Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
Add more filters










Publication year range
1.
eNeuro ; 7(1)2020.
Article in English | MEDLINE | ID: mdl-31996391

ABSTRACT

Visceral sensory neurons encode distinct sensations from healthy organs and initiate pain states that are resistant to common analgesics. Transcriptome analysis is transforming our understanding of sensory neuron subtypes but has generally focused on somatic sensory neurons or the total population of neurons in which visceral neurons form the minority. Our aim was to define transcripts specifically expressed by sacral visceral sensory neurons, as a step towards understanding the unique biology of these neurons and potentially leading to identification of new analgesic targets for pelvic visceral pain. Our strategy was to identify genes differentially expressed between sacral dorsal root ganglia (DRG) that include somatic neurons and sacral visceral neurons, and adjacent lumbar DRG that comprise exclusively of somatic sensory neurons. This was performed in adult and E18.5 male and female mice. By developing a method to restrict analyses to nociceptive Trpv1 neurons, a larger group of genes were detected as differentially expressed between spinal levels. We identified many novel genes that had not previously been associated with pelvic visceral sensation or nociception. Limited sex differences were detected across the transcriptome of sensory ganglia, but more were revealed in sacral levels and especially in Trpv1 nociceptive neurons. These data will facilitate development of new tools to modify mature and developing sensory neurons and nociceptive pathways.


Subject(s)
Ganglia, Spinal , Transcriptome , Animals , Female , Male , Mice , Nociception , Nociceptors , Pain , Sensory Receptor Cells
2.
J Thromb Haemost ; 17(1): 169-182, 2019 01.
Article in English | MEDLINE | ID: mdl-30472780

ABSTRACT

Essentials Tissue factor (TF) isoforms are expressed in pancreatic neuroendocrine tumors (pNET). TF knockdown inhibits proliferation of human pNET cells in vitro. mTOR kinase inhibitor sapanisertib/MLN0128 suppresses TF expression in human pNET cells. Sapanisertib suppresses TF expression and activity and reduces the growth of pNET tumors in vivo. SUMMARY: Background Full-length tissue factor (flTF) and alternatively spliced TF (asTF) contribute to growth and spread of pancreatic ductal adenocarcinoma. It is unknown, however, if flTF and/or asTF contribute to the pathobiology of pancreatic neuroendocrine tumors (pNETs). Objective To assess TF expression in pNETs and the effects of mTOR complex 1/2 (mTORC1/2) inhibition on pNET growth. Methods Human pNET specimens were immunostained for TF. Human pNET cell lines QGP1 and BON were evaluated for TF expression and responsiveness to mTOR inhibition. shRNA were used to knock down TF in BON. TF cofactor activity was assessed using a two-step FXa generation assay. TF promoter activity was assessed using transient transfection of human TF promoter-driven reporter constructs into cells. Mice bearing orthotopic BON tumors were treated with the mTORC1/2 ATP site competitive inhibitor sapanisertib/MLN0128 (3 mg kg-1 , oral gavage) for 34 days. Results Immunostaining of pNET tissue revealed flTF and asTF expression. BON and QGP1 expressed both TF isoforms, with BON exhibiting higher levels. shRNA directed against TF suppressed BON proliferation in vitro. Treatment of BON with sapanisertib inhibited mTOR signaling and suppressed TF levels. BON tumors grown in mice treated with sapanisertib had significantly less TF protein and cofactor activity, and were smaller compared with tumors grown in control mice. Conclusions TF isoforms are expressed in pNETs. Sapanisertib suppresses TF mRNA and protein expression as well as TF cofactor activity in vitro and in vivo. Thus, further studies are warranted to evaluate the clinical utility of TF-suppressing mTORC1/2 inhibitor sapanisertib in pNET management.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Neuroendocrine Tumors/drug therapy , Pancreatic Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Thromboplastin/metabolism , Animals , Cell Line, Tumor , Humans , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Mice, Nude , Neuroendocrine Tumors/enzymology , Neuroendocrine Tumors/genetics , Neuroendocrine Tumors/pathology , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Promoter Regions, Genetic , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Thromboplastin/genetics , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
3.
Nat Commun ; 9(1): 4559, 2018 11 19.
Article in English | MEDLINE | ID: mdl-30455465

ABSTRACT

Epigenetic regulators are attractive anticancer targets, but the promise of therapeutic strategies inhibiting some of these factors has not been proven in vivo or taken into account tumor cell heterogeneity. Here we show that the histone methyltransferase G9a, reported to be a therapeutic target in many cancers, is a suppressor of aggressive lung tumor-propagating cells (TPCs). Inhibition of G9a drives lung adenocarcinoma cells towards the TPC phenotype by de-repressing genes which regulate the extracellular matrix. Depletion of G9a during tumorigenesis enriches tumors in TPCs and accelerates disease progression metastasis. Depleting histone demethylases represses G9a-regulated genes and TPC phenotypes. Demethylase inhibition impairs lung adenocarcinoma progression in vivo. Therefore, inhibition of G9a is dangerous in certain cancer contexts, and targeting the histone demethylases is a more suitable approach for lung cancer treatment. Understanding cellular context and specific tumor populations is critical when targeting epigenetic regulators in cancer for future therapeutic development.


Subject(s)
Disease Progression , Histone Demethylases/metabolism , Histone Methyltransferases/metabolism , Lung Neoplasms/metabolism , Adenocarcinoma of Lung/metabolism , Animals , Carcinogenesis , Cell Line, Tumor/drug effects , Cell Survival , Disease Models, Animal , Extracellular Matrix/genetics , Histone Demethylases/drug effects , Histone-Lysine N-Methyltransferase/drug effects , Histone-Lysine N-Methyltransferase/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Organoids/anatomy & histology , Phenotype , Proto-Oncogene Proteins p21(ras)/genetics
4.
Mol Psychiatry ; 23(2): 271-281, 2018 02.
Article in English | MEDLINE | ID: mdl-27752082

ABSTRACT

The brain-specific tyrosine phosphatase, STEP (STriatal-Enriched protein tyrosine Phosphatase) is an important regulator of synaptic function. STEP normally opposes synaptic strengthening by increasing N-methyl D-aspartate glutamate receptor (NMDAR) internalization through dephosphorylation of GluN2B and inactivation of the kinases extracellular signal-regulated kinase 1/2 and Fyn. Here we show that STEP61 is elevated in the cortex in the Nrg1+/- knockout mouse model of schizophrenia (SZ). Genetic reduction or pharmacological inhibition of STEP prevents the loss of NMDARs from synaptic membranes and reverses behavioral deficits in Nrg1+/- mice. STEP61 protein is also increased in cortical lysates from the central nervous system-specific ErbB2/4 mouse model of SZ, as well as in human induced pluripotent stem cell (hiPSC)-derived forebrain neurons and Ngn2-induced excitatory neurons, from two independent SZ patient cohorts. In these selected SZ models, increased STEP61 protein levels likely reflect reduced ubiquitination and degradation. These convergent findings from mouse and hiPSC SZ models provide evidence for STEP61 dysfunction in SZ.


Subject(s)
Protein Tyrosine Phosphatases/physiology , Schizophrenia/metabolism , Animals , Disease Models, Animal , Female , Humans , Induced Pluripotent Stem Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuregulin-1/genetics , Neurons/metabolism , Phosphorylation , Protein Tyrosine Phosphatases/genetics , Rats , Receptors, N-Methyl-D-Aspartate/metabolism , Schizophrenia/genetics , Ubiquitination
5.
Blood ; 130(10): 1213-1222, 2017 09 07.
Article in English | MEDLINE | ID: mdl-28710059

ABSTRACT

Understanding and blocking the self-renewal pathway of preleukemia stem cells could prevent acute myeloid leukemia (AML) relapse. In this study, we show that increased FOXO1 represents a critical mechanism driving aberrant self-renewal in preleukemic cells expressing the t(8;21)-associated oncogene AML1-ETO (AE). Although generally considered as a tumor suppressor, FOXO1 is consistently upregulated in t(8;21) AML. Expression of FOXO1 in human CD34+ cells promotes a preleukemic state with enhanced self-renewal and dysregulated differentiation. The DNA binding domain of FOXO1 is essential for these functions. FOXO1 activates a stem cell molecular signature that is also present in AE preleukemia cells and preserved in t(8;21) patient samples. Genome-wide binding studies show that AE and FOXO1 share the majority of their binding sites, whereby FOXO1 binds to multiple crucial self-renewal genes and is required for their activation. In agreement with this observation, genetic and pharmacological ablation of FOXO1 inhibited the long-term proliferation and clonogenicity of AE cells and t(8;21) AML cell lines. Targeting of FOXO1 therefore provides a potential therapeutic strategy for elimination of stem cells at both preleukemic and leukemic stages.


Subject(s)
Core Binding Factor Alpha 2 Subunit/metabolism , Forkhead Box Protein O1/metabolism , Gene Regulatory Networks , Leukemia, Myeloid, Acute/genetics , Oncogene Proteins, Fusion/metabolism , Precancerous Conditions/genetics , Animals , Antigens, CD34/metabolism , Cell Line, Tumor , Cell Proliferation , Core Binding Factor Alpha 2 Subunit/genetics , Gene Expression Profiling , Gene Expression Regulation, Leukemic , Genome, Human , Hematopoietic Stem Cells/metabolism , Humans , Leukemia, Myeloid, Acute/pathology , Mice, SCID , Oncogene Proteins, Fusion/genetics , Precancerous Conditions/pathology , RUNX1 Translocation Partner 1 Protein , Up-Regulation/genetics
6.
Transl Psychiatry ; 5: e662, 2015 Oct 20.
Article in English | MEDLINE | ID: mdl-26485546

ABSTRACT

The genetic and epigenetic factors contributing to risk for schizophrenia (SZ) remain unresolved. Here we demonstrate, for the first time, perturbed global protein translation in human-induced pluripotent stem cell (hiPSC)-derived forebrain neural progenitor cells (NPCs) from four SZ patients relative to six unaffected controls. We report increased total protein levels and protein synthesis, together with two independent sets of quantitative mass spectrometry evidence indicating markedly increased levels of ribosomal and translation initiation and elongation factor proteins, in SZ hiPSC NPCs. We posit that perturbed levels of global protein synthesis in SZ hiPSC NPCs represent a novel post-transcriptional mechanism that might contribute to disease progression.


Subject(s)
Induced Pluripotent Stem Cells/metabolism , Neural Stem Cells/metabolism , Neurons/metabolism , Prosencephalon/metabolism , Schizophrenia/metabolism , Cell Differentiation , Cells, Cultured , Humans
7.
Genes Immun ; 15(6): 361-9, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24920534

ABSTRACT

Eosinophilic esophagitis (EoE) is an allergic inflammatory disorder of the esophagus that is compounded by genetic predisposition and hypersensitivity to environmental antigens. Using high-density oligonucleotide expression chips, a disease-specific esophageal transcript signature was identified and was shown to be largely reversible with therapy. In an effort to expand the molecular signature of EoE, we performed RNA sequencing on esophageal biopsies from healthy controls and patients with active EoE and identified a total of 1607 significantly dysregulated transcripts (1096 upregulated, 511 downregulated). When clustered by raw expression levels, an abundance of immune cell-specific transcripts are highly induced in EoE but expressed at low (or undetectable) levels in healthy controls. Moreover, 66% of the gene signature identified by RNA sequencing was previously unrecognized in the EoE transcript signature by microarray-based expression profiling and included several long non-coding RNAs (lncRNA), an emerging class of transcriptional regulators. The lncRNA BRAF-activated non-protein coding RNA (BANCR) was upregulated in EoE and induced in interleukin-13 (IL-13)-treated primary esophageal epithelial cells. Repression of BANCR significantly altered the expression of IL-13-induced proinflammatory genes. Together, these data comprise new potential biomarkers of EoE and demonstrate a novel role for lncRNAs in EoE and IL-13-associated responses.


Subject(s)
Eosinophilic Esophagitis/genetics , Oligonucleotide Array Sequence Analysis/methods , Sequence Analysis, RNA/methods , Transcriptome , Cell Line , Cells, Cultured , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Interleukin-13/pharmacology , RNA Interference , RNA, Untranslated/genetics , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
8.
Placenta ; 34(12): 1150-8, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24119485

ABSTRACT

OBJECTIVE: We tested the effects of selective reduction of placental blood flow by mesenteric uterine artery branch ligation (MUAL) resulting in fetal growth restriction (FGR). METHODS: Timed mated C57BL/6J Day(D) 18 dams were divided into two groups: MUAL (n = 18); and control-sham (n = 18). Pups were delivered on D20, cross-fostered to surrogate CD-1 mothers for 4 weeks, and followed for 8 weeks. Outcome data included birth and placental weight, postnatal growth, placental volume determined by stereology, quantification of placental insulin-like growth factors-1(IGF-1) and IGF-2 and IGF binding proteins(IGFBP 2 and 6) by ELISA and gene expression by qPCR and GeneChip microarray analysis. RESULTS: Compared with control, MUAL had an 11% reduction in mean birth weight (1.06 ± 0.13 g vs. 0.94 ± 0.13 g, p < 0.001) but no difference in placental weight. At 4 weeks of age, mean body weights of MUAL pups were significantly lower than sham. By 8 weeks, males but not females MUAL mice achieved equivalent mean body weight to control. Placental labyrinth depth, volume, and placental gene expression of IGF-1 and 2 were significantly reduced by MUAL. In contrast, placental protein level of IGFBP-2 and 6 were significantly elevated in the MUAL. Genomic expression analysis demonstrated that MUAL pups significantly up-regulated genes that were associated with apoptosis and growth pathways. CONCLUSION: This novel mouse animal model of FGR using selective ligation recapitulates multiple characteristics of placental vascular insufficiency (PI) in humans. This is the first non-genetic mouse model of PI which offers its application in transgenic mice to better study the underlying mechanisms in PI. CONDENSATION: A new mouse model of placental vascular insufficiency by selective ligation of mesenteric uterine artery branch recapitulates multiple findings observed in human placental vascular insufficiency.


Subject(s)
Disease Models, Animal , Fetal Growth Retardation/etiology , Placenta/physiopathology , Placental Circulation , Placental Insufficiency/physiopathology , Animals , Birth Weight , Female , Gene Expression Regulation, Developmental , Insulin-Like Growth Factor Binding Proteins/genetics , Insulin-Like Growth Factor Binding Proteins/metabolism , Ligation , Male , Mice , Mice, Inbred C57BL , Placenta/metabolism , Placenta/pathology , Placental Insufficiency/metabolism , Placental Insufficiency/pathology , Placentation , Pregnancy , Sex Characteristics , Somatomedins/genetics , Somatomedins/metabolism , Uterine Artery/surgery , Weight Gain
9.
Mucosal Immunol ; 5(4): 388-96, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22453679

ABSTRACT

Interleukin 13 (IL-13)-induced epithelial gene and protein expression changes are central to the pathogenesis of multiple allergic diseases. Herein, using human esophageal squamous and bronchial columnar epithelial cells, we identified microRNAs (miRNAs) that were differentially regulated after IL-13 stimulation. Among the IL-13-regulated miRNAs, miR-375 showed a conserved pattern of downregulation. Furthermore, miR-375 was downregulated in the lung of IL-13 lung transgenic mice. We subsequently analyzed miR-375 levels in a human disease characterized by IL-13 overproduction--the allergic disorder eosinophilic esophagitis (EE)--and observed downregulation of miR-375 in EE patient samples compared with control patients. MiR-375 expression levels reflected disease activity, normalized with remission, and inversely correlated with the degree of allergic inflammation. Using a lentiviral strategy and whole-transcriptome analysis in epithelial cells, miR-375 overexpression was sufficient to markedly modify IL-13-associated immunoinflammatory pathways in epithelial cells in vitro, further substantiating interactions between miR-375 and IL-13. Taken together, our results support a key role of miRNAs, particularly miR-375, in regulating and fine-tuning IL-13-mediated responses.


Subject(s)
Epithelial Cells/drug effects , Epithelial Cells/metabolism , Gene Expression Regulation/drug effects , Immunologic Factors/pharmacology , Interleukin-13/pharmacology , MicroRNAs/genetics , Transcriptome , Animals , Cell Line , Cluster Analysis , Eosinophilic Esophagitis/genetics , Esophagus/metabolism , Gene Expression Profiling , Humans , Mice , Mice, Transgenic , Respiratory Mucosa/drug effects , Respiratory Mucosa/metabolism
10.
J Thromb Haemost ; 9(10): 2087-96, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21812913

ABSTRACT

BACKGROUND: TF is highly expressed in cancerous and atherosclerotic lesions. Monocyte recruitment is a hallmark of disease progression in these pathological states. OBJECTIVE: To examine the role of integrin signaling in TF-dependent recruitment of monocytes by endothelial cells. METHODS: The expression of flTF and asTF in cervical cancer and atherosclerotic lesions was examined. Biologic effects of the exposure of primary microvascular endothelial cells (MVEC) to truncated flTF ectodomain (LZ-TF) and recombinant asTF were assessed. RESULTS: flTF and asTF exhibited nearly identical expression patterns in cancer lesions and lipid-rich plaques. Tumor lesions, as well as stromal CD68(+) monocytes/macrophages, expressed both TF forms. Primary MVEC rapidly adhered to asTF and LZ-TF, and this was completely blocked by anti-ß1 integrin antibody. asTF- and LZ-TF-treatment of MVEC promoted adhesion of peripheral blood mononuclear cells (PBMCs) under orbital shear conditions and under laminar flow; asTF-elicited adhesion was more pronounced than that elicited by LZ-TF. Expression profiling and western blotting revealed a broad activation of cell adhesion molecules (CAMs) in MVEC following asTF treatment including E-selectin, ICAM-1 and VCAM-1. In transwell assays, asTF potentiated PMBC migration through MVEC monolayers by ∼3-fold under MCP-1 gradient. CONCLUSIONS: TF splice variants ligate ß1 integrins on MVEC, which induces the expression of CAMs in MVEC and leads to monocyte adhesion and transendothelial migration. asTF appears more potent than flTF in eliciting these effects. Our findings underscore the pathophysiologic significance of non-proteolytic, integrin-mediated signaling by the two naturally occurring TF variants in cancer and atherosclerosis.


Subject(s)
Alternative Splicing , Cell Adhesion Molecules/metabolism , Endothelium, Vascular/cytology , Integrins/metabolism , Monocytes/cytology , Signal Transduction , Thromboplastin/genetics , Blotting, Western , Cells, Cultured , Female , Humans , Reverse Transcriptase Polymerase Chain Reaction , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology
11.
Neuroscience ; 155(3): 714-24, 2008 Aug 26.
Article in English | MEDLINE | ID: mdl-18620027

ABSTRACT

Clusterin (or apolipoprotein J) is a widely distributed multifunctional glycoprotein involved in CNS plasticity and post-traumatic remodeling. Using biochemical and morphological approaches, we investigated the clusterin ontogeny in the CNS of wild-type (WT) mice and explored developmental consequences of clusterin gene knock-out in clusterin null (Clu-/-) mice. A punctiform expression of clusterin mRNA was detected through the hypothalamic region, neocortex and hippocampus at embryonic stages E14/E15. From embryonic stage E16 to the first week of the postnatal life, the vast majority of CNS neurons expressed low levels of clusterin mRNA. In contrast, a very strong hybridizing signal mainly localized in pontobulbar and spinal cord motor nuclei was observed from the end of the first postnatal week to adulthood. Astrocytes expressing clusterin mRNA were often detected through the hippocampus and neocortex in neonatal mice. Real-time polymerase chain amplification and clusterin-immunoreactivity dot-blot analyses indicated that clusterin levels paralleled mRNA expression. Comparative analyses between WT and Clu-/- mice during postnatal development showed no significant differences in brain weight, neuronal, synaptic and astrocyte markers as well myelin basic protein expression. However, quantitative estimation of large motor neuron populations in the facial nucleus revealed a significant deficit in motor cells (-16%) in Clu-/- compared with WT mice. Our data suggest that clusterin expression is already present in fetal life mainly in subcortical structures. Although the lack of this protein does not significantly alter basic aspects of the CNS development, it may have a negative impact on neuronal development in certain motor nuclei.


Subject(s)
Central Nervous System , Clusterin/metabolism , Gene Expression Regulation, Developmental/physiology , Age Factors , Animals , Animals, Newborn , Central Nervous System/embryology , Central Nervous System/growth & development , Central Nervous System/metabolism , Clusterin/deficiency , Clusterin/genetics , Embryo, Mammalian , Gene Expression Regulation, Developmental/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , RNA, Messenger/metabolism
12.
Cancer Gene Ther ; 15(11): 733-41, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18551144

ABSTRACT

Oncolytic herpes simplex viruses (oHSVs) are promising anticancer therapeutics. We sought to characterize the functional genomic response of human cancer cells to oHSV infection using G207, an oHSV previously evaluated in a phase I trial. Five human malignant peripheral nerve sheath tumor cell lines, with differing sensitivity to oHSV, were infected with G207 for 6 h. Functional genomic analysis of virus-infected cells demonstrated large clusters of downregulated cellular mRNAs and smaller clusters of those upregulated, including 21 genes commonly upregulated in all five lines. Of these, 7 are known to be HSV-1 induced and 14 represent novel virus-regulated genes. Gene ontology analysis revealed that a majority of G207-upregulated genes are involved in Janus kinase/signal transducer and activator of transcription signaling, transcriptional regulation, nucleic acid metabolism, protein synthesis and apoptosis. Ingenuity networks highlighted nodes for AP-1 subunits and interferon signaling via STAT1, suppressor of cytokine signaling-1 (SOCS1), SOCS3 and RANTES. As biological confirmation, we found that virus-mediated upregulation of SOCS1 correlated with sensitivity to G207 and that depletion of SOCS1 impaired virus replication by >10-fold. Further characterization of roles provided by oHSV-induced cellular genes during virus replication may be utilized to predict oncolytic efficacy and to provide rational strategies for designing next-generation oncolytic viruses.


Subject(s)
Gene Expression Regulation, Neoplastic/genetics , Herpesvirus 1, Human/physiology , Nerve Sheath Neoplasms/therapy , Nerve Sheath Neoplasms/virology , Oncolytic Virotherapy/methods , Suppressor of Cytokine Signaling Proteins/metabolism , Virus Replication/physiology , Blotting, Western , Herpesvirus 1, Human/genetics , Humans , Oligonucleotide Array Sequence Analysis , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Virus Replication/genetics
13.
Oncogene ; 26(43): 6307-18, 2007 Sep 20.
Article in English | MEDLINE | ID: mdl-17452985

ABSTRACT

Functional inactivation of the retinoblastoma tumor suppressor gene product (RB) is a common event in human cancers. Classically, RB functions to constrain cellular proliferation, and loss of RB is proposed to facilitate the hyperplastic proliferation associated with tumorigenesis. To understand the repertoire of regulatory processes governed by RB, two models of RB loss were utilized to perform microarray analysis. In murine embryonic fibroblasts harboring germline loss of RB, there was a striking deregulation of gene expression, wherein distinct biological pathways were altered. Specifically, genes involved in cell cycle control and classically associated with E2F-dependent gene regulation were upregulated via RB loss. In contrast, a program of gene expression associated with immune function and response to pathogens was significantly downregulated with the loss of RB. To determine the specific influence of RB loss during a defined period and without the possibility of developmental compensation as occurs in embryonic fibroblasts, a second system was employed wherein Rb was acutely knocked out in adult fibroblasts. This model confirmed the distinct regulation of cell cycle and immune modulatory genes through RB loss. Analyses of cis-elements supported the hypothesis that the majority of those genes upregulated with RB loss are regulated via the E2F family of transcription factors. In contrast, those genes whose expression was reduced with the loss of RB harbored different promoter elements. Consistent with these analyses, we found that disruption of E2F-binding function of RB was associated with the upregulation of gene expression. In contrast, cells harboring an RB mutant protein (RB-750F) that retains E2F-binding activity, but is specifically deficient in the association with LXCXE-containing proteins, failed to upregulate these same target genes. However, downregulation of genes involved in immune function was readily observed with disruption of the LXCXE-binding function of RB. Thus, these studies demonstrate that RB plays a significant role in both the positive and negative regulations of transcriptional programs and indicate that loss of RB has distinct biological effects related to both cell cycle control and immune function.


Subject(s)
Cell Cycle , Retinoblastoma Protein/deficiency , Retinoblastoma Protein/metabolism , Retinoblastoma/metabolism , Retinoblastoma/pathology , Transcription, Genetic/genetics , Animals , Cells, Cultured , Down-Regulation , E2F Transcription Factors/genetics , E2F Transcription Factors/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Mice , Mice, Knockout , Promoter Regions, Genetic/genetics , Retinoblastoma/genetics , Retinoblastoma/immunology , Retinoblastoma Protein/genetics , Up-Regulation
14.
Neuroreport ; 15(11): 1735-8, 2004 Aug 06.
Article in English | MEDLINE | ID: mdl-15257138

ABSTRACT

Clusterin accumulates in significant quantity in prion protein lesions associated with bovine spongiform encephalopathy (BSE) and we therefore sought to elucidate its ability to alter BSE pathogenesis and incubation time by comparison of wild type C57BL/6J mice and clusterin knock out (ko) mice. The ko mice had a 40 day increase in mean incubation time compared to wild type mice. PrP deposition in the medulla was less aggregated in clusterin knock out mice when compared to wild type BSE infected mice and a more marked astrocytosis, as determined by GFAP staining, was evident. The vacuolation profiles did not differ between the two strains of mice. Taken together these results suggest that clusterin alters the extracellular deposition of PrP(BSE) and accelerates BSE pathogenesis.


Subject(s)
Encephalopathy, Bovine Spongiform/metabolism , Encephalopathy, Bovine Spongiform/pathology , Glycoproteins/physiology , Molecular Chaperones/physiology , Animals , Cattle , Clusterin , Female , Glycoproteins/deficiency , Glycoproteins/genetics , Male , Medulla Oblongata/metabolism , Medulla Oblongata/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Chaperones/genetics
15.
Rheumatology (Oxford) ; 43(8): 973-9, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15150433

ABSTRACT

OBJECTIVE: To evaluate the ability of microarray-based methods to identify genes with disease-specific expression patterns in peripheral blood mononuclear cells (PBMC) and synovial fluid mononuclear cells (SFMC) of juvenile arthritis patients and healthy controls. METHODS: Microarray data (Affymetrix U95Av2) from 26 PBMC and 20 SFMC samples collected from patients with active disease (classified by course according to ACR criteria) were analysed for expression patterns that correlated with disease characteristics. For comparison, PBMC gene expression profiles were obtained from 15 healthy controls. Real-time PCR was used for confirmation of gene expression differences. RESULTS: Statistical analysis of gene expression patterns in PBMC identified 378 probe sets corresponding to 342 unique genes with differing expression levels between polyarticular course patients and controls (t test, P<0.0001). The genes represented by these probe sets were enriched for functions related to regulation of immune cell functions, receptor signalling as well as protein metabolism and degradation. Included in these probe sets were a group of CXCL chemokines with functions related to angiogenesis. Further analysis showed that, whereas angiogenic CXCL (ELR+) gene expression was elevated in polyarticular PBMC, expression of angiostatic CXCL (ELR-) chemokines was lower in polyarticular SFMC compared with corresponding pauciarticular samples (t test, P<0.05). CONCLUSIONS: This pilot study demonstrates that juvenile arthritis patients exhibit complex patterns of gene expression in PBMC and SFMC. The presence of disease-correlated biologically relevant gene expression patterns suggests that the power of this approach will allow better understanding of disease mechanisms, identify distinct clinical phenotypes in disease subtypes, and suggest new therapeutic approaches.


Subject(s)
Arthritis, Juvenile/genetics , Chemokines, CXC/genetics , Gene Expression/genetics , Leukocytes, Mononuclear/physiology , Spondylarthropathies/genetics , Synovial Fluid/physiology , Adolescent , Adult , Cells, Cultured , Child , Gene Expression Profiling/methods , Humans , Neovascularization, Pathologic/genetics , Oligonucleotide Array Sequence Analysis/methods , Pilot Projects , Protein-Tyrosine Kinases/genetics , Retrospective Studies , Signal Transduction/genetics , Trans-Activators/genetics
16.
J Biol Chem ; 276(45): 42057-62, 2001 Nov 09.
Article in English | MEDLINE | ID: mdl-11572852

ABSTRACT

Many of the functions ascribed to p53 tumor suppressor protein are mediated through transcription regulation. We have shown that p53 represses hepatic-specific alpha-fetoprotein (AFP) gene expression by direct interaction with a composite HNF-3/p53 DNA binding element. Using solid-phase, chromatin-assembled AFP DNA templates and analysis of chromatin structure and transcription in vitro, we find that p53 binds DNA and alters chromatin structure at the AFP core promoter to regulate transcription. Chromatin assembled in the presence of hepatoma extracts is activated for AFP transcription with an open, accessible core promoter structure. Distal (-850) binding of p53 during chromatin assembly, but not post-assembly, reverses transcription activation concomitant with promoter inaccessibility to restriction enzyme digestion. Inhibition of histone deacetylase activity by trichostatin-A (TSA) addition, prior to and during chromatin assembly, activated chromatin transcription in parallel with increased core promoter accessibility. Chromatin immunoprecipitation analyses showed increased H3 and H4 acetylated histones at the core promoter in the presence of TSA, while histone acetylation remained unchanged at the site of distal p53 binding. Our data reveal that p53 targets chromatin structure alteration at the core promoter, independently of effects on histone acetylation, to establish repressed AFP gene expression.


Subject(s)
Chromatin/chemistry , Repressor Proteins/physiology , Tumor Suppressor Protein p53/physiology , alpha-Fetoproteins/genetics , Acetylation , DNA/metabolism , Histones/metabolism , Promoter Regions, Genetic
18.
Physiol Genomics ; 6(2): 105-16, 2001 Jul 17.
Article in English | MEDLINE | ID: mdl-11459926

ABSTRACT

Placental development results from a highly dynamic differentiation program. We used DNA microarray analysis to characterize the process by which human cytotrophoblast cells differentiate into syncytiotrophoblast cells in a purified cell culture system. Of 6,918 genes analyzed, 141 genes were induced and 256 were downregulated by more than 2-fold. Dynamically regulated genes were divided by the K-means algorithm into 9 kinetic pattern groups, then by biologic classification into 6 overall functional categories: cell and tissue structural dynamics, cell cycle and apoptosis, intercellular communication, metabolism, regulation of gene expression, and expressed sequence tag (EST) and function unknown. Gene expression changes within key functional categories were tightly coupled to morphological changes. In several key gene function categories, such as cell and tissue structure, many gene members of the category were strongly activated while others were strongly repressed. These findings suggest that differentiation is augmented by "categorical reprogramming" in which the function of induced genes is enhanced by preventing the further synthesis of categorically related gene products.


Subject(s)
Transcription, Genetic , Trophoblasts/physiology , Cell Differentiation , Cells, Cultured , Chorionic Gonadotropin/pharmacology , Female , Gene Expression Profiling , Gene Expression Regulation , Humans , Kinetics , Models, Biological , Oligonucleotide Array Sequence Analysis , RNA, Messenger/biosynthesis , Trophoblasts/drug effects
19.
Physiol Genomics ; 6(1): 19-28, 2001 Jun 06.
Article in English | MEDLINE | ID: mdl-11395543

ABSTRACT

To define molecular mechanisms of cardiac hypertrophy, genes whose expression was perturbed by any of four different transgenic mouse hypertrophy models [protein kinase C-epsilon activation peptide (PsiepsilonRACK), calsequestrin (CSQ), calcineurin (CN), and Galpha(q)] were compared by DNA microarray analyses using the approximately 8,800 genes present on the Incyte mouse GEM1. The total numbers of regulated genes (tens to hundreds) correlated with phenotypic severity of the model (Galpha(q) > CN > CSQ > PsiepsilonRACK), but demonstrated that no single gene was consistently upregulated. Of the three models exhibiting pathological hypertrophy, only atrial natriuretic peptide was consistently upregulated, suggesting that transcriptional alterations are highly specific to individual genetic causes of hypertrophy. However, hierarchical-tree and K-means clustering analyses revealed that subsets of the upregulated genes did exhibit coordinate regulatory patterns that were unique or overlapping across the different hypertrophy models. One striking set consisted of apoptotic genes uniquely regulated in the apoptosis-prone Galpha(q) model. Thus, rather than identifying a single common hypertrophic cardiomyopathy gene program, these data suggest that extensive groups of genes may be useful for the prediction of specific underlying genetic determinants and condition-specific therapeutic approaches.


Subject(s)
Cardiomegaly/genetics , Animals , Apoptosis/genetics , Calcineurin/genetics , Calsequestrin/genetics , Cardiomegaly/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11 , Gene Expression Profiling , Heterotrimeric GTP-Binding Proteins/genetics , Isoenzymes/metabolism , Mice , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Protein Kinase C/metabolism , Protein Kinase C-epsilon , RNA, Messenger/biosynthesis , Transcription, Genetic , Up-Regulation
20.
Nat Med ; 7(3): 338-43, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11231633

ABSTRACT

Clusterin, also known as apolipoprotein J, is a ubiquitously expressed molecule thought to influence a variety of processes including cell death. In the brain, it accumulates in dying neurons following seizures and hypoxic-ischemic (H-I) injury. Despite this, in vivo evidence that clusterin directly influences cell death is lacking. Following neonatal H-I brain injury in mice (a model of cerebral palsy), there was evidence of apoptotic changes (neuronal caspase-3 activation), as well as accumulation of clusterin in dying neurons. Clusterin-deficient mice had 50% less brain injury following neonatal H-I. Surprisingly, the absence of clusterin had no effect on caspase-3 activation, and clusterin accumulation and caspase-3 activation did not colocalize to the same cells. Studies with cultured cortical neurons demonstrated that exogenous purified astrocyte-secreted clusterin exacerbated oxygen/glucose-deprivation-induced necrotic death. These results indicate that clusterin may be a new therapeutic target to modulate non-caspase-dependent neuronal death following acute brain injury.


Subject(s)
Brain/pathology , Caspases/metabolism , Glycoproteins/physiology , Hypoxia-Ischemia, Brain/pathology , Molecular Chaperones/physiology , Animals , Animals, Newborn , Blotting, Western , Caspase 3 , Cell Death/physiology , Clusterin , Fluorescent Antibody Technique , Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Immunoelectron , Molecular Chaperones/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...