Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Transplant Cell Ther ; 28(1): 30.e1-30.e7, 2022 01.
Article in English | MEDLINE | ID: mdl-34655803

ABSTRACT

Methods that enable monitoring of therapeutic efficacy of autologous chimeric antigen receptor (CAR) T-cell therapy will be clinically useful. The aim of this study is to demonstrate the feasibility of blood-derived cell-free DNA (cfDNA) to predict CAR T-cell therapy response in patients with refractory B-cell lymphomas. Whole blood was collected before and throughout CAR T-cell therapy until day 154. Low-coverage (∼0.4×), genome-wide cfDNA sequencing, similar to that established for noninvasive prenatal testing, was performed. The genomic instability number (GIN) was used to quantify plasma copy number alteration level. Twelve patients were enrolled. Seven (58%) patients achieved a complete response (CR); 2 (25%), a partial response. Median progression-free survival was 99 days; median overall survival was not reached (median follow-up, 247 days). Altogether, 127 blood samples were analyzed (median, 10 samples/patient [range 8-13]). All 5 patients who remained in CR at the time of last measurement had GIN <170 (threshold). Two patients who attained CR, but later relapsed, and all but one patient who had best response other than CR had last GIN measurement of >170. In 5 of 6 patients with relapsed or progressive disease, increasing GIN was observed before the diagnosis by imaging. The abundance of CAR T-cell construct (absolute number of construct copies relative to the number of human genome equivalents) also showed a trend to correlate with outcome (day 10, P = .052). These data describe a proof-of-concept for the use of multiple liquid biopsy technologies to monitor therapeutic response in B-cell lymphoma patients receiving CAR T-cell therapy.


Subject(s)
Cell-Free Nucleic Acids , Lymphoma, B-Cell , Receptors, Chimeric Antigen , Antigens, CD19/genetics , Humans , Immunotherapy, Adoptive , Lymphoma, B-Cell/genetics , Receptors, Chimeric Antigen/genetics
2.
Open Biol ; 4(11): 140180, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25392452

ABSTRACT

Internal ribosome entry sites (IRESs) in cellular mRNAs direct expression of growth-promoting factors through an alternative translation mechanism that has yet to be fully defined. Lymphoid enhancer factor-1 (LEF-1), a Wnt-mediating transcription factor important for cell survival and metastasis in cancer, is produced via IRES-directed translation, and its mRNA is frequently upregulated in malignancies, including chronic myeloid leukaemia (CML). In this study, we determined that LEF1 expression is regulated by Bcr-Abl, the oncogenic protein that drives haematopoietic cell transformation to CML. We have previously shown that the LEF1 5' untranslated region recruits a complex of proteins to its IRES, including the translation initiation factor eIF4A. In this report, we use two small molecule inhibitors, PP242 (dual mTOR (mammalian target of rapamycin) kinase inhibitor) and hippuristanol (eIF4A inhibitor), to define IRES regulation via a Bcr-Abl-mTOR-eIF4A axis in CML cell lines and primary patient leukaemias. We found that LEF1 and other IRESs are uniquely sensitive to the activities of Bcr-Abl/mTOR. Most notably, we discovered that eIF4A, an RNA helicase, elicits potent non-canonical effects on the LEF1 IRES. Hippuristanol inhibition of eIF4A stalls translation of IRES mRNA and triggers dissociation from polyribosomes. We propose that a combination drug strategy which targets mTOR and IRES-driven translation disrupts key factors that contribute to growth and proliferation in CML.


Subject(s)
Eukaryotic Initiation Factor-4A/metabolism , Fusion Proteins, bcr-abl/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Lymphoid Enhancer-Binding Factor 1/metabolism , Protein Biosynthesis , TOR Serine-Threonine Kinases/metabolism , Animals , Cells, Cultured , Eukaryotic Initiation Factor-4A/antagonists & inhibitors , Humans , Indoles/pharmacology , Jurkat Cells , Lymphoid Enhancer-Binding Factor 1/genetics , Mice , Polyribosomes/metabolism , Protein Kinase Inhibitors/pharmacology , Purines/pharmacology , RNA, Messenger/metabolism , Sterols/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors
3.
Virus Res ; 176(1-2): 223-31, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23830997

ABSTRACT

Different types of environmental stress cause mammalian cells to form cytoplasmic foci, termed stress granules, which contain mRNPs that are translationally silenced. These foci are transient and dynamic, and contain components of the cellular translation machinery as well as certain mRNAs and RNA binding proteins. Stress granules are known to be induced by conditions such as hypoxia, nutrient deprivation, and oxidative stress, and a number of cellular factors have been identified that are commonly associated with these foci. More recently it was discovered that poliovirus infection also induces the formation of stress granules, although these cytoplasmic foci appear to be somewhat compositionally unique. Work described here examined the punctate pattern of SRp20 (a host cell mRNA splicing protein) localization in the cytoplasm of poliovirus-infected cells, demonstrating the partial co-localization of SRp20 with the stress granule marker protein TIA-1. We determined that SRp20 does not co-localize with TIA-1, however, under conditions of oxidative stress, indicating that the close association of these two proteins during poliovirus infection is not representative of a general response to cellular stress. We confirmed that the expression of a dominant negative version of TIA-1 (TIA-1-PRD) results in the dissociation of stress granules. Finally, we demonstrated that expression of wild type TIA-1 or dominant negative TIA-1-PRD in cells during poliovirus infection does not dramatically affect viral translation. Taken together, these studies provide a new example of the unique cytoplasmic foci that form during poliovirus infection.


Subject(s)
Cytoplasmic Granules/metabolism , Poliovirus/growth & development , Poly(A)-Binding Proteins/metabolism , Protein Interaction Mapping , RNA-Binding Proteins/metabolism , HeLa Cells , Humans , Serine-Arginine Splicing Factors , T-Cell Intracellular Antigen-1
4.
J Virol ; 87(5): 2390-400, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23255796

ABSTRACT

Infection of mammalian cells by picornaviruses results in the nucleocytoplasmic redistribution of certain host cell proteins. These viruses interfere with import-export pathways, allowing for the cytoplasmic accumulation of nuclear proteins that are then available to function in viral processes. We recently described the cytoplasmic relocalization of cellular splicing factor SRp20 during poliovirus infection. SRp20 is an important internal ribosome entry site (IRES) trans-acting factor (ITAF) for poliovirus IRES-mediated translation; however, it is not known whether other picornaviruses utilize SRp20 as an ITAF and direct its cytoplasmic relocalization. Also, the mechanism by which poliovirus directs the accumulation of SRp20 in the cytoplasm of the infected cell is currently unknown. Work described in this report demonstrated that infection by another picornavirus (coxsackievirus B3) causes SRp20 to relocalize from the nucleus to the cytoplasm of HeLa cells, similar to poliovirus infection; however, SRp20 is relocalized to a somewhat lesser extent in the cytoplasm of HeLa cells during infection by yet another picornavirus (human rhinovirus 16). We show that expression of poliovirus 2A proteinase is sufficient to cause the nucleocytoplasmic redistribution of SRp20. Following expression of poliovirus 2A proteinase in HeLa cells, we detect cleavage of specific nuclear pore proteins known to be cleaved during poliovirus infection. We also find that expression of human rhinovirus 16 2A proteinase alone can cause efficient cytoplasmic relocalization of SRp20, despite the lower levels of SRp20 relocalization observed during rhinovirus infection compared to poliovirus. Taken together, these results further define the mechanism of SRp20 cellular redistribution during picornavirus infections, and they provide additional insight into some of the differences observed between human rhinovirus and other enterovirus infections.


Subject(s)
Cysteine Endopeptidases/metabolism , Picornaviridae Infections/metabolism , RNA-Binding Proteins/metabolism , Viral Proteins/metabolism , 3C Viral Proteases , Active Transport, Cell Nucleus , Cell Line, Tumor , Cell Nucleus/metabolism , Coxsackievirus Infections/metabolism , Cysteine Endopeptidases/biosynthesis , Cysteine Endopeptidases/genetics , Cytoplasm/metabolism , Enterovirus B, Human/metabolism , HeLa Cells , Humans , Membrane Glycoproteins/metabolism , Nuclear Pore Complex Proteins/metabolism , Nuclear Proteins/metabolism , Poliovirus/metabolism , Rhinovirus/metabolism , Serine-Arginine Splicing Factors , Trans-Activators/metabolism , Viral Proteins/biosynthesis , Viral Proteins/genetics
5.
Proc Natl Acad Sci U S A ; 109(36): 14634-9, 2012 Sep 04.
Article in English | MEDLINE | ID: mdl-22908287

ABSTRACT

A previously described mammalian cell activity, called VPg unlinkase, specifically cleaves a unique protein-RNA covalent linkage generated during the viral genomic RNA replication steps of a picornavirus infection. For over three decades, the identity of this cellular activity and its normal role in the uninfected cell had remained elusive. Here we report the purification and identification of VPg unlinkase as the DNA repair enzyme, 5'-tyrosyl-DNA phosphodiesterase-2 (TDP2). Our data show that VPg unlinkase activity in different mammalian cell lines correlates with their differential expression of TDP2. Furthermore, we show that recombinant TDP2 can cleave the protein-RNA linkage generated by different picornaviruses without impairing the integrity of viral RNA. Our results reveal a unique RNA repair-like function for TDP2 and suggest an unusual role in host-pathogen interactions for this cellular enzyme. On the basis of the identification of TDP2 as a potential antiviral target, our findings may lead to the development of universal therapeutics to treat the millions of individuals afflicted annually with diseases caused by picornaviruses, including myocarditis, aseptic meningitis, encephalitis, hepatitis, and the common cold.


Subject(s)
Nuclear Proteins/metabolism , Picornaviridae/metabolism , Ribonucleoproteins/metabolism , Transcription Factors/metabolism , Viral Proteins/metabolism , Amino Acid Sequence , Animals , Blotting, Western , DNA-Binding Proteins , Electrophoresis, Polyacrylamide Gel , Evolution, Molecular , Fluorescent Antibody Technique , HeLa Cells , Host-Pathogen Interactions , Humans , Microscopy, Confocal , Nuclear Proteins/genetics , Phosphoric Diester Hydrolases , Picornaviridae/genetics , Recombinant Proteins/metabolism , Transcription Factors/genetics
6.
PLoS Pathog ; 7(7): e1002127, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21779168

ABSTRACT

Poliovirus IRES-mediated translation requires the functions of certain canonical as well as non-canonical factors for the recruitment of ribosomes to the viral RNA. The interaction of cellular proteins PCBP2 and SRp20 in extracts from poliovirus-infected cells has been previously described, and these two proteins were shown to function synergistically in viral translation. To further define the mechanism of ribosome recruitment for the initiation of poliovirus IRES-dependent translation, we focused on the role of the interaction between cellular proteins PCBP2 and SRp20. Work described here demonstrates that SRp20 dramatically re-localizes from the nucleus to the cytoplasm of poliovirus-infected neuroblastoma cells during the course of infection. Importantly, SRp20 partially co-localizes with PCBP2 in the cytoplasm of infected cells, corroborating our previous in vitro interaction data. In addition, the data presented implicate the presence of these two proteins in viral translation initiation complexes. We show that in extracts from poliovirus-infected cells, SRp20 is associated with PCBP2 bound to poliovirus RNA, indicating that this interaction occurs on the viral RNA. Finally, we generated a mutated version of SRp20 lacking the RNA recognition motif (SRp20ΔRRM) and found that this protein is localized similar to the full length SRp20, and also partially co-localizes with PCBP2 during poliovirus infection. Expression of this mutated version of SRp20 results in a ∼100 fold decrease in virus yield for poliovirus when compared to expression of wild type SRp20, possibly via a dominant negative effect. Taken together, these results are consistent with a model in which SRp20 interacts with PCBP2 bound to the viral RNA, and this interaction functions to recruit ribosomes to the viral RNA in a direct or indirect manner, with the participation of additional protein-protein or protein-RNA interactions.


Subject(s)
Cell Nucleus/metabolism , Models, Biological , Poliomyelitis/metabolism , Poliovirus/metabolism , Protein Biosynthesis , RNA, Viral/metabolism , RNA-Binding Proteins/metabolism , Active Transport, Cell Nucleus/genetics , Amino Acid Motifs , Cell Nucleus/genetics , Cell Nucleus/virology , Cytoplasm/genetics , Cytoplasm/metabolism , Cytoplasm/virology , HeLa Cells , Humans , Mutation , Poliomyelitis/genetics , Poliovirus/genetics , Protein Binding , RNA, Viral/genetics , RNA-Binding Proteins/genetics , Serine-Arginine Splicing Factors
7.
J Virol ; 84(18): 9472-86, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20592073

ABSTRACT

We have previously characterized a 21-kDa protein encoded by UL138 (pUL138) as a viral factor inherent to low-passage strains of human cytomegalovirus (HCMV) that is required for latent infection in vitro. pUL138 is encoded on 3.6-, 2.7-, and 1.4-kb 3' coterminal transcripts that are produced during productive and latent infections. pUL138 is encoded at the 3' end of each transcript and is preceded by an extensive 5' sequence (approximately 0.5 to 2.5 kb) containing several putative open reading frames (ORFs). We determined that three putative ORFs upstream of UL138 (UL133, UL135, and UL136) encode proteins. The UL138 transcripts are polycistronic, such that each transcript expresses pUL138 in addition to the most-5' ORF. The upstream coding sequences (CDS) present a significant challenge for the translation of pUL138 in mammalian cells. We hypothesized that sequences 5' of UL138 mediate translation initiation of pUL138 by alternative strategies. Accordingly, a 663-nucloetide (nt) sequence overlapping the UL136 CDS supported expression of a downstream cistron in a bicistronic reporter system. We did not detect cryptic promoter activity or RNA splicing events that could account for downstream cistron expression. These data are consistent with the sequence element functioning as an internal ribosome entry site (IRES). Interestingly, pUL138 expression from the 3.6- and 2.7-kb transcripts was induced by serum stress, which concomitantly inhibited normal cap-dependent translation. Our work suggests that an alternative and stress-inducible strategy of translation initiation ensures expression of pUL138 under a variety of cellular contexts. The UL138 polycistronic transcripts serve to coordinate the expression of multiple proteins, including a viral determinant of HCMV latency.


Subject(s)
Cytomegalovirus/physiology , Protein Biosynthesis , Stress, Physiological , Viral Proteins/biosynthesis , Virus Latency , Humans , Promoter Regions, Genetic , Ribosomes/metabolism
8.
Biochim Biophys Acta ; 1789(9-10): 518-28, 2009.
Article in English | MEDLINE | ID: mdl-19631772

ABSTRACT

IRES elements are highly structured RNA sequences that function to recruit ribosomes for the initiation of translation. In contrast to the canonical cap-binding, ribosome-scanning model, the mechanism of IRES-mediated translation initiation is not well understood. IRES elements, first discovered in viral RNA genomes, were subsequently found in a subset of cellular RNAs as well. Interestingly, these cellular IRES-containing mRNAs appear to play important roles during conditions of cellular stress, development, and disease (e.g., cancer). It has been shown for viral IRESes that some require specific IRES trans-acting factors (ITAFs), while others require few if any additional proteins and can bind ribosomes directly. Current studies are aimed at elucidating the mechanism of IRES-mediated translation initiation and features that may be common or differ greatly among cellular and viral IRESes. This review will explore IRES elements as important RNA structures that function in both cellular and viral RNA translation and the significance of these structures in providing an alternative mechanism of eukaryotic translation initiation.


Subject(s)
Protein Biosynthesis , RNA, Messenger/genetics , Regulatory Elements, Transcriptional , Ribosomes/metabolism , Animals , Cell Nucleus/metabolism , Cytoplasm/metabolism , Genes, Viral , Guanosine Triphosphate/chemistry , Humans , Hydrolysis , Models, Genetic , Nucleic Acid Conformation , Picornaviridae/genetics , RNA/chemistry , RNA, Messenger/metabolism , RNA, Viral/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...