Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Biologicals ; 51: 18-24, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29225046

ABSTRACT

Currently, porcine circovirus type 2b (PCV2b) is the dominant PCV2 genotype causing postweaning multisystemic wasting disease (PMWS) in pigs worldwide. Efforts have been made to develop various recombinant capsid proteins of PCV2b used in vaccines against PCV2b. However, the nuclear localization signal (NLS) of PCV2b capsid protein (CP) was found to inhibit the expression of the whole length capsid protein in E.coli. Here, we expressed a NLS-deleted capsid protein (ΔCP) of PCV2b in Hansenula polymorpha based on the capsid protein of PCV2b strain Y-7 isolated in China. Comparatively, the ΔCP was expressed at a higher level than the CP. The purified ΔCP could self-assemble into virus like particles (VLPs) with similar morphology of the VLPs formed by CP. The purified ΔCP could be recognized by the anti-sera derived from the mice immunized by inactivated PCV2b particles. Furthermore, it induced higher levels of PCV2b specific antibodies than the purified CP in mice. These results showed that the ΔCP, a recombinant PCV2b capsid protein without nuclear localization signal sequence, could be efficiently expressed in Hansenula polymorpha, and used as a candidate antigen for the development of PCV2b vaccines.


Subject(s)
Capsid Proteins/immunology , Circovirus/immunology , Gene Expression/immunology , Nuclear Localization Signals/immunology , Animals , Antibodies, Viral/immunology , Capsid Proteins/genetics , Capsid Proteins/ultrastructure , Circoviridae Infections/immunology , Circoviridae Infections/virology , Circovirus/genetics , Circovirus/metabolism , Female , Mice, Inbred BALB C , Nuclear Localization Signals/genetics , Pichia/genetics , Recombinant Proteins/immunology , Sequence Deletion , Swine , Vaccination , Viral Vaccines/genetics , Viral Vaccines/immunology
2.
J Immunol ; 195(1): 289-97, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25994966

ABSTRACT

Accurate cellular localization plays a crucial role in the effective function of most signaling proteins, and nuclear trafficking is central to the function of transcription factors. IFN regulatory factor (IRF)3 is a master transcription factor responsible for the induction of type I IFN, which plays a crucial role in host antiviral innate immune responses. However, the mechanisms for control and regulation of IRF3 nuclear import largely remain to be elucidated. In our study, we identified a bipartite nuclear localization signal (NLS) in IRF3, with two interdependent basic clusters separated by a 7-aa linker. Our study further demonstrated that the bipartite NLS of IRF3 is also critical for IRF3 DNA-binding activity, indicating that the two functions of this region are integrated, which is in contrast to other IRFs. Furthermore, the IFN bioassay and infection studies suggest that IRF3 NLS is essential to the IRF3-mediated IFN responses and antiviral immunity. Overall, our results reveal a previously unrecognized bipartite NLS for IRF3 that contains both DNA-binding activity and nuclear import function, and they shed light on the regulatory mechanisms of IRF3 activation and IRF3-mediated antiviral responses.


Subject(s)
Cell Nucleus/immunology , DNA/immunology , Fibroblasts/immunology , Interferon Regulatory Factor-3/immunology , Nuclear Localization Signals/immunology , Active Transport, Cell Nucleus/genetics , Active Transport, Cell Nucleus/immunology , Animals , Binding Sites , Cell Line , Cell Nucleus/metabolism , Cell Nucleus/virology , DNA/metabolism , Fibroblasts/metabolism , Fibroblasts/virology , Gene Expression , Humans , Immunity, Innate , Interferon Regulatory Factor-3/chemistry , Interferon Regulatory Factor-3/classification , Interferon Regulatory Factor-3/genetics , Mice , Models, Molecular , Molecular Sequence Data , Nuclear Localization Signals/genetics , Phylogeny , Protein Binding , Sendai virus/immunology , Sequence Alignment , Signal Transduction , Vesiculovirus/immunology
3.
J Autoimmun ; 55: 33-41, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24786898

ABSTRACT

Non-resolving inflammation is a major contributor to chronic disease pathogenesis, including that of cancer, chronic obstructive pulmonary disease, asthma, arthritis, inflammatory bowel disease, multiple sclerosis and obesity. Some cytokines, such as IL-1α and IL-33, may act as endogenous alarmins that contribute to non-resolving inflammation. These cytokines are constitutively expressed in the nucleus and are thought to promote inflammation only upon release during tissue damage or cell necrosis. However, the importance of their nuclear localization in immune homeostasis is not fully understood. We describe herein a novel mouse model in which the nuclear localization signal of IL-33 is abolished and demonstrate for the first time that, alone, altered subcellular localization of IL-33 dramatically affects immune homeostasis. Heterozygous IL33(tm1/+) mice display elevated serum IL-33 levels, indicating that IL-33 is constitutively released when not actively targeted to the nucleus. IL33(tm1/+) mice succumb to lethal inflammation characterized by eosinophil-dominated immune cell infiltration of multiple organs. The profound inflammatory phenotype is dependent on mediators downstream of ST2 as ST2-null mice are protected in spite of high serum IL-33 levels. Importantly, IL-33 transcript levels in this knock-in mouse model remain under endogenous control. We adopt the term "nuclear alarmin" to describe a danger signal that is primarily regulated by nuclear compartmentalization in this fashion.


Subject(s)
Cell Nucleus/immunology , Homeostasis/immunology , Interleukins/immunology , Nuclear Localization Signals/immunology , Receptors, Interleukin/immunology , Animals , Cell Nucleus/genetics , Cell Nucleus/pathology , Homeostasis/genetics , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Interleukin-1 Receptor-Like 1 Protein , Interleukin-33 , Interleukins/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Nuclear Localization Signals/genetics , Receptors, Interleukin/genetics
4.
J Clin Invest ; 123(3): 1390-401, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23454772

ABSTRACT

Adeno-associated virus (AAV) vectors are attractive for gene delivery-based therapeutics, but data from recent clinical trials have indicated that AAV capsids induce a cytotoxic T lymphocyte (CTL) response that eliminates transduced cells. In this study, we used traditional pharmacological agents and AAV mutants to elucidate the pathway of capsid cross-presentation in AAV-permissive cells. Endosomal acidification inhibitors blocked AAV2 antigen presentation by over 90%, while proteasome inhibitors completely abrogated antigen presentation. Using mutant viruses that are defective for nuclear entry, we observed a 90% decrease in capsid antigen presentation. Different antigen presentation efficiencies were achieved by selectively mutating virion nuclear localization signals. Low antigen presentation was demonstrated with basic region 1 (BR1) mutants, despite relatively high transduction efficiency, whereas there was no difference in antigen presentation between BR2 and BR3 mutants defective for transduction, as compared with wild-type AAV2. These results suggest that effective AAV2 capsid antigen presentation is dependent on AAV virion escape from the endosome/lysosome for antigen degradation by proteasomes, but is independent of nuclear uncoating. These results should facilitate the design of effective strategies to evade capsid-specific CTL-mediated elimination of AAV-transduced target cells in future clinical trials.


Subject(s)
Antigen Presentation , Antigens, Viral/immunology , Capsid Proteins/immunology , Dependovirus/immunology , Endosomes/metabolism , Aminopeptidases/metabolism , Animals , Antigens, Viral/genetics , Antigens, Viral/metabolism , Antiviral Agents/pharmacology , Brefeldin A/pharmacology , COS Cells , Capsid Proteins/genetics , Capsid Proteins/metabolism , Chlorocebus aethiops , Cross-Priming , Dependovirus/genetics , Dependovirus/physiology , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Endosomes/virology , Genetic Therapy , HEK293 Cells , Humans , Mice , Mice, Transgenic , Nuclear Localization Signals/genetics , Nuclear Localization Signals/immunology , Nuclear Localization Signals/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Transport , Proteolysis , Serine Endopeptidases/metabolism , Transduction, Genetic , Virus Assembly
5.
Virol J ; 10: 16, 2013 Jan 07.
Article in English | MEDLINE | ID: mdl-23294939

ABSTRACT

BACKGROUND: PCV ORF2 capsid protein was predicted to contribute to the control of replication via an interaction between the Cap and Rep proteins in the nucleoplasm. We previously showed that the nuclear localization signal (NLS) on the capsid protein plays an accessory role in the replication of PCV in vitro. To further evaluate the in vivo characteristics of NLS-chimeric PCV DNA clones, BALB/C mice were inoculated intranasally and intraperitoneally with the DNA clones. RESULTS: As expected, no gross lesions were detected during the study of the inoculated animals. The chimeric PCV12-, PCV1-NLS2- and PCV2-NLS1-inoculated animals had significantly fewer and less severe histopathological lesions in lymphoid tissues than the PCV2-inoculated animals (P < 0.05). PCV12 induced a specific antibody response against PCV2 ORF2 comparable to that induced by wild-type PCV2 but demonstrated a shorter period of viremia and much lower level of virus loads in sera than those in PCV2-inoculated mice. Remarkably, the PCV2-NLS1 and PCV1-NLS2 chimeras replicated in inoculated mice and induced specific antibody responses but failed to produce viral antigens in the lymph nodes or a detectable viremia. CONCLUSIONS: The chimeric PCV2-NLS1 and PCV1-NLS2 demonstrated a lower replication level as compared with wild type of PCV2 or PCV1 in vivo, suggesting that ORF2 NLSs played an accessory role in PCV replication. The chimeric PCV12 is a good candidate for vaccination against PCV2 infection.


Subject(s)
Capsid Proteins , Circovirus/genetics , Circovirus/pathogenicity , DNA, Viral/genetics , Nuclear Localization Signals , Recombinant Proteins , Animals , Antibodies, Viral/blood , Capsid Proteins/chemistry , Capsid Proteins/genetics , Capsid Proteins/immunology , Cell Line , Circoviridae Infections/immunology , Circoviridae Infections/pathology , Circoviridae Infections/virology , Circovirus/classification , Circovirus/physiology , Mice , Mice, Inbred BALB C , Nuclear Localization Signals/genetics , Nuclear Localization Signals/immunology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Virus Replication
6.
Infect Immun ; 79(4): 1779-88, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21220486

ABSTRACT

Interleukin-4-inducing principle from schistosome eggs (IPSE/alpha-1) is a protein produced exclusively by the eggs of the trematode Schistosoma mansoni. IPSE/alpha-1 is a secretory glycoprotein which activates human basophils via an IgE-dependent but non-antigen-specific mechanism. Sequence analyses revealed a potential nuclear localization signal (NLS) at the C terminus of IPSE/alpha-1. Here we show that this sequence (125-PKRRRTY-131) is both necessary and sufficient for nuclear localization of IPSE or IPSE-enhanced green fluorescent protein (EGFP) fusions. While transiently expressed EGFP-IPSE/alpha-1 was exclusively nuclear in the Huh7 and U-2 OS cell lines, a mutant lacking amino acids 125 to 134 showed both nuclear and cytoplasmic staining. Moreover, insertion of the IPSE/alpha-1 NLS into a tetra-EGFP construct rendered the protein nuclear. Alanine scanning mutagenesis revealed a requirement for the KRRR residues. Fluorescence microscopy depicted, and Western blotting further confirmed, that recombinant IPSE/alpha-1 protein added exogenously is rapidly internalized by CHO cells and accumulates in nuclei in an NLS-dependent manner. A mutant protein in which the NLS motif was disrupted by triple mutation (RRR to AAA) was able to penetrate CHO cells but did not translocate to the nucleus. Furthermore, the uptake of native glycosylated IPSE/alpha-1 was confirmed in human primary monocyte-derived dendritic cells and was found to be a calcium- and temperature-dependent process. Live-cell imaging showed that IPSE/alpha-1 is not targeted to lysosomes. In contrast, peripheral blood basophils do not take up IPSE/alpha-1 and do not require the presence of an intact NLS for activation. Taken together, our results suggest that IPSE/alpha-1 may have additional nuclear functions in host cells.


Subject(s)
Egg Proteins/metabolism , Helminth Proteins/metabolism , Host-Parasite Interactions/physiology , Nuclear Localization Signals/metabolism , Schistosomiasis mansoni/metabolism , Amino Acid Sequence , Animals , Blotting, Western , Cell Line , Cell Nucleus/metabolism , Cell Separation , Egg Proteins/genetics , Egg Proteins/immunology , Flow Cytometry , Fluorescent Antibody Technique , Helminth Proteins/genetics , Helminth Proteins/immunology , Humans , Nuclear Localization Signals/genetics , Nuclear Localization Signals/immunology , Ovum/metabolism , Polymerase Chain Reaction , Protein Transport/physiology , Schistosoma mansoni/genetics , Schistosoma mansoni/immunology , Schistosoma mansoni/metabolism , Schistosomiasis mansoni/genetics , Schistosomiasis mansoni/immunology
7.
Blood ; 111(7): 3599-606, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18223166

ABSTRACT

Regulatory T cells (Tregs) have been shown to play a crucial role in maintaining self-tolerance and suppressing autoimmunity. The forkhead transcription factor, FoxP3, is a key molecule necessary and sufficient for Tregs development and function. However, the molecular mechanisms by which FoxP3 regulates the phenotypic (anergic) and the functional (suppressive) characteristics of Tregs are not well defined. Here we found that the promoter DNA-binding activity of AP-1 transcription factors is selectively inhibited in the naturally occurring CD4+ CD25+ Tregs from mice. The impaired AP-1 DNA binding is not the result of the decreased nuclear translocation of AP-1 family transcription factors, including c-Jun, JunB, and c-Fos. FoxP3 significantly suppresses both the transcriptional activity and promoter DNA-binding of AP-1 by interacting with c-Jun. The N-terminus of FoxP3, but not its C-terminus forkhead domain, specifically interacts with phosphorylated c-Jun and alters c-Jun subnuclear distribution. This N-terminus of FoxP3 with nuclear localization signals (FoxP3N/NLS) is able to suppress AP-1 transcriptional activity. Ectopic expression of FoxP3N/NLS sufficiently induces the unresponsiveness of mouse primary CD4+ CD25- T cells, whereas the full-length FoxP3 is required for the suppressive functions of Tregs. These findings uncover one of the mechanisms underlying how FoxP3 maintains the unresponsiveness of Tregs.


Subject(s)
Cell Nucleus/metabolism , Forkhead Transcription Factors/metabolism , Promoter Regions, Genetic/physiology , T-Lymphocytes, Regulatory/metabolism , Transcription Factor AP-1/metabolism , Transcription, Genetic/physiology , Active Transport, Cell Nucleus/physiology , Animals , Autoimmunity/physiology , Cell Line , Cell Nucleus/genetics , Cell Nucleus/immunology , Clonal Anergy/physiology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Humans , Mice , Nuclear Localization Signals/genetics , Nuclear Localization Signals/immunology , Nuclear Localization Signals/metabolism , Protein Binding/genetics , Protein Binding/immunology , Protein Structure, Tertiary/physiology , Self Tolerance/physiology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Transcription Factor AP-1/genetics , Transcription Factor AP-1/immunology , Transcription Factors/genetics , Transcription Factors/immunology , Transcription Factors/metabolism
8.
J Immunol ; 179(6): 3734-41, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17785810

ABSTRACT

Anergy is an important mechanism of maintaining peripheral immune tolerance. T cells rendered anergic are refractory to further stimulation and are characterized by defective proliferation and IL-2 production. We used a model of in vivo anergy induction in murine CD8+ T cells to analyze the initial signaling events in anergic T cells. Tolerant T cells displayed reduced phospholipase Cgamma activation and calcium mobilization, indicating a defect in calcium signaling. This correlated with a block in nuclear localization of NFAT1 in anergic cells. However, we found that stimulation of anergic, but not naive T cells induced nuclear translocation of NFAT2. This suggested that NFAT2 is activated preferentially by reduced calcium signaling, and we confirmed this hypothesis by stimulating naive T cells under conditions of calcium limitation or partial calcineurin inhibition. Thus, our work provides new insight into how T cell stimulation conditions might dictate specific NFAT isoform activation and implicates NFAT2 involvement in the expression of anergy-related genes.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Calcium Signaling/immunology , Clonal Anergy/immunology , NFATC Transcription Factors/metabolism , Nuclear Localization Signals/metabolism , Active Transport, Cell Nucleus/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Calcium/antagonists & inhibitors , Calcium/physiology , Cells, Cultured , Extracellular Space/immunology , Extracellular Space/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , NFATC Transcription Factors/antagonists & inhibitors , NFATC Transcription Factors/physiology , Nuclear Localization Signals/immunology , Self Tolerance/immunology
9.
Vaccine ; 22(13-14): 1709-16, 2004 Apr 16.
Article in English | MEDLINE | ID: mdl-15068854

ABSTRACT

The low efficacy obtained in large animals makes plasmid-based DNA vaccines commercially unviable. Another concern is the presence of antibiotic resistance markers on virtually all conventional plasmids. Here we describe the use of minimalistic, immunogenically defined gene expression (MIDGE) vectors for DNA vaccination. MIDGE are linear, covalently-closed vectors containing all the essential information for gene expression and none of the non-essential and potentially dangerous plasmid backbone sequences. MIDGE vectors can also be chemically modified on both ends at defined positions allowing targeting of the DNA to specific cell types or cellular compartments. Immunisation of mice with simple and end-modified MIDGE vectors showed that they are efficacious tools to generate and/or manipulate antigen-specific immune responses.


Subject(s)
Plasmids/immunology , Vaccines, DNA/immunology , Animals , Antibodies, Neoplasm/biosynthesis , Antibodies, Neoplasm/immunology , Antibody Formation/immunology , Antibody Specificity , Antigens, Neoplasm/biosynthesis , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Female , Gene Expression , Genetic Vectors , Humans , Immunity, Cellular/immunology , Immunization , Immunoglobulin G/immunology , Interferon-gamma/biosynthesis , K562 Cells , Mice , Mice, Inbred BALB C , Nuclear Localization Signals/genetics , Nuclear Localization Signals/immunology , Th1 Cells/immunology , Transfection
10.
Virology ; 305(1): 77-92, 2003 Jan 05.
Article in English | MEDLINE | ID: mdl-12504543

ABSTRACT

The HIV-1 Vpr protein harbors a nuclear localization signal in its N-terminal domain. A peptide bearing this domain and which is designated VprN has been used as a target to screen a phage display single chain Fv (scFv) library. Here we report the isolation of anti-VprN scFv fragments from this library. The purified scFv fragments were able to bind the VprN peptide in an ELISA-based system and to inhibit VprN-mediated nuclear import in permeabilized as well as in intact microinjected cells. Furthermore, the anti-VprN scFv fragments recognized the full-length recombinant Vpr protein and inhibited its nuclear import. The same scFv fragments did not inhibit nuclear import mediated by the nuclear localization signal of the SV40 large T-antigen demonstrating a specific effect. The use of the described inhibitory anti-VprN scFv fragments to study nuclear import of viral karyophilic proteins and their therapeutic potential is discussed.


Subject(s)
Active Transport, Cell Nucleus , Gene Products, vpr/immunology , HIV Antibodies/immunology , HIV-1/immunology , Nuclear Localization Signals/immunology , Animals , Complementarity Determining Regions , Gene Products, vpr/physiology , Humans , Immunoglobulin Fragments/immunology , Peptide Library , Tumor Cells, Cultured , vpr Gene Products, Human Immunodeficiency Virus
11.
J Neuroimmunol ; 94(1-2): 40-7, 1999 Feb 01.
Article in English | MEDLINE | ID: mdl-10376934

ABSTRACT

The murine T-cell clone, L2, requires both IL2 and PRL to proliferate. Proliferation and selected IL2-driven gene expression are blocked by treatment with rapamycin. Since prolactin translocation to the nucleus is IL2 dependent and required for proliferation, experiments were performed to identify activation pathways that might be involved in nuclear transport and proliferation. L2 cells were stimulated with IL2 in the presence and absence of the mTOR inhibitor rapamycin, PI3-kinase inhibitors (wortmannin, LY294002), the p38 MAP kinase inhibitor SB203580 and the vitamin D analog calcipotriol. The immunosuppressant rapamycin markedly inhibited IL2-induced proliferation and prolactin translocation to the nucleus. Similarly, wortmannin and LY294002 inhibited IL2-induced proliferation and markedly decreased the amount of prolactin transported to the nucleus. SB203580 and calcipotriol partially inhibited IL2-induced proliferation but had no effect on prolactin translocation. None of the inhibitors affected Lucifer Yellow uptake indicating that rapamycin, wortmannin and LY294002 did not inhibit early endosomal formation but rather worked to inhibit prolactin translocation at a later point in the retrograde transport pathway.


Subject(s)
Phosphatidylinositol 3-Kinases/metabolism , Prolactin/metabolism , T-Lymphocytes/enzymology , Androstadienes/pharmacology , Animals , Antineoplastic Agents/pharmacology , Biological Transport/drug effects , Biological Transport/immunology , Calcitriol/analogs & derivatives , Calcitriol/pharmacology , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Cell Division/drug effects , Cell Division/immunology , Cell Nucleus/immunology , Cell Nucleus/metabolism , Chromones/pharmacology , Clone Cells , Enzyme Inhibitors/pharmacology , Fluorescent Dyes/pharmacokinetics , Immunosuppressive Agents/pharmacology , Interleukin-2/immunology , Interleukin-2/pharmacology , Isoquinolines/pharmacokinetics , Mice , Morpholines/pharmacology , Nuclear Localization Signals/immunology , Sirolimus/pharmacology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Wortmannin
12.
J Immunol ; 162(12): 7224-32, 1999 Jun 15.
Article in English | MEDLINE | ID: mdl-10358169

ABSTRACT

A close relationship between Sam68, a tyrosine and proline-rich RNA-binding protein, and Src protein tyrosine kinases (PTK) has already been established, also in T lymphocytes. A constitutive phosphorylation of the molecule has also been documented in various transformed T cells, which probably reflects an increased expression of PTK of the Src family. Using the hybridoma T cell line, T8.1, or Jurkat T cells, we investigated the respective contribution of the two Src kinases Fyn and Lck, expressed in T cells, in this phenomenon. By overexpressing the two proteins, we show that the constitutive phosphorylation of Sam68 in vivo directly correlates with cellular Fyn levels, but not with Lck expression, despite the capacity of the PTK to strongly phosphorylate the molecule in vitro. Overexpressed Fyn is mainly localized at the cell membrane. We find that Sam68 phosphorylation, including in the nuclear fraction in which the molecule is predominantly expressed, is lost with a delocalized Fyn mutant deleted of its N-terminal membrane-anchoring domain. Finally, we demonstrate, using a construct encoding a Sam68 molecule without its nuclear localization signal, that nuclear expression of Sam68 is not required for phosphorylation. We conclude that the constitutive phosphorylation of Sam68 in T cells is a Fyn-dependent process occurring in a cell-membrane compartment from which phospho-Sam68 molecules can thereafter accumulate into the nucleus.


Subject(s)
Cell Nucleus/enzymology , Proto-Oncogene Proteins/metabolism , RNA-Binding Proteins/metabolism , T-Lymphocytes/enzymology , Tyrosine/metabolism , src-Family Kinases/metabolism , Adaptor Proteins, Signal Transducing , Animals , Cell Line , Cell Membrane/enzymology , Cell Nucleus/genetics , Cell Nucleus/metabolism , DNA-Binding Proteins , Humans , Hybridomas , Intracellular Fluid/enzymology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Mice , Nuclear Localization Signals/immunology , Phosphorylation , Proto-Oncogene Proteins c-fyn , RNA-Binding Proteins/genetics , T-Lymphocytes/metabolism
13.
J Immunol ; 161(11): 6084-92, 1998 Dec 01.
Article in English | MEDLINE | ID: mdl-9834092

ABSTRACT

Activation of T lymphocytes by Ags or cytokines results in translocation of the transcription factors NF-kappa B, AP-1, NFAT, and STAT from the cytoplasm into the nucleus. The first step in the nuclear import process is recognition of a nuclear localization sequence (NLS) within the karyophilic protein by a cytoplasmic receptor such as the importin (karyopherin)-alpha subunit. The NLSs of NF-kappa B, AP-1, and NFAT differ and the NLS of STAT1 has not yet been identified. Herein we demonstrate that the inducible nuclear import of NF-kappa B, AP-1, NFAT, and STAT1 in Jurkat T lymphocytes is significantly inhibited by a cell-permeable peptide carrying the NLS of the NF-kappa B p50 subunit. NLS peptide-mediated disruption of the nuclear import of these transcription factors results in inhibition of I kappa B alpha and IL-2 gene expression, processes dependent on NF-kappa B or the combination of NF-kappa B, AP-1, and NFAT. Further, we show that inhibitory NLS peptide interacts in vitro with a cytoplasmic NLS receptor complex comprised of the Rch1/importin (karyopherin)-beta heterodimer expressed in Jurkat T cells. Taken together, these data indicate that the inducible nuclear import of NF-kappa B, AP-1, NFAT, and STAT1 in Jurkat T cells can be regulated by NLS peptide delivered noninvasively to the cytoplasm of Jurkat T cells to target members of the importin (karyopherin)-alpha beta NLS receptor complex.


Subject(s)
DNA-Binding Proteins/metabolism , I-kappa B Proteins , NF-kappa B/metabolism , Nuclear Localization Signals/immunology , Nuclear Proteins , Peptides/metabolism , T-Lymphocytes/metabolism , Trans-Activators/metabolism , Transcription Factor AP-1/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Biological Transport/immunology , Cell Membrane Permeability/immunology , Cell Nucleus/immunology , Cell Nucleus/metabolism , Cytoplasm/metabolism , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic/immunology , Humans , Interleukin-2/antagonists & inhibitors , Interleukin-2/biosynthesis , Interleukin-2/genetics , Jurkat Cells , Kinetics , Molecular Sequence Data , NF-KappaB Inhibitor alpha , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , NF-kappa B p50 Subunit , NFATC Transcription Factors , Nuclear Localization Signals/genetics , Peptides/genetics , Peptides/pharmacology , STAT1 Transcription Factor , Signal Transduction/immunology , Trans-Activators/antagonists & inhibitors , Transcription Factor AP-1/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors
14.
J Immunol ; 161(5): 2642-7, 1998 Sep 01.
Article in English | MEDLINE | ID: mdl-9725267

ABSTRACT

HIV-1 gag p17 protein is an attractive target for molecular intervention, because it is involved in the viral replication cycle at both the pre- and postintegration levels. In the present experiments, we targeted p17 by intracellularly expressing a cDNA encoding an Ab to p17. cDNA from a hybridoma-secreting Ab to p17 was cloned, sequenced, reconstructed as a single-chain Ab fragment (scFv), and expressed in the cytoplasm or nucleus with appropriate retention signals. The expressed scFvs had no effect on T cell growth or CD4 expression and bound specifically to HIV-1 p17. Human CD4+ Jurkat T cells that expressed scFvs and were infected with HIV-1 showed a marked reduction in virus replication compared with cells expressing vector alone. The inhibition of virus replication was more pronounced when scFvs were expressed in the cytoplasm rather than the nucleus. From these studies, we conclude that the intracellular expression of a single-chain Ab to p17 inhibits HIV replication; in addition, the degree of inhibition is related to the intracellular targeting site.


Subject(s)
Anti-HIV Agents/immunology , Antibodies, Monoclonal/genetics , DNA, Complementary/immunology , DNA, Complementary/pharmacology , Gene Products, gag/immunology , HIV Antigens/immunology , HIV-1/immunology , Immunoglobulin Fc Fragments/genetics , Viral Proteins , Virus Replication/drug effects , Amino Acid Sequence , Animals , Anti-HIV Agents/pharmacology , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacology , Binding Sites, Antibody , Cell Line , Cell Nucleus/immunology , Cell Nucleus/metabolism , Cloning, Molecular , Cytoplasm/immunology , Cytoplasm/metabolism , Escherichia coli/genetics , Escherichia coli/immunology , Genetic Vectors , HIV-1/genetics , HIV-1/physiology , Humans , Hybridomas , Immunoglobulin Fc Fragments/biosynthesis , Immunoglobulin Fc Fragments/metabolism , Immunoglobulin Fc Fragments/pharmacology , Immunoglobulin Variable Region/biosynthesis , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/metabolism , Immunoglobulin Variable Region/pharmacology , Intracellular Fluid/immunology , Intracellular Fluid/metabolism , Jurkat Cells , Mice , Molecular Sequence Data , Nuclear Localization Signals/genetics , Nuclear Localization Signals/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/virology , Virus Replication/genetics , Virus Replication/immunology , gag Gene Products, Human Immunodeficiency Virus
15.
J Immunol ; 161(6): 2881-7, 1998 Sep 15.
Article in English | MEDLINE | ID: mdl-9743349

ABSTRACT

Multiple transcription factors are activated in the cytoplasm and translocated to the nucleus where they exert positive or negative control over cellular genes. Such subcellular traffic of transcription factors usually requires the presence of a positively charged nuclear localization sequence (NLS). Upstream stimulating factor 2 (USF2) is one of the few transcription factors that contain two potential domains for nuclear localization. In addition to the conventional basic NLS, USF2 contains a highly conserved USF-specific region that is involved in its nuclear translocation. In the present work, the induction of translocation of USF2 into the mast cell nucleus was observed and found to be dependent on activation of the cells either by IL-3 or IgE-Ag. It was also observed that the prevention of the translocation of USF2 to the nucleus, using a peptide derived from the specific USF-NLS region, significantly inhibited their IL-3-mediated survival. Thus, our findings show a direct connection between mast cell surface receptor-mediated USF2 nuclear translocation and cell viability.


Subject(s)
Cell Nucleus/metabolism , DNA-Binding Proteins , Mast Cells/metabolism , Nuclear Localization Signals/immunology , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Biological Transport/drug effects , Bone Marrow Cells , Carrier Proteins , Cell Survival/drug effects , Cell Survival/immunology , Cells, Cultured , Fibroblast Growth Factor 4 , Fibroblast Growth Factors/pharmacology , Growth Inhibitors/chemical synthesis , Growth Inhibitors/pharmacology , Immune Sera/pharmacology , Interleukin-3/antagonists & inhibitors , Interleukin-3/physiology , Mast Cells/drug effects , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Nuclear Localization Signals/drug effects , Nuclear Localization Signals/physiology , Peptides/chemical synthesis , Peptides/pharmacology , Proto-Oncogene Proteins/pharmacology , Receptors, Cytoplasmic and Nuclear/physiology , Recombinant Proteins , Transcription Factors/antagonists & inhibitors , Transcription Factors/chemical synthesis , Transcription Factors/immunology , Transcription Factors/pharmacology , Upstream Stimulatory Factors
16.
J Steroid Biochem Mol Biol ; 46(3): 309-20, 1993 Sep.
Article in English | MEDLINE | ID: mdl-9831479

ABSTRACT

The binding of cell-free activated glucocorticoid receptor-steroid complexes from HTC cells to various preparations of HTC and rat liver nuclei has been examined under conditions that did or did not support the nuclear translocation of macromolecules via nuclear pores. To the best of our knowledge, this is the first such study with functionally active isolated nuclei. Conventionally prepared HTC nuclei were found to be porous, as determined from their inability to exclude the fluorescent macromolecule phycoerythrin (PE) at 4 degrees C. Thus the nuclear binding of activated complexes to these nuclei can not involve nuclear translocation. Further studies, using established conditions with sealed nuclei prepared from rat liver, revealed that nuclear translocation of PE containing a covalently linked, authentic nuclear translocation sequence could be obtained at 22 degrees C, but not at 4 degrees C. However, under the same conditions, activated glucocorticoid complexes displayed equal levels of nuclear binding at both temperatures. We therefore conclude that the current translocation conditions with intact rat liver nuclei are not sufficient to reproduce the nuclear transport of glucocorticoid complexes observed in intact cells. The nuclear binding that was seen with intact rat liver nuclei was not affected by aurintricarboxylic acid, which selectively inhibits protein-nucleic acid interactions. The antibody AP-64, shown to be specific for amino acids 506-514 of the nuclear translocation sequence of the rat glucocorticoid receptor, inhibited the nuclear binding of activated complexes, apparently by blocking receptor access to the nuclear membrane. Collectively, these data argue that activated complex binding to nuclei capable of nuclear translocation involves only an association with nuclear membrane components such as nuclear pores. Thus this system, and these reagents, may be useful in future studies of activated complex binding to nuclear pores.


Subject(s)
Cell Nucleus/metabolism , Liver/metabolism , Receptors, Glucocorticoid/metabolism , Amino Acid Sequence , Animals , Antibodies/pharmacology , Aurintricarboxylic Acid/pharmacology , Binding, Competitive , Cell Extracts/pharmacology , Dexamethasone/metabolism , Epitopes/chemistry , Epitopes/immunology , Male , Molecular Sequence Data , Nuclear Envelope/physiology , Nuclear Localization Signals/immunology , Nuclear Localization Signals/physiology , Oocytes/metabolism , Peptide Fragments/pharmacology , Phycoerythrin/metabolism , Rats , Spermatozoa/metabolism , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...