Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
NPJ Vaccines ; 9(1): 97, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38821988

ABSTRACT

Current Influenza virus vaccines primarily induce antibody responses against variable epitopes in hemagglutinin (HA), necessitating frequent updates. However, antibodies against neuraminidase (NA) can also confer protection against influenza, making NA an attractive target for the development of novel vaccines. In this study, we aimed to enhance the immunogenicity of recombinant NA antigens by presenting them multivalently on a nanoparticle carrier. Soluble tetrameric NA antigens of the N1 and N2 subtypes, confirmed to be correctly folded by cryo-electron microscopy structural analysis, were conjugated to Mi3 self-assembling protein nanoparticles using the SpyTag-SpyCatcher system. Immunization of mice with NA-Mi3 nanoparticles induced higher titers of NA-binding and -inhibiting antibodies and improved protection against a lethal challenge compared to unconjugated NA. Additionally, we explored the co-presentation of N1 and N2 antigens on the same Mi3 particles to create a mosaic vaccine candidate. These mosaic nanoparticles elicited antibody titers that were similar or superior to the homotypic nanoparticles and effectively protected against H1N1 and H3N2 challenge viruses. The NA-Mi3 nanoparticles represent a promising vaccine candidate that could complement HA-directed approaches for enhanced potency and broadened protection against influenza A virus.

2.
Virol J ; 20(1): 271, 2023 Nov 21.
Article in English | MEDLINE | ID: mdl-37990263

ABSTRACT

BACKGROUND: Generally influenza, a contagious respiratory disease, leads to mild illness, but can present as a severe illness with significant complications for some. It entails significant health challenges and an economic burden. Annual vaccination is considered the most effective preventive measure against influenza, especially in high-risk groups. METHOD: Epidemiological, demographic and vaccination data of influenza from 2009-to-2019 is collected from Sciensano, the Belgian Institute for Health. Sciensano monitors influenza virus through two surveillances: the Influenza-Like Illness (ILI) surveillance in primary care and the Severe Acute Respiratory Infections (SARI) surveillance in hospital settings. RESULTS: 49.6% [± 8.5] of all ILI-samples tested positive in this period. Influenza A was the dominant circulating type, accounting for 73.7% [± 27.5] of positive samples, while influenza B accounted for 24.3% [± 26.7]. For SARI-surveillance, the average rate of samples tested positive was 36.3% [± 9.3]. Influenza A was responsible for respectively 77.7% [± 23.8] of positive samples and influenza B for 22.2% [± 23.7]. Since 2010, epidemics typically lasted about 9.3 weeks [± 2.7]. From 2012 to 2019 the average vaccine effectiveness was 34.9% [± 15.3]. CONCLUSION: Influenza is quickly considered a trivial disease, but can have substantial repercussions. It remains difficult to identify the level of treat of influenza due to antigenic evolution. Measures to prevent, control and treat are needed. Vaccines that provide broader and more durable protection that can be produced more rapidly could be a potential solution.


Subject(s)
Influenza Vaccines , Influenza, Human , Humans , Influenza, Human/epidemiology , Influenza, Human/prevention & control , Belgium/epidemiology , Seasons , Influenza B virus
3.
mBio ; 10(5)2019 10 22.
Article in English | MEDLINE | ID: mdl-31641082

ABSTRACT

Influenza virus neuraminidase (NA) has been under intense study recently as a vaccine antigen, yet there remain unanswered questions regarding the immune response directed toward NA. Antibodies (Abs) that can inhibit NA activity have been shown to aid in the control of disease caused by influenza virus infection in humans and animal models, yet how and if interactions between the Fc portion of anti-NA Abs and Fcγ receptors (FcγR) contribute to protection has not yet been extensively studied. Herein, we show that poly- and monoclonal anti-NA IgG antibodies with NA inhibitory activity can control A(H1N1)pdm09 infection in the absence of FcγRs, but FcγR interaction aided in viral clearance from the lungs. In contrast, a mouse-human chimeric anti-NA IgG1 that was incapable of mediating NA inhibition (NI) solely relied on FcγR interaction to protect transgenic mice (with a humanized FcγR compartment) against A(H1N1)pdm09 infection. As such, this study suggests that NA-specific antibodies contribute to protection against influenza A virus infection even in the absence of NI activity and supports protection through multiple effector mechanisms.IMPORTANCE There is a pressing need for next-generation influenza vaccine strategies that are better able to manage antigenic drift and the cocirculation of multiple drift variants and that consistently improve vaccine effectiveness. Influenza virus NA is a key target antigen as a component of a next-generation vaccine in the influenza field, with evidence for a role in protective immunity in humans. However, mechanisms of protection provided by antibodies directed to NA remain largely unexplored. Herein, we show that antibody Fc interaction with Fcγ receptors (FcγRs) expressed on effector cells contributes to viral control in a murine model of influenza. Importantly, a chimeric mouse-human IgG1 with no direct antiviral activity was demonstrated to solely rely on FcγRs to protect mice from disease. Therefore, antibodies without NA enzymatic inhibitory activity may also play a role in controlling influenza viruses and should be of consideration when designing NA-based vaccines and assessing immunogenicity.


Subject(s)
Antibodies, Viral/pharmacology , Antibodies, Viral/therapeutic use , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Influenza A Virus, H1N1 Subtype/drug effects , Neuraminidase/immunology , Orthomyxoviridae Infections/drug therapy , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antibodies, Viral/immunology , Antiviral Agents/immunology , Female , Immunoglobulin Fc Fragments/immunology , Male , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/immunology
4.
NPJ Vaccines ; 3: 55, 2018.
Article in English | MEDLINE | ID: mdl-30510776

ABSTRACT

Split inactivated influenza vaccines remain one of the primary preventative strategies against severe influenza disease in the population. However, current vaccines are only effective against a limited number of matched strains. The need for broadly protective vaccines is acute due to the high mutational rate of influenza viruses and multiple strain variants in circulation at any one time. The neuraminidase (NA) glycoprotein expressed on the influenza virion surface has recently regained recognition as a valuable vaccine candidate. We sought to broaden the protection provided by NA within the N1 subtype by computationally engineering consensus NA sequences. Three NA antigens (NA5200, NA7900, NA9100) were designed based on sequence clusters encompassing three major groupings of NA sequence space; (i) H1N1 2009 pandemic and Swine H1N1, (ii) historical seasonal H1N1 and (iii) H1N1 viruses ranging from 1933 till current times. Recombinant NA proteins were produced as a vaccine and used in a mouse challenge model. The design of the protein dictated the protection provided against the challenge strains. NA5200 protected against H1N1 pdm09, a Swine isolate from 1998 and NIBRG-14 (H5N1). NA7900 protected against all seasonal H1N1 viruses tested, and NA9100 showed the broadest range of protection covering all N1 viruses tested. By passive transfer studies and serological assays, the protection provided by the cluster-based consensus (CBC) designs correlated to antibodies capable of mediating NA inhibition. Importantly, sera raised to the consensus NAs displayed a broader pattern of reactivity and protection than naturally occurring NAs, potentially supporting a predictive approach to antigen design.

5.
Mucosal Immunol ; 11(1): 273-289, 2018 01.
Article in English | MEDLINE | ID: mdl-28295019

ABSTRACT

Matrix protein 2 ectodomain (M2e) is considered an attractive component of a broadly protective, universal influenza A vaccine. Here we challenge the canonical view that antibodies against M2e are the prime effectors of protection. Intranasal immunizations of Balb/c mice with CTA1-3M2e-DD-generated M2e-specific memory CD4 T cells that were I-Ad restricted and critically protected against infection, even in the complete absence of antibodies, as observed in JhD mice. Whereas some M2e-tetramer-specific memory CD4 T cells resided in spleen and lymph nodes, the majority were lung-resident Th17 cells, that rapidly expanded upon a viral challenge infection. Indeed, immunized IL-17A-/- mice were significantly less well protected compared with wild-type mice despite exhibiting comparable antibody levels. Similarly, poor protection was also observed in congenic Balb/B (H-2b) mice, which failed to develop M2e-specific CD4 T cells, but exhibited comparable antibody levels. Lung-resident CD69+ CD103low M2e-specific memory CD4 T cells were αß TCR+ and 50% were Th17 cells that were associated with an early influx of neutrophils after virus challenge. Adoptively transferred M2e memory CD4 T cells were strong helper T cells, which accelerated M2e- but more importantly also hemagglutinin-specific IgG production. Thus, for the first time we demonstrate that M2e-specific memory CD4 T cells are broadly protective.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Influenza A virus/immunology , Orthomyxoviridae Infections/immunology , Viral Matrix Proteins/immunology , Animals , Antibodies, Viral/blood , Histocompatibility Antigens Class II/metabolism , Hybridomas , Immunologic Memory , Interleukin-17/genetics , Interleukin-17/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Protein Binding , Protein Domains/genetics , Vaccination , Viral Matrix Proteins/genetics
6.
J Virol ; 92(4)2018 02 15.
Article in English | MEDLINE | ID: mdl-29167342

ABSTRACT

There is increasing evidence to suggest that antibodies directed toward influenza A virus (IAV) neuraminidase (NA) are an important correlate of protection against influenza in humans. Moreover, the potential of NA-specific antibodies to provide broader protection than conventional hemagglutinin (HA) antibodies has been recognized. Here, we describe the isolation of two monoclonal antibodies, N1-7D3 and N1-C4, directed toward the N1 NA. N1-7D3 binds to a conserved linear epitope in the membrane-distal, carboxy-terminal part of the NA and reacted with the NA of seasonal H1N1 isolates ranging from 1977 to 2007 and the 2009 H1N1pdm virus, as well as A/Vietnam/1194/04 (H5N1). However, N1-7D3 lacked NA inhibition (NI) activity and the ability to protect BALB/c mice against a lethal challenge with a range of H1N1 viruses. Conversely, N1-C4 bound to a conformational epitope that is conserved between two influenza virus subtypes, 2009 H1N1pdm and H5N1 IAV, and displayed potent in vitro antiviral activity mediating both NI and plaque size reduction. Moreover, N1-C4 could provide heterosubtypic protection in BALB/c mice against a lethal challenge with 2009 H1N1pdm or H5N1 virus. Glutamic acid residue 311 in the NA was found to be critical for the NA binding and antiviral activity of monoclonal antibody N1-C4. Our data provide further evidence for cross-protective epitopes within the N1 subtype and highlight the potential of NA as an important target for vaccine and therapeutic approaches.IMPORTANCE Influenza remains a worldwide burden on public health. As such, the development of novel vaccines and therapeutics against influenza virus is crucial. Human challenge studies have recently highlighted the importance of antibodies directed toward the viral neuraminidase (NA) as an important correlate of reduced influenza-associated disease severity. Furthermore, there is evidence that anti-NA antibodies can provide broader protection than antibodies toward the viral hemagglutinin. Here, we describe the isolation and detailed characterization of two N1 NA-specific monoclonal antibodies. One of these monoclonal antibodies broadly binds N1-type NAs, and the second displays NA inhibition and in vitro and in vivo antiviral activity against 2009 H1N1pdm and H5N1 influenza viruses. These two new anti-NA antibodies contribute to our understanding of the antigenic properties and protective potential of the influenza virus NA antigen.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies, Viral/therapeutic use , Neuraminidase/immunology , Orthomyxoviridae Infections/prevention & control , Viral Proteins/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Cross Protection , Disease Models, Animal , Female , Immunization, Passive , Influenza A Virus, H1N1 Subtype , Influenza A Virus, H5N1 Subtype , Mice , Mice, Inbred BALB C
7.
Sci Rep ; 7(1): 5986, 2017 07 20.
Article in English | MEDLINE | ID: mdl-28729715

ABSTRACT

Due to their capacity to skew T cell responses towards Th1 oriented immunity, oligonucleotides containing unmethylated CpG motifs (CpG) have emerged as interesting adjuvants for vaccination. Whereas the signalling pathways in response to CpG mediated TLR9 activation have been extensively documented at the level of the individual cell, little is however known on the precise identity of the innate immune cells that govern T cell priming and polarisation to CpG adjuvanted protein antigens in vivo. In this study, we demonstrate that optimal induction of Th1 oriented immunity to CpG adjuvanted protein vaccines requires the coordinated actions of conventional DCs and of monocytes. Whilst conventional DCs were required for antigen presentation and initial T cell priming, monocytes constitute the main source of the Th1 polarising cytokine IL-12.


Subject(s)
Adjuvants, Immunologic/pharmacology , Immunity, Cellular , Inflammation/pathology , Interleukin-12/biosynthesis , Monocytes/pathology , Oligodeoxyribonucleotides/pharmacology , Th1 Cells/immunology , Vaccines/immunology , Animals , Antigen Presentation/drug effects , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/metabolism , Antigens/metabolism , Antigens, Ly/metabolism , Cell Movement/drug effects , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Immunity, Cellular/drug effects , Lymph Nodes/drug effects , Lymph Nodes/metabolism , Lymph Nodes/pathology , Membrane Proteins/metabolism , Mice, Inbred C57BL , Monocytes/drug effects , Monocytes/metabolism , Phenotype , Receptors, CCR2/metabolism , Vaccination
8.
Chem Commun (Camb) ; 52(16): 3352-5, 2016 Feb 25.
Article in English | MEDLINE | ID: mdl-26823186

ABSTRACT

We report on a straightforward strategy to fabricate bioactive glycosylated gold nanoparticles via a combination of RAFT polymerization, carbohydrate ligation through reductive amination and thiol-gold self-assembly. This approach is used for the design of gold nanoparticles decorated with the complex sialylated glycan Neu5Ac-α-2-6-Gal, and we demonstrate multivalent and specific recognition between the nanoparticles, lectins and hemagglutinin on the surface of the influenza virus.


Subject(s)
Gold/chemistry , Metal Nanoparticles/chemistry , Polymers/chemistry , Amination , Oxidation-Reduction , Polymerization
9.
J Microsc ; 259(2): 80-96, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25623622

ABSTRACT

When electron microscopy (EM) was introduced in the 1930s it gave scientists their first look into the nanoworld of cells. Over the last 80 years EM has vastly increased our understanding of the complex cellular structures that underlie the diverse functions that cells need to maintain life. One drawback that has been difficult to overcome was the inherent lack of volume information, mainly due to the limit on the thickness of sections that could be viewed in a transmission electron microscope (TEM). For many years scientists struggled to achieve three-dimensional (3D) EM using serial section reconstructions, TEM tomography, and scanning EM (SEM) techniques such as freeze-fracture. Although each technique yielded some special information, they required a significant amount of time and specialist expertise to obtain even a very small 3D EM dataset. Almost 20 years ago scientists began to exploit SEMs to image blocks of embedded tissues and perform serial sectioning of these tissues inside the SEM chamber. Using first focused ion beams (FIB) and subsequently robotic ultramicrotomes (serial block-face, SBF-SEM) microscopists were able to collect large volumes of 3D EM information at resolutions that could address many important biological questions, and do so in an efficient manner. We present here some examples of 3D EM taken from the many diverse specimens that have been imaged in our core facility. We propose that the next major step forward will be to efficiently correlate functional information obtained using light microscopy (LM) with 3D EM datasets to more completely investigate the important links between cell structures and their functions.


Subject(s)
Histocytological Preparation Techniques/methods , Imaging, Three-Dimensional/methods , Microscopy, Electron, Scanning/methods , Animals , Brain/ultrastructure , Electron Microscope Tomography/methods , Lung/cytology , Lung/ultrastructure , Mice , Microscopy, Electron , Microscopy, Electron, Scanning/instrumentation , Microtomy , Plant Roots/ultrastructure
10.
Cell Death Dis ; 4: e742, 2013 Jul 25.
Article in English | MEDLINE | ID: mdl-23887633

ABSTRACT

ABT-263 and its structural analogues ABT-199 and ABT-737 inhibit B-cell lymphoma 2 (Bcl-2), BCL2L1 long isoform (Bcl-xL) and BCL2L2 (Bcl-w) proteins and promote cancer cell death. Here, we show that at non-cytotoxic concentrations, these small molecules accelerate the deaths of non-cancerous cells infected with influenza A virus (IAV) or other viruses. In particular, we demonstrate that ABT-263 altered Bcl-xL interactions with Bcl-2 antagonist of cell death (Bad), Bcl-2-associated X protein (Bax), uveal autoantigen with coiled-coil domains and ankyrin repeats protein (UACA). ABT-263 thereby activated the caspase-9-mediated mitochondria-initiated apoptosis pathway, which, together with the IAV-initiated caspase-8-mediated apoptosis pathway, triggered the deaths of IAV-infected cells. Our results also indicate that Bcl-xL, Bcl-2 and Bcl-w interact with pattern recognition receptors (PRRs) that sense virus constituents to regulate cellular apoptosis. Importantly, premature killing of IAV-infected cells by ABT-263 attenuated the production of key pro-inflammatory and antiviral cytokines. The imbalance in cytokine production was also observed in ABT-263-treated IAV-infected mice, which resulted in an inability of the immune system to clear the virus and eventually lowered the survival rates of infected animals. Thus, the results suggest that the chemical inhibition of Bcl-xL, Bcl-2 and Bcl-w could potentially be hazardous for cancer patients with viral infections.


Subject(s)
Aniline Compounds/pharmacology , Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Sulfonamides/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cytokines/biosynthesis , Disease Models, Animal , Influenza A virus/physiology , Macrophages/metabolism , Mice , Neoplasms/pathology , Neoplasms/virology , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/pathology
11.
Mucosal Immunol ; 6(2): 276-87, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22806098

ABSTRACT

Influenza is a global health concern. Licensed influenza vaccines induce strain-specific virus-neutralizing antibodies but hamper the induction of possibly cross-protective T-cell responses upon subsequent infection.(1) In this study, we compared protection induced by a vaccine based on the conserved extracellular domain of matrix 2 protein (M2e) with that of a conventional whole inactivated virus (WIV) vaccine using single as well as consecutive homo- and heterosubtypic challenges. Both vaccines protected against a primary homologous (with respect to hemagglutinin and neuraminidase in WIV) challenge. Functional T-cell responses were induced after primary challenge of M2e-immune mice but were absent in WIV-vaccinated mice. M2e-immune mice displayed limited inducible bronchus-associated lymphoid tissue, which was absent in WIV-immune animals. Importantly, M2e- but not WIV-immune mice were protected from a primary as well as a secondary, severe heterosubtypic challenge, including challenge with pandemic H1N1 2009 virus. Our findings advocate the use of infection-permissive influenza vaccines, such as those based on M2e, in immunologically naive individuals. The combined immune response induced by M2e-vaccine and by clinically controlled influenza virus replication results in strong and broad protection against pandemic influenza. We conclude that the challenge of the M2e-immune host induces strong and broadly reactive immunity against influenza virus infection.


Subject(s)
Immunity, Cellular , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , Viral Matrix Proteins/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Cross Reactions/immunology , Disease Models, Animal , Female , Host-Pathogen Interactions/immunology , Lung/immunology , Lung/virology , Mice , Orthomyxoviridae Infections/virology
12.
Anim Genet ; 41(2): 169-78, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19917049

ABSTRACT

Shadow of prion protein (SPRN) is an interesting candidate gene thought to be involved in prion pathogenesis. In humans, an association has already been discovered between mutations in SPRN and the incidence of variant and sporadic Creutzfeldt-Jakob disease. However, in sheep, the effect of mutations in SPRN is largely unknown. Therefore, we analysed the presence of mutations in the entire ovine SPRN gene, their association with scrapie susceptibility and their effect on SPRN promoter activity. In total, 26 mutations were found: seven in the promoter region, four in intron 1, seven in the coding sequence and eight in the 3' untranslated region. The mutations detected in the coding sequence and the promoter region were subsequently analysed in more detail. In the coding sequence, a polymorphism causing a deletion of two alanines was found to be associated with susceptibility for classical scrapie in sheep. Furthermore, a functional analysis of deletion constructs of the ovine SPRN promoter revealed that the region 464 to 230 bp upstream of exon 1 (containing a putative AP-2 and putative Sp1 binding sites) is of functional importance for SPRN transcription. Six mutations in the SPRN promoter were also found to alter the promoter activity in vitro. However, no association between any of these promoter mutations and susceptibility for classical scrapie was found.


Subject(s)
Genetic Predisposition to Disease , Nerve Tissue Proteins/genetics , Scrapie/genetics , Sheep/genetics , Animals , Base Sequence , Molecular Sequence Data , Mutation , Polymorphism, Genetic , Promoter Regions, Genetic
13.
Vaccine ; 27(8): 1279-86, 2009 Feb 18.
Article in English | MEDLINE | ID: mdl-19114075

ABSTRACT

Mucosal vaccination has several advantages over parenteral vaccination. In this study, viscosity-enhancing mucosal delivery systems for the induction of an adaptive immune response against viral antigen were investigated. Powder formulations based on spray-dried mixtures of starch (Amioca)/poly(acrylic acid) (Carbopol 974P) in different ratios were used as carriers of the viral antigen. A comparison of these formulations for intranasal delivery of heat-inactivated influenza virus combined with LTR192G adjuvant was made in vivo in a rabbit model. Individual rabbit sera were tested for seroconversion against hemagglutinin (HA), the major surface antigen of influenza. The powder vaccine formulations were able to induce systemic anti-HA IgG responses. The presence of Carbopol 974P improved the kinetics of the immune responses and the level of IgG titers in a dose-dependent way which was correlated with moderately irritating capacities of the formulation. In contrast, mucosal IgA responses were not detected. In conclusion, it was demonstrated that the use of bioadhesive carriers based on Amioca starch and poly(acrylic acid) facilitates the induction of a systemic anti-HA antibody response after intranasal vaccination with a whole virus influenza vaccine.


Subject(s)
Acrylic Resins/administration & dosage , Influenza Vaccines/administration & dosage , Powders/administration & dosage , Powders/chemistry , Starch/administration & dosage , Animals , Antibodies, Viral/blood , Immunity, Mucosal , Immunoglobulin A/analysis , Immunoglobulin G/blood , Influenza Vaccines/immunology , Mucous Membrane/chemistry , Rabbits , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/immunology
14.
Cell Death Differ ; 14(5): 1050-9, 2007 May.
Article in English | MEDLINE | ID: mdl-17318221

ABSTRACT

We have studied the involvement of receptor interacting protein kinase-1 (RIP1) and dsRNA-activated protein kinase (PKR) in external dsRNA-induced apoptotic and necrotic cell death in Jurkat T cell lymphoma. Our results suggest that RIP1 plays an imported role in dsRNA-induced apoptosis and necrosis. We demonstrated that contrary to necrosis, protein synthesis is inhibited in apoptosis. Here, we show that phosphorylation of translation initiation factor 2-alpha (eukaryotic initiation factor 2-alpha (eIF2-alpha)) and its kinase, PKR, occur in dsRNA-induced apoptosis but not in necrosis. These events are caspase-dependent and coincide with the appearance of the caspase-mediated PKR fragments, N-terminal domain (ND) and kinase domain (KD). Our immunoprecipitation experiments demonstrated that both fragments could independently co-precipitate with full-length PKR. Expression of PKR-KD leads to PKR and eIF2-alpha phosphorylation and inhibits protein translation, whereas that of PKR-ND does not. Co-expression of PKR-ND and PKR-KD promotes their interaction with PKR, PKR and eIF2-alpha phosphorylation and suppresses protein translation better than PKR-KD alone. Our findings suggest a caspase-dependent mode of activation of PKR in apoptosis in which the PKR-KD fragment interacts with and activates intact PKR. PKR-ND facilitates the interaction of PKR-KD with full-length PKR and thus the activation of the kinase and amplifies the translation inhibitory signal.


Subject(s)
Caspases/metabolism , Peptide Fragments/metabolism , Protein Biosynthesis , eIF-2 Kinase/metabolism , Apoptosis/drug effects , Caspase Inhibitors , Cell Line , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Humans , Immunoprecipitation , Jurkat Cells , Necrosis , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Biosynthesis/drug effects , Protein Processing, Post-Translational/drug effects , Protein Structure, Tertiary/drug effects , RNA, Double-Stranded/pharmacology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , eIF-2 Kinase/biosynthesis
16.
Cell Death Differ ; 10(5): 570-9, 2003 May.
Article in English | MEDLINE | ID: mdl-12728255

ABSTRACT

Recent studies have implicated the dying cell as a potential reservoir of modified autoantigens that might initiate and drive systemic autoimmunity in susceptible hosts. The spliceosomal Sm proteins are recognized by the so-called anti-Sm autoantibodies, an antibody population found exclusively in patients suffering from systemic lupus erythematosus. We have studied the effects of apoptosis on the Sm proteins and demonstrate that one of the Sm proteins, the Sm-F protein, is proteolytically cleaved in apoptotic cells. Cleavage of the Sm-F protein generates a 9-kDa apoptotic fragment, which remains associated with the U snRNP complexes in apoptotic cells. Sm-F cleavage is dependent on caspase activation and the cleavage site has been located near the C-terminus, EEED(81) downward arrow G. Use of different caspase inhibitors suggests that besides caspase-8 other caspases are implicated in Sm-F cleavage. A C-terminally truncated mutant of the Sm-F protein, representing the modified form of the protein, is capable of forming an Sm E-F-G complex in vitro that is recognized by many anti-Sm patient sera.


Subject(s)
Apoptosis , Caspases/metabolism , Ribonucleoprotein, U1 Small Nuclear/metabolism , Ribonucleoproteins, Small Nuclear/metabolism , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Murine-Derived , Apoptosis/drug effects , Apoptosis/immunology , Autoantibodies/blood , Autoantigens , Blotting, Western , Caspase Inhibitors , Cell Line, Tumor , Cysteine Proteinase Inhibitors/pharmacology , Electrophoresis, Polyacrylamide Gel , HeLa Cells , Humans , Jurkat Cells , Lupus Erythematosus, Systemic/blood , Lupus Erythematosus, Systemic/immunology , Ribonucleoproteins, Small Nuclear/genetics , Ribonucleoproteins, Small Nuclear/immunology , Time Factors , fas Receptor/immunology , snRNP Core Proteins
17.
Cell Death Differ ; 9(11): 1207-11, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12404119

ABSTRACT

Caspases are crucial for the initiation, propagation and execution of apoptosis. They normally exist as proenzymes, which can be activated through recruitment into activating complexes and by proteolytic cleavage by other caspases or proteases. Perturbation of organelles such as nuclei, endoplasmatic reticulum and mitochondria results in the activation of caspases. A number of caspases (-2, -3, -8 and -9) were published as being localized in the intermembrane space of mitochondria. However, in three different models of apoptosis (anti-Fas-induced cell death in murine hepatocytes, Fas ligand-induced apoptosis in Jurkat cells and apoptosis induced by growth factor withdrawal in Ba/F3 cells) we could not identify a mitochondrial location of caspases, neither under control nor under apoptotic conditions. In all three apoptotic models caspases were found in the cytosolic (caspases-2, -3, -6, -7, -8, -9) and nuclear subcellular fractions (caspases-2, -3). In another approach we treated isolated liver mitochondria with truncated Bid. Although tBid-dependent release of Cytochrome c, AIF, adenylate kinase, Smac/DIABLO and Omi/HtrA2 could be demonstrated, none of the caspases were detectable both in the supernatant and the mitochondrial fraction after treatment. Our results demonstrate that, in contrast to previous studies, no caspases-2, -3, -8 and -9 are associated with the mitochondrial fraction. These findings support the concept of a separate compartmentalization between proapoptotic cofactors in the mitochondria and silent precursor caspases in the cytosol.


Subject(s)
Apoptosis/physiology , Caspases/metabolism , Mitochondria/metabolism , Animals , Biomarkers , Caspase 2 , Enzyme Precursors/metabolism , Humans , Jurkat Cells , Mice , Mice, Inbred C57BL
18.
Cell Death Differ ; 9(10): 1031-42, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12232790

ABSTRACT

Mitochondria are 'life-essential' organelles for the production of metabolic energy in the form of ATP. Paradoxically mitochondria also play a key role in controlling the pathways that lead to cell death. This latter role of mitochondria is more than just a 'loss of function' resulting in an energy deficit but is an active process involving different mitochondrial proteins. Cytochrome c was the first characterised mitochondrial factor shown to be released from the mitochondrial intermembrane space and to be actively implicated in apoptotic cell death. Since then, other mitochondrial proteins, such as AIF, Smac/DIABLO, endonuclease G and Omi/HtrA2, were found to undergo release during apoptosis and have been implicated in various aspects of the cell death process. Members of the Bcl-2 protein family control the integrity and response of mitochondria to apoptotic signals. The molecular mechanism by which mitochondrial intermembrane space proteins are released and the regulation of mitochondrial homeostasis by Bcl-2 proteins is still elusive. This review summarises and evaluates the current knowledge concerning the complex role of released mitochondrial proteins in the apoptotic process.


Subject(s)
Apoptosis/physiology , Eukaryotic Cells/metabolism , Intracellular Membranes/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Signal Transduction/physiology , Animals , Humans
19.
Cell Death Differ ; 9(9): 981-94, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12181749

ABSTRACT

Interferons enhance the cellular antiviral response by inducing expression of protective proteins. Many of these proteins are activated by dsRNA, a typical by-product of viral infection. Here we show that type-I and type-II interferons can sensitize cells to dsRNA-induced cytotoxicity. In caspase-8- or FADD-deficient Jurkat cells dsRNA induces necrosis, instead of apoptosis. In L929sA cells dsRNA-induced necrosis involves high reactive oxygen species production. The antioxidant butylated hydroxyanisole protects cells from necrosis, but shifts the response to apoptosis. Treatment with the caspase inhibitor benzyloxycarbonyl-Val-Ala-DL-Asp(OMe)-fluoromethylketone or overexpression of Bcl-2 prevent this shift and promote necrosis. Our results suggest that a single stimulus can initiate different death-signaling pathways, leading to either necrotic or apoptotic cell death. Inhibition of key events in these signaling pathways, such as caspase activation, cytochrome c release or mitochondrial reactive oxygen species production, tips the balance between necrosis and apoptosis, leading to dominance of one of these death programs.


Subject(s)
Adaptor Proteins, Signal Transducing , Apoptosis/drug effects , Interferons/pharmacology , Jurkat Cells/drug effects , Necrosis , RNA Virus Infections/drug therapy , RNA Viruses/drug effects , RNA, Double-Stranded/drug effects , Signal Transduction/drug effects , Animals , Antioxidants/pharmacology , Apoptosis/genetics , Butylated Hydroxyanisole/pharmacology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Caspase 8 , Caspase 9 , Caspase Inhibitors , Caspases/deficiency , Caspases/genetics , Caspases/metabolism , Enzyme Inhibitors/pharmacology , Fas-Associated Death Domain Protein , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Humans , Interferon-beta/pharmacology , Interferon-gamma/pharmacology , Jurkat Cells/metabolism , Jurkat Cells/virology , Mice , Proto-Oncogene Proteins c-bcl-2/genetics , RNA Virus Infections/genetics , RNA Viruses/genetics , RNA, Double-Stranded/genetics , RNA, Double-Stranded/pharmacology , Reactive Oxygen Species/metabolism , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL