Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Cell ; 180(6): 1115-1129.e13, 2020 03 19.
Article in English | MEDLINE | ID: mdl-32200799

ABSTRACT

Influenza A virus (IAV) is a lytic RNA virus that triggers receptor-interacting serine/threonine-protein kinase 3 (RIPK3)-mediated pathways of apoptosis and mixed lineage kinase domain-like pseudokinase (MLKL)-dependent necroptosis in infected cells. ZBP1 initiates RIPK3-driven cell death by sensing IAV RNA and activating RIPK3. Here, we show that replicating IAV generates Z-RNAs, which activate ZBP1 in the nucleus of infected cells. ZBP1 then initiates RIPK3-mediated MLKL activation in the nucleus, resulting in nuclear envelope disruption, leakage of DNA into the cytosol, and eventual necroptosis. Cell death induced by nuclear MLKL was a potent activator of neutrophils, a cell type known to drive inflammatory pathology in virulent IAV disease. Consequently, MLKL-deficient mice manifest reduced nuclear disruption of lung epithelia, decreased neutrophil recruitment into infected lungs, and increased survival following a lethal dose of IAV. These results implicate Z-RNA as a new pathogen-associated molecular pattern and describe a ZBP1-initiated nucleus-to-plasma membrane "inside-out" death pathway with potentially pathogenic consequences in severe cases of influenza.


Subject(s)
Influenza A virus/genetics , Necroptosis/genetics , RNA-Binding Proteins/metabolism , Animals , Apoptosis/genetics , Cell Death/genetics , Cell Line, Tumor , Female , Influenza A virus/metabolism , Male , Mice , Mice, Inbred C57BL , Necrosis/metabolism , Phosphorylation , Protein Kinases/metabolism , RNA/metabolism , RNA, Double-Stranded/genetics , RNA, Double-Stranded/metabolism , RNA-Binding Proteins/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/physiology
2.
Nature ; 628(8009): 835-843, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38600381

ABSTRACT

Severe influenza A virus (IAV) infections can result in hyper-inflammation, lung injury and acute respiratory distress syndrome1-5 (ARDS), for which there are no effective pharmacological therapies. Necroptosis is an attractive entry point for therapeutic intervention in ARDS and related inflammatory conditions because it drives pathogenic lung inflammation and lethality during severe IAV infection6-8 and can potentially be targeted by receptor interacting protein kinase 3 (RIPK3) inhibitors. Here we show that a newly developed RIPK3 inhibitor, UH15-38, potently and selectively blocked IAV-triggered necroptosis in alveolar epithelial cells in vivo. UH15-38 ameliorated lung inflammation and prevented mortality following infection with laboratory-adapted and pandemic strains of IAV, without compromising antiviral adaptive immune responses or impeding viral clearance. UH15-38 displayed robust therapeutic efficacy even when administered late in the course of infection, suggesting that RIPK3 blockade may provide clinical benefit in patients with IAV-driven ARDS and other hyper-inflammatory pathologies.


Subject(s)
Lung Injury , Necroptosis , Orthomyxoviridae Infections , Protein Kinase Inhibitors , Receptor-Interacting Protein Serine-Threonine Kinases , Animals , Female , Humans , Male , Mice , Alveolar Epithelial Cells/pathology , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/virology , Alveolar Epithelial Cells/metabolism , Influenza A virus/classification , Influenza A virus/drug effects , Influenza A virus/immunology , Influenza A virus/pathogenicity , Lung Injury/complications , Lung Injury/pathology , Lung Injury/prevention & control , Lung Injury/virology , Mice, Inbred C57BL , Necroptosis/drug effects , Orthomyxoviridae Infections/complications , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/mortality , Orthomyxoviridae Infections/virology , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Respiratory Distress Syndrome/complications , Respiratory Distress Syndrome/pathology , Respiratory Distress Syndrome/prevention & control , Respiratory Distress Syndrome/virology
3.
Nature ; 606(7914): 594-602, 2022 06.
Article in English | MEDLINE | ID: mdl-35614224

ABSTRACT

Only a small proportion of patients with cancer show lasting responses to immune checkpoint blockade (ICB)-based monotherapies. The RNA-editing enzyme ADAR1 is an emerging determinant of resistance to ICB therapy and prevents ICB responsiveness by repressing immunogenic double-stranded RNAs (dsRNAs), such as those arising from the dysregulated expression of endogenous retroviral elements (EREs)1-4. These dsRNAs trigger an interferon-dependent antitumour response by activating A-form dsRNA (A-RNA)-sensing proteins such as MDA-5 and PKR5. Here we show that ADAR1 also prevents the accrual of endogenous Z-form dsRNA elements (Z-RNAs), which were enriched in the 3' untranslated regions of interferon-stimulated mRNAs. Depletion or mutation of ADAR1 resulted in Z-RNA accumulation and activation of the Z-RNA sensor ZBP1, which culminated in RIPK3-mediated necroptosis. As no clinically viable ADAR1 inhibitors currently exist, we searched for a compound that can override the requirement for ADAR1 inhibition and directly activate ZBP1. We identified a small molecule, the curaxin CBL0137, which potently activates ZBP1 by triggering Z-DNA formation in cells. CBL0137 induced ZBP1-dependent necroptosis in cancer-associated fibroblasts and reversed ICB unresponsiveness in mouse models of melanoma. Collectively, these results demonstrate that ADAR1 represses endogenous Z-RNAs and identifies ZBP1-mediated necroptosis as a new determinant of tumour immunogenicity masked by ADAR1. Therapeutic activation of ZBP1-induced necroptosis provides a readily translatable avenue for rekindling the immune responsiveness of ICB-resistant human cancers.


Subject(s)
Adenosine Deaminase , Necroptosis , Neoplasms , RNA-Binding Proteins , 3' Untranslated Regions , Adenosine Deaminase/metabolism , Animals , Cancer-Associated Fibroblasts , Carbazoles/pharmacology , Humans , Immunotherapy/trends , Interferons/metabolism , Melanoma , Mice , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , RNA, Double-Stranded/immunology , RNA-Binding Proteins/metabolism
4.
J Immunol ; 203(5): 1348-1355, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31358656

ABSTRACT

Receptor-interacting protein kinase 1 (RIPK1) regulates cell fate and proinflammatory signaling downstream of multiple innate immune pathways, including those initiated by TNF-α, TLR ligands, and IFNs. Genetic ablation of Ripk1 results in perinatal lethality arising from both RIPK3-mediated necroptosis and FADD/caspase-8-driven apoptosis. IFNs are thought to contribute to the lethality of Ripk1-deficient mice by activating inopportune cell death during parturition, but how IFNs activate cell death in the absence of RIPK1 is not understood. In this study, we show that Z-form nucleic acid binding protein 1 (ZBP1; also known as DAI) drives IFN-stimulated cell death in settings of RIPK1 deficiency. IFN-activated Jak/STAT signaling induces robust expression of ZBP1, which complexes with RIPK3 in the absence of RIPK1 to trigger RIPK3-driven pathways of caspase-8-mediated apoptosis and MLKL-driven necroptosis. In vivo, deletion of either Zbp1 or core IFN signaling components prolong viability of Ripk1-/- mice for up to 3 mo beyond parturition. Together, these studies implicate ZBP1 as the dominant activator of IFN-driven RIPK3 activation and perinatal lethality in the absence of RIPK1.


Subject(s)
Cell Death/physiology , RNA-Binding Proteins/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Apoptosis/physiology , Caspase 8/metabolism , Cell Line , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Signal Transduction/physiology
5.
J Immunol ; 200(10): 3626-3634, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29654208

ABSTRACT

The cytokine IFN-γ has well-established antibacterial properties against the bacterium Salmonella enterica in phagocytes, but less is known about the effects of IFN-γ on Salmonella-infected nonphagocytic cells, such as intestinal epithelial cells (IECs) and fibroblasts. In this article, we show that exposing human and murine IECs and fibroblasts to IFN-γ following infection with Salmonella triggers a novel form of cell death that is neither pyroptosis nor any of the major known forms of programmed cell death. Cell death required IFN-γ-signaling via STAT1-IRF1-mediated induction of guanylate binding proteins and the presence of live Salmonella in the cytosol. In vivo, ablating IFN-γ signaling selectively in murine IECs led to higher bacterial burden in colon contents and increased inflammation in the intestine of infected mice. Together, these results demonstrate that IFN-γ signaling triggers release of Salmonella from the Salmonella-containing vacuole into the cytosol of infected nonphagocytic cells, resulting in a form of nonpyroptotic cell death that prevents bacterial spread in the gut.


Subject(s)
Cell Death/immunology , Interferon-gamma/immunology , Phagocytes/immunology , Pyroptosis/immunology , Salmonella Infections/immunology , Salmonella enterica/immunology , 3T3 Cells , Animals , Cell Line , Cytosol/immunology , Cytosol/microbiology , Epithelial Cells/immunology , Epithelial Cells/microbiology , Fibroblasts/immunology , Fibroblasts/microbiology , Humans , Inflammation/immunology , Inflammation/microbiology , Interferon Regulatory Factor-1/immunology , Intestines/immunology , Intestines/microbiology , Mice , Phagocytes/microbiology , STAT1 Transcription Factor/immunology , Salmonella Infections/microbiology
6.
Mol Ther ; 25(5): 1168-1186, 2017 05 03.
Article in English | MEDLINE | ID: mdl-28366764

ABSTRACT

CRISPR-associated protein 9 (Cas9)-mediated genome editing provides a promising cure for HIV-1/AIDS; however, gene delivery efficiency in vivo remains an obstacle to overcome. Here, we demonstrate the feasibility and efficiency of excising the HIV-1 provirus in three different animal models using an all-in-one adeno-associated virus (AAV) vector to deliver multiplex single-guide RNAs (sgRNAs) plus Staphylococcus aureus Cas9 (saCas9). The quadruplex sgRNAs/saCas9 vector outperformed the duplex vector in excising the integrated HIV-1 genome in cultured neural stem/progenitor cells from HIV-1 Tg26 transgenic mice. Intravenously injected quadruplex sgRNAs/saCas9 AAV-DJ/8 excised HIV-1 proviral DNA and significantly reduced viral RNA expression in several organs/tissues of Tg26 mice. In EcoHIV acutely infected mice, intravenously injected quadruplex sgRNAs/saCas9 AAV-DJ/8 reduced systemic EcoHIV infection, as determined by live bioluminescence imaging. Additionally, this quadruplex vector induced efficient proviral excision, as determined by PCR genotyping in the liver, lungs, brain, and spleen. Finally, in humanized bone marrow/liver/thymus (BLT) mice with chronic HIV-1 infection, successful proviral excision was detected by PCR genotyping in the spleen, lungs, heart, colon, and brain after a single intravenous injection of quadruplex sgRNAs/saCas9 AAV-DJ/8. In conclusion, in vivo excision of HIV-1 proviral DNA by sgRNAs/saCas9 in solid tissues/organs can be achieved via AAV delivery, a significant step toward human clinical trials.


Subject(s)
Endonucleases/genetics , Genetic Therapy/methods , Genome, Viral , HIV Infections/therapy , HIV-1/genetics , Proviruses/genetics , RNA, Guide, Kinetoplastida/genetics , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Base Sequence , CRISPR-Cas Systems , Clustered Regularly Interspaced Short Palindromic Repeats , Dependovirus/genetics , Dependovirus/metabolism , Disease Models, Animal , Endonucleases/metabolism , Gene Editing/methods , Genetic Vectors/chemistry , Genetic Vectors/metabolism , HIV Infections/pathology , HIV Infections/virology , HIV Long Terminal Repeat , HIV-1/metabolism , Humans , Mice , Mice, Transgenic , Oligonucleotides/genetics , Oligonucleotides/metabolism , Proviruses/metabolism , RNA, Guide, Kinetoplastida/metabolism , Staphylococcus aureus/chemistry , Staphylococcus aureus/enzymology , gag Gene Products, Human Immunodeficiency Virus/genetics , gag Gene Products, Human Immunodeficiency Virus/metabolism , pol Gene Products, Human Immunodeficiency Virus/genetics , pol Gene Products, Human Immunodeficiency Virus/metabolism
7.
Biochem Biophys Res Commun ; 483(3): 923-929, 2017 02 12.
Article in English | MEDLINE | ID: mdl-27746176

ABSTRACT

GATA transcription factors regulate an array of genes important in cell proliferation and differentiation. Here we report the identification of regulator of G protein signaling 4 (RGS4) as a novel target for GATA-6 transcription factor. Although three sites (a, b, c) within the proximal region of rabbit RGS4 promoter for GATA transcription factors were predicted by bioinformatics analysis, only GATA-a site (16 bp from the core TATA box) is essential for RGS4 transcriptional regulation. RT-PCR analysis demonstrated that only GATA-6 was highly expressed in rabbit colonic smooth muscle cells but GATA-4/6 were expressed in cardiac myocytes and GATA-1/2/3 expressed in blood cells. Adenovirus-mediated expression of GATA-6 but not GATA-1 significantly increased the constitutive and IL-1ß-induced mRNA expression of the endogenous RGS4 in colonic smooth muscle cells. IL-1ß stimulation induced GATA-6 nuclear translocation and increased GATA-6 binding to RGS4 promoter. These data suggest that GATA factor could affect G protein signaling through regulating RGS4 expression, and GATA signaling may develop as a future therapeutic target for RGS4-related diseases.


Subject(s)
GATA6 Transcription Factor/metabolism , RGS Proteins/metabolism , Active Transport, Cell Nucleus/drug effects , Animals , Base Sequence , Binding Sites/genetics , Cells, Cultured , Colon/cytology , Colon/drug effects , Colon/metabolism , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/metabolism , GATA6 Transcription Factor/genetics , Gene Expression Regulation/drug effects , Interleukin-1beta/metabolism , Interleukin-1beta/pharmacology , Mutagenesis, Site-Directed , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Promoter Regions, Genetic , RGS Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rabbits , Signal Transduction
8.
Development ; 141(22): 4332-42, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25371367

ABSTRACT

Recently, emerging evidence has shown that Stat3 controls tumor cell migration and invasion. However, the molecular mechanisms by which Stat3 controls the cell movement remain largely unknown. Embryonic gastrula progenitors display coordinated and orientated migration, called collective cell migration. Collective cell migration is the simultaneous movement of multiple cells and is universally involved in physiological and pathological programs. Stat3 activity is required for the migration of gastrula progenitors, but it does not affect cell specification, thus suggesting that gastrula movements are an excellent model to provide insight into Stat3 control of cell migration in vivo. In this study, we reveal a novel mechanism by which Stat3 modulates extracellular matrix (ECM) assembly to control the coherence of collective migration of prechordal plate progenitors during zebrafish embryonic gastrulation. We show that Stat3 regulates the expression of Efemp2a in the prechordal plate progenitors that migrate anteriorly during gastrulation. Alteration of Stat3-Efemp2a signaling activity disrupted the configuration of fibronectin (FN) and laminin (LM) matrices, resulting in defective coherence of prechordal plate progenitor movements in zebrafish embryos. We demonstrate that Efemp2a acts as a downstream effector of Stat3 to promote ECM configuration for coherent collective cell migrations in vivo.


Subject(s)
Cell Movement/physiology , Endoderm/cytology , Extracellular Matrix Proteins/metabolism , Extracellular Matrix/physiology , Gastrulation/physiology , STAT3 Transcription Factor/metabolism , Stem Cells/physiology , Zebrafish Proteins/metabolism , Zebrafish/embryology , Adenoviridae , Animals , Chromatin Immunoprecipitation , Cloning, Molecular , DNA Primers/genetics , Dogs , Extracellular Matrix Proteins/genetics , Fluorescent Antibody Technique , Gene Knockdown Techniques , HeLa Cells , Human Umbilical Vein Endothelial Cells , Humans , Immunoblotting , Immunoprecipitation , In Situ Hybridization , Madin Darby Canine Kidney Cells , Morpholinos/genetics , Mutagenesis , STAT3 Transcription Factor/genetics , Time-Lapse Imaging , Zebrafish/genetics , Zebrafish Proteins/genetics
9.
Dig Dis Sci ; 61(7): 1925-40, 2016 07.
Article in English | MEDLINE | ID: mdl-26879904

ABSTRACT

BACKGROUND: The contractility of colonic smooth muscle is dysregulated due to immune/inflammatory responses in inflammatory bowel diseases. Inflammation in vitro induces up-regulation of regulator of G-protein signaling 4 (RGS4) expression in colonic smooth muscle cells. AIMS: To characterize the immune/inflammatory responses and RGS4 expression pattern in colonic smooth muscle after induction of colitis. METHODS: Colitis was induced in rabbits by intrarectal instillation of 2,4,6-trinitrobenzene sulfonic acid (TNBS). Innate/adaptive immune response RT-qPCR array was performed using colonic circular muscle strips. At 1-9 weeks after colonic intramuscular microinjection of lentivirus, the distal and proximal colons were collected, and muscle strips and dispersed muscle cells were prepared from circular muscle layer. Expression levels of RGS4 and NFκB signaling components were determined by Western blot analysis. The biological consequences of RGS4 knockdown were assessed by measurement of muscle contraction and phospholipase C (PLC)-ß activity in response to acetylcholine (ACh). RESULTS: Contraction in response to ACh was significantly inhibited in the inflamed colonic circular smooth muscle cells. RGS4, IL-1, IL-6, IL-8, CCL3, CD1D, and ITGB2 were significantly up-regulated, while IL-18, CXCR4, CD86, and C3 were significantly down-regulated in the inflamed muscle strips. RGS4 protein expression in the inflamed smooth muscles was dramatically increased. RGS4 stable knockdown in vivo augmented ACh-stimulated PLC-ß activity and contraction in colonic smooth muscle cells. CONCLUSION: Inflamed smooth muscle exhibits up-regulation of IL-1-related signaling components, Th1 cytokines and RGS4, and inhibition of contraction. Stable knockdown of endogenous RGS4 in colonic smooth muscle increases PLC-ß activity and contractile responses.


Subject(s)
Colon/drug effects , Inflammation/chemically induced , Muscle Contraction/immunology , Muscle, Smooth/pathology , Trinitrobenzenesulfonic Acid/toxicity , Animals , Cytokines/genetics , Cytokines/metabolism , Gene Expression Regulation/immunology , Inflammation/pathology , Rabbits , Up-Regulation
10.
Methods Mol Biol ; 2651: 277-284, 2023.
Article in English | MEDLINE | ID: mdl-36892774

ABSTRACT

Eukaryotic cells sense and respond to virus infections by detecting conserved virus-generated molecular structures, called pathogen-associated molecular patterns (PAMPs). PAMPs are usually produced by replicating viruses, but not typically seen in uninfected cells. Double-stranded RNA (dsRNA) is a common PAMP produced by most, if not all, RNA viruses, as well as by many DNA viruses. DsRNA can adopt either the right-handed (A-RNA) or the left-handed (Z-RNA) double-helical conformation. A-RNA is sensed by cytosolic pattern recognition receptors (PRRs) such as RIG-1-like receptor MDA-5 and the dsRNA-dependent protein kinase PKR. Z-RNA is detected by Zα domain containing PRRs, including Z-form nucleic acid binding protein 1 (ZBP1) and the p150 subunit of adenosine deaminase RNA specific 1 (ADAR1). We have recently shown that Z-RNA is generated during orthomyxovirus (e.g., influenza A virus) infections and serves as activating ligand for ZBP1. In this chapter, we describe our procedure for detecting Z-RNA in influenza A virus (IAV)-infected cells. We also outline how this procedure can be used to detect Z-RNA produced during vaccinia virus infection, as well as Z-DNA induced by a small-molecule DNA intercalator.


Subject(s)
DNA, Z-Form , RNA Viruses , Animals , Pathogen-Associated Molecular Pattern Molecules , RNA, Double-Stranded , Vaccinia virus/genetics , RNA Viruses/genetics , Receptors, Pattern Recognition/genetics , Mammals/genetics
11.
J Biol Chem ; 286(11): 9514-25, 2011 Mar 18.
Article in English | MEDLINE | ID: mdl-21212265

ABSTRACT

The high density lipoprotein (HDL) represents a class of lipid- and protein-containing particles and consists of two major apolipoproteins apoA-I and apoA-II. ApoA-II has been shown to be involved in the pathogenesis of insulin resistance, adiposity, diabetes, and metabolic syndrome. In embryo, apoa2 mRNAs are abundant in the liver, brain, lung, placenta, and in fish yolk syncytial layer (YSL), suggesting that apoa2 may perform a function during embryonic development. Here we find out that apoa2 modulates zebrafish embryonic development by regulating the organization of YSL. Disruption of apoa2 function in zebrafish caused chromosome fusing, which strongly blocked YSL nuclear division, inducing disorders in YSL organization and finally disturbing the embryonic epiboly. Purified native human apoA-II was able specifically to rescue the defects and induced nuclear division in zebrafish embryos and in human HeLa cells. The C terminus of apoA-II was required for the proper chromosome separation during nuclear division of YSL in zebrafish embryos and in human HeLa cells. Our data indicate that organization of YSL is required for blastoderm patterning and morphogenesis and suggest that apolipoprotein apoA-II is a novel factor of nuclear division in YSL involved in the regulation of early zebrafish embryonic morphogenesis and in mammalian cells for proliferation.


Subject(s)
Apolipoprotein A-II/metabolism , Blastoderm/metabolism , Body Patterning/physiology , Cell Nucleus Division/physiology , Giant Cells/metabolism , Morphogenesis/physiology , Animals , Apolipoprotein A-II/genetics , Apolipoprotein A-II/pharmacology , Blastoderm/cytology , Body Patterning/drug effects , Cell Nucleus Division/drug effects , Chromosomes/genetics , Chromosomes/metabolism , Giant Cells/cytology , HeLa Cells , Humans , Morphogenesis/drug effects , Zebrafish
12.
J Exp Med ; 217(11)2020 11 02.
Article in English | MEDLINE | ID: mdl-32797196

ABSTRACT

Influenza A virus (IAV) activates ZBP1-initiated RIPK3-dependent parallel pathways of necroptosis and apoptosis in infected cells. Although mice deficient in both pathways fail to control IAV and succumb to lethal respiratory infection, RIPK3-mediated apoptosis by itself can limit IAV, without need for necroptosis. However, whether necroptosis, conventionally considered a fail-safe cell death mechanism to apoptosis, can restrict IAV-or indeed any virus-in the absence of apoptosis is not known. Here, we use mice selectively deficient in IAV-activated apoptosis to show that necroptosis drives robust antiviral immune responses and promotes effective virus clearance from infected lungs when apoptosis is absent. We also demonstrate that apoptosis and necroptosis are mutually exclusive fates in IAV-infected cells. Thus, necroptosis is an independent, "stand-alone" cell death mechanism that fully compensates for the absence of apoptosis in antiviral host defense.


Subject(s)
Caspase 8/genetics , Host Microbial Interactions/genetics , Influenza A virus/immunology , Necroptosis/genetics , Orthomyxoviridae Infections/immunology , Adaptive Immunity , Animals , Apoptosis/genetics , Apoptosis/immunology , Caspase 8/metabolism , Female , Gene Knock-In Techniques , Host Microbial Interactions/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Necroptosis/immunology , Orthomyxoviridae Infections/virology , RNA-Binding Proteins/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
13.
Cell Res ; 33(5): 333-334, 2023 05.
Article in English | MEDLINE | ID: mdl-36792808

Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Lung
14.
J Mol Histol ; 47(5): 437-44, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27554369

ABSTRACT

The differentiation potentials of stem cells have been evaluated by various in vivo and in vitro assays. However, these assays have different limitations hindering efficient study of mammalian stem cells. Here we describe a rapid and powerful mosaic assay to study the differentiation potentials of stem cells in real time in vivo by using zebrafish embryo. We transplanted mouse neural stem cells into zebrafish embryos at different developmental stages and found that they mainly formed neural tissues while occasionally trans-differentiated into mesoderm- and endoderm-derived tissues. Because zebrafish embryo is transparent, the behaviors of transplanted mouse stem cells can be easily tracked in a real-time manner and at single-cell resolution. We expect that this assay may be widely applied to explore the in vivo behaviors of any stem cells available.


Subject(s)
Mosaicism , Stem Cells/cytology , Animals , Animals, Genetically Modified , Cell Culture Techniques , Cell Differentiation , Cell Transdifferentiation , Endoderm/cytology , Mesoderm/cytology , Mice , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Stem Cells/metabolism , Zebrafish
15.
AIDS ; 30(8): 1163-74, 2016 05 15.
Article in English | MEDLINE | ID: mdl-26990633

ABSTRACT

OBJECTIVE: There is an urgent need for the development of HIV-1 genome eradication strategies that lead to a permanent cure for HIV-1/AIDS. We previously reported that four guide RNAs (gRNAs) targeting HIV-1 long terminal repeats (LTR) effectively eradicated the entire HIV-1 genome. In this study, we sought to identify the best gRNAs targeting HIV-1 LTR and viral structural region and optimize gRNA pairing that can efficiently eradicate the HIV-1 genome. DESIGN: Highly specific gRNAs were designed using bioinformatics tools, and their capacity of guiding CRISPR-associated system 9 to cleave HIV-1 proviral DNA was evaluated using high-throughput HIV-1 luciferase reporter assay and rapid Direct-PCR genotyping. METHODS: The target seed sequences for each gRNA were cloned into lentiviral vectors. HEK293T cells were cotransfected with a pEcoHIV-NL4-3-firefly-luciferase reporter vector (1 : 20) over lentiviral vectors carrying CRISPR-associated system 9 and single gRNA or various combinations of gRNAs. The EcoHIV DNA cleaving efficiency was evaluated by Direct-PCR genotyping, and the EcoHIV transcription/replication activity was examined by a luciferase reporter assay. RESULTS: Most of the designed gRNAs are effective to eliminate the predicted HIV-1 genome sequence between the selected two target sites. This is evidenced by the presence of PCR genotypic deletion/insertion and the decrease of luciferase reporter activity. In particular, a combination of viral structural gRNAs with LTR gRNAs provided a higher efficiency of genome eradication and an easier approach for PCR genotyping. CONCLUSION: Our screening strategy can specifically and effectively identify gRNAs targeting HIV-1 LTR and structural region to excise proviral HIV-1 from the host genome.


Subject(s)
Acquired Immunodeficiency Syndrome/virology , Anti-HIV Agents/metabolism , Biological Products/metabolism , Gene Expression Regulation, Viral , Genetic Testing , HIV-1/drug effects , RNA, Guide, Kinetoplastida/metabolism , Acquired Immunodeficiency Syndrome/therapy , Computational Biology , Genes, Reporter , Genome, Viral , Genotyping Techniques , HEK293 Cells , HIV-1/genetics , Humans , Luciferases/analysis , Luciferases/genetics , Polymerase Chain Reaction , Proviruses/drug effects , Proviruses/genetics , RNA, Guide, Kinetoplastida/genetics , Recombination, Genetic , Virus Latency/drug effects
16.
Sci Rep ; 5: 16277, 2015 Nov 05.
Article in English | MEDLINE | ID: mdl-26538064

ABSTRACT

Current antiretroviral therapy does not eliminate the integrated and transcriptionally silent HIV-1 provirus in latently infected cells. Recently, a "shock and kill" strategy has been extensively explored to eradicate the HIV-1 latent reservoirs for a permanent cure of AIDS. The therapeutic efficacy of currently used agents remains disappointing because of low efficiency, non-specificity and cellular toxicity. Here we present a novel catalytically-deficient Cas9-synergistic activation mediator (dCas9-SAM) technology to selectively, potently and persistently reactivate the HIV-1 latent reservoirs. By screening 16 MS2-mediated single guide RNAs, we identified long terminal repeat (LTR)-L and O that surround the enhancer region (-165/-145 for L and -92/-112 for O) and induce robust reactivation of HIV-1 provirus in HIV-1 latent TZM-bI epithelial, Jurkat T lymphocytic and CHME5 microglial cells. This compulsory reactivation induced cellular suicide via toxic buildup of viral proteins within HIV-1 latent Jurkat T and CHME5 microglial cells. These results suggest that this highly effective and target-specific dCas9-SAM system can serve as a novel HIV-latency-reversing therapeutic tool for the permanent elimination of HIV-1 latent reservoirs.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats/genetics , HIV Infections/virology , HIV-1/genetics , RNA, Guide, Kinetoplastida/genetics , Virus Activation/genetics , Virus Latency/genetics , Cell Line, Tumor , HIV Long Terminal Repeat/genetics , Humans , Jurkat Cells , Proviruses/genetics , T-Lymphocytes/virology
17.
Sci Rep ; 4: 5831, 2014 Jul 25.
Article in English | MEDLINE | ID: mdl-25060222

ABSTRACT

Normal interfollicular epidermis (IFE) homeostasis is maintained throughout the entire life by its own stem cells that self-renew and generate progeny that undergo terminal differentiation. However, the fine markers of the stem cells in interfollicular epidermis are not well defined yet. Here we found that TLR7 identified the existence of progenitors and interfollicular epidermal stem cells in murine skin. In vitro, TLR7-expressing cells comprised of two subpopulations that were competent to proliferate and exhibited distinct differentiation potentials. Three-dimensional (3D) organotypic culture and skin reconstitution assays showed that TLR7-expressing cells were able to reconstruct the interfollicular epidermis. Finally, TLR7-expressing cells maintained the intact interfollicular epidermal structures revealed in serial transplantation assays in vivo in mice. Taken together, our results suggest that TLR7-expressing cells comprise an interfollicular epidermal stem cell population.


Subject(s)
Epidermal Cells , Stem Cells/cytology , Toll-Like Receptor 7/metabolism , Aminoquinolines/pharmacology , Animals , Cell Culture Techniques , Cell Proliferation/drug effects , Cells, Cultured , Epidermis/metabolism , Epidermis/pathology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hair Follicle/metabolism , Imiquimod , Keratinocytes/cytology , Keratinocytes/drug effects , Keratinocytes/transplantation , Mice , Mice, Inbred C57BL , Mice, Transgenic , Regeneration , Skin/metabolism , Skin/pathology , Stem Cell Transplantation , Stem Cells/metabolism , Tissue Engineering , Toll-Like Receptor 7/agonists , Toll-Like Receptor 7/genetics
SELECTION OF CITATIONS
SEARCH DETAIL