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










Publication year range
1.
bioRxiv ; 2024 Jan 08.
Article in English | MEDLINE | ID: mdl-38260325

ABSTRACT

Type I interferon (IFN) is induced in virus infected cells, secreted and it inhibits viral replication in neighboring cells. IFN is also an important player in many non-viral diseases and in the development of normal immune cells. Although the signaling pathways for IFN induction by viral RNA or DNA have been extensively studied, its mode of induction in uninfected cells remains obscure. Here, we report that inflammatory cytokines, such as TNF-α and IL-1ß, can induce IFN-ß through activation of the cytoplasmic RIG-I signaling pathway. However, RIG-I is activated not by RNA, but by PACT, the protein activator of PKR. In cell lines or primary cells expressing RIG-I and PACT, activation of the MAPK, p38, by cytokine signaling, leads to phosphorylation of PACT, which binds to primed RIG-I and activates its signaling pathway. Thus, a new mode of type I IFN induction by ubiquitous inflammatory cytokines has been revealed. Key points: Cytochalasin D followed by TNF-α / IL-1ß treatment activates IFN-ß expression.IFN-ß expression happens due to activation of RIG-I signaling.Interaction between RIG-I and PACT activates IFN-ß expression.

2.
J Virol ; 97(12): e0130423, 2023 Dec 21.
Article in English | MEDLINE | ID: mdl-37982645

ABSTRACT

IMPORTANCE: Interferon-stimulated genes (ISGs) are induced in response to interferon expression due to viral infections. Role of these ISGs can be variable in different cells or organs. Our study highlights such cell-specific role of an ISG, Ddx3, which regulates the translation of mRNAs essential for interferon induction (PACT) and interferon signaling (STAT1) in a cell-specific manner. Our study also highlights the role of PACT in RNA virus-induced RLR signaling. Our study depicts how Ddx3 regulates innate immune signaling pathways in an indirect manner. Such cell-specific behavior of ISGs helps us to better understand viral pathogenesis and highlights the complexities of viral tropism and innate immune responses.


Subject(s)
Immunity, Innate , Interferons , RNA Viruses , Immunity, Innate/immunology , Interferons/biosynthesis , Interferons/immunology , RNA Viruses/immunology , RNA Viruses/pathogenicity , Signal Transduction , Humans , Animals , Mice
3.
Traffic ; 24(12): 576-586, 2023 12.
Article in English | MEDLINE | ID: mdl-37658794

ABSTRACT

To combat microbial infections, mammalian cells use a variety of innate immune response pathways to induce synthesis of anti-microbial proteins. The cGAS/STING pathway recognizes cytoplasmic viral or cellular DNA to elicit signals that lead to type I interferon and other cytokine synthesis. cGAMP, synthesized by DNA-activated cGAS, activates the ER-associated protein, STING, which oligomerizes and translocates to other intracellular membrane compartments to trigger different branches of signaling. We have reported that, in the ER, EGFR-mediated phosphorylation of Tyr245 of STING is required for its transit to the late endosomes, where it recruits and activates the transcription factor IRF3 required for IFN induction. In the current study, we inquired whether STING Tyr245 phosphorylation per se or STING's location in the late endosomes was critical for its ability to recruit IRF3 and induce IFN. Using pharmacological inhibitors or genetic ablation of proteins that are essential for specific steps of STING trafficking, we demonstrated that the presence of STING in the late endosomal membranes, even without Tyr245 phosphorylation, was sufficient for IRF3-mediated IFN induction.


Subject(s)
Interferons , Protein Serine-Threonine Kinases , Animals , Protein Serine-Threonine Kinases/genetics , Membrane Proteins/metabolism , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Immunity, Innate/genetics , DNA , Endosomes/metabolism , Mammals/genetics , Mammals/metabolism
4.
J Interferon Cytokine Res ; 43(5): 189-193, 2023 05.
Article in English | MEDLINE | ID: mdl-37093156

ABSTRACT

Many pattern recognition receptors in mammalian cells initiate signaling processes that culminate in mounting an innate protective response mediated by induced synthesis of a large number of proteins including type I interferons and other cytokines. Many of these receptors are not located on the plasma membrane but on the membranes of intracellular organelles such as endosomes, mitochondria, and the endoplasmic reticulum; they primarily recognize microbial or cellular nucleic acids. In the course of biochemical analyses of the signaling pathways triggered by these receptors, we discovered that they require tyrosine phosphorylation by the protein kinase activity of the epidermal growth factor receptor (EGFR), which is located not only on the plasma membrane but also on the intracellular membranes. Here, we discuss how specific members of this family of receptors, such as TLR3, TLR9, or STING, interact with EGFR and other protein tyrosine kinases and what are the functional consequences of their post-translational modifications. The article highlights an unexpected functional link between a growth factor receptor and cellular innate immune response.


Subject(s)
ErbB Receptors , Tyrosine , Animals , Phosphorylation , Tyrosine/metabolism , ErbB Receptors/metabolism , Protein-Tyrosine Kinases/metabolism , Cytokines/metabolism , Protein Processing, Post-Translational , Receptors, Pattern Recognition/metabolism , Mammals/metabolism
5.
mBio ; 12(6): e0322821, 2021 12 21.
Article in English | MEDLINE | ID: mdl-34933455

ABSTRACT

The nature and the intensity of innate immune response to virus infection determine the course of pathogenesis in the host. Among the many pathogen-associated molecular pattern recognition receptors, STING, an endoplasmic reticulum (ER)-associated protein, plays a pivotal role in triggering responses to microbial or cellular cytoplasmic DNA. Herpes simplex virus 1 (HSV-1), a common human pathogen, activates STING signaling, and the resultant induction of type I interferon causes inhibition of virus replication. In this context, we have observed that phosphorylation of Tyr245 of STING by epidermal growth factor receptor kinase is necessary for interferon induction. Here, we report that phosphorylation of Tyr240 by the tyrosine kinase Syk is essential for all signaling activities of STING. Our analysis showed that upon ligand-binding, STING dimerizes and interacts with membrane-bound EGFR, which autophosphorylates and provides the platform for the recruitment of cytoplasmic Syk to the signaling complex and its activation. Activated Syk phosphorylates Tyr240 of STING, followed by phosphorylation of Tyr245 by epidermal growth factor receptor (EGFR). Pharmacological or genetic ablation of Syk activity resulted in an arrest of STING in the ER compartment and a complete block of gene induction. Consequently, in the absence of Syk, HSV-1 could not induce interferon, and it replicated more robustly. IMPORTANCE The innate immune response to virus infection leads to interferon production and inhibition of viral replication. STING, an ER-bound protein, mediates such a response to cytoplasmic cellular or microbial DNA. HSV-1, a DNA virus, activates STING, and it replicates more efficiently in the absence of STING signaling. We demonstrate that phosphorylation of Tyr240 of STING by the protein tyrosine kinase Syk is essential for STING-mediated gene induction. To signal, ligand-activated STING recruits two kinases, Syk and EGFR, which phosphorylate Tyr240 and Tyr245, respectively. The dependence of STING signaling on Syk has broad significance, because STING plays a major role in many microbial, mitochondrial, and autoimmune diseases as well as in cancer development and therapy.


Subject(s)
Herpes Simplex/metabolism , Herpesvirus 1, Human/physiology , Interferon-beta/metabolism , Membrane Proteins/metabolism , Syk Kinase/metabolism , Amino Acid Motifs , ErbB Receptors/genetics , ErbB Receptors/metabolism , Herpes Simplex/genetics , Herpes Simplex/virology , Herpesvirus 1, Human/genetics , Humans , Interferon-beta/genetics , Membrane Proteins/chemistry , Membrane Proteins/genetics , Phosphorylation , Syk Kinase/genetics , Virus Replication
6.
PLoS Pathog ; 17(9): e1009950, 2021 09.
Article in English | MEDLINE | ID: mdl-34591940

ABSTRACT

STING is a nodal point for cellular innate immune response to microbial infections, autoimmunity and cancer; it triggers the synthesis of the antiviral proteins, type I interferons. Many DNA viruses, including Herpes Simplex Virus 1 (HSV1), trigger STING signaling causing inhibition of virus replication. Here, we report that HSV1 evades this antiviral immune response by inducing a cellular microRNA, miR-24, which binds to the 3' untranslated region of STING mRNA and inhibits its translation. Expression of the gene encoding miR-24 is induced by the transcription factor AP1 and activated by MAP kinases in HSV1-infected cells. Introduction of exogenous miR-24 or prior activation of MAPKs, causes further enhancement of HSV1 replication in STING-expressing cells. Conversely, transfection of antimiR-24 inhibits virus replication in those cells. HSV1 infection of mice causes neuropathy and death; using two routes of infection, we demonstrated that intracranial injection of antimiR-24 alleviates both morbidity and mortality of the infected mice. Our studies reveal a new immune evasion strategy adopted by HSV1 through the regulation of STING and demonstrates that it can be exploited to enhance STING's antiviral action.


Subject(s)
Herpes Simplex/immunology , Immune Evasion/immunology , Membrane Proteins/immunology , MicroRNAs/immunology , Animals , Gene Expression Regulation/immunology , Herpes Simplex/metabolism , Herpesvirus 1, Human/immunology , Herpesvirus 1, Human/metabolism , Host-Pathogen Interactions/immunology , Humans , Immunity, Cellular/immunology , Membrane Proteins/metabolism , Mice
7.
EMBO J ; 39(22): e104106, 2020 11 16.
Article in English | MEDLINE | ID: mdl-32926474

ABSTRACT

STING (STimulator of INterferon Genes) mediates protective cellular response to microbial infection and tissue damage, but its aberrant activation can lead to autoinflammatory diseases. Upon ligand stimulation, the endoplasmic reticulum (ER) protein STING translocates to endosomes for induction of interferon production, while an alternate trafficking route delivers it directly to the autophagosomes. Here, we report that phosphorylation of a specific tyrosine residue in STING by the epidermal growth factor receptor (EGFR) is required for directing STING to endosomes, where it interacts with its downstream effector IRF3. In the absence of EGFR-mediated phosphorylation, STING rapidly transits into autophagosomes, and IRF3 activation, interferon production, and antiviral activity are compromised in cell cultures and mice, while autophagic activity is enhanced. Our observations illuminate a new connection between the tyrosine kinase activity of EGFR and innate immune functions of STING and suggest new experimental and therapeutic approaches for selective regulation of STING functions.


Subject(s)
ErbB Receptors/metabolism , Immunity, Innate , Membrane Proteins/metabolism , Protein Transport/physiology , Tyrosine/metabolism , Animals , Cell Line , Endoplasmic Reticulum/metabolism , Endosomes/metabolism , ErbB Receptors/genetics , Gene Knockdown Techniques , HeLa Cells , Humans , Immunity, Innate/genetics , Interferon Regulatory Factor-3/genetics , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Phosphorylation , RAW 264.7 Cells , Signal Transduction , Transcriptome
8.
J Virol ; 94(6)2020 02 28.
Article in English | MEDLINE | ID: mdl-31896590

ABSTRACT

The innate immune response is crucial for defense against viral infections. Cells recognize virus infection through pattern recognition receptors and induce type I interferons as well as proinflammatory cytokines to orchestrate an innate immune response. Herpes simplex virus 1 (HSV-1) triggers both the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) and Toll-like receptor 3 (TLR3) pathways. It is well known that TLR3 uses the adaptor protein Toll/interleukin-1 receptor (IL-1R) domain-containing adaptor-inducing beta interferon (TRIF) for signaling, but we recently reported that STING signaling also requires TRIF. Because STING directly binds to TRIF, we identified the STING-interacting domain of TRIF and generated STING-noninteracting mutants of human and mouse TRIFs. The mutant TRIFs were unable to support STING signaling, although they were fully functional in the TLR3 pathway. These mutants were used to assess the relative contributions of the TLR3 and STING pathways to the attenuation of HSV-1 replication in mouse and human cell lines. For this purpose, the mouse L929 and NB41A3 cell lines and the human HT1080 and HeLa-M cell lines, in which both the TLR3 and the STING pathways are operational, were used. The TRIF gene was disrupted in these lines by CRISPR/Cas9, before reconstituting them with mutant and wild-type TRIF expression vectors. Infection of the reconstituted cells with HSV-1 revealed that both the cGAS-STING and the TLR3 signaling pathways are required for the attenuation of virus replication, but their relative contributions in attenuating HSV-1 replication were found to be different in mouse versus human cell lines. Thus, our study suggests that the relative contributions of the cGAS-STING and the TLR3 pathways in the attenuation of viral infection may be species specific.IMPORTANCE The magnitude of fatal infections caused by all different viruses in human and animal populations justifies a better understanding of the host innate immune response process that attenuates virus replication. In particular, the relative contributions of different signaling pathways which are responsible for the generation of the innate immune response are still largely unknown. In this study, we used STING-noninteracting TRIF mutants to decipher the relative contributions of the TLR3 and cGAS-STING signaling pathways to the attenuation of HSV-1 infection. We show that the relative contributions of the two pathways to the attenuation of viral infection are different in mouse versus human cell lines. Together, our results provide new insights into the relative contributions of two different signaling pathways in the attenuation of viral infection and may lead to the development of new antiviral strategies aimed at blocking viral infection at very early stages.


Subject(s)
Herpesvirus 1, Human/physiology , Membrane Proteins/metabolism , Nucleotidyltransferases/metabolism , Signal Transduction , Toll-Like Receptor 3/metabolism , Virus Replication , Animals , HEK293 Cells , HeLa Cells , Humans , Membrane Proteins/genetics , Mice , Toll-Like Receptor 3/genetics
9.
J Immunol ; 200(8): 2809-2818, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29531172

ABSTRACT

Mammalian TLRs recognize microbial infection or cell death-associated danger signals and trigger the appropriate cellular response. These responses determine the strength and the outcome of the host-microbe interaction. TLRs are transmembrane proteins located on the plasma or the endosomal membrane. Their ectodomains recognize specific microbial or endogenous ligands, and the cytoplasmic domains interact with specific proteins to activate intracellular signaling pathways. TLR9, an endosomal TLR, is activated by endocytosed DNA. Activated TLR9 recruits the cytoplasmic adapter MyD88 and other signaling proteins to induce the synthesis of inflammatory cytokines and IFN. Uncontrolled activation of TLR9 leads to the undesired overproduction of inflammatory cytokines and consequent pathogenesis. Therefore, appropriate activation and the regulation of TLR9 signaling are critical. Tyrosine (Tyr) phosphorylation of TLR9 is essential for its activation; however, the role of specific Tyr kinases is not clear. In this article, we report that epidermal growth factor receptor (EGFR), a membrane-bound protein Tyr kinase, is essential for TLR9 signaling. Genetic ablation of EGFR or pharmacological inhibition of its kinase activity attenuates TLR9-mediated induction of genes in myeloid and nonmyeloid cell types. EGFR is constitutively bound to TLR9; upon ligand stimulation, it mediates TLR9 Tyr phosphorylation, which leads to the recruitment of MyD88, activation of the signaling kinases and transcription factors, and gene induction. In mice, TLR9-mediated liver injury and death are blocked by an EGFR inhibitor or deletion of the EGFR gene from myeloid cells, which are the major producers of inflammatory cytokines.


Subject(s)
ErbB Receptors/metabolism , Macrophages/metabolism , Signal Transduction/immunology , Toll-Like Receptor 9/metabolism , Animals , ErbB Receptors/immunology , Humans , Macrophages/immunology , Mice , Phosphorylation , Toll-Like Receptor 9/immunology , Tyrosine/metabolism
11.
Cell Host Microbe ; 20(3): 329-341, 2016 Sep 14.
Article in English | MEDLINE | ID: mdl-27631700

ABSTRACT

The intracellular microbial nucleic acid sensors, TLR3 and STING, recognize pathogen molecules and signal to activate the interferon pathway. The TIR-domain containing protein TRIF is the sole adaptor of TLR3. Here, we report an essential role for TRIF in STING signaling: various activators of STING could not induce genes in the absence of TRIF. TRIF and STING interacted directly, through their carboxy-terminal domains, to promote STING dimerization, intermembrane translocation, and signaling. Herpes simplex virus (HSV), which triggers the STING signaling pathway and is controlled by it, replicated more efficiently in the absence of TRIF, and HSV-infected TRIF(-/-) mice displayed pronounced pathology. Our results indicate that defective STING signaling may be responsible for the observed genetic association between TRIF mutations and herpes simplex encephalitis in patients.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Herpesvirus 1, Human/immunology , Immunity, Innate , Membrane Proteins/metabolism , Animals , Cell Line , Disease Models, Animal , Herpes Simplex/pathology , Herpes Simplex/virology , Herpesvirus 1, Human/pathogenicity , Humans , Mice , Mice, Knockout , Protein Binding , Protein Interaction Mapping , Protein Multimerization , Protein Transport , Signal Transduction
12.
J Biol Chem ; 291(26): 13535-45, 2016 Jun 24.
Article in English | MEDLINE | ID: mdl-27137933

ABSTRACT

The chromosomally clustered interferon-induced with tetratricopeptide repeat motif (IFIT) gene family members share structural features at the gene and protein levels. Despite these similarities, different IFIT genes have distinct inducer- and cell type-specific induction patterns. Here, we investigated the mechanism for the observed differential induction of the mouse Ifit1, Ifit2, and Ifit3 genes in B cells and demonstrated that the repressive effect of the transcription factor interferon regulatory factor 8 (IRF8), which is highly expressed in B cells, played an essential role in this regulation. Although IRF8 could impair induction of all three IFIT genes following stimulation of retinoic acid-inducible gene I (RIG-I), it could selectively impair the induction of the Ifit1 gene following IFN stimulation. The above properties could be imparted to IRF8-non-expressing cells by ectopic expression of the protein. Induction of reporter genes, driven by truncated Ifit1 promoters, identified the regions that mediate the repression, and a chromatin immunoprecipitation assay revealed that more IRF8 bound to the IFN-stimulated response element of the Ifit1 gene than to those of the Ifit2 and the Ifit3 genes. Mutational analyses of IRF8 showed that its ability to bind DNA, interact with other proteins, and undergo sumoylation were all necessary to selectively repress Ifit1 gene induction in response to IFN. Our study revealed a new role for IRFs in differentially regulating the induction patterns of closely related IFN-stimulated genes that are located adjacent to one another in the mouse genome.


Subject(s)
B-Lymphocytes/metabolism , Carrier Proteins/metabolism , Interferon Regulatory Factors/metabolism , Proteins/metabolism , Response Elements/physiology , Adaptor Proteins, Signal Transducing , Animals , Apoptosis Regulatory Proteins , B-Lymphocytes/cytology , Carrier Proteins/genetics , Interferon Regulatory Factors/genetics , Intracellular Signaling Peptides and Proteins , Mice , Multigene Family , Mutation , Proteins/genetics , RNA-Binding Proteins
13.
FEBS J ; 282(24): 4766-81, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26414443

ABSTRACT

The murine double-stranded RNA-binding protein termed protein kinase R (PKR)-associated protein X (RAX) and the human homolog, protein activator of PKR (PACT), were originally characterized as activators of PKR. Mice deficient in RAX show reproductive and developmental defects, including reduced body size, craniofacial defects and anterior pituitary hypoplasia. As these defects are not observed in PKR-deficient mice, the phenotype has been attributed to PKR-independent activities of RAX. Here we further investigated the involvement of PKR in the physiological function of RAX, by generating rax(-/-) mice deficient in PKR, or carrying a kinase-inactive mutant of PKR (K271R) or an unphosphorylatable mutant of the PKR substrate eukaryotic translation initiation factor 2 α subunit (eIF2α) (S51A). Ablating PKR expression rescued the developmental and reproductive deficiencies in rax(-/-) mice. Generating rax(-/-) mice with a kinase-inactive mutant of PKR resulted in similar rescue, confirming that the rax(-/-) defects are PKR dependent; specifically that the kinase activity of PKR was required for these defects. Moreover, generating rax(-/-) mice that were heterozygous for an unphosphorylatable mutant eIF2α provides partial rescue of the rax(-/-) defect, consistent with mutation of one copy of the Eif2s1 gene. These observations were further investigated in vitro by reducing RAX expression in anterior pituitary cells, resulting in increased PKR activity and induction of the PKR-regulated cyclin-dependent kinase inhibitor p21(WAF1/CIP1). These results demonstrate that PKR kinase activity is required for onset of the rax(-/-) phenotype, implying an unexpected function for RAX as a negative regulator of PKR in the context of postnatal anterior pituitary tissue, and identify a critical role for the regulation of PKR activity for normal development.


Subject(s)
Pituitary Gland, Anterior/metabolism , RNA-Binding Proteins/metabolism , eIF-2 Kinase/antagonists & inhibitors , Amino Acid Substitution , Animals , Cell Cycle , Cell Line , Cell Proliferation , Crosses, Genetic , Enzyme Activation , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism , Female , Humans , Male , Mice , Mice, Knockout , Mice, Transgenic , Mutation , Organ Size , Phosphorylation , Pituitary Gland, Anterior/cytology , Pituitary Gland, Anterior/enzymology , Pituitary Gland, Anterior/growth & development , Protein Processing, Post-Translational , RNA Interference , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/genetics , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
14.
J Biol Chem ; 286(30): 26319-26, 2011 Jul 29.
Article in English | MEDLINE | ID: mdl-21636578

ABSTRACT

RNase L and RNA-dependent protein kinase (PKR) are effectors of the interferon antiviral response that share homology in their pseudokinase and protein kinase domains, respectively. Sunitinib is an orally available, ATP-competitive inhibitor of VEGF and PDGF receptors used clinically to suppress angiogenesis and tumor growth. Sunitinib also impacts IRE1, an endoplasmic reticulum protein involved in the unfolded protein response that is closely related to RNase L. Here, we report that sunitinib is a potent inhibitor of both RNase L and PKR with IC(50) values of 1.4 and 0.3 µM, respectively. In addition, flavonol activators of IRE1 inhibited RNase L. Sunitinib treatment of wild type (WT) mouse embryonic fibroblasts resulted in about a 12-fold increase in encephalomyocarditis virus titers. However, sunitinib had no effect on encephalomyocarditis virus growth in cells lacking both PKR and RNase L. Furthermore, oral delivery of sunitinib in WT mice resulted in 10-fold higher viral titers in heart tissues while suppressing by about 2-fold the IFN-ß levels. In contrast, sunitinib had no effect on viral titers in mice deficient in both RNase L and PKR. Also, sunitinib reduced mean survival times from 12 to 6 days in virus-infected WT mice while having no effect on survival of mice lacking both RNase L and PKR. Results indicate that sunitinib treatments prevent antiviral innate immune responses mediated by RNase L and PKR.


Subject(s)
Antineoplastic Agents/pharmacology , Cardiovirus Infections/immunology , Endoribonucleases/antagonists & inhibitors , Immunity, Innate/drug effects , Indoles/pharmacology , Membrane Proteins/immunology , Protein Serine-Threonine Kinases/immunology , Pyrroles/pharmacology , eIF-2 Kinase/antagonists & inhibitors , Animals , Cardiovirus Infections/enzymology , Cardiovirus Infections/genetics , Encephalomyocarditis virus , Endoribonucleases/genetics , Endoribonucleases/immunology , Endoribonucleases/metabolism , Humans , Immunity, Innate/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/metabolism , Sunitinib , eIF-2 Kinase/genetics , eIF-2 Kinase/immunology
15.
Mol Vis ; 15: 162-7, 2009.
Article in English | MEDLINE | ID: mdl-19158959

ABSTRACT

PURPOSE: Microphthalmia, anophthalmia, and coloboma are ocular malformations with a significant genetic component. Rx is a homeobox gene expressed early in the developing retina and is important in retinal cell fate specification as well as stem cell proliferation. We screened a group of 24 patients with microphthalmia, coloboma, and/or anophthalmia for RX mutations. METHODS: We used standard PCR and automated sequencing techniques to amplify and sequence each of the three RX exons. Patients' charts were reviewed for clinical information. The pathologic impact of the identified sequence variant was analyzed by computational methods using PolyPhen and PMut algorithms. RESULTS: In addition to the polymorphisms we identified a single patient with coloboma having a heterozygous nucleotide change (g.197G>C) in the first exon that results in a missense mutation of arginine to threonine at amino acid position 66 (R66T). In silico analysis predicted R66T to be a deleterious mutation. CONCLUSIONS: Sequence variations in RX are uncommon in patients with congenital ocular malformations, but may play a role in disease pathogenesis. We observed a missense mutation in RX in a patient with a small, typical chorioretinal coloboma, and postulate that the mutation is responsible for the patient's phenotype.


Subject(s)
Anophthalmos/genetics , Coloboma/genetics , Eye Proteins/genetics , Homeodomain Proteins/genetics , Microphthalmos/genetics , Transcription Factors/genetics , Amino Acid Substitution , Base Sequence , DNA Mutational Analysis , Fundus Oculi , Heterozygote , Humans , Molecular Sequence Data
16.
Exp Cell Res ; 314(20): 3663-8, 2008 Dec 10.
Article in English | MEDLINE | ID: mdl-18955045

ABSTRACT

Cellular retinoic acid binding protein II (CRABP-II) is overexpressed in a wide variety of cancers. Previously we have shown that CRABP-II expression levels are also elevated in neuroblastoma and Wilms tumors. To elucidate the molecular mechanisms underlying the abnormal expression of CRABP-II in Wilms tumor, we studied the expression of MycN and CRABP-II in these tumor samples. Our data revealed that CRABP-II is overexpressed in Wilms tumor compared to normal adjacent non-neoplastic tissue and its levels are even higher in late stage tumors. Its expression correlates with MycN expression in tumors. The tumors that do not express MycN have no CRABP-II expression. The expression of CRABP-II is also regulated by methylation and its promoter is unmethylated in tumors. Knockdown of MycN by small interfering RNA leads to downregulation of CRABP-II. Thus our results suggest that both MycN and DNA methylation are responsible for CRABP-II expression in pediatric tumors and demethylation of CRABP-II may be an early event in tumor development.


Subject(s)
Kidney Neoplasms/genetics , Nuclear Proteins/physiology , Oncogene Proteins/physiology , Receptors, Retinoic Acid/genetics , Wilms Tumor/genetics , CpG Islands , DNA Methylation , Fetus/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , N-Myc Proto-Oncogene Protein , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Oncogene Proteins/antagonists & inhibitors , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Promoter Regions, Genetic , RNA, Small Interfering/pharmacology , Receptors, Retinoic Acid/metabolism , Tumor Cells, Cultured
17.
Br J Ophthalmol ; 91(10): 1385-92, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17475713

ABSTRACT

AIM: In an effort to identify patients with uveal melanoma at high risk of metastasis, the authors undertook correlation of gene expression profiles with histopathology data and tumour-related mortality. METHODS: The RNA was isolated from 27 samples of uveal melanoma from patients who had consented to undergo enucleation, and transcripts profiled using a cDNA array comprised of sequence-verified cDNA clones representing approximately 4000 genes implicated in cancer development. Two multivariate data mining techniques--hierarchical cluster analysis and multidimensional scaling--were used to investigate the grouping structure in the gene expression data. Cluster analysis was performed with a subset of 10,000 randomly selected genes and the cumulative contribution of all the genes in making the correct grouping was recorded. RESULTS: Hierarchical cluster analysis and multidimensional scaling revealed two distinct classes. When correlated with the data on metastasis, the two molecular classes corresponded very well to the survival data for the 27 patients. Thirty two discrete genes (corresponding to 44 probe sets) that correctly defined the molecular classes were selected. A single gene (ectonucleotide pyrophosphatase/phosphodiesterase 2; autotaxin) could classify the molecular types. The expression pattern was confirmed using real-time quantitative PCR. CONCLUSIONS: Gene expression profiling identifies two distinct prognostic classes of uveal melanoma. Underexpression of autotaxin in class 2 uveal melanoma with a poor prognosis needs to be explored further.


Subject(s)
Melanoma/mortality , Multienzyme Complexes/genetics , Phosphodiesterase I/genetics , Pyrophosphatases/genetics , Uveal Neoplasms/mortality , Cluster Analysis , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/genetics , Genes, Neoplasm/genetics , Humans , Kaplan-Meier Estimate , Male , Melanoma/genetics , Melanoma/pathology , Middle Aged , Neoplasm Metastasis/genetics , Neoplasm Proteins/genetics , Phosphoric Diester Hydrolases , Prognosis , Uveal Neoplasms/genetics , Uveal Neoplasms/pathology
18.
Cancer Res ; 65(7): 2592-601, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15805255

ABSTRACT

Anaplastic histology and metastasis are each associated with higher relapse and mortality rates in Wilms tumor patients. However, not all anaplastic tumors relapse and some nonanaplastic tumors relapse unexpectedly. To identify more accurate early prognostic indicators, we analyzed expression of 4,900 cancer-related genes in 26 primary Wilms tumors. This analysis revealed that expression of a set of four genes predicts future relapse of primary Wilms tumors with high accuracy, independent of anaplasia. Random permutation testing of this prognostic gene expression signature yielded P = 0.003. Real-time reverse transcription-PCR analysis of the four genes in an independent primary tumor set resulted in correct prediction of future relapse with an accuracy of 92%. One of the four genes in the prognostic signature, CCAAT/enhancer binding protein beta (C/EBPB), is expressed at higher levels in both primary relapsing tumors and metastatic tumors than in primary nonrelapsing tumors. Short interfering RNA-mediated down-regulation of C/EBPB expression in WiT49, a cell line derived from a metastatic Wilms tumor, resulted in spontaneous apoptosis. These findings suggest that C/EBPB is a critical survival factor for Wilms tumor cells and that its expression contributes to the prognosis of Wilms tumor patients.


Subject(s)
Kidney Neoplasms/genetics , Wilms Tumor/genetics , CCAAT-Enhancer-Binding Protein-beta/biosynthesis , CCAAT-Enhancer-Binding Protein-beta/genetics , Gene Expression Profiling , Humans , Kidney Neoplasms/metabolism , Oligonucleotide Array Sequence Analysis , Prognosis , Recurrence , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Wilms Tumor/metabolism
19.
Oncogene ; 24(3): 457-68, 2005 Jan 13.
Article in English | MEDLINE | ID: mdl-15531917

ABSTRACT

Anaplasia (unfavorable histology) is associated with therapy resistance and poor prognosis of Wilms tumor, but the molecular basis for this phenotype is unclear. Here, we used a cDNA array with 9240 clones relevant to cancer biology and/or kidney development to examine the expression profiles of 54 Wilms tumors, five normal kidneys and fetal kidney. By linking genes differentially expressed between fetal kidney and Wilms tumors to kidney morphogenesis, we found that genes expressed at a higher level in Wilms tumors tend to be expressed more in uninduced metanephrogenic mesenchyme or blastema than in their differentiated structures. Conversely, genes expressed at a lower level in Wilms tumors tend to be expressed less in uninduced metanephrogenic mesenchyme or blastema. We also identified 97 clones representing 76 Unigenes or unclustered ESTs that clearly separate anaplastic Wilms tumors from tumors with favorable histology. Genes in this set provide insight into the nature of the abnormal nuclear morphology of anaplastic tumors and may facilitate identification of molecular targets to improve their responsiveness to treatment.


Subject(s)
Cell Transformation, Neoplastic/pathology , Gene Expression Profiling/methods , Kidney Neoplasms/genetics , Kidney/embryology , Wilms Tumor/genetics , Fetus , Gene Expression Regulation, Developmental , Humans , Kidney/growth & development , Kidney/physiology , Morphogenesis , Reference Values
20.
J Biol Chem ; 279(32): 33575-85, 2004 Aug 06.
Article in English | MEDLINE | ID: mdl-15155752

ABSTRACT

Wilms tumors are a heterogeneous class of tumors in which Wilms tumor suppressor-1 (WT1) and the p53 tumor suppressor may be variously inactivated by mutation, reduced in expression, or even overexpressed in the wild-type state. The downstream transcriptional targets of WT1 and p53 that are critical for mediating their roles in Wilms tumorigenesis are not well defined. The WiT49 cell line is characteristic of anaplastic Wilms tumors that are refractory to treatment and expresses wild-type WT1 and mutant p53. We have used the small molecule compound CP-31398 (Pfizer) to restore wild-type p53 function to the codon 248 mutant p53 present in WiT49 cells. In these cells, CP-31398 activated transcription of p53-regulated promoters and enhanced UV light-induced apoptosis without altering the overall p53 protein level. These phenotypes were accompanied by restored binding of the p53 protein to promoter sequences in vivo. Gene expression profiling of CP-31398-treated WiT49 cells revealed subsets of putative p53 target genes that were up- or down-regulated. A preferred target of p53-mediated repression in this system is the podocalyxin (PODXL) gene. PODXL is also transcriptionally regulated by WT1 and has roles in cell adhesion and anti-adhesion. Our results show that PODXL is a bona fide target of p53-mediated transcriptional repression while being positively regulated by WT1. We propose that inappropriate expression of PODXL due to changes in WT1 and/or p53 activity may contribute to Wilms tumorigenesis.


Subject(s)
Gene Expression Regulation , Sialoglycoproteins/genetics , Tumor Suppressor Protein p53/physiology , WT1 Proteins/genetics , Apoptosis , Blotting, Western , DNA Damage , Humans , Immunosorbent Techniques , In Situ Nick-End Labeling , Mutation , Nucleic Acid Hybridization , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic/genetics , Pyrimidines/pharmacology , RNA, Messenger/analysis , Repressor Proteins , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic , Transfection , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , WT1 Proteins/physiology , Wilms Tumor
SELECTION OF CITATIONS
SEARCH DETAIL
...