Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nucleic Acids Res ; 50(8): e44, 2022 05 06.
Article in English | MEDLINE | ID: mdl-34967412

ABSTRACT

Many pathological processes are driven by RNA-protein interactions, making such interactions promising targets for molecular interventions. HIV-1 assembly is one such process, in which the viral genomic RNA interacts with the viral Gag protein and serves as a scaffold to drive Gag multimerization that ultimately leads to formation of a virus particle. Here, we develop self-assembled RNA nanostructures that can inhibit HIV-1 virus assembly, achieved through hybridization of multiple artificial small RNAs with a stem-loop structure (STL) that we identify as a prominent ligand of Gag that can inhibit virus particle production via STL-Gag interactions. The resulting STL-decorated nanostructures (double and triple stem-loop structures denoted as Dumbbell and Tribell, respectively) can elicit more pronounced viral blockade than their building blocks, with the inhibition arising as a result of nanostructures interfering with Gag multimerization. These findings could open up new avenues for RNA-based therapy.


Subject(s)
HIV-1 , Nanostructures , HIV-1/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , Virion/metabolism , Virus Assembly/physiology , gag Gene Products, Human Immunodeficiency Virus/genetics , gag Gene Products, Human Immunodeficiency Virus/metabolism
2.
Cell Immunol ; 360: 104275, 2021 02.
Article in English | MEDLINE | ID: mdl-33421676

ABSTRACT

The liver contains a rich mix of T cells, including activated T cells, tissue-resident memory T cells and cells undergoing apoptosis. When antigens are presented in this milieu the default result is functional tolerance. T cell tolerance in the liver could be constitutive, or it could be adaptive, in which case liver cells would become unresponsive after encountering antigen in the liver context. To test this model, we evaluated the potential of human liver T cells to respond to T cell receptor ligation in liver tissue slice cultures. These T cells contained an actively motile subset of CD4+ T cells marked by CCR7 and CD62L, and fully functional subsets of CD4+ and CD8+ T cells that synthesized effector cytokines but subsequently assumed an exhausted phenotype. These data favor the model that human liver T cells are not constitutively tolerant but undergo adaptive tolerance after activation.


Subject(s)
Immune Tolerance/immunology , Liver/immunology , T-Lymphocytes/metabolism , Adaptive Immunity/immunology , Antigens , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Humans , Immunologic Memory/immunology , Liver/pathology , Lymphocyte Activation/immunology , Phenotype , T-Lymphocyte Subsets/immunology , T-Lymphocytes/immunology
3.
Nucleic Acids Res ; 47(20): e131, 2019 11 18.
Article in English | MEDLINE | ID: mdl-31504824

ABSTRACT

Clustered regularly interspaced short palindromic repeats (CRISPR)-based genomic imaging systems predominantly rely on fluorescent protein reporters, which lack the optical properties essential for sensitive dynamic imaging. Here, we modified the CRISPR single-guide RNA (sgRNA) to carry two distinct molecular beacons (MBs) that can undergo fluorescence resonance energy transfer (FRET) and demonstrated that the resulting system, CRISPR/dual-FRET MB, enables dynamic imaging of non-repetitive genomic loci with only three unique sgRNAs.


Subject(s)
CRISPR-Cas Systems , Fluorescence Resonance Energy Transfer/methods , Genetic Loci , Fluorescent Dyes/chemistry , HeLa Cells , Humans , RNA, Guide, Kinetoplastida/chemistry , RNA, Guide, Kinetoplastida/metabolism
4.
Proc Natl Acad Sci U S A ; 115(26): 6721-6726, 2018 06 26.
Article in English | MEDLINE | ID: mdl-29891653

ABSTRACT

During HIV-1 assembly, the retroviral structural protein Gag forms an immature capsid, containing thousands of Gag molecules, at the plasma membrane (PM). Interactions between Gag nucleocapsid (NC) and viral RNA (vRNA) are thought to drive assembly, but the exact roles of these interactions have remained poorly understood. Since previous studies have shown that Gag dimer- or trimer-forming mutants (GagZiL) lacking an NC domain can form immature capsids independent of RNA binding, it is often hypothesized that vRNA drives Gag assembly by inducing Gag to form low-ordered multimers, but is dispensable for subsequent assembly. In this study, we examined the role of vRNA in HIV-1 assembly by characterizing the distribution and mobility of Gag and Gag NC mutants at the PM using photoactivated localization microscopy (PALM) and single-particle tracking PALM (spt-PALM). We showed that both Gag and GagZiL assembly involve a similar basic assembly unit, as expected. Unexpectedly, the two proteins underwent different subsequent assembly pathways, with Gag cluster density increasing asymptotically, while GagZiL cluster density increased linearly. Additionally, the directed movement of Gag, but not GagZiL, was maintained at a constant speed, suggesting that the two proteins experience different external driving forces. Assembly was abolished when Gag was rendered monomeric by NC deletion. Collectively, these results suggest that, beyond inducing Gag to form low-ordered multimer basic assembly units, vRNA is essential in scaffolding and maintaining the stability of the subsequent assembly process. This finding should advance the current understanding of HIV-1 and, potentially, other retroviruses.


Subject(s)
RNA, Viral/metabolism , Single Molecule Imaging , Virus Assembly/physiology , gag Gene Products, Human Immunodeficiency Virus/metabolism , Animals , COS Cells , Chlorocebus aethiops , Diffusion , HIV-1/metabolism , Nucleocapsid/metabolism , Protein Binding , Protein Domains , Proviruses/metabolism , Recombinant Proteins/metabolism
5.
J Virol ; 93(4)2019 02 15.
Article in English | MEDLINE | ID: mdl-30518652

ABSTRACT

Distinct populations of hepatocytes infected with hepatitis B virus (HBV) or only harboring HBV DNA integrations coexist within an HBV chronically infected liver. These hepatocytes express HBV antigens at different levels and with different intracellular localizations, but it is not known whether this heterogeneity of viral antigen expression could result in an uneven hepatic presentation of distinct HBV epitopes/HLA class I complexes triggering different levels of activation of HBV-specific CD8+ T cells. Using antibodies specific to two distinct HLA-A*02:01/HBV epitope complexes of HBV nucleocapsid and envelope proteins, we mapped their topological distributions in liver biopsy specimens of two anti-hepatitis B e antigen-positive (HBe+) chronic HBV (CHB) patients. We demonstrated that the core and envelope CD8+ T cell epitopes were not uniformly distributed in the liver parenchyma but preferentially located in distinct and sometimes mutually exclusive hepatic zones. The efficiency of HBV epitope presentation was then tested in vitro utilizing HLA-A*02:01/HBV epitope-specific antibodies and the corresponding CD8+ T cells in primary human hepatocyte and hepatoma cell lines either infected with HBV or harboring HBV DNA integration. We confirmed the existence of a marked variability in the efficiency of HLA class I/HBV epitope presentation among the different targets that was influenced by the presence of gamma interferon (IFN-γ) and availability of newly translated viral antigens. In conclusion, HBV antigen presentation can be heterogeneous within an HBV-infected liver. As a consequence, CD8+ T cells of different HBV specificities might have different antiviral efficacies.IMPORTANCE The inability of patients with chronic HBV infection to clear HBV is associated with defective HBV-specific CD8+ T cells. Hence, the majority of immunotherapy developments focus on HBV-specific T cell function restoration. However, knowledge of whether distinct HBV-specific T cells can equally target all the HBV-infected hepatocytes of a chronically infected liver is lacking. In this work, analysis of CHB patient liver parenchyma and in vitro HBV infection models shows a nonuniform distribution of HBV CD8+ T cell epitopes that is influenced by the presence of IFN-γ and availability of newly translated viral antigens. These results suggest that CD8+ T cells recognizing different HBV epitopes can be necessary for efficient immune therapeutic control of chronic HBV infection.


Subject(s)
CD8-Positive T-Lymphocytes/physiology , Hepatitis B virus/immunology , Hepatitis B/immunology , CD8-Positive T-Lymphocytes/immunology , Epitopes/immunology , Epitopes, T-Lymphocyte/immunology , Hep G2 Cells , Hepatitis B/metabolism , Hepatitis B Core Antigens/metabolism , Hepatitis B virus/genetics , Hepatitis B virus/pathogenicity , Humans , Interferon-gamma/metabolism , Liver/immunology , Spatio-Temporal Analysis
6.
Nucleic Acids Res ; 46(13): e80, 2018 07 27.
Article in English | MEDLINE | ID: mdl-29718399

ABSTRACT

The clustered regularly interspersed short palindromic repeat (CRISPR) gene-editing system has been repurposed for live-cell genomic imaging, but existing approaches rely on fluorescent protein reporters, making sensitive and continuous imaging difficult. Here, we present a fluorophore-based live-cell genomic imaging system that consists of a nuclease-deactivated mutant of the Cas9 protein (dCas9), a molecular beacon (MB), and an engineered single-guide RNA (sgRNA) harboring a unique MB target sequence (sgRNA-MTS), termed CRISPR/MB. Specifically, dCas9 and sgRNA-MTS are first co-expressed to target a specific locus in cells, followed by delivery of MBs that can then hybridize to MTS to illuminate the target locus. We demonstrated the feasibility of this approach for quantifying genomic loci, for monitoring chromatin dynamics, and for dual-color imaging when using two orthogonal MB/MTS pairs. With flexibility in selecting different combinations of fluorophore/quencher pairs and MB/MTS sequences, our CRISPR/MB hybrid system could be a promising platform for investigating chromatin activities.


Subject(s)
CRISPR-Cas Systems , Fluorescent Dyes , Microscopy, Fluorescence , Oligonucleotide Probes , CRISPR-Associated Protein 9/genetics , CRISPR-Associated Protein 9/metabolism , Chromatin/metabolism , Genomics , HEK293 Cells , HeLa Cells , Humans
7.
Proc Natl Acad Sci U S A ; 114(47): E10056-E10065, 2017 11 21.
Article in English | MEDLINE | ID: mdl-29114055

ABSTRACT

The packaging and budding of Gag polyprotein and viral RNA is a critical step in the HIV-1 life cycle. High-resolution structures of the Gag polyprotein have revealed that the capsid (CA) and spacer peptide 1 (SP1) domains contain important interfaces for Gag self-assembly. However, the molecular details of the multimerization process, especially in the presence of RNA and the cell membrane, have remained unclear. In this work, we investigate the mechanisms that work in concert between the polyproteins, RNA, and membrane to promote immature lattice growth. We develop a coarse-grained (CG) computational model that is derived from subnanometer resolution structural data. Our simulations recapitulate contiguous and hexameric lattice assembly driven only by weak anisotropic attractions at the helical CA-SP1 junction. Importantly, analysis from CG and single-particle tracking photoactivated localization (spt-PALM) trajectories indicates that viral RNA and the membrane are critical constituents that actively promote Gag multimerization through scaffolding, while overexpression of short competitor RNA can suppress assembly. We also find that the CA amino-terminal domain imparts intrinsic curvature to the Gag lattice. As a consequence, immature lattice growth appears to be coupled to the dynamics of spontaneous membrane deformation. Our findings elucidate a simple network of interactions that regulate the early stages of HIV-1 assembly and budding.


Subject(s)
Cell Membrane/chemistry , Gene Products, gag/chemistry , HIV-1/physiology , RNA, Viral/chemistry , Virus Assembly/physiology , Virus Release/physiology , Binding Sites , Cell Membrane/metabolism , Gene Expression , Gene Products, gag/genetics , Gene Products, gag/metabolism , HEK293 Cells , Host-Pathogen Interactions , Humans , Kinetics , Molecular Dynamics Simulation , Protein Binding , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Structure, Secondary , RNA, Viral/genetics , RNA, Viral/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Thermodynamics
8.
Proc Natl Acad Sci U S A ; 111(26): E2676-83, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24938790

ABSTRACT

MicroRNAs (miRNAs) are small, 18-22 nt long, noncoding RNAs that act as potent negative gene regulators in a variety of physiological and pathological processes. To repress gene expression, miRNAs are packaged into RNA-induced silencing complexes (RISCs) that target mRNAs for degradation and/or translational repression in a sequence-specific manner. Recently, miRNAs have been shown to also interact with proteins outside RISCs, impacting cellular processes through mechanisms not involving gene silencing. Here, we define a previously unappreciated activity of miRNAs in inhibiting RNA-protein interactions that in the context of HIV-1 biology blocks HIV virus budding and reduces virus infectivity. This occurs by miRNA binding to the nucleocapsid domain of the Gag protein, the main structural component of HIV-1 virions. The resulting miRNA-Gag complexes interfere with viral-RNA-mediated Gag assembly and viral budding at the plasma membrane, with imperfectly assembled Gag complexes endocytosed and delivered to lysosomes. The blockade of virus production by miRNA is reversed by adding the miRNA's target mRNA and stimulated by depleting Argonaute-2, suggesting that when miRNAs are not mediating gene silencing, they can block HIV-1 production through disruption of Gag assembly on membranes. Overall, our findings have significant implications for understanding how cells modulate HIV-1 infection by miRNA expression and raise the possibility that miRNAs can function to disrupt RNA-mediated protein assembly processes in other cellular contexts.


Subject(s)
HIV-1/physiology , MicroRNAs/metabolism , Multiprotein Complexes/metabolism , Protein Precursors/metabolism , Virus Assembly/physiology , Virus Release/physiology , Argonaute Proteins/metabolism , Cluster Analysis , DNA Primers/genetics , HEK293 Cells , Humans , Microscopy, Fluorescence , RNA, Small Interfering/genetics
9.
J Immunol ; 189(8): 3925-35, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22972930

ABSTRACT

A regulatory subset of B cells has been found to modulate immune responses in autoimmunity, infection, and cancer, but it has not been investigated in the setting of human persistent viral infection. IL-10 is elevated in patients with chronic hepatitis B virus infection (CHB), but its cellular sources and impact on antiviral T cells have not been addressed. We investigated the role of IL-10 and regulatory B cells in the pathogenesis of CHB. Serum IL-10 levels were studied longitudinally in patients with CHB undergoing spontaneous disease flares. There was a close temporal correlation between IL-10 levels and fluctuations in viral load or liver inflammation. Blockade of IL-10 in vitro rescued polyfunctional virus-specific CD8 T cell responses. To investigate the potential contribution of regulatory B cells, their frequency was measured directly ex vivo and after exposure to stimuli relevant to hepatitis B virus (HBV) (CpG or HBV Ags). IL-10-producing B cells were enriched in patients, and their frequency correlated temporally with hepatic flares, both after stimulation and directly ex vivo. Phenotypically, these cells were predominantly immature (CD19(+)CD24(hi)CD38(hi)) ex vivo; sorted CD19(+)CD24(hi)CD38(hi) cells suppressed HBV-specific CD8 T cell responses in an IL-10-dependent manner. In summary, these data reveal a novel IL-10-producing subset of B cells able to regulate T cell immunity in CHB.


Subject(s)
B-Lymphocytes, Regulatory/immunology , B-Lymphocytes, Regulatory/metabolism , Hepatitis B virus/immunology , Hepatitis B, Chronic/immunology , Hepatitis B, Chronic/pathology , Interleukin-10/biosynthesis , Adult , Aged , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , B-Lymphocyte Subsets/virology , B-Lymphocytes, Regulatory/virology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/virology , Cell Differentiation/immunology , Cells, Cultured , Female , Hepatitis B, Chronic/etiology , Humans , Interleukin-10/antagonists & inhibitors , Interleukin-10/blood , Longitudinal Studies , Male , Middle Aged , Young Adult
10.
Sci Adv ; 10(8): eadk8297, 2024 Feb 23.
Article in English | MEDLINE | ID: mdl-38394201

ABSTRACT

HIV-1 Gag proteins can multimerize upon the viral genomic RNA or multiple random cellular messenger RNAs to form a virus particle or a virus-like particle, respectively. To date, whether the two types of particles form via the same Gag multimerization process has remained unclarified. Using photoactivated localization microscopy to illuminate Gag organizations and dynamics at the nanoscale, here, we showed that genomic RNA mediates Gag multimerization in a more cluster-centric, cooperative, and spatiotemporally coordinated fashion, with the ability to drive dense Gag clustering dependent on its ability to act as a long-stranded scaffold not easily attainable by cellular messenger RNAs. These differences in Gag multimerization were further shown to affect downstream selective protein sorting into HIV membranes, indicating that the choice of RNA for packaging can modulate viral membrane compositions. These findings should advance the understanding of HIV assembly and further benefit the development of virus-like particle-based therapeutics.


Subject(s)
HIV Infections , RNA, Viral , Humans , RNA, Viral/genetics , RNA, Viral/metabolism , Cell Membrane/metabolism , Gene Products, gag/genetics , Gene Products, gag/metabolism , RNA, Messenger/metabolism , HIV Infections/metabolism , Protein Multimerization
11.
Hepatology ; 56(6): 2027-38, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22684948

ABSTRACT

UNLABELLED: During antiviral therapy, specific delivery of interferon-α (IFNα) to infected cells may increase its antiviral efficacy, trigger a localized immune reaction, and reduce the side effects caused by systemic administration. Two T-cell receptor-like antibodies (TCR-L) able to selectively bind hepatitis B virus (HBV)-infected hepatocytes of chronic hepatitis B patients and recognize core (HBc18-27) and surface (HBs183-91) HBV epitopes associated with different human leukocyte antigen (HLA)-A*02 alleles (A*02:01, A*02:02, A*02:07, A*02:11) were generated. Each antibody was genetically linked to two IFNα molecules to produce TCR-L/IFNα fusion proteins. We demonstrate that the fusion proteins triggered an IFNα response preferentially on the hepatocytes presenting the correct HBV-peptide HLA-complex and that the mechanism of the targeted IFNα response was dependent on the specific binding of the fusion proteins to the HLA/HBV peptide complexes through the TCR-like variable regions of the antibodies. CONCLUSION: TCR-L antibodies can be used to target cytokines to HBV-infected hepatocytes in vitro. Fusion of IFNα to TCR-L decreased the intrinsic biological activity of IFNα but preserved the overall specificity of the protein for the cognate HBV peptide/HLA complexes. This induction of an effective IFNα response selectively in HBV-infected cells might have a therapeutic advantage in comparison to the currently used native or pegylated IFNα.


Subject(s)
Antibodies, Viral/pharmacology , Antiviral Agents/pharmacology , HLA-A Antigens/immunology , Hepatitis B virus/immunology , Hepatitis B/immunology , Interferon-alpha/pharmacology , Recombinant Fusion Proteins/pharmacology , Animals , Antibodies, Viral/immunology , Artificial Gene Fusion , CD8-Positive T-Lymphocytes/drug effects , Chemokines/metabolism , Drug Carriers/pharmacology , Hep G2 Cells , Hepatitis B/drug therapy , Hepatitis B/virology , Hepatitis B virus/genetics , Humans , Lymphocyte Activation/drug effects , Mice
12.
Biotechnol Bioeng ; 110(10): 2731-41, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23568715

ABSTRACT

The extracellular matrix (ECM) consists of a complex mixture of biochemical and physical stimuli that together regulate cell behavior. In this study, we engineer a model ECM consisting of fibrillar Type-1 collagen plus fibronectin that allows systematic examination of the effects of matrix composition and mechanics on cells. On this combined protein matrix, cells exhibit intermediate degrees of spreading and proliferation compared to their responses on collagen or fibronectin alone. Adhesion to the combination matrix could be blocked by peptides containing the sequence arginine-glycine-aspartic acid (RGD) and by antibodies against α1 integrin, suggesting cell-matrix engagement was mediated by a combination of integrin receptors that recognize fibronectin and collagen. Regardless of integrin engagement, cells were sensitive to the mechanical properties of the combination ECM, suggesting that cells could process biochemical and mechanical cues simultaneously and independently.


Subject(s)
Cell Proliferation/drug effects , Cell Shape/drug effects , Collagen Type I/pharmacology , Fibronectins/pharmacology , Adsorption , Analysis of Variance , Animals , Biomechanical Phenomena/physiology , Cattle , Cell Culture Techniques , Cell Line , Collagen Type I/chemistry , Elasticity , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Fibronectins/chemistry , Integrin alpha1/metabolism , Oligopeptides/metabolism , Rats
13.
J Immunol ; 186(9): 5107-18, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21430225

ABSTRACT

The hepatitis C virus (HCV)-specific T cell response in patients with chronic HCV is dysfunctional. In this study, we aimed at restoring immunological function through therapeutic vaccination in a transgenic mouse model with impaired HCV-specific T cell responses due to a persistent presence of hepatic HCV nonstructural (NS)3/4A Ags. The HCV-specific T cells have an actively maintained dysfunction reflected in reduced frequency, impaired cytokine production, and impaired effector function in vivo, which can be partially restored by blocking regulatory T cells or programmed cell death ligand 1. We hypothesized that the impairment could be corrected by including sequences that created a normal priming environment by recruiting "healthy" heterologous T cells and by activating innate signaling. Endogenously expressed hepatitis B core Ag (HBcAg) can recruit heterologous T cells and activate TLR (TLR7) signaling. Hence, by combining HCV NS3/4A with different forms of HBcAg we found that heterologous sequences somewhat improved activation and expansion of NS3/4A-specific T cells in a wild-type host. Importantly, the signals provided by HBcAg effectively restored the activation of HCV-specific T cells in a tolerant NS3/4A-transgenic mouse model. The adjuvant effect could also be transferred to the priming of dysfunctional HLA-A2-restricted NS3-specific T cells in vivo. Thus, recruiting healthy heterologous T cells to the site of priming may also help restore HCV-specific responses present in a chronically infected host.


Subject(s)
Hepatitis C, Chronic/immunology , T-Lymphocytes/immunology , Vaccination/methods , Viral Hepatitis Vaccines/immunology , Viral Nonstructural Proteins/immunology , Animals , Animals, Genetically Modified , Antigens, Viral/immunology , Antigens, Viral/therapeutic use , Blotting, Western , Cell Line, Tumor , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Hepacivirus/immunology , Hepatitis B Core Antigens/immunology , Hepatitis B Core Antigens/therapeutic use , Hepatitis C, Chronic/therapy , Humans , Immunoprecipitation , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic
14.
Protein Cell ; 14(6): 433-447, 2023 06 07.
Article in English | MEDLINE | ID: mdl-37402315

ABSTRACT

Molecular knowledge of human gastric corpus epithelium remains incomplete. Here, by integrated analyses using single-cell RNA sequencing (scRNA-seq), spatial transcriptomics, and single-cell assay for transposase accessible chromatin sequencing (scATAC-seq) techniques, we uncovered the spatially resolved expression landscape and gene-regulatory network of human gastric corpus epithelium. Specifically, we identified a stem/progenitor cell population in the isthmus of human gastric corpus, where EGF and WNT signaling pathways were activated. Meanwhile, LGR4, but not LGR5, was responsible for the activation of WNT signaling pathway. Importantly, FABP5 and NME1 were identified and validated as crucial for both normal gastric stem/progenitor cells and gastric cancer cells. Finally, we explored the epigenetic regulation of critical genes for gastric corpus epithelium at chromatin state level, and identified several important cell-type-specific transcription factors. In summary, our work provides novel insights to systematically understand the cellular diversity and homeostasis of human gastric corpus epithelium in vivo.


Subject(s)
Epigenesis, Genetic , Gastric Mucosa , Humans , Gastric Mucosa/metabolism , Chromatin/metabolism , Stem Cells , Epithelium/metabolism , Fatty Acid-Binding Proteins/metabolism
15.
AIDS ; 37(3): 401-411, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36384811

ABSTRACT

OBJECTIVE: Liver disease is accelerated in people with HIV (PWH) with hepatitis B virus (HBV) coinfection. We hypothesized that liver fibrosis in HIV-HBV is triggered by increased hepatocyte apoptosis, microbial translocation and/or HIV/HBV viral products. DESIGN: Sera from PWH with HBV coinfection versus from those with HBV only or putative mediators were used to examine the pathogenesis of liver disease in HIV-HBV. METHODS: We applied sera from PWH and HBV coinfection versus HBV alone, or putative mediators (including HMGB1), to primary human hepatic stellate cells (hHSC) and examined pro-fibrogenic changes at the single cell level using flow cytometry. High mobility group box 1 (HMGB1) levels in the applied sera were assessed according to donor fibrosis stage. RESULTS: Quantitative flow cytometric assessment of pro-fibrogenic and inflammatory changes at the single cell level revealed an enhanced capacity for sera from PWH with HBV coinfection to activate hHSC. This effect was recapitulated by lipopolysaccharide, HIV-gp120, hepatocyte conditioned-media and the alarmin HMGB1. Induction of hepatocyte cell death increased their pro-fibrogenic potential, an effect blocked by HMGB1 antagonist glycyrrhizic acid. Consistent with a role for this alarmin, HMGB1 levels were elevated in sera from PWH and hepatitis B coinfection compared to HBV alone and higher in those with HIV-HBV with liver fibrosis compared to those without. CONCLUSIONS: Sera from PWH and HBV coinfection have an enhanced capacity to activate primary hHSC. We identified an increase in circulating HMGB1 which, in addition to HIV-gp120 and translocated microbial products, drove pro-fibrogenic changes in hHSC, as mechanisms contributing to accelerated liver disease in HIV-HBV.


Subject(s)
Coinfection , HIV Infections , HMGB1 Protein , Hepatitis B , Humans , Hepatitis B virus , Alarmins , Hepatitis B/complications , Liver Cirrhosis/pathology
16.
J Gen Virol ; 93(Pt 8): 1680-1686, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22592266

ABSTRACT

It has been well established that immunological escape mutations within the hepatitis C virus genotype (gt) 1a non-structural (NS) 3/4A protease are partly prevented by a reduction in viral protease fitness. Surprisingly little is known about whether similar mutations affect proteases from other genotypes. In the present study, we assessed both the HLA-A2-restricted CTL response and gt3a NS3/4A protease fitness. Similar to gt1, the 1073-1081 epitope was immunodominant within the gt3a-specific HLA-A2-restricted CTL response, despite sequence similarity of only 56 % between the gt1a and gt3a genes. However, unlike the gt1a NS3/4A protease, all residues within the gt3a 1073-1081 epitope could be replaced sequentially by alanine while retaining protease activity, at least in part.


Subject(s)
Alanine/chemistry , Gene Expression Regulation, Viral/physiology , Hepacivirus/genetics , Immunodominant Epitopes/genetics , Viral Nonstructural Proteins/metabolism , Amino Acid Sequence , Amino Acid Substitution , Cloning, Molecular , Genotype , Humans , Molecular Sequence Data , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics
17.
J Virol ; 85(20): 10432-9, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21849442

ABSTRACT

Pigs are considered to be intermediate hosts and "mixing vessels," facilitating the genesis of pandemic influenza viruses, as demonstrated by the emergence of the 2009 H1N1 pandemic (pdm/09) virus. The prevalence and repeated introduction of the pdm/09 virus into pigs raises the possibility of generating novel swine influenza viruses with the potential to infect humans. To address this, an active influenza surveillance program was conducted with slaughtered pigs in abattoirs in southern China. Over 50% of the pigs tested were found to be seropositive for one or more H1 influenza viruses, most commonly pdm/09-like viruses. Out of 36 virus isolates detected, one group of novel reassortants had Eurasian avian-like swine H1N1 surface genes and pdm/09 internal genes. Animal experiments showed that this virus transmitted effectively from pig to pig and from pig to ferret, and it could also replicate in ex vivo human lung tissue. Immunization against the 2009 pandemic virus gave only partial protection to ferrets. The continuing prevalence of the pdm/09 virus in pigs could lead to the genesis of novel swine reassortant viruses with the potential to infect humans.


Subject(s)
Influenza A virus/genetics , Orthomyxoviridae Infections/virology , Reassortant Viruses/genetics , Swine/virology , Animals , China , Ferrets , Humans , In Vitro Techniques , Influenza A virus/classification , Influenza A virus/isolation & purification , Influenza A virus/pathogenicity , Lung/virology , Male , Molecular Sequence Data , Orthomyxoviridae Infections/transmission , RNA, Viral/genetics , Reassortant Viruses/classification , Reassortant Viruses/isolation & purification , Reassortant Viruses/pathogenicity , Sequence Analysis, DNA
18.
Scand J Infect Dis ; 44(1): 55-9, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21933033

ABSTRACT

A major problem in chronic hepatitis B virus (HBV) infection is that treatment with specific antivirals is life-long since they rarely induce a sustained response. An attractive option is therefore to combine antiviral therapy with some type of immune stimulator, such as a therapeutic vaccine. Several lines of evidence suggest that a key target for the cellular immune response is the HBV core antigen (HBcAg). However, it may also be of advantage to simultaneously improve the neutralizing antibody response to the surface (S) region of HBV. We therefore generated chimeric HBcAg particles expressing preS1 residues 1-42 at the tip of the spike region. We could show that this chimeric HBcAg-preS1 protein primed both HBcAg-specific T cells and antibodies to preS1. This strongly suggests that this may be a viable approach to develop an effective bi-functional therapeutic vaccine as an add-on for the treatment of chronic HBV infections.


Subject(s)
Hepatitis B Core Antigens/therapeutic use , Hepatitis B Surface Antigens/therapeutic use , Hepatitis B virus/immunology , Hepatitis B, Chronic/drug therapy , T-Lymphocytes/immunology , Vaccination/methods , Animals , Antibodies, Neutralizing , Female , Hepatitis B Core Antigens/immunology , Hepatitis B Surface Antigens/immunology , Hepatitis B, Chronic/immunology , Immunity, Cellular/immunology , Mice , Mice, Inbred C57BL , Vaccines, Synthetic/immunology , Vaccines, Synthetic/therapeutic use
19.
Nucleic Acids Res ; 38(14): e148, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20507905

ABSTRACT

Numerous studies have utilized molecular beacons (MBs) to image RNA expression in living cells; however, there is growing evidence that the sensitivity of RNA detection is significantly hampered by their propensity to emit false-positive signals. To overcome these limitations, we have developed a new RNA imaging probe called ratiometric bimolecular beacon (RBMB), which combines functional elements of both conventional MBs and siRNA. Analogous to MBs, RBMBs elicit a fluorescent reporter signal upon hybridization to complementary RNA. In addition, an siRNA-like double-stranded domain is used to facilitate nuclear export. Accordingly, live-cell fluorescent imaging showed that RBMBs are localized predominantly in the cytoplasm, whereas MBs are sequestered into the nucleus. The retention of RBMBs within the cytoplasmic compartment led to >15-fold reduction in false-positive signals and a significantly higher signal-to-background compared with MBs. The RBMBs were also designed to possess an optically distinct reference fluorophore that remains unquenched regardless of probe confirmation. This reference dye not only provided a means to track RBMB localization, but also allowed single cell measurements of RBMB fluorescence to be corrected for variations in probe delivery. Combined, these attributes enabled RBMBs to exhibit an improved sensitivity for RNA detection in living cells.


Subject(s)
Fluorescent Dyes/chemistry , Oligonucleotide Probes/chemistry , RNA/analysis , Animals , Cells, Cultured , Electroporation , Humans , Microscopy, Fluorescence , Oligonucleotide Probes/analysis
20.
Nat Commun ; 13(1): 205, 2022 01 11.
Article in English | MEDLINE | ID: mdl-35017543

ABSTRACT

The genome exists as an organized, three-dimensional (3D) dynamic architecture, and each cell type has a unique 3D genome organization that determines its cell identity. An unresolved question is how cell type-specific 3D genome structures are established during development. Here, we analyzed 3D genome structures in muscle cells from mice lacking the muscle lineage transcription factor (TF), MyoD, versus wild-type mice. We show that MyoD functions as a "genome organizer" that specifies 3D genome architecture unique to muscle cell development, and that H3K27ac is insufficient for the establishment of MyoD-induced chromatin loops in muscle cells. Moreover, we present evidence that other cell lineage-specific TFs might also exert functional roles in orchestrating lineage-specific 3D genome organization during development.


Subject(s)
Genome , Histones/genetics , Muscle, Skeletal/metabolism , MyoD Protein/genetics , Myoblasts/metabolism , Animals , Binding Sites , CCCTC-Binding Factor/genetics , CCCTC-Binding Factor/metabolism , Cell Line , Cell Lineage/genetics , Chromatin Assembly and Disassembly , Chromosomes/chemistry , Chromosomes/metabolism , Gene Expression Regulation, Developmental , Gene Library , Histones/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/cytology , MyoD Protein/metabolism , Myoblasts/cytology , Myogenin/genetics , Myogenin/metabolism , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL