Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 15(1): 730, 2024 Jan 25.
Article in English | MEDLINE | ID: mdl-38272925

ABSTRACT

Stimulating the innate immune system has been explored as a therapeutic option for the treatment of gliomas. Inactivating mutations in ATRX, defining molecular alterations in IDH-mutant astrocytomas, have been implicated in dysfunctional immune signaling. However, little is known about the interplay between ATRX loss and IDH mutation on innate immunity. To explore this, we generated ATRX-deficient glioma models in the presence and absence of the IDH1R132H mutation. ATRX-deficient glioma cells are sensitive to dsRNA-based innate immune agonism and exhibit impaired lethality and increased T-cell infiltration in vivo. However, the presence of IDH1R132H dampens baseline expression of key innate immune genes and cytokines in a manner restored by genetic and pharmacological IDH1R132H inhibition. IDH1R132H co-expression does not interfere with the ATRX deficiency-mediated sensitivity to dsRNA. Thus, ATRX loss primes cells for recognition of dsRNA, while IDH1R132H reversibly masks this priming. This work reveals innate immunity as a therapeutic vulnerability of astrocytomas.


Subject(s)
Astrocytoma , Brain Neoplasms , Glioma , Humans , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , X-linked Nuclear Protein/genetics , Glioma/genetics , Glioma/metabolism , Astrocytoma/genetics , Mutation , Immunity, Innate/genetics , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism
2.
bioRxiv ; 2023 Apr 21.
Article in English | MEDLINE | ID: mdl-37131619

ABSTRACT

Stimulating the innate immune system has been explored as a therapeutic option for the treatment of gliomas. Inactivating mutations in ATRX , defining molecular alterations in IDH -mutant astrocytomas, have been implicated in dysfunctional immune signaling. However, little is known about the interplay between ATRX loss and IDH mutation on innate immunity. To explore this, we generated ATRX knockout glioma models in the presence and absence of the IDH1 R 132 H mutation. ATRX-deficient glioma cells were sensitive to dsRNA-based innate immune agonism and exhibited impaired lethality and increased T-cell infiltration in vivo . However, the presence of IDH1 R 132 H dampened baseline expression of key innate immune genes and cytokines in a manner restored by genetic and pharmacological IDH1 R132H inhibition. IDH1 R132H co-expression did not interfere with the ATRX KO-mediated sensitivity to dsRNA. Thus, ATRX loss primes cells for recognition of dsRNA, while IDH1 R132H reversibly masks this priming. This work reveals innate immunity as a therapeutic vulnerability of astrocytoma.

3.
Sci Transl Med ; 15(682): eabn5649, 2023 02 08.
Article in English | MEDLINE | ID: mdl-36753564

ABSTRACT

D2C7-immunotoxin (IT), a dual-specific IT targeting wild-type epidermal growth factor receptor (EGFR) and mutant EGFR variant III (EGFRvIII) proteins, demonstrates encouraging survival outcomes in a subset of patients with glioblastoma. We hypothesized that immunosuppression in glioblastoma limits D2C7-IT efficacy. To improve the response rate and reverse immunosuppression, we combined D2C7-IT tumor cell killing with αCD40 costimulation of antigen-presenting cells. In murine glioma models, a single intratumoral injection of D2C7-IT+αCD40 treatment activated a proinflammatory phenotype in microglia and macrophages, promoted long-term tumor-specific CD8+ T cell immunity, and generated cures. D2C7-IT+αCD40 treatment increased intratumoral Slamf6+CD8+ T cells with a progenitor phenotype and decreased terminally exhausted CD8+ T cells. D2C7-IT+αCD40 treatment stimulated intratumoral CD8+ T cell proliferation and generated cures in glioma-bearing mice despite FTY720-induced peripheral T cell sequestration. Tumor transcriptome profiling established CD40 up-regulation, pattern recognition receptor, cell senescence, and immune response pathway activation as the drivers of D2C7-IT+αCD40 antitumor responses. To determine potential translation, immunohistochemistry staining confirmed CD40 expression in human GBM tissue sections. These promising preclinical data allowed us to initiate a phase 1 study with D2C7-IT+αhCD40 in patients with malignant glioma (NCT04547777) to further evaluate this treatment in humans.


Subject(s)
Brain Neoplasms , Glioblastoma , Glioma , Immunotoxins , Humans , Animals , Mice , Glioblastoma/pathology , Immunotoxins/genetics , CD8-Positive T-Lymphocytes , Adaptive Immunity , ErbB Receptors/metabolism , Cell Line, Tumor , Brain Neoplasms/therapy
5.
PLoS Pathog ; 15(11): e1007634, 2019 11.
Article in English | MEDLINE | ID: mdl-31682641

ABSTRACT

Induction of vast transcriptional programs is a central event of innate host responses to viral infections. Here we report a transcriptional program with potent antiviral activity, driven by E74-like ETS transcription factor 1 (ELF1). Using microscopy to quantify viral infection over time, we found that ELF1 inhibits eight diverse RNA and DNA viruses after multi-cycle replication. Elf1 deficiency results in enhanced susceptibility to influenza A virus infections in mice. ELF1 does not feed-forward to induce interferons, and ELF1's antiviral effect is not abolished by the absence of STAT1 or by inhibition of JAK phosphorylation. Accordingly, comparative expression analyses by RNA-seq revealed that the ELF1 transcriptional program is distinct from interferon signatures. Thus, ELF1 provides an additional layer of the innate host response, independent from the action of type I interferons.


Subject(s)
Antiviral Agents/pharmacology , Gene Expression Regulation/drug effects , Influenza A virus/immunology , Interferon Type I/pharmacology , Nuclear Proteins/metabolism , Orthomyxoviridae Infections/immunology , Transcription Factors/metabolism , Virus Replication/immunology , A549 Cells , Animals , Female , Humans , Immunity, Innate , Influenza A virus/drug effects , Mice , Mice, Inbred C57BL , NF-kappa B/genetics , NF-kappa B/metabolism , Nuclear Proteins/genetics , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/virology , Phosphorylation , STAT1 Transcription Factor , Signal Transduction , Transcription Factors/genetics , Virus Replication/drug effects
6.
Neuron ; 90(1): 43-55, 2016 Apr 06.
Article in English | MEDLINE | ID: mdl-26971948

ABSTRACT

Transcription Factor 4 (TCF4) is a clinically pleiotropic gene associated with schizophrenia and Pitt-Hopkins syndrome (PTHS). To gain insight about the neurobiology of TCF4, we created an in vivo model of PTHS by suppressing Tcf4 expression in rat prefrontal neurons immediately prior to neurogenesis. This cell-autonomous genetic insult attenuated neuronal spiking by increasing the afterhyperpolarization. At the molecular level, using a novel technique called iTRAP that combined in utero electroporation and translating ribosome affinity purification, we identified increased translation of two ion channel genes, Kcnq1 and Scn10a. These ion channel candidates were validated by pharmacological rescue and molecular phenocopy. Remarkably, similar excitability deficits were observed in prefrontal neurons from a Tcf4(+/tr) mouse model of PTHS. Thus, we identify TCF4 as a regulator of neuronal intrinsic excitability in part by repression of Kcnq1 and Scn10a and suggest that this molecular function may underlie pathophysiology associated with neuropsychiatric disorders.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , DNA-Binding Proteins/genetics , Hyperventilation/genetics , Intellectual Disability/genetics , KCNQ1 Potassium Channel/genetics , NAV1.8 Voltage-Gated Sodium Channel/genetics , Prefrontal Cortex/metabolism , RNA, Messenger/metabolism , Transcription Factors/genetics , Animals , CRISPR-Cas Systems , Disease Models, Animal , Electroporation , Facies , Female , Gene Expression Regulation, Developmental , Genetic Predisposition to Disease , Haploinsufficiency , Mice , Patch-Clamp Techniques , Prefrontal Cortex/embryology , Pregnancy , RNA, Small Interfering , Rats , Rats, Wistar , Schizophrenia/genetics , Transcription Factor 4
7.
Antimicrob Agents Chemother ; 58(6): 3399-410, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24709254

ABSTRACT

A small-molecule inhibitor of hepatitis C virus (HCV) designated AP89652 was identified by screening a compound library with an HCV genotype 1b subgenomic replicon assay. AP89652 contains two chiral centers, and testing of two syn enantiomers revealed that activity in the replicon assay resided with only one, AP80978, whose 50% effective concentration (EC50) (the concentration at which a 50% reduction in Renilla luciferase levels was observed relative to an untreated control) was 630 nM. AP80978 was inhibitory against HCV genotypes 1a and 1b but not genotype 2a. In a replicon clearance assay, the potency and clearance rate of AP80978 were similar to those of telaprevir (VX950) and cyclosporine (CsA). AP80978 was nontoxic when tested against a panel of human cell lines, and inhibitory activity was HCV specific in that there was limited activity against negative-strand viruses, an alphavirus, and flaviviruses. By selection of resistant replicons and assessment of activity in genotype 1b/2a intergenotypic replicons, the viral protein target of this compound was identified as NS4B. NS4B F98V/L substitutions were confirmed by site-directed mutagenesis as AP80978 resistance-associated mutations. When tested against HCV produced in cell culture, the compound was significantly more potent than other HCV inhibitors, including VX950, CsA, and 2'-C-methyladenosine (2'C-meA). In addition, AP80977, the enantiomer that was inactive in the replicon assay, had activity against the virus, although it was lower than the activity of AP80978. These results suggest that AP80978 has the potential to be optimized into an effective antiviral drug and is a useful tool to further study the role of NS4B in HCV replication.


Subject(s)
Antiviral Agents/pharmacology , Furans/pharmacology , Hepacivirus/drug effects , Hepatitis C/drug therapy , Pyridines/pharmacology , Thiophenes/pharmacology , Viral Nonstructural Proteins/antagonists & inhibitors , Adenosine/analogs & derivatives , Adenosine/pharmacology , Biological Assay , Cell Culture Techniques , Cell Line , Cell Survival/drug effects , Cyclosporine/pharmacology , Genotype , Hepacivirus/genetics , Hepatitis C/virology , High-Throughput Screening Assays , Humans , Luciferases, Renilla , Mutagenesis, Site-Directed , Oligopeptides/pharmacology , Replicon/drug effects , Virus Replication/drug effects
8.
Semin Hematol ; 50(1): 38-47, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23507482

ABSTRACT

In lymphoid malignancies, aberrant epigenetic mechanisms such as DNA methylation and histone modifications influence chromatin architecture and can result in altered gene expression. These alterations commonly affect genes that play important roles in the cell cycle, apoptosis, and DNA repair in non-Hodgkin lymphoma (NHL). The ability to identify epigenetic modifications to these important genes has increased exponentially due to advances in technology. As a result, there are well-defined, gene-specific epigenetic aberrations associated with NHL comprising follicular lymphoma (FL), mantle cell lymphoma (MCL), chronic lymphocytic leukemia (CLL), and diffuse large B-cell lymphoma (DLBCL). The identification of these genes is important because they may be used as biomarkers for prognosis, diagnosis and in developing improved treatment strategies. Also important, in the control of gene expression, is the packaging of DNA within the nucleus of a cell. This packaging can be distorted by epigenetic alterations and may alter the accessibility of certain regions of the genome in cancer cells. This review discusses the impact of known epigenetic aberration on the regulation of gene expression in NHL and provides insight into the spatial conformation of the genome (DNA packaging) in acute lymphoblastic leukemia.


Subject(s)
Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Lymphoma, Non-Hodgkin/genetics , Animals , DNA/genetics , DNA Methylation , Histones/genetics , Humans
9.
PLoS One ; 8(3): e58793, 2013.
Article in English | MEDLINE | ID: mdl-23536826

ABSTRACT

The spatial conformation of a genome plays an important role in the long-range regulation of genome-wide gene expression and methylation, but has not been extensively studied due to lack of genome conformation data. The recently developed chromosome conformation capturing techniques such as the Hi-C method empowered by next generation sequencing can generate unbiased, large-scale, high-resolution chromosomal interaction (contact) data, providing an unprecedented opportunity to investigate the spatial structure of a genome and its applications in gene regulation, genomics, epigenetics, and cell biology. In this work, we conducted a comprehensive, large-scale computational analysis of this new stream of genome conformation data generated for three different human leukemia cells or cell lines by the Hi-C technique. We developed and applied a set of bioinformatics methods to reliably generate spatial chromosomal contacts from high-throughput sequencing data and to effectively use them to study the properties of the genome structures in one-dimension (1D) and two-dimension (2D). Our analysis demonstrates that Hi-C data can be effectively applied to study tissue-specific genome conformation, chromosome-chromosome interaction, chromosomal translocations, and spatial gene-gene interaction and regulation in a three-dimensional genome of primary tumor cells. Particularly, for the first time, we constructed genome-scale spatial gene-gene interaction network, transcription factor binding site (TFBS) - TFBS interaction network, and TFBS-gene interaction network from chromosomal contact information. Remarkably, all these networks possess the properties of scale-free modular networks.


Subject(s)
Epistasis, Genetic , Gene Regulatory Networks , Genome, Human , Nucleic Acid Conformation , Binding Sites , Cell Line , Chromosome Mapping , Computational Biology/methods , Genomics/methods , Humans , Neoplasms/genetics , Transcription Factors , Translocation, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...