Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
1.
J Virol ; 93(21)2019 11 01.
Article in English | MEDLINE | ID: mdl-31375595

ABSTRACT

Influenza A virus (IAV) nonstructural protein 1 (NS1), a potent antagonist of the host immune response, is capable of interacting with RNA and a wide range of cellular proteins. NS1 consists of an RNA-binding domain (RBD) and an effector domain (ED) separated by a flexible linker region (LR). H5N1-NS1 has a characteristic 5-residue deletion in the LR, with either G (minor group) or E (major group) at the 71st position, and non-H5N1-NS1 contains E71 with an intact linker. Based on the orientation of the ED with respect to the RBD, previous crystallographic studies have shown that minor group H5N1-NS1(G71), a non-H5N1-NS1 [H6N6-NS1(E71)], and the LR deletion mutant H6N6-NS1(Δ80-84/E71) mimicking the major group H5N1-NS1 exhibit "open," "semiopen," and "closed" conformations, respectively, suggesting that NS1 exhibits a strain-dependent conformational preference. Here we report the first crystal structure of a naturally occurring H5N1-NS1(E71) and show that it adopts an open conformation similar to that of the minor group of H5N1-NS1 [H5N1-NS1(G71)]. We also show that H6N6-NS1(Δ80-84/E71) under a different crystallization condition and H6N6-NS1(Δ80-84/G71) also exhibit open conformations, suggesting that NS1 can adopt an open conformation irrespective of E or G at the 71st position. Our single-molecule fluorescence resonance energy transfer (FRET) analysis to investigate the conformational preference of NS1 in solution showed that all NS1 constructs predominantly exist in an open conformation. Further, our coimmunoprecipitation and binding studies showed that they all bind to cellular factors with similar affinities. Taken together, our studies suggest that NS1 exhibits strain-independent structural plasticity that allows it to interact with a wide variety of cellular ligands during viral infection.IMPORTANCE IAV is responsible for several pandemics over the last century and continues to infect millions annually. The frequent rise in drug-resistant strains necessitates exploring novel targets for developing antiviral drugs that can reduce the global burden of influenza infection. Because of its critical role in the replication and pathogenesis of IAV, nonstructural protein 1 (NS1) is a potential target for developing antivirals. Previous studies suggested that NS1 adopts strain-dependent "open," "semiopen," and "closed" conformations. Here we show, based on three crystal structures, that NS1 irrespective of strain differences can adopt an open conformation. We further show that NS1 from different strains primarily exists in an open conformation in solution and binds to cellular proteins with a similar affinity. Together, our findings suggest that conformational polymorphism facilitated by a flexible linker is intrinsic to NS1, and this may be the underlying factor allowing NS1 to bind several cellular factors during IAV replication.


Subject(s)
Influenza A virus/chemistry , Viral Nonstructural Proteins/chemistry , Amino Acid Sequence , Crystallography, X-Ray , Fluorescence Resonance Energy Transfer , Influenza A virus/classification , Influenza A virus/genetics , Ligands , Mutation , Protein Binding , Protein Conformation , Structure-Activity Relationship , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism
2.
Virol J ; 16(1): 22, 2019 02 20.
Article in English | MEDLINE | ID: mdl-30786885

ABSTRACT

BACKGROUND: The regulatory cyclin, Cyclin T1 (CycT1), is a host factor essential for HIV-1 replication in CD4 T cells and macrophages. The importance of CycT1 and the Positive Transcription Elongation Factor b (P-TEFb) complex for HIV replication is well-established, but regulation of CycT1 expression and protein levels during HIV replication and latency establishment in CD4 T cells is less characterized. METHODS: To better define the regulation of CycT1 levels during HIV replication in CD4 T cells, multiparameter flow cytometry was utilized to study the interaction between HIV replication (intracellular p24) and CycT1 of human peripheral blood memory CD4 T cells infected with HIV in vitro. CycT1 was further examined in CD4 T cells of human lymph nodes. RESULTS: In activated (CD3+CD28 costimulation) uninfected blood memory CD4 T cells, CycT1 was most significantly upregulated in maximally activated (CD69+CD25+ and HLA.DR+CD38+) cells. In memory CD4 T cells infected with HIV in vitro, two distinct infected populations of p24+CycT1+ and p24+CycT1- cells were observed during 7 days infection, suggestive of different phases of productive HIV replication and subsequent latency establishment. Intriguingly, p24+CycT1- cells were the predominant infected population in activated CD4 T cells, raising the possibility that productively infected cells may transition into latency subsequent to CycT1 downregulation. Additionally, when comparing infected p24+ cells to bystander uninfected p24- cells (after bulk HIV infections), HIV replication significantly increased T cell activation (CD69, CD25, HLA.DR, CD38, and Ki67) without concomitantly increasing CycT1 protein levels, possibly due to hijacking of P-TEFb by the viral Tat protein. Lastly, CycT1 was constitutively expressed at higher levels in lymph node CD4 T cells compared to blood T cells, potentially enhancing latency generation in lymphoid tissues. CONCLUSIONS: CycT1 is most highly upregulated in maximally activated memory CD4 T cells as expected, but may become less associated with T cell activation during HIV replication. The progression into latency may further be predicated by substantial generation of p24+CycT1- cells during HIV replication.


Subject(s)
CD4-Positive T-Lymphocytes/virology , Cyclin T/genetics , HIV Infections/immunology , Virus Latency/physiology , Virus Replication/physiology , CD4-Positive T-Lymphocytes/immunology , Flow Cytometry , Gene Expression Regulation , HIV-1/physiology , Host Microbial Interactions , Humans , Positive Transcriptional Elongation Factor B/genetics , Transcriptional Activation
3.
Nucleic Acids Res ; 45(16): 9348-9360, 2017 Sep 19.
Article in English | MEDLINE | ID: mdl-28637181

ABSTRACT

In eukaryotic cells, the gene expression status is strictly controlled by epigenetic modifications on chromatin. The repressive status of chromatin largely contributes to HIV latency. Studies have shown that modification of histone H3K27 acts as a key molecular switch for activation or suppression of many cellular genes. In this study, we found that K27-acetylated histone H3 specifically recruited Super Elongation Complex (SEC), the transcriptional elongation complex essential for HIV-1 long terminal repeat (LTR)-mediated and general cellular transcription. Interestingly, H3K27 acetylation further stimulates H3R26 methylation, which subsequently abrogates the recruitment of SEC, forming a negative feedback regulatory loop. Importantly, by inhibiting methyltransferase activity of CARM1, the enzyme responsible for H3R26 methylation, HIV-1 transcription is reactivated in several HIV latency cell models, including a primary resting CD4+ T cell model. When combined with other latency disrupting compounds such as JQ1 or vorinostat/SAHA, the CARM1 inhibitor achieved synergistic effects on HIV-1 activation. This study suggests that coordinated and dynamic modifications at histone H3K27 and H3R26 orchestrate HIV-1 LTR-mediated transcription, and potentially opens a new avenue to disrupt latent HIV-1 infection by targeting specific epigenetic enzymes.


Subject(s)
Gene Expression Regulation, Viral , HIV-1/genetics , Histone Code , Histones/metabolism , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Virus Latency/genetics , Acetylation , Benzylidene Compounds/chemistry , Benzylidene Compounds/pharmacology , Cell Line , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , HIV Long Terminal Repeat , HIV-1/drug effects , HIV-1/metabolism , HIV-1/physiology , Humans , Methylation , Piperidones/chemistry , Piperidones/pharmacology , Transcription Elongation, Genetic , Transcription Factors/metabolism , Virus Latency/drug effects
4.
BMC Bioinformatics ; 18(1): 74, 2017 Jan 31.
Article in English | MEDLINE | ID: mdl-28143596

ABSTRACT

BACKGROUND: Gene regulatory interactions are of fundamental importance to various biological functions and processes. However, only a few previous computational studies have claimed success in revealing genome-wide regulatory landscapes from temporal gene expression data, especially for complex eukaryotes like human. Moreover, recent work suggests that these methods still suffer from the curse of dimensionality if a network size increases to 100 or higher. RESULTS: Here we present a novel scalable algorithm for identifying genome-wide gene regulatory network (GRN) structures, and we have verified the algorithm performances by extensive simulation studies based on the DREAM challenge benchmark data. The highlight of our method is that its superior performance does not degenerate even for a network size on the order of 104, and is thus readily applicable to large-scale complex networks. Such a breakthrough is achieved by considering both prior biological knowledge and multiple topological properties (i.e., sparsity and hub gene structure) of complex networks in the regularized formulation. We also validate and illustrate the application of our algorithm in practice using the time-course gene expression data from a study on human respiratory epithelial cells in response to influenza A virus (IAV) infection, as well as the CHIP-seq data from ENCODE on transcription factor (TF) and target gene interactions. An interesting finding, owing to the proposed algorithm, is that the biggest hub structures (e.g., top ten) in the GRN all center at some transcription factors in the context of epithelial cell infection by IAV. CONCLUSIONS: The proposed algorithm is the first scalable method for large complex network structure identification. The GRN structure identified by our algorithm could reveal possible biological links and help researchers to choose which gene functions to investigate in a biological event. The algorithm described in this article is implemented in MATLAB Ⓡ , and the source code is freely available from https://github.com/Hongyu-Miao/DMI.git .


Subject(s)
Algorithms , Gene Regulatory Networks , Epithelial Cells/metabolism , Epithelial Cells/virology , Humans , Influenza A virus/physiology , Transcription Factors/metabolism
5.
J Virol ; 89(7): 3557-67, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25589658

ABSTRACT

UNLABELLED: By recruiting the host protein XPO1 (CRM1), the HIV-1 Rev protein mediates the nuclear export of incompletely spliced viral transcripts. We mined data from the recently described human nuclear complexome to identify a host protein, RBM14, which associates with XPO1 and Rev and is involved in Rev function. Using a Rev-dependent p24 reporter plasmid, we found that RBM14 depletion decreased Rev activity and Rev-mediated enhancement of the cytoplasmic levels of unspliced viral transcripts. RBM14 depletion also reduced p24 expression during viral infection, indicating that RBM14 is limiting for Rev function. RBM14 has previously been shown to localize to nuclear paraspeckles, a structure implicated in retaining unspliced HIV-1 transcripts for either Rev-mediated nuclear export or degradation. We found that depletion of NEAT1 RNA, a long noncoding RNA required for paraspeckle integrity, abolished the ability of overexpressed RBM14 to enhance Rev function, indicating the dependence of RBM14 function on paraspeckle integrity. Our study extends the known host cell interactome of Rev and XPO1 and further substantiates a critical role for paraspeckles in the mechanism of action of Rev. Our study also validates the nuclear complexome as a database from which viral cofactors can be mined. IMPORTANCE: This study mined a database of nuclear protein complexes to identify a cellular protein named RBM14 that is associated with XPO1 (CRM1), a nuclear protein that binds to the HIV-1 Rev protein and mediates nuclear export of incompletely spliced viral RNAs. Functional assays demonstrated that RBM14, a protein found in paraspeckle structures in the nucleus, is involved in HIV-1 Rev function. This study validates the nuclear complexome database as a reference that can be mined to identify viral cofactors.


Subject(s)
HIV-1/physiology , Host-Pathogen Interactions , Intracellular Signaling Peptides and Proteins/metabolism , Karyopherins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , rev Gene Products, Human Immunodeficiency Virus/metabolism , Active Transport, Cell Nucleus , Cell Line , Humans , RNA, Viral/metabolism , Exportin 1 Protein
6.
J Virol ; 88(8): 4113-22, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24478439

ABSTRACT

UNLABELLED: NS1 of influenza A virus is a potent antagonist of host antiviral interferon responses. This multifunctional protein with two distinctive domains, an RNA-binding domain (RBD) and an effector domain (ED) separated by a linker region (LR), is implicated in replication, pathogenesis, and host range. Although the structures of individual domains of NS1 from different strains of influenza viruses have been reported, the only structure of full-length NS1 available to date is from an H5N1 strain (A/Vietnam/1203/2004). By carrying out crystallographic analyses of full-length H6N6-NS1 (A/blue-winged teal/MN/993/1980) and an LR deletion mutant, combined with mutational analysis, we show here that these full-length NS1 structures provide an exquisite structural sampling of various conformational states of NS1 that based on the orientation of the ED with respect to RBD can be summarized as "open," "semi-open," and "closed" conformations. Our studies show that preference for these states is clearly dictated by determinants such as linker length, residue composition at position 71, and a mechanical hinge, providing a structural basis for strain-dependent functional variations in NS1. Because of the flexibility inherent in the LR, any particular NS1 could sample the conformational space around these states to engage ED in different quaternary interactions so that it may participate in specific protein-protein or protein-RNA interactions to allow for the known multifunctionality of NS1. We propose that such conformational plasticity provides a mechanism for autoregulating NS1 functions, depending on its temporal distribution, posttranslational modifications, and nuclear or cellular localization, during the course of virus infection. IMPORTANCE: NS1 of influenza A virus is a multifunctional protein associated with numerous strain-specific regulatory functions during viral infection, including conferring resistance to antiviral interferon induction, replication, pathogenesis, virulence, and host range. NS1 has two domains, an RNA-binding domain and an effector domain separated by a linker. To date, the only full-length NS1 structure available is that from an H5N1 strain (A/Vietnam/1203/2004). Here, we determined crystal structures of the wild type and a linker region mutant of the H6N6 NS1 (A/blue-winged teal/MN/993/1980), which together with the previously determined H5N1 NS1 structure show that NS1 exhibits significant strain-dependent structural polymorphism due to variations in linker length, residue composition at position 71, and a mechanical hinge. Such a structural polymorphism may be the basis for strain-specific functions associated with NS1.


Subject(s)
Influenza A virus/metabolism , Influenza in Birds/virology , Viral Nonstructural Proteins/chemistry , Animals , Birds , Influenza A Virus, H5N1 Subtype/chemistry , Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/metabolism , Influenza A virus/chemistry , Influenza A virus/genetics , Models, Molecular , Protein Conformation , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism
7.
J Virol ; 87(2): 1211-20, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23152527

ABSTRACT

P-TEFb, a cellular kinase composed of Cyclin T1 and CDK9, is essential for processive HIV-1 transcription. P-TEFb activity is dependent on phosphorylation of Thr186 in the CDK9 T loop. In resting CD4(+) T cells which are nonpermissive for HIV-1 replication, the levels of Cyclin T1 and T-loop-phosphorylated CDK9 are very low but increase significantly upon cellular activation. Little is known about how P-TEFb activity and expression are regulated in resting central memory CD4(+) T cells, one of the main reservoirs of latent HIV-1. We used an in vitro primary cell model of HIV-1 latency to show that P-TEFb availability in resting memory CD4(+) T cells is governed by the differential expression and phosphorylation of its subunits. This is in contrast to previous observations in dividing cells, where P-TEFb can be regulated by its sequestration in the 7SK RNP complex. We find that resting CD4(+) T cells, whether naïve or memory and independent of their infection status, have low levels of Cyclin T1 and T-loop-phosphorylated CDK9, which increase upon activation. We also show that the decrease in Cyclin T1 protein upon the acquisition of a memory phenotype is in part due to proteasome-mediated proteolysis and likely also to posttranscriptional downregulation by miR-150. We also found that HEXIM1 levels are very low in ex vivo- and in vitro-generated resting memory CD4(+) T cells, thus limiting the sequestration of P-TEFb in the 7SK RNP complex, indicating that this mechanism is unlikely to be a driver of viral latency in this cell type.


Subject(s)
CD4-Positive T-Lymphocytes/virology , Cyclin T/metabolism , Cyclin-Dependent Kinase 9/metabolism , HIV-1/pathogenicity , Virus Latency , Cells, Cultured , Gene Expression , HIV-1/physiology , Humans , Phosphorylation
8.
J Virol ; 86(6): 3244-52, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22205749

ABSTRACT

The replication of integrated human immunodeficiency virus type 1 (HIV-1) is dependent on the cellular cofactor cyclin T1, which binds the viral Tat protein and activates the RNA polymerase II transcription of the integrated provirus. The activation of resting CD4(+) T cells upregulates cyclin T1 protein levels independently of an increase in cyclin T1 mRNA levels, suggesting a translational repression of cyclin T1 in resting CD4(+) T cells. Hypothesizing that microRNAs (miRNAs) repress cyclin T1 translation in resting CD4(+) T cells and that this inhibition is lifted upon cell activation, we used microarray expression analysis to identify miRNAs miR-27b, miR-29b, miR-150, and miR-223 as being significantly downregulated upon CD4(+) T cell activation. The overexpression of these miRNAs decreased endogenous cyclin T1 protein levels, while treatment with the corresponding antagomiRs increased cyclin T1 protein levels. An miR-27b binding site within the cyclin T1 3' untranslated region (3'UTR) was identified and confirmed to be functional after the mutation of key resides abrogated the ability of miR-27b to decrease the expression of a luciferase reporter upstream of the cyclin T1 3'UTR. Ago2 immunoprecipitation revealed an association with cyclin T1 mRNA that was decreased following treatment with miR-27b and miR-29b antagomiRs. Cells overexpressing miR-27b showed decreased viral gene expression levels of the HIV-1 reporter virus and a decreased replication of strain NL4.3; a partial rescue of viral transcription could be seen following the transfection of cyclin T1. These results implicate miR-27b as a novel regulator of cyclin T1 protein levels and HIV-1 replication, while miR-29b, miR-223, and miR-150 may regulate cyclin T1 indirectly.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Cyclin T/genetics , HIV Infections/genetics , HIV-1/physiology , MicroRNAs/metabolism , Virus Replication , CD4-Positive T-Lymphocytes/virology , Cell Line , Cells, Cultured , Cyclin T/metabolism , Down-Regulation , HIV Infections/metabolism , HIV Infections/virology , HIV-1/genetics , Humans , MicroRNAs/genetics , Protein Biosynthesis , Up-Regulation
9.
J Acquir Immune Defic Syndr ; 94(2S): S80-S85, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37707853

ABSTRACT

BACKGROUND: The Texas Developmental Center for AIDS Research (D-CFAR) diversity program, termed the CFAR Diversity, Equity, and Inclusion Pathway Initiative (CDEIPI), was created in 2021 to engage high school students and graduate students from Underrepresented Minorities/Black, Indigenous, and People of Color populations. SETTING: The Texas D-CFAR CDEIPI has partnered with 2 Texas high schools with predominantly economically disadvantaged and minority student populations-Michael E. DeBakey High School for Health Professions in Houston, TX, and the South Texas Independent School District Medical Professions High School in Olmito, TX in the Rio Grande Valley. METHODS: A total of 370 high school student learners at both partner schools participated in presentations of research and career paths related to HIV-1 and SARS-CoV-2 during the 2021-2022 academic year. Afterward, learners completed anonymous surveys to share their self-reported interest in research degrees and careers. RESULTS: Learners reported increased knowledge of related science content and interest in research careers, including HIV-1 research, after each of the sessions. CONCLUSIONS: The programming has been of interest to student learners, and future additions intend to build upon the Texas D-CFAR CDEIPI.


Subject(s)
Acquired Immunodeficiency Syndrome , COVID-19 , HIV Infections , HIV Seropositivity , HIV-1 , Humans , Acquired Immunodeficiency Syndrome/epidemiology , Acquired Immunodeficiency Syndrome/prevention & control , Diversity, Equity, Inclusion , Texas/epidemiology , SARS-CoV-2 , HIV Infections/epidemiology , HIV Infections/prevention & control
10.
J Acquir Immune Defic Syndr ; 94(2S): S5-S12, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37707842

ABSTRACT

BACKGROUND: There is an urgent need to increase diversity among scientific investigators in the HIV research field to be more reflective of communities highly affected by the HIV epidemic. Thus, it is critical to promote the inclusion and advancement of early-stage scholars from racial and ethnic groups underrepresented in HIV science and medicine. METHODS: To widen the HIV research career pathway for early-stage scholars from underrepresented minority groups, the National Institutes of Health supported the development of the Centers for AIDS Research (CFAR) Diversity, Equity, and Inclusion Pathway Initiative (CDEIPI). This program was created through partnerships between CFARs and Historically Black Colleges and Universities and other Minority Serving Institutions throughout the United States. RESULTS: Seventeen CFARs and more than 20 Historically Black Colleges and Universities and Minority Serving Institutions have participated in this initiative to date. Programs were designed for the high school (8), undergraduate (13), post baccalaureate (2), graduate (12), and postdoctoral (4) levels. Various pedagogical approaches were used including didactic seminar series, intensive multiday workshops, summer residential programs, and mentored research internship opportunities. During the first 18 months of the initiative, 257 student scholars participated in CDEIPI programs including 150 high school, 73 undergraduate, 3 post baccalaureate, 27 graduate, and 4 postdoctoral students. CONCLUSION: Numerous student scholars from a wide range of educational levels, geographic backgrounds, and racial and ethnic minority groups have engaged in CDEIPI programs. Timely and comprehensive program evaluation data will be critical to support a long-term commitment to this unique training initiative.


Subject(s)
Acquired Immunodeficiency Syndrome , HIV Infections , United States , Humans , Ethnicity , Diversity, Equity, Inclusion , Minority Groups
11.
J Cell Physiol ; 227(2): 609-17, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21448926

ABSTRACT

Eukaryotic RNA polymerase II transcriptional elongation is a tightly regulated process and is dependent upon positive transcription elongation factor-b (P-TEFb). The core P-TEFb complex is composed of Cdk9 and Cyclin T and is essential for the expression of most protein coding genes. Cdk9 kinase function is dependent upon phosphorylation of Thr186 in its T-loop. In this study, we examined kinases and signaling pathways that influence Cdk9 T-loop phosphorylation. Using an RNAi screen in HeLa cells, we found that Cdk9 T-loop phosphorylation is regulated by Ca(2+)/calmodulin-dependent kinase 1D (CaMK1D). Using small molecules inhibitors in HeLa cells and primary CD4(+) T lymphocytes, we found that the Ca(2+) signaling pathway is required for Cdk9 T-loop phosphorylation. Inhibition of Ca(2+) signaling led to dephosphorylation of Thr186 on Cdk9. In reporter plasmid assays, inhibition of the Ca(2+) signaling pathway repressed the PCNA promoter and HIV-1 Tat transactivation of the HIV-1 LTR, but not HTLV-1 Tax transactivation of the HTLV-1 LTR, suggesting that perturbation of the Ca(2+) pathway and reduction of Cdk9 T-loop phosphorylation inhibits transcription units that have a rigorous requirement for P-TEFb function.


Subject(s)
Calcium Signaling/physiology , Cyclin-Dependent Kinase 9/metabolism , CD4-Positive T-Lymphocytes/physiology , Calcium-Calmodulin-Dependent Protein Kinase Type 1/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 1/metabolism , Cyclin-Dependent Kinase 9/chemistry , Cyclin-Dependent Kinase 9/genetics , Gene Expression Regulation/physiology , HeLa Cells , Humans , Positive Transcriptional Elongation Factor B/metabolism , Proteasome Endopeptidase Complex , RNA Interference , Signal Transduction/physiology
12.
J Am Chem Soc ; 134(17): 7459-66, 2012 May 02.
Article in English | MEDLINE | ID: mdl-22486461

ABSTRACT

The anthracene cyclophane bis-anthracene (BA) can undergo a [4 + 4] photocycloaddition reaction that results in a photodimer with two cyclobutane rings. We find that the subsequent dissociation of the dimer, which involves the rupture of two carbon-carbon bonds, is strongly accelerated by the application of mild pressures. The reaction kinetics of the dimer dissociation in a Zeonex (polycycloolefin) polymer matrix were measured at various pressures and temperatures. Biexponential reaction kinetics were observed for all pressures, consistent with the presence of two different isomers of bis(anthracene). One of the rates showed a strong dependence on pressure, yielding a negative activation volume for the dissociation reaction of ΔV(++) = -16 Å(3). The 93 kJ/mol activation energy for the dissociation reaction at ambient pressure is lowered by more than an order of magnitude from 93 to 7 kJ/mol with the application of modest pressure (0.9 GPa). Both observations are consistent with a transition state that is stabilized at higher pressures, and a mechanism for this is proposed in terms of a two-step process where a flattening of the anthracene rings precedes rupture of the cyclobutane rings. The ability to catalyze covalent bond breakage in isolated small molecules using compressive forces may present opportunities for the development of materials that can be activated by acoustic shock or stress.

13.
Retrovirology ; 9: 52, 2012 Jun 22.
Article in English | MEDLINE | ID: mdl-22727189

ABSTRACT

BACKGROUND: Processive elongation of the integrated HIV-1 provirus is dependent on recruitment of P-TEFb by the viral Tat protein to the viral TAR RNA element. P-TEFb kinase activity requires phosphorylation of Thr186 in the T-loop of the CDK9 subunit. In resting CD4+T cells, low levels of T-loop phosphorylated CDK9 are found, which increase significantly upon activation. This suggests that the phosphorylation status of the T-loop is actively regulated through the concerted actions of cellular proteins such as Ser/Thr phosphatases. We investigated the role of phosphatase PPM1A in regulating CDK9 T-loop phosphorylation and its effect on HIV-1 proviral transcription. RESULTS: We found that overexpression of PPM1A inhibits HIV-1 gene expression during viral infection and this required PPM1A catalytic function. Using an artificial CDK tethering system, we further found that PPM1A inhibits CDK9, but not CDK8 mediated activation of the HIV-1 LTR. SiRNA depletion of PPM1A in resting CD4+T cells increased the level of CDK9 T-loop phosphorylation and enhanced HIV-1 gene expression. We also observed that PPM1A protein levels are relatively high in resting CD4+T cells and are not up-regulated upon T cell activation. CONCLUSIONS: Our results establish a functional link between HIV-1 replication and modulation of CDK9 T-loop phosphorylation by PPM1A. PPM1A represses HIV-1 gene expression by inhibiting CDK9 T-loop phosphorylation, thus reducing the amount of active P-TEFb available for recruitment to the viral LTR. We also infer that PPM1A enzymatic activity in resting and activated CD4+ T cells are likely regulated by as yet undefined factors.


Subject(s)
CD4-Positive T-Lymphocytes/virology , Gene Expression Regulation, Viral , HIV-1/genetics , Phosphoprotein Phosphatases/metabolism , Positive Transcriptional Elongation Factor B/metabolism , CD4-Positive T-Lymphocytes/metabolism , Cyclin-Dependent Kinase 8/genetics , Cyclin-Dependent Kinase 8/metabolism , Cyclin-Dependent Kinase 9/genetics , Cyclin-Dependent Kinase 9/metabolism , Gene Knockdown Techniques , HEK293 Cells , HIV Infections/virology , HIV Long Terminal Repeat , HIV-1/metabolism , HeLa Cells , Humans , Phosphoprotein Phosphatases/genetics , Phosphorylation , Positive Transcriptional Elongation Factor B/genetics , Protein Phosphatase 2C , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Transfection
14.
Retrovirology ; 9: 90, 2012 Oct 30.
Article in English | MEDLINE | ID: mdl-23110726

ABSTRACT

BACKGROUND: HIV-1 Tat activates RNA Polymerase II (RNAP II) elongation of the integrated provirus by recruiting a protein kinase known as P-TEFb to TAR RNA at the 5' end of nascent viral transcripts. The catalytic core of P-TEFb contains CDK9 and Cyclin T1 (CCNT1). A human endogenous complexome has recently been described - the set of multi-protein complexes in HeLa cell nuclei. We mined this complexome data set and identified 12 distinct multi-protein complexes that contain both CDK9 and CCNT1. We have termed these complexes CCAPs for CDK9/CCNT1-associated protein complexes. Nine CCAPs are novel, while three were previously identified as Core P-TEFb, the 7SK snRNP, and the Super-Elongation Complex. We have investigated the role of five newly identified CCAPs in Tat function and viral gene expression. RESULTS: We examined five CCAPs that contain: 1) PPP1R10/TOX3/WDR82; 2) TTF2; 3) TPR; 4) WRNIP1; 5) FBXO11/CUL1/SKP1. SiRNA depletions of protein subunits of the five CCAPs enhanced Tat activation of an integrated HIV-1 LTR-Luciferase reporter in TZM-bl cells. Using plasmid transfection assays in HeLa cells, we also found that siRNA depletions of TTF2, FBXO11, PPP1R10, WDR82, and TOX3 enhanced Tat activation of an HIV-1 LTR-luciferase reporter, but the depletions did not enhance expression of an NF-κB reporter plasmid with the exception of PPP1R10. We found no evidence that depletion of CCAPs perturbed the level of CDK9/CCNT1 in the 7SK snRNP. We also found that the combination of siRNA depletions of both TTF2 and FBXO11 sensitized a latent provirus in Jurkat cells to reactivation by sub-optimal amounts of αCD3/CD28 antibodies. CONCLUSIONS: Our results identified five novel CDK9/CCNT1 complexes that are capable of negative regulation of HIV-1 Tat function and viral gene expression. Because siRNA depletions of CCAPs enhance Tat function, it is possible that these complexes reduce the level of CDK9 and CCNT1 available for Tat, similar to the negative regulation of Tat by the 7SK snRNP. Our results highlight the complexity in the biological functions of CDK9 and CCNT1.


Subject(s)
Cyclin T/metabolism , Cyclin-Dependent Kinase 9/metabolism , HIV-1/pathogenicity , Host-Pathogen Interactions , Multiprotein Complexes/metabolism , RNA, Small Interfering/metabolism , tat Gene Products, Human Immunodeficiency Virus/metabolism , Gene Expression Regulation, Viral , Humans , Transcription, Genetic , Virulence Factors/metabolism
15.
J Virol ; 85(22): 11544-56, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21775458

ABSTRACT

More than a decade ago, three viral oncoproteins, adenovirus type 9 E4-ORF1, human T-lymphotropic virus type 1 Tax, and high-risk human papillomavirus E6, were found to encode a related carboxyl-terminal PDZ domain-binding motif (PBM) that mediates interactions with a select group of cellular PDZ proteins. Recent studies have shown that many other viruses also encode PBM-containing proteins that bind to cellular PDZ proteins. Interestingly, these recently recognized viruses include not only some with oncogenic potential (hepatitis B virus, rhesus papillomavirus, cottontail rabbit papillomavirus) but also many without this potential (influenza virus, Dengue virus, tick-borne encephalitis virus, rabies virus, severe acute respiratory syndrome coronavirus, human immunodeficiency virus). Examination of the cellular PDZ proteins that are targets of viral PBMs reveals that the viral proteins often interact with the same or similar types of PDZ proteins, most notably Dlg1 and other members of the membrane-associated guanylate kinase protein family, as well as Scribble. In addition, cellular PDZ protein targets of viral PBMs commonly control tight junction formation, cell polarity establishment, and apoptosis. These findings reveal a new theme in virology wherein many different virus families encode proteins that bind and perturb the function of cellular PDZ proteins. The inhibition or perturbation of the function of cellular PDZ proteins appears to be a widely used strategy for viruses to enhance their replication, disseminate in the host, and transmit to new hosts.


Subject(s)
Host-Pathogen Interactions , Viral Proteins/metabolism , Viruses/pathogenicity , Animals , Humans , Protein Binding , Protein Interaction Domains and Motifs
16.
J Virol ; 85(20): 10639-48, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21849460

ABSTRACT

The influenza A virus NS1 protein contains a conserved 4-amino-acid-residue PDZ-ligand binding motif (PBM) at the carboxyl terminus that can function as a virulence determinant by targeting cellular PDZ proteins. The NS1 proteins from avian and human viral isolates have consensus PBM sequences ESEV and RSKV, respectively. Currently circulating highly pathogenic H5N1 viruses contain the ESEV PBM which specifically associates with the PDZ proteins Scribble, Dlg1, MAGI-1, MAGI-2, and MAGI-3. In this study, we found NS1 proteins from viral isolates that contain the PBM sequence RSKV, KSEV, or EPEV are unable to associate with these PDZ proteins. Other results showed that the ESEV PBM mediates an indirect association with PDZ protein, Lin7C, via an interaction with Dlg1. Infection with a virus that expresses a NS1 protein with the ESEV PBM results in colocalization of NS1, Scribble, and Dlg1 within perinuclear puncta and mislocalization of plasma membrane-associated Lin7C to the cytoplasm. Infection of polarized MDCK cells with the ESEV virus additionally results in functional disruption of the tight junction (TJ) as measured by altered localization of TJ markers ZO-1 and Occludin, decreased transepithelial electrical resistance, and increased fluorescein isothiocyanate (FITC)-inulin diffusion across the polarized cell monolayer. A similar effect on the TJ was observed in MDCK cells depleted for either Scribble or Dlg1 by small interfering RNA (siRNA). These findings indicate that ESEV PBM-mediated binding of NS1 to Scribble and Dlg1 functions to disrupt the cellular TJ and that this effect likely contributes to the severe disease associated with highly pathogenic H5N1 influenza A viruses.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Host-Pathogen Interactions , Influenza A Virus, H5N1 Subtype/pathogenicity , Membrane Proteins/metabolism , Tight Junctions/physiology , Tumor Suppressor Proteins/metabolism , Viral Nonstructural Proteins/metabolism , Animals , Cell Line , Discs Large Homolog 1 Protein , Humans , Protein Binding
17.
J Leukoc Biol ; 112(5): 1245-1259, 2022 11.
Article in English | MEDLINE | ID: mdl-35362118

ABSTRACT

The reservoirs of the HIV display cellular properties resembling long-lived immune memory cells that could be exploited for viral clearance. Our interest in developing a cure for HIV stems from the studies of immunologic memory against infections. We and others have found that long-lived immune memory cells employ prosurvival autophagy and antiapoptotic mechanisms to protect their longevity. Here, we describe the rationale for the development of an approach to clear HIV-1 by selective elimination of host cells harboring replication-competent HIV (SECH). While reactivation of HIV-1 in the host cells with latency reversing agents (LRAs) induces viral gene expression leading to cell death, LRAs also simultaneously up-regulate prosurvival antiapoptotic molecules and autophagy. Mechanistically, transcription factors that promote HIV-1 LTR-directed gene expression, such as NF-κB, AP-1, and Hif-1α, can also enhance the expression of cellular genes essential for cell survival and metabolic regulation, including Bcl-xL, Mcl-1, and autophagy genes. In the SECH approach, we inhibit the prosurvival antiapoptotic molecules and autophagy induced by LRAs, thereby allowing maximum killing of host cells by the induced HIV-1 proteins. SECH treatments cleared HIV-1 infections in humanized mice in vivo and in HIV-1 patient PBMCs ex vivo. SECH also cleared infections by the SIV in rhesus macaque PBMCs ex vivo. Research efforts are underway to improve the efficacy and safety of SECH and to facilitate the development of SECH as a therapeutic approach for treating people with HIV.


Subject(s)
HIV Infections , HIV-1 , Mice , Animals , Virus Latency , NF-kappa B , Macaca mulatta , Myeloid Cell Leukemia Sequence 1 Protein/therapeutic use , Transcription Factor AP-1 , Autophagy , Apoptosis , CD4-Positive T-Lymphocytes , Virus Activation/genetics
18.
J Virol ; 84(21): 11164-74, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20702615

ABSTRACT

The NS1 protein from influenza A viruses contains a four-amino-acid sequence at its carboxyl terminus that is termed the PDZ-binding motif (PBM). The NS1 PBM is predicted to bind to cellular PDZ proteins and functions as a virulence determinant in infected mice. ESEV is the consensus PBM sequence of avian influenza viruses, while RSKV is the consensus sequence of human viruses. Currently circulating highly pathogenic H5N1 influenza viruses encode an NS1 protein with the ESEV PBM. We identified cellular targets of the avian ESEV PBM and identified molecular mechanisms involved in its function. Using glutathione S-transferase (GST) pull-down assays, we found that the ESEV PBM enables NS1 to associate with the PDZ proteins Scribble, Dlg1, MAGI-1, MAGI-2, and MAGI-3. Because Scribble possesses a proapoptotic activity, we investigated the interaction between NS1 and Scribble. The association between NS1 and Scribble is direct and requires the ESEV PBM and two Scribble PDZ domains. We constructed recombinant H3N2 viruses that encode an H6N6 avian virus NS1 protein with either an ESEV or mutant ESEA PBM, allowing an analysis of the ESEV PBM in infections in mammalian cells. The ESEV PBM enhanced viral replication up to 4-fold. In infected cells, NS1 with the ESEV PBM relocalized Scribble into cytoplasmic puncta concentrated in perinuclear regions and also protected cells from apoptosis. In addition, the latter effect was eliminated by small interfering RNA (siRNA)-mediated Scribble depletion. This study shows that one function of the avian ESEV PBM is to reduce apoptosis during infection through disruption of Scribble's proapoptotic function.


Subject(s)
Apoptosis , Influenza A virus/physiology , Influenza in Birds/pathology , Membrane Proteins/physiology , Tumor Suppressor Proteins/physiology , Viral Nonstructural Proteins/physiology , Animals , Binding Sites , Birds , Cell Line , Humans , Influenza A Virus, H3N2 Subtype , Influenza A virus/chemistry , PDZ Domains
19.
PLoS Pathog ; 5(1): e1000263, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19148268

ABSTRACT

Cyclin T1 is a regulatory subunit of a general RNA polymerase II elongation factor known as P-TEFb. Cyclin T1 is also required for Tat transactivation of HIV-1 LTR-directed gene expression. Translation of Cyclin T1 mRNA has been shown to be repressed in human monocytes, and this repression is relieved when cells differentiate to macrophages. We identified miR-198 as a microRNA (miRNA) that is strongly down-regulated when monocytes are induced to differentiate. Ectopic expression of miR-198 in tissue culture cells reduced Cyclin T1 protein expression, and plasmid reporter assays verified miR-198 target sequences in the 3' untranslated region (3'UTR) of Cyclin T1 mRNA. Cyclin T1 protein levels increased when an inhibitor of miR-198 was transfected into primary monocytes, and overexpression of miR-198 in primary monocytes repressed the normal up-regulation of Cyclin T1 during differentiation. Expression of an HIV-1 proviral plasmid and HIV-1 replication were repressed in a monocytic cell line upon overexpression of miR-198. Our data indicate that miR-198 functions to restrict HIV-1 replication in monocytes, and its mechanism of action appears to involve repression of Cyclin T1 expression.


Subject(s)
Cyclins/drug effects , HIV-1/genetics , MicroRNAs/physiology , Monocytes/virology , 3' Untranslated Regions/drug effects , Cell Differentiation , Cell Line , Cyclin T , Down-Regulation , HIV Infections/genetics , HIV Long Terminal Repeat , Humans , Macrophages/virology , Monocytes/drug effects , Monocytes/metabolism , RNA, Messenger/metabolism , Virus Replication/genetics
20.
PLoS One ; 16(4): e0244771, 2021.
Article in English | MEDLINE | ID: mdl-33914760

ABSTRACT

Although current antiretroviral therapies (ART) are successful in controlling HIV-1 infection, a stable viral reservoir reactivates when ART is discontinued. Consequently, there is a major research effort to develop approaches to disrupt the latent viral reservoir and enhance the immune system's ability to clear HIV-1. A number of small molecules, termed latency reversal agents (LRAs), have been identified which can reactivate latent HIV-1 in cell lines and patients' cells ex vivo. However, clinical trials have suggested that combinations of LRAs will be required to efficiently reactivate HIV-1 in vivo, especially LRAs that act synergistically by functioning through distinct pathways. To identify novel LRAs, we used an image-based assay to screen a natural compound library for the ability to induce a low level of aggregation of resting primary CD4+ T cells from healthy donors. We identified celastrol as a novel LRA. Celastrol functions synergistically with other classes of LRA to reactivate latent HIV-1 in a Jurkat cell line, suggesting a novel mechanism in its LRA activity. Additionally, celastrol does not appear to activate resting CD4+ T cells at levels at which it can reactivate latent HIV-1. Celastrol appears to represent a novel class of LRAs and it therefore can serve as a lead compound for LRA development.


Subject(s)
Anti-HIV Agents/pharmacology , HIV Infections/drug therapy , HIV-1/drug effects , Pentacyclic Triterpenes/pharmacology , Virus Latency/drug effects , CD4-Positive T-Lymphocytes/drug effects , Cells, Cultured , Drug Discovery , HIV-1/physiology , Humans , Jurkat Cells
SELECTION OF CITATIONS
SEARCH DETAIL