Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
PLoS One ; 18(8): e0283489, 2023.
Article in English | MEDLINE | ID: mdl-37639428

ABSTRACT

The oral cholera vaccine WC-rBS consists of 4 different inactivated strains of Vibrio cholerae (LPS source) admixed with recombinant cholera toxin B subunit. Because of its unique composition and anti-inflammatory properties reported for both CTB and low doses of LPS from other Gram-negative bacteria, we speculated that WC-rBS might have anti-inflammatory potential in a chronic autoimmune disease such as inflammatory bowel diseases. First in vitro endotoxin tolerance experiments showed the surprising WC-rBS potential in the modulation of inflammatory responses on both PBMCs and THP1 cells. WC-rBS was further evaluated in the Dextran Sodium Sulfate colitis mouse model. Administrated orally at different dosages, WC-rBS vaccine was safe and showed immunomodulatory properties when administered in a preventive mode (before and during the induction of DSS colitis) as well as in a curative mode (after colitis induction); with improvement of disease activity index (from 27 to 73%) and histological score (from 65 to 88%). Interestingly, the highest therapeutic effect of WC-rBS vaccine was observed with the lowest dosage, showing even better anti-inflammatory properties than mesalamine; an approved 5-aminosalicylic acid drug for treating IBD patients. In summary, this is the first time that a prophylactic medicine, safe and approved for prevention of an infectious disease, showed a benefit in an inflammatory bowel disease model, potentially offering a novel therapeutic modality for IBD patients.


Subject(s)
Cholera Vaccines , Colitis , Inflammatory Bowel Diseases , Vibrio cholerae , Animals , Mice , Lipopolysaccharides , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/prevention & control , Colitis/chemically induced , Colitis/drug therapy , Colitis/prevention & control , Mesalamine
2.
Vaccine ; 40(27): 3797-3801, 2022 06 15.
Article in English | MEDLINE | ID: mdl-35610103

ABSTRACT

Repurposing of existing drugs and vaccines for diseases that they were not originally intended for is a promising research field. Recently there has been evidence that oral cholera vaccine might be used in the treatment of inflammatory disease and some common cancers. Specifically, Ji et al showed that the administration of cholera vaccine after a prostate cancer diagnosis reduced prostate cancer specific mortality rates by almost 50%. In a cohort of men from Stockholm, Sweden, with more detailed cancer data and a higher coverage of exposure to vaccine, we replicated these findings using a marginal structural Cox model. We showed that administration of cholera vaccine after prostate cancer diagnosis is associated with a significant reduction in mortality (HR 0.46, 95% CI 0.31-0.69, p-value 0.0001) even after adjusting for all known confounders. However, the same effect (or even stronger) could be seen for several other traveling vaccines and malaria prophylaxis. Therefore, we conclude that this effect is most likely due to a healthy traveler bias and is an example of residual confounding.


Subject(s)
Cholera Vaccines , Cholera , Prostatic Neoplasms , Administration, Oral , Cholera/prevention & control , Cohort Studies , Disease Progression , Humans , Male , Sweden/epidemiology , Travel
4.
Biologicals ; 61: 22-31, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31474416

ABSTRACT

During the establishment of the duck EB66® cell line as a new cell substrate for vaccine production in industry, a very low level of reverse transcriptase (RT) activity was detected in the culture supernatant by product-enhanced RT assay but a whole battery of tests failed to evidence infectious particles. Results from extensive biochemical and physical testing demonstrated that RT activity was associated to an intracellular, non-enveloped and dense structure different from an infectious retroviruses. In silico analysis of Anas platyrhynchos genome revealed that the most likely candidates for encoding a ribonucleoprotein (RNP)-associated RT were nine copies of chicken repeat 1 (CR1)-like elements, belonging to the non-long terminal repeat retrotransposons. The presence of the full length Anas platyrhynchos chicken repeat 1-like sequence (APCR1) was confirmed in EB66® cells and the related ribonucleic acid was present in the RT-containing fraction of EB66® cells.


Subject(s)
Avian Proteins/genetics , Ducks/genetics , Genome , RNA-Directed DNA Polymerase/genetics , Retroelements , Sequence Analysis, DNA , Animals , Cell Line
5.
Appl Microbiol Biotechnol ; 102(20): 8725-8737, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30091043

ABSTRACT

A live-attenuated, human vaccine against mosquito-borne yellow fever virus has been available since the 1930s. The vaccine provides long-lasting immunity and consistent mass vaccination campaigns counter viral spread. However, traditional egg-based vaccine manufacturing requires about 12 months and vaccine supplies are chronically close to shortages. In particular, for urban outbreaks, vaccine demand can be covered rarely by global stockpiling. Thus, there is an urgent need for an improved vaccine production platform, ideally transferable to other flaviviruses including Zika virus. Here, we present a proof-of-concept study regarding cell culture-based yellow fever virus 17D (YFV) and wild-type Zika virus (ZIKV) production using duck embryo-derived EB66® cells. Based on comprehensive studies in shake flasks, 1-L bioreactor systems were operated with scalable hollow fiber-based tangential flow filtration (TFF) and alternating tangential flow filtration (ATF) perfusion systems for process intensification. EB66® cells grew in chemically defined medium to cell concentrations of 1.6 × 108 cells/mL. Infection studies with EB66®-adapted virus led to maximum YFV titers of 7.3 × 108 PFU/mL, which corresponds to about 10 million vaccine doses for the bioreactor harvest. For ZIKV, titers of 1.0 × 1010 PFU/mL were achieved. Processes were automated successfully using a capacitance probe to control perfusion rates based on on-line measured cell concentrations. The use of cryo-bags for direct inoculation of production bioreactors facilitates pre-culture preparation contributing to improved process robustness. In conclusion, this platform is a powerful option for next generation cell culture-based flavivirus vaccine manufacturing.


Subject(s)
Cell Culture Techniques/methods , Virus Cultivation/methods , Yellow fever virus/growth & development , Zika Virus/growth & development , Animals , Bioreactors/virology , Cell Culture Techniques/instrumentation , Cell Line , Ducks/virology , Viral Vaccines/immunology , Virus Cultivation/instrumentation , Yellow fever virus/immunology , Zika Virus/immunology
7.
Vaccine ; 34(48): 5878-5885, 2016 11 21.
Article in English | MEDLINE | ID: mdl-27997338

ABSTRACT

The selection of a cell substrate is a critical step for the development and manufacturing of a viral vaccine candidate. Several parameters such as cell susceptibility and permissiveness to the viral pathogens but also performance in terms of viral antigens quality and production yields are important considerations when identifying the ideal match between a viral vaccine and cell substrate. The modified vaccinia virus Ankara (MVA) is a replication-deficient viral vector that holds great promise as a vaccine platform, however only limited cell substrates have been tested or are available for industrialization. Here we evaluate the duck embryo-derived EB66® cell line as potential cell substrate for MVA production. To this end, we used two recombinant MVA constructs and demonstrated that EB66® cells are propagating the tested MVA viruses very efficiently, while preserving viral attenuation and transgene expression for up to 20 serial passages. Furthermore we developed upstream and downstream processes that enable industrialization of the virus production. In conclusion, we showed that EB66® cells can be used as potent cell substrate for MVA-based vaccines and represent therefore an attractive alternative for vaccine production.


Subject(s)
Vaccinia virus/genetics , Vaccinia virus/physiology , Viral Vaccines , Virus Cultivation , Virus Replication , Animals , Antigens, Viral , Cell Line , Ducks , Embryo, Nonmammalian/cytology , Humans , Serial Passage , Transgenes , Vaccines, Attenuated , Vaccines, DNA , Vaccinia virus/immunology , Vaccinia virus/pathogenicity
8.
Proc Natl Acad Sci U S A ; 113(44): 12532-12537, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27791128

ABSTRACT

Therapeutic targeting of the VEGF signaling axis by the VEGF-neutralizing monoclonal antibody bevacizumab has clearly demonstrated clinical benefit in cancer patients. To improve this strategy using a polyclonal approach, we developed a vaccine targeting VEGF using 3D-structured peptides that mimic the bevacizumab binding site. An in-depth study on peptide optimization showed that the antigen's 3D structure is essential to achieve neutralizing antibody responses. Peptide 1 adopts a clear secondary, native-like structure, including the typical cysteine-knot fold, as evidenced by CD spectroscopy. Binding and competition studies with bevacizumab in ELISA and surface plasmon resonance analysis revealed that peptide 1 represents the complete bevacizumab binding site, including the hairpin loop (ß5-turn-ß6) and the structure-supporting ß2-α2-ß3 loop. Vaccination with peptide 1 elicited high titers of cross-reactive antibodies to VEGF, with potent neutralizing activity. Moreover, vaccination-induced antisera displayed strong angiostatic and tumor-growth-inhibiting properties in a preclinical mouse model for colorectal carcinoma, whereas antibodies raised with peptides exclusively encompassing the ß5-turn-ß6 loop (peptides 15 and 20) did not. Immunization with peptide 1 or 7 (murine analog of 1) in combination with the potent adjuvant raffinose fatty acid sulfate ester (RFASE) showed significant inhibition of tumor growth in the B16F10 murine melanoma model. Based on these data, we conclude that this vaccination technology, which is currently being investigated in a phase I clinical trial (NCT02237638), can potentially outperform currently applied anti-VEGF therapeutics.


Subject(s)
Bevacizumab/therapeutic use , Colonic Neoplasms/drug therapy , Peptides/therapeutic use , Vaccination/methods , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Amino Acid Sequence , Angiogenesis Inhibitors/immunology , Angiogenesis Inhibitors/therapeutic use , Animals , Antibodies, Neutralizing/immunology , Bevacizumab/immunology , Binding Sites/immunology , Cell Line, Tumor , Colonic Neoplasms/immunology , Cross Reactions/immunology , Humans , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Molecular Targeted Therapy/methods , Peptides/chemistry , Peptides/immunology , Rats, Wistar , Vascular Endothelial Growth Factor A/immunology , Xenograft Model Antitumor Assays
9.
Vaccine ; 33(44): 5905-12, 2015 Nov 04.
Article in English | MEDLINE | ID: mdl-26187258

ABSTRACT

Vaccines have been used for centuries to protect people and animals against infectious diseases. For vaccine production, it has become evident that cell culture technology can be considered as a key milestone and has been the result of decades of progress. The development and implementation of cell substrates have permitted massive and safe production of viral vaccines. The demand in new vaccines against emerging viral diseases, the increasing vaccine production volumes, and the stringent safety rules for manufacturing have made cell substrates mandatory viral vaccine producer factories. In this review, we focus on cell substrates for the production of vaccines against human viral diseases. Depending on the nature of the vaccine, choice of the cell substrate is critical. Each manufacturer intending to develop a new vaccine candidate should assess several cell substrates during the early development phase in order to select the most convenient for the application. First, as vaccine safety is quite naturally a central concern of Regulatory Agencies, the cell substrate has to answer the regulatory rules stringency. In addition, the cell substrate has to be competitive in terms of viral-specific production yields and manufacturing costs. No cell substrate, even the so-called "designer" cell lines, is able to fulfil all the requested criteria for all viral vaccines. Therefore, the availability of a variety of cell substrates for vaccine production is essential because it improves the chance to successfully respond to the current and future needs of vaccines linked to new emerging or re-emerging infectious diseases (e.g. pandemic flu, Ebola, and Chikungunya outbreaks).


Subject(s)
Technology, Pharmaceutical/methods , Viral Vaccines/isolation & purification , Viral Vaccines/metabolism , Virus Diseases/prevention & control , Cell Culture Techniques/methods , Cell Line , Humans
10.
MAbs ; 6(6): 1415-24, 2014.
Article in English | MEDLINE | ID: mdl-25484047

ABSTRACT

BACKGROUND: Development of functional monoclonal antibodies against intractable GPCR targets. RESULTS: Identification of structured peptides mimicking the ligand binding site, their use in panning to enrich for a population of binders, and the subsequent challenge of this population with receptor overexpressing cells leads to functional monoclonal antibodies. CONCLUSION: The combination of techniques provides a successful strategic approach for the development of functional monoclonal antibodies against CXCR2 in a relatively small campaign. SIGNIFICANCE: The presented combination of techniques might be applicable for other, notoriously difficult, GPCR targets. SUMMARY: The CXC chemokine receptor-2 (CXCR2) is a member of the large 'family A' of G-protein-coupled-receptors and is overexpressed in various types of cancer cells. CXCR2 is activated by binding of a number of ligands, including interleukin 8 (IL-8) and growth-related protein α (Gro-α). Monoclonal antibodies capable of blocking the ligand-receptor interaction are therefore of therapeutic interest; however, the development of biological active antibodies against highly structured GPCR proteins is challenging. Here we present a combination of techniques that improve the discovery of functional monoclonal antibodies against the native CXCR2 receptor. The IL-8 binding site of CXCR2 was identified by screening peptide libraries with the IL-8 ligand, and then reconstructed as soluble synthetic peptides. These peptides were used as antigens to probe an antibody fragment phage display library to obtain subpopulations binding to the IL-8 binding site of CXCR2. Further enrichment of the phage population was achieved by an additional selection round with CXCR2 overexpressing cells as a different antigen source. The scFvs from the CXCR2 specific phage clones were sequenced and converted into monoclonal antibodies. The obtained antibodies bound specifically to CXCR2 expressing cells and inhibited the IL-8 and Gro-α induced ß-arrestin recruitment with IC50 values of 0.3 and 0.2 nM, respectively, and were significantly more potent than the murine monoclonal antibodies (18 and 19 nM, respectively) obtained by the classical hybridoma technique, elicited with the same peptide antigen. According to epitope mapping studies, the antibody efficacy is largely defined by N-terminal epitopes comprising the IL-8 and Gro-α binding sites. The presented strategic combination of in vitro techniques, including the use of different antigen sources, is a powerful alternative for the development of functional monoclonal antibodies by the classical hybridoma technique, and might be applicable to other GPCR targets.


Subject(s)
Antibodies, Monoclonal/immunology , Cell Surface Display Techniques/methods , Epitope Mapping/methods , Receptors, Interleukin-8B/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/metabolism , Antibody Specificity/immunology , Binding Sites/genetics , Binding Sites/immunology , Chemokine CXCL1/immunology , Chemokine CXCL1/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , HEK293 Cells , Humans , Immunization , Interleukin-8/immunology , Interleukin-8/metabolism , Mice, Inbred BALB C , Molecular Sequence Data , Peptide Library , Protein Binding/immunology , Receptors, Interleukin-8B/genetics , Receptors, Interleukin-8B/metabolism
11.
Proc Natl Acad Sci U S A ; 109(14): E812-20, 2012 Apr 03.
Article in English | MEDLINE | ID: mdl-22411803

ABSTRACT

Wnt binding to members of the seven-span transmembrane Frizzled (Fz) receptor family controls essential cell fate decisions and tissue polarity during development and in adulthood. The Fz-mediated membrane recruitment of the cytoplasmic effector Dishevelled (Dvl) is a critical step in Wnt/ß-catenin signaling initiation, but how Fz and Dvl act together to drive downstream signaling events remains largely undefined. Here, we use an Fz peptide-based microarray to uncover a mechanistically important role of the bipartite Dvl DEP domain and C terminal region (DEP-C) in binding a three-segmented discontinuous motif in Fz. We show that cooperative use of two conserved motifs in the third intracellular loop and the classic C-terminal motif of Fz is required for DEP-C binding and Wnt-induced ß-catenin activation in cultured cells and Xenopus embryos. Within the complex, the Dvl DEP domain mainly binds the Fz C-terminal tail, whereas a short region at the Dvl C-terminal end is required to bind the Fz third loop and stabilize the Fz-Dvl interaction. We conclude that Dvl DEP-C binding to Fz is a key event in Wnt-mediated signaling relay to ß-catenin. The discontinuous nature of the Fz-Dvl interface may allow for precise regulation of the interaction in the control of Wnt-dependent cellular responses.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Frizzled Receptors/metabolism , Phosphoproteins/metabolism , Signal Transduction , Wnt Proteins/metabolism , beta Catenin/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Amino Acid Sequence , Cell Line , Dishevelled Proteins , Fluorescence Polarization , Frizzled Receptors/chemistry , Humans , Microscopy, Confocal , Molecular Sequence Data , Phosphoproteins/chemistry , Protein Binding , Xenopus Proteins
12.
EMBO J ; 28(22): 3534-48, 2009 Nov 18.
Article in English | MEDLINE | ID: mdl-19779455

ABSTRACT

Heat shock and other environmental stresses rapidly induce transcriptional responses subject to regulation by a variety of post-translational modifications. Among these, poly(ADP-ribosyl)ation and sumoylation have received growing attention. Here we show that the SUMO E3 ligase PIASy interacts with the poly(ADP-ribose) polymerase PARP-1, and that PIASy mediates heat shock-induced poly-sumoylation of PARP-1. Furthermore, PIASy, and hence sumoylation, appears indispensable for full activation of the inducible HSP70.1 gene. Chromatin immunoprecipitation experiments show that PIASy, SUMO and the SUMO-conjugating enzyme Ubc9 are rapidly recruited to the HSP70.1 promoter upon heat shock, and that they are subsequently released with kinetics similar to PARP-1. Finally, we provide evidence that the SUMO-targeted ubiquitin ligase RNF4 mediates heat-shock-inducible ubiquitination of PARP-1, regulates the stability of PARP-1, and, like PIASy, is a positive regulator of HSP70.1 gene activity. These results, thus, point to a novel mechanism for regulating PARP-1 transcription function, and suggest crosstalk between sumoylation and RNF4-mediated ubiquitination in regulating gene expression in response to heat shock.


Subject(s)
Heat-Shock Response/physiology , Poly(ADP-ribose) Polymerases/metabolism , Poly(ADP-ribose) Polymerases/physiology , SUMO-1 Protein/metabolism , Transcriptional Activation , Animals , Cells, Cultured , Gene Knockdown Techniques , HeLa Cells , Humans , Jurkat Cells , Mice , Models, Biological , Poly (ADP-Ribose) Polymerase-1 , Poly-ADP-Ribose Binding Proteins , Protein Inhibitors of Activated STAT/metabolism , Protein Inhibitors of Activated STAT/physiology , Protein Processing, Post-Translational/physiology , Spodoptera , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/physiology
13.
J Biochem ; 144(1): 39-49, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18344540

ABSTRACT

The modification of proteins by SUMO (small ubiquitin-like modifier) regulates various cellular processes. Sumoylation often occurs on a specific lysine residue within the consensus motif psiKxE/D. However, little is known about the specificity and selectivity of SUMO target sites. We describe here a SUMO assay with peptide array on solid support for the simultaneous characterization of hundreds of different SUMO target sites. This approach was used to characterize known SUMO substrates. The position of the motif within the peptide and the amino acids flanking the acceptor site affected the efficiency of SUMO modification. Interestingly, a sequence of only four amino acids, corresponding to the SUMO consensus motif without flanking amino acids, was a bona fide target site. Analysis of a peptide library for all variants of the psiKxE/D consensus motif revealed that the first and third positions in the tetrapeptide preferably contain aromatic amino acid residues. Furthermore, by adding the SUMO E3 ligase PIAS1 to the reaction mixture, we show specific enhancement of the modification of a PIAS1-dependent SUMO substrate in this system. Overall, our results demonstrate that the sumoylation assay with peptide array on solid support can be used for the high-throughput characterization of SUMO target sites, and provide new insights into the composition, selectivity and specificity of SUMO target sites.


Subject(s)
Peptides/chemistry , Protein Array Analysis/methods , Protein Processing, Post-Translational , Small Ubiquitin-Related Modifier Proteins/metabolism , Amino Acid Motifs , Amino Acid Sequence , Consensus Sequence , GTPase-Activating Proteins/chemistry , GTPase-Activating Proteins/metabolism , HeLa Cells , Humans , Molecular Sequence Data , Peptide Library , Peptides/chemical synthesis , Small Ubiquitin-Related Modifier Proteins/analysis , Substrate Specificity
14.
Mol Cell Biol ; 28(8): 2771-81, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18285457

ABSTRACT

The members of the protein inhibitor of activated STAT (PIAS) family of proteins are implicated in fundamental cellular processes, including transcriptional regulation, either through action as E3 SUMO ligases or through SUMO-independent effects. We report here the identification of FIP200 (focal adhesion kinase family-interacting protein of 200 kDa) as a new PIASy-interacting protein. We show that the interaction depends on the integrity of the RING finger of PIASy and the carboxy terminus of FIP200. Both in vitro and in vivo sumoylation assays failed to reveal any sumoylation of FIP200, suggesting that FIP200 is not a bona fide SUMO substrate. Immunofluorescence microscopy and subcellular fractionation, either upon forced PIASy expression or in the absence of PIASy, revealed that interaction with PIASy redistributes FIP200 from the cytoplasm to the nucleus, correlating with abrogation of FIP200 regulation of TSC/S6K signaling. Conversely, FIP200 enhances the transcriptional activation of the p21 promoter by PIASy whereas PIASy transcription activity is severely reduced upon FIP200 depletion by RNA interference. Chromatin immunoprecipitation analysis demonstrates that endogenous PIASy and FIP200 are corecruited to the p21 promoter. Altogether, these results provide the first evidence for the existence of a close-spatially controlled-mode of regulation of FIP200 and PIASy nucleocytoplasmic functions.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Protein Inhibitors of Activated STAT/metabolism , Protein-Tyrosine Kinases/metabolism , Signal Transduction , Transcription, Genetic/genetics , Animals , Autophagy-Related Proteins , Calcium-Binding Proteins/metabolism , Cells, Cultured , Gene Expression Regulation , Humans , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Mice , Poly-ADP-Ribose Binding Proteins , Promoter Regions, Genetic/genetics , Protein Binding , Protein Inhibitors of Activated STAT/deficiency , Protein Inhibitors of Activated STAT/genetics , Protein Transport , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism
15.
Mol Cell ; 22(6): 783-794, 2006 06 23.
Article in English | MEDLINE | ID: mdl-16793547

ABSTRACT

Cellular senescence and apoptosis have evolved to restrain unwarranted proliferation of potentially tumorigenic cells. Here we show that overexpression of the E3 SUMO ligase PIASy in normal human fibroblasts recruits the p53 and Rb tumor suppressor pathways to provoke a senescence arrest. By contrast, in Rb-deficient fibroblasts, expression of PIASy leads to p53-dependent apoptosis. Induction of senescence requires PIASy E3 activity and is specific for this member of the PIAS ligase family. PIASy stimulates sumoylation and transcriptional activity of p53 and increases Rb-dependent corepression through recruitment to E2F-responsive promoters. Viral oncoprotein E6 suppresses both PIASy-induced senescence and sumoylation of PIASy substrates. Finally, we show that fibroblasts lacking PIASy exhibit a highly reduced propensity to undergo senescence in response to a prosenescence stimulus. Altogether, these data provide the first evidence for a direct role of an E3 SUMO ligase, and by implication of the SUMO pathway, in cellular senescence and apoptosis.


Subject(s)
Apoptosis/physiology , Cellular Senescence/physiology , Fibroblasts/physiology , Protein Inhibitors of Activated STAT/metabolism , Retinoblastoma Protein/metabolism , Cell Transformation, Viral/genetics , E2F Transcription Factors/genetics , E2F Transcription Factors/metabolism , Fibroblasts/cytology , Gene Expression/genetics , Humans , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/metabolism , Poly-ADP-Ribose Binding Proteins , Protein Inhibitors of Activated STAT/deficiency , Protein Processing, Post-Translational , Response Elements/genetics , Retinoblastoma Protein/genetics , SUMO-1 Protein/genetics , SUMO-1 Protein/metabolism , Transfection , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
16.
J Biol Chem ; 278(29): 26612-9, 2003 Jul 18.
Article in English | MEDLINE | ID: mdl-12736252

ABSTRACT

We report that the product of the inducible gene encoding the kinase known as IKKi/IKKepsilon (IKKi) is required for expression of a group of genes up-regulated by pro-inflammatory stimuli such as bacterial endotoxin (lipopolysaccharide (LPS)). Here, using murine embryonic fibroblasts obtained from mice bearing deletions in IKK2, p65, and IKKi genes, we provide evidence to support a link between signaling through the NF-kappaB and CCAAA/enhancer-binding protein (C/EBP) pathways. This link includes an NF-kappaB-dependent regulation of C/EBPbeta and C/EBPdelta gene transcription and IKKi-mediated activation of C/EBP. Disruption of the NF-kappaB pathway results in the blockade of the inducible up-regulation of C/EBPbeta, C/EBPdelta, and IKKi genes. Cells lacking IKKi are normal in activation of the canonical NF-kappaB pathway but fail to induce C/EBPdelta activity and transcription of C/EBP and C/EBP-NF-kappaB target genes in response to LPS. In addition we show that, in response to LPS or tumor necrosis factor alpha, both beta and delta subunits of C/EBP interact with IKKi promoter, suggesting a feedback mechanism in the regulation of IKKi-dependent cellular processes. These data are among the first to provide insights into the biological function of IKKi.


Subject(s)
Inflammation/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Base Sequence , CCAAT-Enhancer-Binding Proteins/metabolism , Cells, Cultured , Feedback , Humans , I-kappa B Kinase , Inflammation/genetics , Lipopolysaccharides/pharmacology , Mice , Mice, Knockout , NF-kappa B/metabolism , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/genetics , RNA Processing, Post-Transcriptional , RNA, Small Interfering/genetics , Signal Transduction , Tumor Necrosis Factor-alpha/pharmacology
17.
Immunity ; 18(1): 13-26, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12530972

ABSTRACT

The mechanism by which TCR signaling activates NF-kappaB is poorly understood. We demonstrate here that the IKK kinase complex is recruited to the immunological synapse and can be coprecipitated with the TCR after T cell activation. Using ZAP-70-deficient T cells expressing a hybrid molecule between the SH2 domain of ZAP-70 and NEMO/IKKgamma, we showed that targeting NEMO to the immunological synapse, and more specifically its 120 N-terminal amino acids, was sufficient to selectively restore NF-kappaB activation in response to TCR ligation. Finally, we demonstrated that targeting of NEMO to the membrane of T cells was sufficient to induce constitutive NF-kappaB activation. This study shows that the localization of NEMO to the immunological synapse is important for TCR-induced NF-kappaB activation and offers a powerful system to dissect the NF-kappaB cascade in T cells.


Subject(s)
NF-kappa B/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Antigen, T-Cell/metabolism , Animals , Antibodies, Monoclonal/pharmacology , CD28 Antigens/metabolism , CD3 Complex/metabolism , Humans , I-kappa B Kinase , Jurkat Cells , Lymphocyte Activation , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Deletion , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , ZAP-70 Protein-Tyrosine Kinase , src Homology Domains
18.
Expert Opin Biol Ther ; 2(6): 621-32, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12171506

ABSTRACT

Altered gene expression contributes to the aetiology of inflammatory disease by modulation of the concentration of disease-related proteins. The expression of inflammatory genes is controlled through the concerted actions of specific transcription factors. Signal transduction networks positively or negatively regulate the activity of these transcription factors. Key components of these networks are protein kinases, which phosphorylate substrates on tyrosine, threonine or serine residues. During the disease process, pro-inflammatory signalling at the cell surface leads to a cascade of kinase activation, which ultimately culminates in modulation of the activity of transcription factors. Thus, pharmacological inhibition of protein kinases is a potential therapeutic strategy to treat inflammation. There are approximately 500 protein kinases in the human genome. Targeted small molecule inhibitors of these kinases should allow for tissue- and disease-specific therapies of unprecedented selectivity. Heralding this new era in molecular medicine is imatinib (Gleevec, Norvartis) a recently marketed tyrosine kinase inhibitor. This review focuses on kinase inhibitors that are currently in development for inflammatory diseases and the transcription factors that are involved.


Subject(s)
Enzyme Inhibitors/pharmacology , Inflammation/therapy , Phosphotransferases/antagonists & inhibitors , Transcription, Genetic , Animals , Drug Design , Gene Expression Regulation , Humans , Models, Biological , Models, Chemical , Phosphorylation , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...