Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
JCI Insight ; 3(14)2018 07 26.
Article in English | MEDLINE | ID: mdl-30046005

ABSTRACT

Resistance to current therapies still impacts a significant number of melanoma patients and can be regulated by epigenetic alterations. Analysis of global cytosine methylation in a cohort of primary melanomas revealed a pattern of early demethylation associated with overexpression of oncogenic transcripts. Loss of methylation and associated overexpression of the CSF 1 receptor (CSF1R) was seen in a majority of tumors and was driven by an alternative, endogenous viral promoter in a subset of samples. CSF1R was particularly elevated in melanomas with BRAF and other MAPK activating mutations. Furthermore, rebound ERK activation after BRAF inhibition was associated with RUNX1-mediated further upregulation of CSF-1R and its ligand IL-34. Importantly, increased CSF-1R and IL-34 overexpression were detected in an independent cohort of resistant melanomas. Inhibition of CSF-1R kinase or decreased CSF-1R expression by RNAi reduced 3-D growth and invasiveness of melanoma cells. Coinhibition of CSF-1R and BRAF resulted in synergistic efficacy in vivo. To our knowledge, our data unveil a previously unknown role for the autocrine-regulated CSF-1R in BRAF V600E resistance and provide a preclinical rationale for targeting this pathway in melanoma.


Subject(s)
Core Binding Factor Alpha 2 Subunit/metabolism , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Interleukins/metabolism , Melanoma/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , DNA Methylation , Drug Synergism , Female , Humans , MAP Kinase Signaling System , Melanoma/genetics , Melanoma/pathology , Mice , Mice, Inbred BALB C , Mitogen-Activated Protein Kinase Kinases/drug effects , Mutation , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , THP-1 Cells , Transplantation, Heterologous , U937 Cells
2.
Microbiologyopen ; 3(4): 470-83, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24916637

ABSTRACT

The bacteriophage Mu Mor activator protein is absolutely required for transcription from the Mu middle promoter P(m). However, when RNA polymerase (RNAP) was incubated with P(m) DNA in the absence of Mor, a band at promoter position -51 was hypersensitive to DNase I cleavage, demonstrating an interaction of RNAP with the promoter DNA. The hypersensitivity was similar at four different lengths of P(m) DNA assayed from -62 to +10, -62 to +46, -96 to +10, and -96 to +46. The hypersensitivity occurred equally well at 5 °C, 15 °C, and 30 °C, indicating that it did not require open complex formation, which only occurred at 30 °C. The -51 hypersensitivity at 5 °C and 15 °C was eliminated by the addition of heparin, consistent with the possibility that it arose by formation of unstable closed complexes of RNAP bound to P(m) DNA. Generation of the hypersensitive band required the complete RNAP with its αCTDs, but neither the αCTD nor intact α were sufficient for the interaction and resulting hypersensitivity. There was no correlation between the level of hypersensitivity observed in vitro and the level of Pm activity in vivo, as assayed by the Mor-dependent production of ß-galactosidase from a P(m)-lacZ fusion. In an "order of addition" experiment, preincubation of P(m) DNA with Mor followed by addition of RNAP led to the fastest open complex formation, whereas preincubation of P(m) DNA with RNAP gave the slowest. These results support the conclusion that Mor recruits RNAP to P(m) rather than reposition a prebound RNAP, as occurs for C-dependent repositioning of RNAP at the Mu late promoter Pmom .


Subject(s)
Bacteriophage mu/enzymology , DNA-Directed RNA Polymerases/metabolism , Promoter Regions, Genetic , Trans-Activators/metabolism , Gene Expression Profiling , Genes, Reporter , Protein Binding , Temperature , Transcription, Genetic , beta-Galactosidase/analysis , beta-Galactosidase/genetics
3.
J Clin Invest ; 124(3): 1158-67, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24487588

ABSTRACT

Acute myeloid leukemia (AML) is characterized by disruption of HSC and progenitor cell differentiation. Frequently, AML is associated with mutations in genes encoding epigenetic modifiers. We hypothesized that analysis of alterations in DNA methylation patterns during healthy HSC commitment and differentiation would yield epigenetic signatures that could be used to identify stage-specific prognostic subgroups of AML. We performed a nano HpaII-tiny-fragment-enrichment-by-ligation-mediated-PCR (nanoHELP) assay to compare genome-wide cytosine methylation profiles between highly purified human long-term HSC, short-term HSC, common myeloid progenitors, and megakaryocyte-erythrocyte progenitors. We observed that the most striking epigenetic changes occurred during the commitment of short-term HSC to common myeloid progenitors and these alterations were predominantly characterized by loss of methylation. We developed a metric of the HSC commitment­associated methylation pattern that proved to be highly prognostic of overall survival in 3 independent large AML patient cohorts, regardless of patient treatment and epigenetic mutations. Application of the epigenetic signature metric for AML prognosis was superior to evaluation of commitment-based gene expression signatures. Together, our data define a stem cell commitment­associated methylome that is independently prognostic of poorer overall survival in AML.


Subject(s)
Cell Differentiation , Epigenesis, Genetic , Hematopoietic Stem Cells/physiology , Leukemia, Myeloid, Acute/pathology , DNA Methylation , Humans , Kaplan-Meier Estimate , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/mortality , Prognosis , Proportional Hazards Models , Transcriptome
4.
J Biol Chem ; 288(13): 8805-14, 2013 Mar 29.
Article in English | MEDLINE | ID: mdl-23306203

ABSTRACT

Differentiation of hematopoietic stem cells to red cells requires coordinated expression of numerous erythroid genes and is characterized by nuclear condensation and extrusion during terminal development. To understand the regulatory mechanisms governing these widespread phenotypic changes, we conducted a high resolution methylomic and transcriptomic analysis of six major stages of human erythroid differentiation. We observed widespread epigenetic differences between early and late stages of erythropoiesis with progressive loss of methylation being the dominant change during differentiation. Gene bodies, intergenic regions, and CpG shores were preferentially demethylated during erythropoiesis. Epigenetic changes at transcription factor binding sites correlated significantly with changes in gene expression and were enriched for binding motifs for SCL, MYB, GATA, and other factors not previously implicated in erythropoiesis. Demethylation at gene promoters was associated with increased expression of genes, whereas epigenetic changes at gene bodies correlated inversely with gene expression. Important gene networks encoding erythrocyte membrane proteins, surface receptors, and heme synthesis proteins were found to be regulated by DNA methylation. Furthermore, integrative analysis enabled us to identify novel, potential regulatory areas of the genome as evident by epigenetic changes in a predicted PU.1 binding site in intron 1 of the GATA1 gene. This intronic site was found to be conserved across species and was validated to be a novel PU.1 binding site by quantitative ChIP in erythroid cells. Altogether, our study provides a comprehensive analysis of methylomic and transcriptomic changes during erythroid differentiation and demonstrates that human terminal erythropoiesis is surprisingly associated with hypomethylation of the genome.


Subject(s)
Erythropoiesis/physiology , Gene Expression Profiling , Gene Expression Regulation , Antigens, CD34/biosynthesis , Binding Sites , Cell Differentiation , CpG Islands , DNA Methylation , Epigenesis, Genetic , Epigenomics , Erythrocytes/cytology , Flow Cytometry/methods , Genome, Human , Genomics , Humans , Introns , Methylation , Oligonucleotide Array Sequence Analysis , Stem Cells/chemistry
5.
Cancer Res ; 73(3): 1076-85, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23066032

ABSTRACT

Even though mutations in epigenetic regulators frequently occur in myeloproliferative neoplasms, their effects on the epigenome have not been well studied. Furthermore, even though primary myelofibrosis (PMF) has a markedly worse prognosis than essential thrombocytosis or polycythemia vera, the molecular distinctions between these subgroups are not well elucidated. We conducted the HELP (HpaII tiny fragment enriched by LM-PCR) assay to study genome-wide methylation in polycythemia vera, essential thrombocytosis, and PMF samples compared with healthy controls. We determined that polycythemia vera and essential thrombocytosis are characterized by aberrant promoter hypermethylation, whereas PMF is an epigenetically distinct subgroup characterized by both aberrant hyper- and hypomethylation. Aberrant hypomethylation in PMF was seen to occur in non-CpG island loci, showing further qualitative differences between the disease subgroups. The differentially methylated genes in polycythemia vera and essential thrombocytosis were involved predominantly in cell signaling pathways and were enriched for binding sites of GATA1 and other transcription factors. In contrast, aberrantly methylated genes in PMF were involved in inflammatory pathways and were enriched for NF1, LEF1, and other transcription factors. Within the PMF subgroup, cases with ASXL1 disruptions formed an epigenetically distinct subgroup with relatively increased methylation. Cases of myeloproliferative neoplasms (MPN) with TET2 mutations showed decreased levels of hydroxymethylation and distinct set of hypermethylated genes. In contrast, the JAK2V617F mutation did not drive epigenetic clustering within MPNs. Finally, the significance of aberrant methylation was shown by sensitivity of MPN-derived cell lines to decitabine. These results show epigenetic differences between PMF and polycythemia vera/essential thrombocytosis and reveal methylomic signatures of ASXL1 and TET2 mutations.


Subject(s)
DNA Methylation , Mutation , Polycythemia Vera/genetics , Primary Myelofibrosis/genetics , Thrombocythemia, Essential/genetics , Adult , Aged , Aged, 80 and over , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Cell Line, Tumor , DNA-Binding Proteins/genetics , Decitabine , Dioxygenases , Female , Humans , Janus Kinase 2/genetics , Male , Middle Aged , Proto-Oncogene Proteins/genetics , Repressor Proteins/genetics
6.
J Biol Chem ; 286(28): 25211-23, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21532034

ABSTRACT

Myelodysplastic syndromes (MDS) are characterized by abnormal and dysplastic maturation of all blood lineages. Even though epigenetic alterations have been seen in MDS marrow progenitors, very little is known about the molecular alterations in dysplastic peripheral blood cells. We analyzed the methylome of MDS leukocytes by the HELP assay and determined that it was globally distinct from age-matched controls and was characterized by numerous novel, aberrant hypermethylated marks that were located mainly outside of CpG islands and preferentially affected GTPase regulators and other cancer-related pathways. Additionally, array comparative genomic hybridization revealed that novel as well as previously characterized deletions and amplifications could also be visualized in peripheral blood leukocytes, thus potentially reducing the need for bone marrow samples for future studies. Using integrative analysis, potentially pathogenic genes silenced by genetic deletions and aberrant hypermethylation in different patients were identified. DOCK4, a GTPase regulator located in the commonly deleted 7q31 region, was identified by this unbiased approach. Significant hypermethylation and reduced expression of DOCK4 in MDS bone marrow stem cells was observed in two large independent datasets, providing further validation of our findings. Finally, DOCK4 knockdown in primary marrow CD34(+) stem cells led to decreased erythroid colony formation and increased apoptosis, thus recapitulating the bone marrow failure seen in MDS. These findings reveal widespread novel epigenetic alterations in myelodysplastic leukocytes and implicate DOCK4 as a pathogenic gene located on the 7q chromosomal region.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 7/metabolism , Epigenesis, Genetic , GTPase-Activating Proteins/biosynthesis , Leukocytes/metabolism , Myelodysplastic Syndromes/metabolism , Apoptosis/genetics , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Chromosomes, Human, Pair 7/genetics , CpG Islands/genetics , DNA Methylation/genetics , Female , GTPase-Activating Proteins/genetics , Genetic Markers , Humans , Leukocytes/pathology , Male , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , Stem Cells/metabolism , Stem Cells/pathology
7.
PLoS Genet ; 7(3): e1001356, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21483804

ABSTRACT

Although a combination of genomic and epigenetic alterations are implicated in the multistep transformation of normal squamous esophageal epithelium to Barrett esophagus, dysplasia, and adenocarcinoma, the combinatorial effect of these changes is unknown. By integrating genome-wide DNA methylation, copy number, and transcriptomic datasets obtained from endoscopic biopsies of neoplastic progression within the same individual, we are uniquely able to define the molecular events associated progression of Barrett esophagus. We find that the previously reported global hypomethylation phenomenon in cancer has its origins at the earliest stages of epithelial carcinogenesis. Promoter hypomethylation synergizes with gene amplification and leads to significant upregulation of a chr4q21 chemokine cluster and other transcripts during Barrett neoplasia. In contrast, gene-specific hypermethylation is observed at a restricted number of loci and, in combination with hemi-allelic deletions, leads to downregulatation of selected transcripts during multistep progression. We also observe that epigenetic regulation during epithelial carcinogenesis is not restricted to traditionally defined "CpG islands," but may also occur through a mechanism of differential methylation outside of these regions. Finally, validation of novel upregulated targets (CXCL1 and 3, GATA6, and DMBT1) in a larger independent panel of samples confirms the utility of integrative analysis in cancer biomarker discovery.


Subject(s)
Barrett Esophagus/genetics , Biomarkers, Tumor/genetics , Cell Transformation, Neoplastic/genetics , Chemokines/genetics , DNA Methylation , Esophageal Neoplasms/genetics , Gene Amplification , Barrett Esophagus/pathology , Biomarkers, Tumor/metabolism , Calcium-Binding Proteins , Cell Line, Tumor , Cell Transformation, Neoplastic/pathology , Chemokine CXCL1/genetics , Chemokine CXCL1/metabolism , Chemokines/metabolism , Chemokines, CXC/genetics , Chemokines, CXC/metabolism , DNA-Binding Proteins , Esophageal Neoplasms/pathology , GATA6 Transcription Factor/genetics , GATA6 Transcription Factor/metabolism , Gene Expression Profiling , Humans , Neoplasm Staging , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Tumor Suppressor Proteins
8.
Cancer Res ; 71(3): 955-63, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21189329

ABSTRACT

Even though myelodysplastic syndromes (MDS) are characterized by ineffective hematopoiesis, the molecular alterations that lead to marrow failure have not been well elucidated. We have previously shown that the myelosuppressive TGF-ß pathway is constitutively activated in MDS progenitors. Because there is conflicting data about upregulation of extracellular TGF-ß levels in MDS, we wanted to determine the molecular basis of TGF-ß pathway overactivation and consequent hematopoietic suppression in this disease. We observed that SMAD7, a negative regulator of TGF-ß receptor I (TBRI) kinase, is markedly decreased in a large meta-analysis of gene expression studies from MDS marrow-derived CD34(+) cells. SMAD7 protein was also found to be significantly decreased in MDS marrow progenitors when examined immunohistochemically in a bone marrow tissue microarray. Reduced expression of SMAD7 in hematopoietic cells led to increased TGF-ß-mediated gene transcription and enhanced sensitivity to TGF-ß-mediated suppressive effects. The increased TGF-ß signaling due to SMAD7 reduction could be effectively inhibited by a novel clinically relevant TBRI (ALK5 kinase) inhibitor, LY-2157299. LY-2157299 could inhibit TGF-ß-mediated SMAD2 activation and hematopoietic suppression in primary hematopoietic stem cells. Furthermore, in vivo administration of LY-2157299 ameliorated anemia in a TGF-ß overexpressing transgenic mouse model of bone marrow failure. Most importantly, treatment with LY-2157199 stimulated hematopoiesis from primary MDS bone marrow specimens. These studies demonstrate that reduction in SMAD7 is a novel molecular alteration in MDS that leads to ineffective hematopoiesis by activating of TGF-ß signaling in hematopoietic cells. These studies also illustrate the therapeutic potential of TBRI inhibitors in MDS.


Subject(s)
Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyrazoles/pharmacology , Quinolines/pharmacology , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Smad7 Protein/metabolism , Transforming Growth Factor beta/metabolism , Anemia/drug therapy , Anemia/metabolism , Anemia/pathology , Hematopoiesis/drug effects , Hematopoietic Stem Cells/metabolism , Humans , K562 Cells , Myelodysplastic Syndromes/blood , Myelodysplastic Syndromes/enzymology , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/drug effects , Transforming Growth Factor beta/antagonists & inhibitors
9.
Leuk Lymphoma ; 51(6): 1108-14, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20536349

ABSTRACT

The binding of all-trans retinoic acid (ATRA) to retinoid receptor-alpha (RAR-alpha) relieves transcriptional repression induced by the promyelocytic leukemia-retinoic acid receptor (PML-RAR) oncoprotein. The ATRA molecule contains a cyclohexenyl ring, a polyene chain containing conjugated double alkene bonds, and a terminal carboxyl group. To determine the contributions of these structural components of ATRA to its clinical efficacy, we synthesized three novel retinoids. These consisted of either a modified conjugated alkene backbone with an intact acid moiety (13a) or a modified conjugated alkene backbone and conversion of the acid group to either an ester (13b) or an aromatic amide (13c). Reporter assays demonstrated that compound 13a successfully relieved transcriptional repression by RAR-alpha, while 13b and 13c could not, demonstrating the critical role of the acid moiety in this binding. However, only ATRA was able to significantly inhibit the proliferation of APL cells while 13a, 13b, or 13c was not. Furthermore, only 13a led to partial non-significant differentiation of NB4 cells, demonstrating the importance of C9-C10 double bonds in differentiation induced CD11 expression. Our results demonstrate that both the acid moiety and conjugated double bonds present in the ATRA molecule are important for its biological activity in APL and have important implications for the design of future novel retinoids.


Subject(s)
Cell Proliferation/drug effects , Oncogene Proteins, Fusion/metabolism , Tretinoin/pharmacology , Apoptosis/drug effects , CD11 Antigens/analysis , Cell Differentiation/drug effects , Cell Line, Tumor , Drug Design , Flow Cytometry , Gene Expression Regulation/drug effects , Humans , Infant , Leukemia, Promyelocytic, Acute/genetics , Leukemia, Promyelocytic, Acute/metabolism , Leukemia, Promyelocytic, Acute/pathology , Luciferases/genetics , Luciferases/metabolism , Molecular Structure , Oncogene Proteins, Fusion/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Response Elements/genetics , Transfection , Tretinoin/chemical synthesis , Tretinoin/metabolism
10.
Melanoma Res ; 20(4): 253-65, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20418788

ABSTRACT

Malignant melanoma remains one of the most deadly human cancers with no effective cures for metastatic disease. The poor efficacy of current therapy in advanced melanoma highlights the need for better understanding of molecular mechanisms contributing to the disease. Recent work has shown that epigenetic changes, including aberrant DNA methylation, lead to alterations in gene expression and are as important in the development of malignant melanoma as the specific and well-characterized genetic events. Reversion of these methylation patterns could thus lead to a more targeted therapy and are currently under clinical investigation. The purpose of this review is to compile recent information on aberrant DNA methylation of melanoma, to highlight key genes and molecular pathways in melanoma development, which have been found to be epigenetically altered and to provide insight as to how DNA methylation might serve as targeted treatment option as well as a molecular and prognostic marker in malignant melanoma.


Subject(s)
DNA Methylation , Melanoma/genetics , Skin Neoplasms/genetics , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Melanoma/metabolism , Skin Neoplasms/metabolism
11.
Nucleic Acids Res ; 37(12): 3829-39, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19386619

ABSTRACT

Many genome-wide assays involve the generation of a subset (or representation) of the genome following restriction enzyme digestion. The use of enzymes sensitive to cytosine methylation allows high-throughput analysis of this epigenetic regulatory process. We show that the use of a dual-adapter approach allows us to generate genomic representations that includes fragments of <200 bp in size, previously not possible when using the standard approach of using a single adapter. By expanding the representation to smaller fragments using HpaII or MspI, we increase the representation by these isoschizomers to more than 1.32 million loci in the human genome, representing 98.5% of CpG islands and 91.1% of refSeq promoters. This advance allows the development of a new, high-resolution version of our HpaII-tiny fragment Enrichment by Ligation-mediated PCR (HELP) assay to study cytosine methylation. We also show that the MspI representation generates information about copy-number variation, that the assay can be used on as little as 10 ng of DNA and that massively parallel sequencing can be used as an alternative to microarrays to read the output of the assay, making this a powerful discovery platform for studies of genomic and epigenomic abnormalities.


Subject(s)
Cytosine/metabolism , DNA Methylation , DNA/analysis , Polymerase Chain Reaction/methods , Cells, Cultured , DNA/chemistry , Deoxyribonuclease HpaII , Genome, Human , Humans
12.
Leuk Lymphoma ; 49(10): 1963-75, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18949619

ABSTRACT

Myelodysplastic syndromes (MDS) are common causes of ineffective hematopoiesis and cytopenias in the elderly. Various myelosuppressive and proinflammatory cytokines have been implicated in the high rates of apoptosis and hematopoietic suppression seen in MDS. We have previously shown that p38 MAPK is overactivated in MDS hematopoietic progenitors, which led to current clinical studies of the selective p38alpha inhibitor, SCIO-469, in this disease. We now demonstrate that the myelosuppressive cytokines TNFalpha and IL-1beta are secreted by bone marrow (BM) cells in a p38 MAPK-dependent manner. Their secretion is stimulated by paracrine interactions between BM stromal and mononuclear cells and cytokine induction correlates with CD34+ stem cell apoptosis in an inflammation-simulated in vitro bone marrow microenvironment. Treatment with SCIO-469 inhibits TNF secretion in primary MDS bone marrow cells and protects cytogenetically normal progenitors from apoptosis ex vivo. Furthermore, p38 inhibition diminishes the expression of TNFalpha or IL-1beta-induced proinflammatory chemokines in BM stromal cells. These data indicate that p38 inhibition has anti-inflammatory effects on the bone marrow microenvironment that complements its cytoprotective effect on progenitor survival. These findings support clinical investigation of p38alpha as a potential therapeutic target in MDS and other related diseases characterised by inflammatory bone marrow failure.


Subject(s)
Bone Marrow/pathology , Inflammation Mediators/antagonists & inhibitors , Myelodysplastic Syndromes/pathology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Aged , Cytokines/antagonists & inhibitors , Cytokines/biosynthesis , Humans , Indoles/pharmacology , Inflammation/etiology , Interleukin-1beta/metabolism , Myelodysplastic Syndromes/drug therapy , Paracrine Communication/drug effects , Protein Kinase Inhibitors/pharmacology , Tumor Necrosis Factor-alpha/metabolism
13.
PLoS One ; 3(8): e2965, 2008 Aug 13.
Article in English | MEDLINE | ID: mdl-18698424

ABSTRACT

Microarray-based studies of global gene expression (GE) have resulted in a large amount of data that can be mined for further insights into disease and physiology. Meta-analysis of these data is hampered by technical limitations due to many different platforms, gene annotations and probes used in different studies. We tested the feasibility of conducting a meta-analysis of GE studies to determine a transcriptional signature of hematopoietic progenitor and stem cells. Data from studies that used normal bone marrow-derived hematopoietic progenitors was integrated using both RefSeq and UniGene identifiers. We observed that in spite of variability introduced by experimental conditions and different microarray platforms, our meta-analytical approach can distinguish biologically distinct normal tissues by clustering them based on their cell of origin. When studied in terms of disease states, GE studies of leukemias and myelodysplasia progenitors tend to cluster with normal progenitors and remain distinct from other normal tissues, further validating the discriminatory power of this meta-analysis. Furthermore, analysis of 57 normal hematopoietic stem and progenitor cell GE samples was used to determine a gene expression signature characteristic of these cells. Genes that were most uniformly expressed in progenitors and at the same time differentially expressed when compared to other normal tissues were found to be involved in important biological processes such as cell cycle regulation and hematopoiesis. Validation studies using a different microarray platform demonstrated the enrichment of several genes such as SMARCE, Septin 6 and others not previously implicated in hematopoiesis. Most interestingly, alpha-integrin, the only common stemness gene discovered in a recent comparative murine analysis (Science 302(5644):393) was also enriched in our dataset, demonstrating the usefulness of this analytical approach.


Subject(s)
Gene Expression Profiling/methods , Gene Expression , Hematopoietic Stem Cells/physiology , Oligonucleotide Array Sequence Analysis , Antigens, CD/analysis , Bone Marrow Cells/physiology , Cells, Cultured/pathology , Cells, Cultured/physiology , Databases, Genetic , Female , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/pathology , Humans , Male , Meta-Analysis as Topic , Organ Specificity , Reference Values
14.
Blood ; 112(8): 3434-43, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18474728

ABSTRACT

MDS is characterized by ineffective hematopoiesis that leads to peripheral cytopenias. Development of effective treatments has been impeded by limited insight into pathogenic pathways governing dysplastic growth of hematopoietic progenitors. We demonstrate that smad2, a downstream mediator of transforming growth factor-beta (TGF-beta) receptor I kinase (TBRI) activation, is constitutively activated in MDS bone marrow (BM) precursors and is overexpressed in gene expression profiles of MDS CD34(+) cells, providing direct evidence of overactivation of TGF-beta pathway in this disease. Suppression of the TGF-beta signaling by lentiviral shRNA-mediated down-regulation of TBRI leads to in vitro enhancement of hematopoiesis in MDS progenitors. Pharmacologic inhibition of TBRI (alk5) kinase by a small molecule inhibitor, SD-208, inhibits smad2 activation in hematopoietic progenitors, suppresses TGF-beta-mediated gene activation in BM stromal cells, and reverses TGF-beta-mediated cell-cycle arrest in BM CD34(+) cells. Furthermore, SD-208 treatment alleviates anemia and stimulates hematopoiesis in vivo in a novel murine model of bone marrow failure generated by constitutive hepatic expression of TGF-beta1. Moreover, in vitro pharmacologic inhibition of TBRI kinase leads to enhancement of hematopoiesis in varied morphologic MDS subtypes. These data directly implicate TGF-beta signaling in the pathobiology of ineffective hematopoiesis and identify TBRI as a potential therapeutic target in low-risk MDS.


Subject(s)
Hematopoiesis , Myelodysplastic Syndromes/metabolism , Myelodysplastic Syndromes/pathology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Aged , Aged, 80 and over , Animals , Antigens, CD34/biosynthesis , Bone Marrow/drug effects , Bone Marrow/pathology , Female , Humans , Lentivirus/genetics , Male , Mice , Mice, Transgenic , Middle Aged , Models, Biological , Protein Serine-Threonine Kinases/metabolism , Pteridines/pharmacology , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/metabolism
15.
Infect Immun ; 75(10): 4804-16, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17664267

ABSTRACT

Mycobacteria target and persist within phagocytic monocytes and are strong adjuvants, making them attractive candidate vectors for DNA vaccines. We characterized the ability of mycobacteria to deliver transgenes to mammalian cells and the effects of various bacterial chromosomal mutations on the efficiency of transfer in vivo and in vitro. First, we observed green fluorescent protein expression via microscopy and fluorescence-activated cell sorting analysis after infection of phagocytic and nonphagocytic cell lines by Mycobacterium smegmatis or M. bovis BCG harboring a plasmid encoding the fluorescence gene under the control of a eukaryotic promoter. Next, we compared the efficiencies of gene transfer using M. smegmatis or BCG containing chromosomal insertions or deletions that cause early lysis, hyperconjugation, or an increased plasmid copy number. We observed a significant-albeit only 1.7-fold-increase in the level of plasmid transfer to eukaryotic cells infected with M. smegmatis hyperconjugation mutants. M. smegmatis strains that overexpressed replication proteins (Rep) of pAL5000, a plasmid whose replicon is incorporated in many mycobacterial constructs, generated a 10-fold increase in plasmid copy number and 3.5-fold and 3-fold increases in gene transfer efficiency to HeLa cells and J774 cells, respectively. Although BCG strains overexpressing Rep could not be recovered, BCG harboring a plasmid with a copy-up mutation in oriM resulted in a threefold increase in gene transfer to J774 cells. Moreover, M. smegmatis strains overexpressing Rep enhanced gene transfer in vivo compared with a wild-type control. Immunization of mice with mycobacteria harboring a plasmid (pgp120(h)(E)) encoding human immunodeficiency virus gp120 elicited gp120-specific CD8 T-cell responses among splenocytes and peripheral blood mononuclear cells that were up to twofold (P < 0.05) and threefold (P < 0.001) higher, respectively, in strains supporting higher copy numbers. The magnitude of these responses was approximately one-half of that observed after intramuscular immunization with pgp120(h)(E). M. smegmatis and other nonpathogenic mycobacteria are promising candidate vectors for DNA vaccine delivery.


Subject(s)
Bacterial Vaccines/genetics , Mycobacterium smegmatis/genetics , Plasmids , Transformation, Genetic , Vaccination/methods , Vaccines, DNA/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Line , HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp120/immunology , Humans , Mice , Mice, Inbred BALB C , Mycobacterium bovis/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...