of the ERK MAPK signalling pathway was sufficient to activate Scx in mouse limb mesodermal progenitors and mesenchymal stem cells.


Subject(s)
Extremities/physiology , Gene Expression Regulation, Developmental/physiology , Signal Transduction/physiology , Tendons/cytology , Transcriptome/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Computational Biology , Gene Expression Profiling , Gene Expression Regulation, Developmental/genetics , In Situ Hybridization , Mesenchymal Stem Cells/metabolism , Mice , Microarray Analysis , Mitogen-Activated Protein Kinases/metabolism , Real-Time Polymerase Chain Reaction , Tendons/metabolism , Transcriptome/genetics , Transforming Growth Factor beta/metabolism
3.
J Neuropathol Exp Neurol ; 73(10): 908-15, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25192052

ABSTRACT

Gemistocytic astrocytoma (World Health Organization grade II) is a rare variant of diffuse astrocytoma that is characterized by the presence of neoplastic gemistocytes and has a significantly less favorable prognosis. Other than frequent TP53 mutations (>80%), little is known about its molecular profile. Here, we show that gemistocytic astrocytomas carry a lower frequency of IDH mutations than fibrillary astrocytomas (74% vs 92%; p = 0.0255) but have profiles similar to those of fibrillary astrocytomas with respect to TERT promoter mutations (5% vs 0%), 1p/19q loss (10% vs 8%), and loss of heterozygosity 10q (10% vs 12%). Exome sequencing in 5 gemistocytic astrocytomas revealed homozygous deletion of genes at 19q13 (i.e. RRAS [related RAS viral oncogene homolog; 2 cases] and ERCC1 [excision repair cross-complementing rodent repair deficiency, complementation group 1; 1 case]). Further screening showed RRAS homozygous deletion in 7 of 42 (17%) gemistocytic astrocytomas and in 3 of 24 (13%) IDH1 mutated secondary glioblastomas. Patients with gemistocytic astrocytoma and secondary glioblastoma with an RRAS deletion tended to have shorter survival rates than those without deletion. Differential polymerase chain reaction and methylation-specific polymerase chain reaction revealed an ERCC1 homozygous deletion or promoter methylation in 10 of 42 (24%) gemistocytic astrocytomas and in 8 of 24 (33%) secondary glioblastomas. Alterations in RRAS and ERCC1 appear to be typical in gemistocytic astrocytomas and secondary glioblastomas, since they were not present in 49 fibrillary astrocytomas or 30 primary glioblastomas.


Subject(s)
Astrocytoma/genetics , Brain Neoplasms/genetics , Chromosomes, Human, Pair 19/genetics , DNA-Binding Proteins/genetics , Endonucleases/genetics , ras Proteins/genetics , Adult , Astrocytoma/diagnosis , Astrocytoma/mortality , Brain Neoplasms/diagnosis , Brain Neoplasms/mortality , Female , Humans , Male , Middle Aged , Mutation/genetics , Survival Rate/trends
4.
Cancer Res ; 74(21): 6082-93, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25205102

ABSTRACT

Viruses disrupt the host cell microRNA (miRNA) network to facilitate their replication. Human T-cell leukemia virus type I (HTLV-1) replication relies on the clonal expansion of its host CD4(+) and CD8(+) T cells, yet this virus causes adult T-cell leukemia/lymphoma (ATLL) that typically has a CD4(+) phenotype. The viral oncoprotein Tax, which is rarely expressed in ATLL cells, has long been recognized for its involvement in tumor initiation by promoting cell proliferation, genetic instability, and miRNA dysregulation. Meanwhile, HBZ is expressed in both untransformed infected cells and ATLL cells and is involved in sustaining cell proliferation and silencing virus expression. Here, we show that an HBZ-miRNA axis promotes cell proliferation and genetic instability, as indicated by comet assays that showed increased numbers of DNA-strand breaks. Expression profiling of miRNA revealed that infected CD4(+) cells, but not CD8(+) T cells, overexpressed oncogenic miRNAs, including miR17 and miR21. HBZ activated these miRNAs via a posttranscriptional mechanism. These effects were alleviated by knocking down miR21 or miR17 and by ectopic expression of OBFC2A, a DNA-damage factor that is downregulated by miR17 and miR21 in HTLV-1-infected CD4(+) T cells. These findings extend the oncogenic potential of HBZ and suggest that viral expression might be involved in the remarkable genetic instability of ATLL cells.


Subject(s)
Basic-Leucine Zipper Transcription Factors/genetics , Cell Proliferation/genetics , Genomic Instability , Leukemia-Lymphoma, Adult T-Cell/genetics , Viral Proteins/genetics , Adult , Basic-Leucine Zipper Transcription Factors/metabolism , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Gene Expression Regulation, Viral , Genes, pX/genetics , Human T-lymphotropic virus 1/genetics , Humans , Leukemia-Lymphoma, Adult T-Cell/immunology , Leukemia-Lymphoma, Adult T-Cell/pathology , Retroviridae Proteins , Viral Proteins/metabolism
5.
Virol J ; 11: 32, 2014 Feb 20.
Article in English | MEDLINE | ID: mdl-24555484

ABSTRACT

BACKGROUND: Hantaviruses are single-stranded RNA viruses, which are transmitted to humans primarily via inhalation of aerosolised virus in contaminated rodent urine and faeces. Whilst infected reservoir hosts are asymptomatic, human infections can lead to two clinical manifestations, haemorrhagic fever with renal syndrome (HFRS) and hantavirus cardiopulmonary syndrome (HCPS), with varying degrees of clinical severity. The incidence of rodent and human cases of Seoul virus (SEOV) in Europe has been considered to be low, and speculated to be driven by the sporadic introduction of infected brown rats (Rattus norvegicus) via ports. METHODS: Between October 2010 and March 2012, 128 brown rats were caught at sites across the Lyon region in France. RESULTS: SEOV RNA was detected in the lungs of 14% (95% CI 8.01-20.11) of brown rats tested using a nested pan-hantavirus RT-PCR (polymerase gene). Phylogenetic analysis supports the inclusion of the Lyon SEOV within Lineage 7 with SEOV strains originating from SE Asia and the previously reported French & Belgian SEOV strains. Sequence data obtained from the recent human SEOV case (Replonges) was most similar to that obtained from one brown rat trapped in a public park in Lyon city centre. We obtained significantly improved recovery of virus genome sequence directly from SEOV infected lung material using a simple viral enrichment approach and NGS technology. CONCLUSIONS: The detection of SEOV in two wild caught brown rats in the UK and the multiple detection of SEOV infected brown rats in the Lyon region of France, suggests that SEOV is circulating in European brown rats. Under-reporting and difficulties in identifying the hantaviruses associated with HFRS may mask the public health impact of SEOV in Europe.


Subject(s)
Carrier State/veterinary , Disease Reservoirs , Rats/virology , Seoul virus/isolation & purification , Animals , Carrier State/epidemiology , Carrier State/virology , Cluster Analysis , France/epidemiology , Lung/virology , Molecular Sequence Data , Phylogeny , Polymerase Chain Reaction , RNA, Viral/genetics , Sequence Analysis, DNA , Sequence Homology
6.
PLoS One ; 8(6): e67313, 2013.
Article in English | MEDLINE | ID: mdl-23805307

ABSTRACT

BRCA1 (Breast Cancer 1) has been implicated in a number of cellular processes, including transcription regulation, DNA damage repair and protein ubiquitination. We previously demonstrated that BRCA1 interacts with PABP1 (Poly(A)-Binding Protein 1) and that BRCA1 modulates protein synthesis through this interaction. To identify the mRNAs that are translationally regulated by BRCA1, we used a microarray analysis of polysome-bound mRNAs in BRCA1-depleted and non-depleted MCF7 cells. Our findings show that BRCA1 modifies the translational efficiency of approximately 7% of the mRNAs expressed in these cells. Further analysis revealed that several processes contributing to cell surveillance such as cell cycle arrest, cell death, cellular growth and proliferation, DNA repair and gene expression, are largely enriched for the mRNAs whose translation is impacted by BRCA1. The BRCA1-dependent translation of these species of mRNAs therefore uncovers a novel mechanism through which BRCA1 exerts its onco-suppressive role. In addition, the BRCA1-dependent translation of mRNAs participating in unexpected functions such as cellular movement, nucleic acid metabolism or protein trafficking is indicative of novel functions for BRCA1. Finally, this study contributes to the identification of several markers associated with BRCA1 deficiency and to the discovery of new potential anti-neoplastic therapeutic targets.


Subject(s)
BRCA1 Protein/metabolism , Biomarkers, Tumor/biosynthesis , Breast Neoplasms/metabolism , Poly(A)-Binding Protein I/metabolism , Protein Biosynthesis , RNA, Messenger/metabolism , RNA, Neoplasm/metabolism , BRCA1 Protein/genetics , Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Female , Humans , MCF-7 Cells , Poly(A)-Binding Protein I/genetics , RNA, Messenger/genetics , RNA, Neoplasm/genetics
7.
J Virol ; 87(1): 234-42, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23077304

ABSTRACT

Monocyte-derived dendritic cells (MDDCs) play a key role in the regulation of the immune system and are the target of numerous gene therapy applications. The genetic modification of MDDCs is possible with human immunodeficiency virus type 1 (HIV-1)-derived lentiviral vectors (LVs) but requires high viral doses to bypass their natural resistance to viral infection, and this in turn affects their physiological properties. To date, a single viral protein is able to counter this restrictive phenotype, Vpx, a protein derived from members of the HIV-2/simian immunodeficiency virus SM lineage that counters at least two restriction factors present in myeloid cells. By tagging Vpx with a short heterologous membrane-targeting domain, we have obtained HIV-1 LVs incorporating high levels of this protein (HIV-1-Src-Vpx). These vectors efficiently transduce differentiated MDDCs and monocytes either as previously purified populations or as populations within unsorted peripheral blood mononuclear cells (PBMCs). In addition, these vectors can be efficiently pseudotyped with receptor-specific envelopes, further restricting their cellular tropism almost uniquely to MDDCs. Compared to conventional HIV-1 LVs, these novel vectors allow for an efficient genetic modification of MDDCs and, more importantly, do not cause their maturation or affect their survival, which are unwanted side effects of the transduction process. This study describes HIV-1-Src-Vpx LVs as a novel potent tool for the genetic modification of differentiated MDDCs and of circulating monocyte precursors with strong potential for a wide range of gene therapy applications.


Subject(s)
Dendritic Cells/virology , Genetic Vectors , HIV-1/immunology , Monocytes/virology , Transduction, Genetic , Viral Regulatory and Accessory Proteins/metabolism , Cells, Cultured , HIV-1/genetics , HIV-2/genetics , Humans , Simian Immunodeficiency Virus/genetics , Viral Regulatory and Accessory Proteins/genetics
8.
EMBO J ; 31(10): 2296-308, 2012 May 16.
Article in English | MEDLINE | ID: mdl-22522705

ABSTRACT

RNA interference (RNAi) silences gene expression by acting both at the transcriptional and post-transcriptional levels in a broad range of eukaryotes. In the fission yeast Schizosaccharomyces pombe the RNA-Induced Transcriptional Silencing (RITS) RNAi complex mediates heterochromatin formation at non-coding and repetitive DNA. However, the targeting and role of RITS at other genomic regions, including protein-coding genes, remain unknown. Here we show that RITS localizes to specific meiotic genes and mRNAs. Remarkably, RITS is guided to these meiotic targets by the RNA-binding protein Mmi1 and its associated RNA surveillance machinery that together degrade selective meiotic mRNAs during vegetative growth. Upon sexual differentiation, RITS localization to the meiotic genes and mRNAs is lost. Large-scale identification of Mmi1 RNA targets reveals that RITS subunit Chp1 associates with the vast majority of them. In addition, loss of RNAi affects the effective repression of sexual differentiation mediated by the Mmi1 RNA surveillance machinery. These findings uncover a new mechanism for recruiting RNAi to specific meiotic genes and suggest that RNAi participates in the control of sexual differentiation in fission yeast.


Subject(s)
Gene Expression Regulation, Fungal , Genes, Fungal , RNA-Induced Silencing Complex/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , mRNA Cleavage and Polyadenylation Factors/metabolism , Models, Biological , Protein Binding , RNA, Fungal/metabolism
9.
Nutr Res ; 32(2): 124-32, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22348461

ABSTRACT

Folate is essential for purine and thymidylate biosynthesis and in methyl transfer for DNA methylation. Folate deficiency alters the secretion of melatonin, a hormone involved in circadian rhythm entrainment, and causes hyperhomocysteinemia because of disruption of homocysteine metabolism. Adverse effects of homocysteine include the generation of free radicals, activation of proliferation or apoptosis, and alteration of gene expression. The liver is an important organ for folate metabolism, and its genome analysis has revealed numerous clock-regulated genes. The variations at the level of their expression during folate deficiency are not known. The aim of our study was to investigate the effects of folate deficiency on gene expression in the mouse liver. A control group receiving a synthetic diet and a folate-depleted group were housed for 4 weeks on a 12-hour/12-hour light/dark cycle. Three mice from each group were euthanized under dim red light at the beginning of the light cycle, and 3, at the beginning of the dark period. Gene expression was studied in a microarray analysis. Of the 53 genes showing modified daily expression in the controls, 52 showed a less marked or no difference after folate depletion. Only 1, lpin1, showed a more marked difference. Ten genes coding for proteins involved in lipid metabolism did not show a morning/evening difference in controls but did after folate depletion. This study shows that, in the mouse liver, dietary folate depletion leads to major changes in expression of several genes involved in fatty acid metabolism, DNA synthesis, and expression of circadian genes.


Subject(s)
Circadian Rhythm/genetics , DNA Replication/genetics , Folic Acid Deficiency/genetics , Folic Acid/administration & dosage , Gene Expression , Lipid Metabolism/genetics , Vitamin B Complex/administration & dosage , Animals , DNA/biosynthesis , DNA Methylation , Darkness , Fatty Acids , Folic Acid Deficiency/complications , Homocysteine/metabolism , Hyperhomocysteinemia/etiology , Light , Liver/metabolism , Male , Melatonin/metabolism , Mice , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphatidate Phosphatase/genetics , Phosphatidate Phosphatase/metabolism
10.
Epigenetics ; 6(11): 1295-307, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22048253

ABSTRACT

Methyl-CpG Binding Domain (MBD) proteins are thought to be key molecules in the interpretation of DNA methylation signals leading to gene silencing through recruitment of chromatin remodeling complexes. In cancer, the MBD-family member, MBD2, may be primarily involved in the repression of genes exhibiting methylated CpG at their 5' end. Here we ask whether MBD2 randomly associates methylated sequences, producing chance effects on transcription, or exhibits a more specific recognition of some methylated regions. Using chromatin and DNA immunoprecipitation, we analyzed MBD2 and RNA polymerase II deposition and DNA methylation in HeLa cells on arrays representing 25,500 promoter regions. This first whole-genome mapping revealed the preferential localization of MBD2 near transcription start sites (TSSs), within the region analyzed, 7.5 kb upstream through 2.45 kb downstream of 5' transcription start sites. Probe by probe analysis correlated MBD2 deposition and DNA methylation. Motif analysis did not reveal specific sequence motifs; however, CCG and CGC sequences seem to be overrepresented. Nonrandom association (multiple correspondence analysis, p < 0.0001) between silent genes, DNA methylation and MBD2 binding was observed. The association between MBD2 binding and transcriptional repression weakened as the distance between binding site and TSS increased, suggesting that MBD2 represses transcriptional initiation. This hypothesis may represent a functional explanation for the preferential binding of MBD2 at methyl-CpG in TSS regions.


Subject(s)
DNA-Binding Proteins/genetics , Transcription Initiation Site , Binding Sites , Chromatin/metabolism , CpG Islands/genetics , DNA/metabolism , DNA Methylation , HeLa Cells , Humans , Promoter Regions, Genetic , RNA Polymerase II/genetics , RNA Polymerase II/metabolism
11.
Mol Cell Biol ; 31(7): 1459-69, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21262769

ABSTRACT

CCN5 is a member of the CCN (connective tissue growth factor/cysteine-rich 61/nephroblastoma overexpressed) family and was identified as an estrogen-inducible gene in estrogen receptor-positive cell lines. However, the role of CCN5 in breast carcinogenesis remains unclear. We report here that the CCN5 protein is localized mostly in the cytoplasm and in part in the nucleus of human tumor breast tissue. Using a heterologous transcription assay, we demonstrate that CCN5 can act as a transcriptional repressor presumably through association with histone deacetylase 1 (HDAC1). Microarray gene expression analysis showed that CCN5 represses expression of genes associated with epithelial-mesenchymal transition (EMT) as well as expression of key components of the transforming growth factor ß (TGF-ß) signaling pathway, prominent among them TGF-ßRII receptor. We show that CCN5 is recruited to the TGF-ßRII promoter, thereby providing a mechanism by which CCN5 restricts transcription of the TGF-ßRII gene. Consistent with this finding, CCN5, we found, functions to suppress TGF-ß-induced transcriptional responses and invasion that is concomitant with EMT. Thus, our data uncovered CCN5 as a novel transcriptional repressor that plays an important role in regulating tumor progression functioning, at least in part, by inhibiting the expression of genes involved in the TGF-ß signaling cascade that is known to promote EMT.


Subject(s)
Intercellular Signaling Peptides and Proteins/metabolism , Repressor Proteins/metabolism , Signal Transduction , Transcription Factors/metabolism , Transcription, Genetic , Transforming Growth Factor beta/metabolism , CCN Intercellular Signaling Proteins , Cadherins/metabolism , Cell Line, Tumor , Down-Regulation/genetics , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Silencing , Histone Deacetylases/metabolism , Humans , Intercellular Signaling Peptides and Proteins/chemistry , Neoplasm Invasiveness , Promoter Regions, Genetic/genetics , Protein Binding , Protein Serine-Threonine Kinases/genetics , Protein Structure, Tertiary , Protein Transport , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Repressor Proteins/chemistry , Subcellular Fractions/metabolism , Transcription Factors/chemistry , Transforming Growth Factor beta/genetics
12.
Brain Pathol ; 21(5): 533-43, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21251114

ABSTRACT

Integrative genomics approaches associating DNA structure and transcriptomic analysis should allow the identification of cascades of events relating to tumor aggressiveness. While different genome alterations have been identified in pituitary tumors, none have ever been correlated with the aggressiveness. This study focused on one subtype of pituitary tumor, the prolactin (PRL) pituitary tumors, to identify molecular events associated with the aggressive and malignant phenotypes. We combined a comparative genomic hybridization and transcriptomic analysis of 13 PRL tumors classified as nonaggressive or aggressive. Allelic loss within the p arm region of chromosome 11 was detected in five of the aggressive tumors. Allelic loss in the 11q arm was observed in three of these five tumors, all three of which were considered as malignant based on the occurrence of metastases. Comparison of genomic and transcriptomic data showed that allelic loss impacted upon the expression of genes located in the imbalanced region. Data filtering allowed us to highlight five deregulated genes (DGKZ, CD44, TSG101, GTF2H1, HTATIP2), within the missing 11p region, potentially responsible for triggering the aggressive and malignant phenotypes of PRL tumors. Our combined genomic and transcriptomic analysis underlines the importance of chromosome allelic loss in determining the aggressiveness and malignancy of tumors.


Subject(s)
Chromosome Aberrations , Chromosomes, Human, Pair 11 , Pituitary Neoplasms/genetics , Prolactinoma/genetics , Acetyltransferases/genetics , Adult , Aged , Aged, 80 and over , Cell Cycle/genetics , Comparative Genomic Hybridization , DNA-Binding Proteins/genetics , Diacylglycerol Kinase/genetics , Diacylglycerol Kinase/metabolism , Down-Regulation/genetics , Endosomal Sorting Complexes Required for Transport/genetics , Female , Gene Expression Profiling/methods , Genetic Predisposition to Disease/genetics , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Male , Middle Aged , Phosphoproteins/genetics , Signal Transduction/genetics , Transcription Factor TFIIH , Transcription Factors/genetics , Transcription Factors, TFII/genetics
13.
Mol Cell Endocrinol ; 326(1-2): 30-9, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20211685

ABSTRACT

Predicting pituitary tumor behavior remains a challenge, since the prognostic value of identified pathological markers has not yet been evaluated. Genome-wide transcriptome analyses allow the identification of molecular markers to highlight global changes in gene expression and enable the discovery of signaling pathways within complex biological networks. While several transcriptome studies comparing normal with tumoral pituitary as a whole or with respect to subtype have given interesting data concerning pituitary pathogenesis, none have considered pituitary tumor prognosis. Only one study to date, focusing on the pathological classification and progression of prolactin tumors, has identified some molecular markers with diagnostic and prognostic value. We reviewed data in the literature on human pituitary tumor transcriptome and conducted a meta-analysis on the expression of genes involved in cell growth, proliferation and the cell cycle. Based on our expertise, we evaluate the interests and the limitations of using this approach with human pituitary tumors.


Subject(s)
Biomarkers, Tumor/genetics , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Pituitary Neoplasms/genetics , Biomarkers, Tumor/analysis , Cell Proliferation , Disease Progression , Genome , Humans , Oligonucleotide Array Sequence Analysis , Pituitary Gland/chemistry , Pituitary Gland/pathology , Pituitary Hormones/genetics , Pituitary Neoplasms/metabolism , Pituitary Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...