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.
Microbiol Spectr ; 9(1): e0039221, 2021 09 03.
Article in English | MEDLINE | ID: mdl-34431687

ABSTRACT

Tests to diagnose acute SARS-CoV-2 infection are at the center of controlling the COVID-19 pandemic. Rapid tests benefit from providing quick results but suffer from lower sensitivity, while PCR tests usually take longer to provide more reliable results and can be difficult to scale to meet population needs. We evaluated the diagnostic efficacy of a Molecular Mirror assay (MMA) using nucleic acid extraction and a nucleic acid extraction-free method to determine its ability to identify SARS-CoV-2 in nasal specimens from individuals suspected of having SARS-CoV-2. We compared the MMA using nucleic acid extraction to the emergency use authorization (EUA)-approved TaqPath reverse transcriptase PCR (RT-PCR) assay to determine its performance characteristics. From 412 total specimens (including 115 previous positives and 297 previous negatives), we found that the positive percent agreement (PPA) was 99.1% (confidence interval [CI], 97.4% to 100.0%) and the negative percent agreement (NPA) was 99.3% (95% CI, 98.4% to 100.0%) for SARS-CoV-2 detection. Using the extraction-free method, we analyzed 109 specimens (51 previous positives and 58 previous negatives) and found that the PPA for the more rapid version of the assay was 87.8% (95% CI, 78.5% to 96.9%) and the NPA was 100.0% (95% CI, 100.0%) for virus detection. The extraction method has performance comparable to what is observed in many PCR-based assays. The extraction-free method has lower PPA but has the advantage of being more rapid and having a higher throughput. Our data offer a proof of concept that nuclear magnetic resonance (NMR) detection can be used in SARS-CoV-2 diagnostic testing and may allow for alternative supply chains to increase testing options. IMPORTANCE Accurate diagnostics for SARS-CoV-2 infections have been critical for responding to the COVID-19 pandemic. Both high-sensitivity/specificity PCR-based tests and lower-sensitivity/specificity rapid antigen assays have been the subject of worldwide supply chain limitations as individual facilities and countries have struggled to meet their population testing needs. We evaluated the diagnostic efficacy of a Molecular Mirror assay (MMA), which uses nuclear magnetic resonance to detect the presence of SARS-CoV-2 nucleic acids both with and without full nucleic acid extractions. We found that compared to a U.S. emergency use authorization (EUA) approved assay (TaqPath) that uses reverse transcriptase PCR (RT-PCR), the MMA had high PPA and NPA with full nucleic acid extractions, and acceptable positive percent agreement (PPA) and negative percent agreement (NPA) with an extraction-free protocol. In a landscape marred by supply chain shortages across the world, altered SARS-CoV-2 detection methods such as the MMA can add to testing supplies while providing quality SARS-CoV-2 testing results.


Subject(s)
COVID-19 Nucleic Acid Testing/methods , COVID-19/diagnosis , High-Throughput Screening Assays/methods , SARS-CoV-2/isolation & purification , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Female , Humans , Infant , Male , Middle Aged , Molecular Diagnostic Techniques/methods , Pandemics , Polymerase Chain Reaction , Sensitivity and Specificity , Technology , Young Adult
2.
Sci Adv ; 7(13)2021 03.
Article in English | MEDLINE | ID: mdl-33771858

ABSTRACT

Inhibition of type 1 interferon (IFN-I) signaling promotes the control of persistent virus infection, but the underlying mechanisms remain poorly understood. Here, we report that genetic ablation of Ifnar1 specifically in natural killer (NK) cells led to elevated numbers of T follicular helper cells, germinal center B cells, and plasma cells and improved antiviral T cell function, resulting in hastened virus clearance that was comparable to IFNAR1 neutralizing antibody treatment. Antigen-specific B cells and antiviral antibodies were essential for the accelerated control of LCMV Cl13 infection following IFNAR1 blockade. IFNAR1 signaling in NK cells promoted NK cell function and general killing of antigen-specific CD4 and CD8 T cells. Therefore, inhibition of IFN-I signaling in NK cells enhances CD4 and CD8 T cell responses, promotes humoral immune responses, and thereby facilitates the control of persistent virus infection.


Subject(s)
Interferon Type I , Virus Diseases , Animals , Antiviral Agents , CD8-Positive T-Lymphocytes , Humans , Killer Cells, Natural , Mice , Mice, Inbred C57BL , Receptor, Interferon alpha-beta/genetics
3.
PLoS Genet ; 13(2): e1006623, 2017 02.
Article in English | MEDLINE | ID: mdl-28241004

ABSTRACT

MicroRNAs (miRNAs) are thought to exert their functions by modulating the expression of hundreds of target genes and each to a small degree, but it remains unclear how small changes in hundreds of target genes are translated into the specific function of a miRNA. Here, we conducted an integrated analysis of transcriptome and translatome of primary B cells from mutant mice expressing miR-17~92 at three different levels to address this issue. We found that target genes exhibit differential sensitivity to miRNA suppression and that only a small fraction of target genes are actually suppressed by a given concentration of miRNA under physiological conditions. Transgenic expression and deletion of the same miRNA gene regulate largely distinct sets of target genes. miR-17~92 controls target gene expression mainly through translational repression and 5'UTR plays an important role in regulating target gene sensitivity to miRNA suppression. These findings provide molecular insights into a model in which miRNAs exert their specific functions through a small number of key target genes.


Subject(s)
B-Lymphocytes/metabolism , Gene Expression Regulation , MicroRNAs/genetics , Protein Biosynthesis/genetics , Transcriptome/genetics , 5' Untranslated Regions/genetics , Animals , B-Lymphocytes/cytology , Base Sequence , Bcl-2-Like Protein 11/genetics , Bcl-2-Like Protein 11/metabolism , Cells, Cultured , Flow Cytometry , Gene Expression Profiling/methods , Immunoblotting , Mice, Knockout , Mice, Transgenic , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Ribosomes/genetics , Ribosomes/metabolism
4.
Nat Commun ; 7: 12207, 2016 08 02.
Article in English | MEDLINE | ID: mdl-27481093

ABSTRACT

The molecular mechanisms that regulate B-cell development and tolerance remain incompletely understood. In this study, we identify a critical role for the miR-17∼92 microRNA cluster in regulating B-cell central tolerance and demonstrate that these miRNAs control early B-cell development in a cell-intrinsic manner. While the cluster member miR-19 suppresses the expression of Pten and plays a key role in regulating B-cell tolerance, miR-17 controls early B-cell development through other molecular pathways. These findings demonstrate differential control of two closely linked B-cell developmental stages by different members of a single microRNA cluster through distinct molecular pathways.


Subject(s)
B-Lymphocytes/physiology , Immune Tolerance/genetics , Lymphocyte Activation/genetics , MicroRNAs/physiology , PTEN Phosphohydrolase/genetics , Animals , Cell Differentiation/genetics , Cells, Cultured , Female , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Models, Animal
5.
J Exp Med ; 213(9): 1901-19, 2016 08 22.
Article in English | MEDLINE | ID: mdl-27481129

ABSTRACT

MicroRNA (miRNA) deficiency impairs the generation of T follicular helper (Tfh) cells, but the contribution of individual miRNAs to this phenotype remains poorly understood. In this study, we performed deep sequencing analysis of miRNAs expressed in Tfh cells and identified a five-miRNA signature. Analyses of mutant mice deficient of these miRNAs revealed that miR-22 and miR-183/96/182 are dispensable, but miR-155 is essential for the generation and function of Tfh cells. miR-155 deficiency led to decreased proliferation specifically at the late stage of Tfh cell differentiation and reduced CD40 ligand (CD40L) expression on antigen-specific CD4(+) T cells. Mechanistically, miR-155 repressed the expression of Peli1, a ubiquitin ligase that promotes the degradation of the NF-κB family transcription factor c-Rel, which controls cellular proliferation and CD40L expression. Therefore, our study identifies a novel miR-155-Peli1-c-Rel pathway that specifically regulates Tfh cell generation and function.


Subject(s)
MicroRNAs/physiology , Nuclear Proteins/physiology , Proto-Oncogene Proteins c-rel/physiology , Signal Transduction/physiology , T-Lymphocytes, Helper-Inducer/immunology , Ubiquitin-Protein Ligases/physiology , Animals , CD40 Ligand/analysis , Cell Differentiation , Mice , Mice, Inbred C57BL , NF-kappa B/physiology , T-Lymphocytes, Cytotoxic/immunology
6.
Nat Immunol ; 17(4): 433-40, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26901150

ABSTRACT

Autoreactive B cells have critical roles in a large diversity of autoimmune diseases, but the molecular pathways that control these cells remain poorly understood. We performed an in vivo functional screen of a lymphocyte-expressed microRNA library and identified miR-148a as a potent regulator of B cell tolerance. Elevated miR-148a expression impaired B cell tolerance by promoting the survival of immature B cells after engagement of the B cell antigen receptor by suppressing the expression of the autoimmune suppressor Gadd45α, the tumor suppressor PTEN and the pro-apoptotic protein Bim. Furthermore, increased expression of miR-148a, which occurs frequently in patients with lupus and lupus-prone mice, facilitated the development of lethal autoimmune disease in a mouse model of lupus. Our studies demonstrate a function for miR-148a as a regulator of B cell tolerance and autoimmunity.


Subject(s)
Apoptosis/genetics , Autoimmunity/genetics , B-Lymphocytes/immunology , Immune Tolerance/genetics , MicroRNAs/genetics , Animals , Apoptosis/immunology , Apoptosis Regulatory Proteins/metabolism , Autoimmunity/immunology , Bcl-2-Like Protein 11 , Bone Marrow Transplantation , Cell Cycle Proteins/metabolism , Cell Proliferation/genetics , Disease Models, Animal , HEK293 Cells , Humans , Immune Tolerance/immunology , Immunoblotting , Lupus Erythematosus, Systemic/immunology , Membrane Proteins/metabolism , Mice , Mice, Inbred MRL lpr , MicroRNAs/immunology , Nuclear Proteins/metabolism , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, RNA
7.
PLoS One ; 10(4): e0124661, 2015.
Article in English | MEDLINE | ID: mdl-25884630

ABSTRACT

Cre-recombinase mediated conditional deletion of Lox-P site flanked ("floxed") genes is widely used for functional gene annotation in mice. Many different Cre-transgenic mouse lines have been developed for cell-type specific gene disruption. But often, the precise tissue-patterns of Cre activity remain incompletely characterized. Two widely used transgenes for conditional gene recombination in hematopoietic cells are Vav-iCre driven from the murine Vav1 promotor, and hCD2-iCre driven from the human CD2 promotor. Vav-iCre expresses active Cre in fetal and adult hematopoietic stem cells and all descendants, hCD2-iCre in immature and mature B and T lymphocytes. To better characterize which hematopoietic cells contain hCD2-iCre activity, we compared EYFP fluorescence in hCD2-iCre+/- R26-stop-EYFP+/- and Vav-iCre+/- R26-stop-EYFP+/-mice. R26-stop-EYFP ubiquitously encodes EYFP preceded by a floxed stop cassette. By removing it, Cre activity induces measurable EYFP expression. Our results confirm the known activity patterns for both Cre transgenes and unveil additional hCD2-iCre mediated reporter gene recombination in common lymphoid progenitors, in natural killer cells and their progenitors, and in plasmacytoid and conventional dendritic cells. This supports previously proposed common lymphoid origins for natural killer cells and subsets of dendritic cells, and indicates the need to consider pleiotropic effects when studying hCD2-iCre mediated conditional knockout mice. Vav-iCre+/- R26-stop-EYFP+/-mice did not show the non-hematopoietic recombination in vascular endothelial cells seen in other Vav-Cre mouse lines, but displayed an unexpected Vav-iCre mediated recombination in a bone cell subset lacking hematopoietic markers. This pinpoints the need to consider stromal cell contributions to phenotypes of Vav-iCre mediated conditional knockout mice. Altogether, our data provide the first detailed assessment of hCD2-iCre and Vav-iCre mediated deletion of floxed genes during lymphocyte development from hematopoietic stem cells and open up novel applications for either Cre-transgenic mouse line.


Subject(s)
Bone Marrow Cells/metabolism , CDC2 Protein Kinase/genetics , Integrases/genetics , Recombination, Genetic , Animals , Mice
8.
EMBO J ; 32(17): 2377-91, 2013 Aug 28.
Article in English | MEDLINE | ID: mdl-23921550

ABSTRACT

MicroRNAs (miRNAs) have been broadly implicated in cancer, but their exact function and mechanism in carcinogenesis remain poorly understood. Elevated miR-17~92 expression is frequently found in human cancers, mainly due to gene amplification and Myc-mediated transcriptional upregulation. Here we show that B cell-specific miR-17~92 transgenic mice developed lymphomas with high penetrance and that, conversely, Myc-driven lymphomagenesis stringently requires two intact alleles of miR-17~92. We experimentally identified miR-17~92 target genes by PAR-CLIP and validated select target genes in miR-17~92 transgenic mice. These analyses demonstrate that miR-17~92 drives lymphomagenesis by suppressing the expression of multiple negative regulators of the PI3K and NFκB pathways and by inhibiting the mitochondrial apoptosis pathway. Accordingly, miR-17~92-driven lymphoma cells exhibited constitutive activation of the PI3K and NFκB pathways and chemical inhibition of either pathway reduced tumour size and prolonged the survival of lymphoma-bearing mice. These findings establish miR-17~92 as a powerful cancer driver that coordinates the activation of multiple oncogenic pathways, and demonstrate for the first time that chemical inhibition of miRNA downstream pathways has therapeutic value in treating cancers caused by miRNA dysregulation.


Subject(s)
Gene Expression Regulation, Neoplastic , Lymphoma/genetics , MicroRNAs/physiology , Animals , B-Lymphocytes/pathology , B-Lymphocytes/physiology , Burkitt Lymphoma/genetics , Burkitt Lymphoma/pathology , Cell Proliferation , Cell Survival/genetics , Homeodomain Proteins/genetics , Humans , Imidazoles/pharmacology , Lymphoma/metabolism , Lymphoma/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Morpholines/pharmacology , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/administration & dosage , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Quinoxalines/pharmacology , RNA, Long Noncoding , Reproducibility of Results
9.
Nat Immunol ; 14(8): 849-57, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23812097

ABSTRACT

Follicular helper T cells (T(FH) cells) provide critical help to B cells during humoral immune responses. Here we report that mice with T cell-specific deletion of the miR-17∼92 family of microRNAs (miRNAs) had substantially compromised T(FH) differentiation, germinal-center formation and antibody responses and failed to control chronic viral infection. Conversely, mice with T cell-specific expression of a transgene encoding miR-17∼92 spontaneously accumulated T(FH) cells and developed a fatal immunopathology. Mechanistically, the miR-17∼92 family controlled the migration of CD4(+) T cells into B cell follicles by regulating signaling intensity from the inducible costimulator ICOS and kinase PI(3)K by suppressing expression of the phosphatase PHLPP2. Our findings demonstrate an essential role for the miR-17∼92 family in T(FH) differentiation and establish PHLPP2 as an important mediator of their function in this process.


Subject(s)
B-Lymphocytes/immunology , Cell Differentiation/immunology , Germinal Center/immunology , MicroRNAs/immunology , Nuclear Proteins/immunology , Phosphoprotein Phosphatases/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Flow Cytometry , Germinal Center/cytology , Immunity, Humoral/immunology , Immunohistochemistry , Inducible T-Cell Co-Stimulator Protein/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Phosphatidylinositol 3-Kinases/immunology , Signal Transduction/immunology , Specific Pathogen-Free Organisms , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...