Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Biomolecules ; 13(11)2023 10 31.
Article in English | MEDLINE | ID: mdl-38002281

ABSTRACT

We recently identified protein kinase N1 (PKN1) as a negative gatekeeper of neuronal AKT protein kinase activity during postnatal cerebellar development. The developing cerebellum is specifically vulnerable to hypoxia-ischemia (HI), as it occurs during hypoxic-ischemic encephalopathy, a condition typically caused by oxygen deprivation during or shortly after birth. In that context, activation of the AKT cell survival pathway has emerged as a promising new target for neuroprotective interventions. Here, we investigated the role of PKN1 in an in vitro model of HI, using postnatal cerebellar granule cells (Cgc) derived from Pkn1 wildtype and Pkn1-/- mice. Pkn1-/- Cgc showed significantly higher AKT phosphorylation, resulting in reduced caspase-3 activation and improved survival after HI. Pkn1-/- Cgc also showed enhanced axonal outgrowth on growth-inhibitory glial scar substrates, further pointing towards a protective phenotype of Pkn1 knockout after HI. The specific PKN1 phosphorylation site S374 was functionally relevant for the enhanced axonal outgrowth and AKT interaction. Additionally, PKN1pS374 shows a steep decrease during cerebellar development. In summary, we demonstrate the pathological relevance of the PKN1-AKT interaction in an in vitro HI model and establish the relevant PKN1 phosphorylation sites, contributing important information towards the development of specific PKN1 inhibitors.


Subject(s)
Hypoxia-Ischemia, Brain , Neuroprotective Agents , Animals , Mice , Hypoxia-Ischemia, Brain/pathology , Proto-Oncogene Proteins c-akt/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Hypoxia , Cerebellum/metabolism , Animals, Newborn
2.
Am J Hematol ; 95(12): 1562-1571, 2020 12.
Article in English | MEDLINE | ID: mdl-32936982

ABSTRACT

Hyperdiploidy (HRD) and specific immunoglobulin heavy locus (IGH) translocations are primary chromosomal abnormalities (CA) in multiple myeloma (MM). In this retrospective study of 794 MM patients we aimed to investigate clinical features and common CA including gain(1q) in separate subgroups defined by primary CA. In the entire group, we confirmed that gain(1q) was associated with short time to next treatment and adverse overall survival (OS). The impact was worse for four or more copies of 1q21 as compared to three copies. However, in a subgroup of patients with clonal gain(11q) and without known primary IGH translocations (CG11q), already three copies of 1q21 were associated with a poor outcome; in the absence of gain(1q), patients in this subgroup had a remarkably long median OS of more than nine years. These cases were associated with HRD, coexpression of CD56 and CD117, male gender, and IgG subtype. In non-CG11q patients, four or more copies of 1q21 (but not three copies) had a significant adverse impact on outcome. Several associations with CA and clinical findings were observed for the defined subgroups. As an example, we found a predominance of early tetraploidy, plasma cell leukemia, and female gender in the t(14;16) subgroup. Our results underscore the importance of subgrouping in MM.


Subject(s)
Chromosomes, Human, Pair 1/genetics , Genetic Loci , Immunoglobulin Heavy Chains/genetics , Multiple Myeloma/genetics , Multiple Myeloma/mortality , Neoplasm Proteins/genetics , Translocation, Genetic , Adult , Aged , Aged, 80 and over , CD56 Antigen/genetics , Disease-Free Survival , Female , Humans , Immunoglobulin G/genetics , Male , Middle Aged , Multiple Myeloma/therapy , Proto-Oncogene Proteins c-kit/genetics , Survival Rate
3.
Cell Commun Signal ; 17(1): 56, 2019 05 28.
Article in English | MEDLINE | ID: mdl-31138259

ABSTRACT

BACKGROUND: The protein kinase C theta (PKCθ) has an important and non-redundant function downstream of the antigen receptor and co-receptor complex in T lymphocytes. PKCθ is not only essential for activation of NF-κB, AP-1 and NFAT and subsequent interleukin-2 expression, but also critical for positive selection and development of regulatory T lymphocytes in the thymus. Several domains regulate its activity, such as a pseudosubstrate sequence mediating an auto-inhibitory intramolecular interaction, the tandem C1 domains binding diacylglycerol, and phosphorylation at conserved tyrosine, threonine as well as serine residues throughout the whole length of the protein. To address the importance of the variable domain V1 at the very N-terminus, which is encoded by exon 2, a mutated version of PKCθ was analyzed for its ability to stimulate T lymphocyte activation. METHODS: T cell responses were analyzed with promoter luciferase reporter assays in Jurkat T cells transfected with PKCθ expression constructs. A mouse line expressing mutated instead of wild type PKCθ was analyzed in comparison to PKCθ-deficient and wild type mice for thymic development and T cell subsets by flow cytometry and T cell activation by quantitative RT-PCR, luminex analysis and flow cytometry. RESULTS: In cell lines, the exon 2-replacing mutation impaired the transactivation of interleukin-2 expression by constitutively active mutant form of PKCθ. Moreover, analysis of a newly generated exon 2-mutant mouse line (PKCθ-E2mut) revealed that the N-terminal replacement mutation results in an hypomorph mutant of PKCθ combined with reduced PKCθ protein levels in CD4+ T lymphocytes. Thus, PKCθ-dependent functions in T lymphocytes were affected resulting in impaired thymic development of single positive T lymphocytes in vivo. In particular, there was diminished generation of regulatory T lymphocytes. Furthermore, early activation responses such as interleukin-2 expression of CD4+ T lymphocytes were significantly reduced even though cell viability was not affected. Thus, PKCθ-E2mut mice show a phenotype similar to conventional PKCθ-deficient mice. CONCLUSION: Taken together, PKCθ-E2mut mice show a phenotype similar to conventional PKCθ-deficient mice. Both our in vitro T cell culture experiments and ex vivo analyses of a PKCθ-E2-mutant mouse line independently validate the importance of PKCθ downstream of the antigen-receptor complex for activation of CD4+ T lymphocytes.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Lymphocyte Activation , Mutation , Protein Kinase C-theta/genetics , Animals , HEK293 Cells , Humans , Jurkat Cells , Mice , Mice, Inbred C57BL , Phenotype , Protein Kinase C-theta/metabolism
4.
J Clin Invest ; 128(5): 2076-2088, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29494346

ABSTRACT

Increasing evidence suggests that synapse dysfunctions are a major determinant of several neurodevelopmental and neurodegenerative diseases. Here we identify protein kinase N1 (PKN1) as a novel key player in fine-tuning the balance between axonal outgrowth and presynaptic differentiation in the parallel fiber-forming (PF-forming) cerebellar granule cells (Cgcs). Postnatal Pkn1-/- animals showed a defective PF-Purkinje cell (PF-PC) synapse formation. In vitro, Pkn1-/- Cgcs exhibited deregulated axonal outgrowth, elevated AKT phosphorylation, and higher levels of neuronal differentiation-2 (NeuroD2), a transcription factor preventing presynaptic maturation. Concomitantly, Pkn1-/- Cgcs had a reduced density of presynaptic sites. By inhibiting AKT with MK-2206 and siRNA-mediated knockdown, we found that AKT hyperactivation is responsible for the elongated axons, higher NeuroD2 levels, and reduced density of presynaptic specifications in Pkn1-/- Cgcs. In line with our in vitro data, Pkn1-/- mice showed AKT hyperactivation, elevated NeuroD2 levels, and reduced expression of PF-PC synaptic markers during stages of PF maturation in vivo. The long-term effect of Pkn1 knockout was further seen in cerebellar atrophy and mild ataxia. In summary, our results demonstrate that PKN1 functions as a developmentally active gatekeeper of AKT activity, thereby fine-tuning axonal outgrowth and presynaptic differentiation of Cgcs and subsequently the correct PF-PC synapse formation.


Subject(s)
Axons/enzymology , Neuronal Outgrowth , Protein Kinase C/metabolism , Purkinje Cells/enzymology , Synapses/metabolism , Animals , Heterocyclic Compounds, 3-Ring/pharmacology , Mice , Mice, Knockout , Protein Kinase C/genetics , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Purkinje Cells/cytology , Synapses/genetics
5.
J Proteome Res ; 17(3): 1269-1277, 2018 03 02.
Article in English | MEDLINE | ID: mdl-29441788

ABSTRACT

Afamin is an 87 kDa glycoprotein with five predicted N-glycosylation sites. Afamin's glycan abundance contributes to conformational and chemical inhomogeneity presenting great challenges for molecular structure determination. For the purpose of studying the structure of afamin, various forms of recombinantly expressed human afamin (rhAFM) with different glycosylation patterns were thus created. Wild-type rhAFM and various hypoglycosylated forms were expressed in CHO, CHO-Lec1, and HEK293T cells. Fully nonglycosylated rhAFM was obtained by transfection of point-mutated cDNA to delete all N-glycosylation sites of afamin. Wild-type and hypo/nonglycosylated rhAFM were purified from cell culture supernatants by immobilized metal ion affinity and size exclusion chromatography. Glycan analysis of purified proteins demonstrated differences in micro- and macro-heterogeneity of glycosylation enabling the comparison between hypoglycosylated, wild-type rhAFM, and native plasma afamin. Because antibody fragments can work as artificial chaperones by stabilizing the structure of proteins and consequently enhance the chance for successful crystallization, we incubated a Fab fragment of the monoclonal anti-afamin antibody N14 with human afamin and obtained a stoichiometric complex. Subsequent results showed sufficient expression of various partially or nonglycosylated forms of rhAFM in HEK293T and CHO cells and revealed that glycosylation is not necessary for expression and secretion.


Subject(s)
Antibodies, Monoclonal/chemistry , Antigen-Antibody Complex/chemistry , Carrier Proteins/chemistry , Glycoproteins/chemistry , Immunoglobulin Fab Fragments/chemistry , Protein Processing, Post-Translational , Serum Albumin, Human/chemistry , Amino Acid Sequence , Animals , Antibodies, Monoclonal/metabolism , Antigen-Antibody Complex/metabolism , CHO Cells , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cloning, Molecular , Cricetulus , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Glycoproteins/genetics , Glycoproteins/metabolism , Glycosylation , HEK293 Cells , Humans , Immunoglobulin Fab Fragments/metabolism , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/chemistry , Polysaccharides/chemistry , Polysaccharides/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serum Albumin, Human/genetics , Serum Albumin, Human/metabolism
6.
J Biol Chem ; 291(45): 23557-23568, 2016 Nov 04.
Article in English | MEDLINE | ID: mdl-27624939

ABSTRACT

Dysfunctional mitochondria contribute to the development of many diseases and pathological conditions through the excessive production of reactive oxygen species (ROS), and, where studied, ablation of p66Shc (p66) was beneficial. p66 translocates to the mitochondria and oxidizes cytochrome c to yield H2O2, which in turn initiates cell death. PKCß-mediated phosphorylation of serine 36 in p66 has been implicated as a key regulatory step preceding mitochondrial translocation, ROS production, and cell death, and PKCß thus may provide a target for therapeutic intervention. We performed a reassessment of PKCß regulation of the oxidoreductase activity of p66. Although our experiments did not substantiate Ser36 phosphorylation by PKCß, they instead provided evidence for Ser139 and Ser213 as PKCß phosphorylation sites regulating the pro-oxidant and pro-apoptotic function of p66. Mutation of another predicted PKCß phosphorylation site also located in the phosphotyrosine binding domain, threonine 206, had no phenotype. Intriguingly, p66 with Thr206 and Ser213 mutated to glutamic acid showed a gain-of-function phenotype with significantly increased ROS production and cell death induction. Taken together, these data argue for a complex mechanism of PKCß-dependent regulation of p66 activation involving Ser139 and a motif surrounding Ser213.


Subject(s)
Protein Kinase C beta/metabolism , Reactive Oxygen Species/metabolism , Src Homology 2 Domain-Containing, Transforming Protein 1/metabolism , 3T3 Cells , Animals , Cell Death , Gene Deletion , HEK293 Cells , Humans , Hydrogen Peroxide/metabolism , Mice , Mitochondria/genetics , Mitochondria/metabolism , Models, Molecular , Oxidative Stress , Oxidoreductases/genetics , Oxidoreductases/metabolism , Phosphorylation , Point Mutation , Protein Kinase C beta/genetics , RNA Interference , RNA, Small Interfering/genetics , Serine/genetics , Serine/metabolism , Src Homology 2 Domain-Containing, Transforming Protein 1/genetics
7.
Nat Med ; 22(8): 915-23, 2016 08.
Article in English | MEDLINE | ID: mdl-27428901

ABSTRACT

Fungal infections claim an estimated 1.5 million lives each year. Mechanisms that protect from fungal infections are still elusive. Recognition of fungal pathogens relies on C-type lectin receptors (CLRs) and their downstream signaling kinase SYK. Here we report that the E3 ubiquitin ligase CBLB controls proximal CLR signaling in macrophages and dendritic cells. We show that CBLB associates with SYK and ubiquitinates SYK, dectin-1, and dectin-2 after fungal recognition. Functionally, CBLB deficiency results in increased inflammasome activation, enhanced reactive oxygen species production, and increased fungal killing. Genetic deletion of Cblb protects mice from morbidity caused by cutaneous infection and markedly improves survival after a lethal systemic infection with Candida albicans. On the basis of these findings, we engineered a cell-permeable CBLB inhibitory peptide that protects mice from lethal C. albicans infections. We thus describe a key role for Cblb in the regulation of innate antifungal immunity and establish a novel paradigm for the treatment of fungal sepsis.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Candidiasis, Invasive/immunology , Dendritic Cells/immunology , Lectins, C-Type/metabolism , Macrophages/immunology , Peptides/pharmacology , Phagocytosis/immunology , Proto-Oncogene Proteins c-cbl/immunology , Reactive Oxygen Species/immunology , Sepsis/immunology , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Animals , Blotting, Western , Candida albicans , Caspase 8 , Cytokines/immunology , Dendritic Cells/drug effects , Enzyme-Linked Immunosorbent Assay , Immunoprecipitation , Kidney , Lectins, C-Type/drug effects , Macrophages/drug effects , Mice , Mice, Knockout , Monocytes/drug effects , Monocytes/immunology , Neutrophils/drug effects , Neutrophils/immunology , Phagocytosis/drug effects , Phagocytosis/genetics , Polymerase Chain Reaction , Proto-Oncogene Proteins c-cbl/antagonists & inhibitors , Proto-Oncogene Proteins c-cbl/genetics , Ubiquitination
8.
Sci Rep ; 6: 20930, 2016 Feb 12.
Article in English | MEDLINE | ID: mdl-26868434

ABSTRACT

p66Shc-dependent ROS production contributes to many pathologies including ischemia/reperfusion injury (IRI) during solid organ transplantation. Inhibiting p66Shc activation may provide a novel therapeutic approach to prevent damage, which is poorly managed by antioxidants in vivo. Previous work suggested that pro-oxidant and a pro-apoptotic function of p66Shc required mitochondrial import, which depended on serine 36 phosphorylation. PKCß has been proposed as S36 kinase but cJun N-terminal kinases (JNKs) may also phosphorylate this residue. To simulate the early stages of ischemia/reperfusion (IR) we either used H2O2 treatment or hypoxia/reoxygenation (HR). As during reperfusion in vivo, we observed increased JNK and p38 activity in mouse embryonic fibroblasts (MEFs) and HL-1 cardiomyocytes along with significantly increased p66ShcS36 phosphorylation, ROS production and cell damage. Application of specific inhibitors caused a pronounced decrease in p66ShcS36 phosphorylation only in the case of JNK1/2. Moreover, S36 phosphorylation of recombinant p66Shc by JNK1 but not PKCß was demonstrated. We further confirmed JNK1/2-dependent regulation of p66ShcS36 phosphorylation, ROS production and cell death using JNK1/2 deficient MEFs. Finally, the low ROS phenotype of JNK1/2 knockout MEFs was reversed by the phosphomimetic p66ShcS36E mutant. Inhibiting JNK1/2-regulated p66Shc activation may thus provide a therapeutic approach for the prevention of oxidative damage.


Subject(s)
JNK Mitogen-Activated Protein Kinases/metabolism , Phosphoserine/metabolism , Src Homology 2 Domain-Containing, Transforming Protein 1/metabolism , Animals , Cell Death/drug effects , Cell Hypoxia/drug effects , DNA Damage , Enzyme Activation/drug effects , Gene Knockout Techniques , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Mutant Proteins/metabolism , Oxidants/toxicity , Oxidative Stress/drug effects , Oxygen/pharmacology , Phenotype , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Kinase C beta/metabolism , Protein Kinase Inhibitors/pharmacology , Reactive Oxygen Species/metabolism , Recombinant Proteins/metabolism
9.
Cell Commun Signal ; 13: 22, 2015 Mar 31.
Article in English | MEDLINE | ID: mdl-25889880

ABSTRACT

BACKGROUND: Protein kinase C-θ (PKCθ) plays an important role in signal transduction down-stream of the T cell receptor and T cells deficient of PKCθ show impaired NF-κB as well as NFAT/AP-1 activation resulting in strongly decreased IL-2 expression and proliferation. However, it is not yet entirely clear, how the function of PKCθ - upon T cell activation - is regulated on a molecular level. FINDINGS: Employing a yeast two-hybrid screen and co-immunoprecipitation analyses, we here identify coronin 1A (Coro1A) as a novel PKCθ-interacting protein. We show that the NH2-terminal WD40 domains of Coro1A and the C2-like domain of PKCθ are sufficient for the interaction. Furthermore, we confirm a physical interaction by GST-Coro1A mediated pull-down of endogenous PKCθ protein. Functionally, wild-type but not Coro1A lacking its actin-binding domain negatively interferes with PKCθ-dependent NF-κB, Cyclin D1 and IL-2 transactivation when analysed with luciferase promoter activation assays in Jurkat T cells. This could be phenocopied by pharmacological inhibitors of actin polymerization and PKC, respectively. Mechanistically, Coro1A overexpression attenuates both lipid raft and plasma membrane recruitment of PKCθ in CD3/CD28-activated T cells. Using primary CD3(+) T cells, we observed that (opposite to PKCθ) Coro1A does not localize preferentially to the immunological synapse. In addition, we show that CD3(+) T cells isolated from Coro1A-deficient mice show impaired IKK/NF-κB transactivation. CONCLUSIONS: Together, these findings both in Jurkat T cells as well as in primary T cells indicate a regulatory role of Coro1A on PKCθ recruitment and function downstream of the TCR leading to NF-κB transactivation.


Subject(s)
Isoenzymes/metabolism , Microfilament Proteins/metabolism , Multiprotein Complexes/metabolism , Protein Kinase C/metabolism , T-Lymphocytes/metabolism , Animals , Humans , Isoenzymes/genetics , Jurkat Cells , Mice , Microfilament Proteins/genetics , Multiprotein Complexes/genetics , NF-kappa B/genetics , NF-kappa B/metabolism , Protein Kinase C/genetics , Protein Kinase C-theta , Protein Transport/physiology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/cytology , Transcriptional Activation/physiology
10.
PLoS One ; 10(3): e0121582, 2015.
Article in English | MEDLINE | ID: mdl-25822457

ABSTRACT

Amazingly little sequence variation is reported for the kringle IV 2 copy number variation (KIV 2 CNV) in the human LPA gene. Apart from whole genome sequencing projects, this region has only been analyzed in some detail in samples of European populations. We have performed a systematic resequencing study of the exonic and flanking intron regions within the KIV 2 CNV in 90 alleles from Asian, European, and four different African populations. Alleles have been separated according to their CNV length by pulsed field gel electrophoresis prior to unbiased specific PCR amplification of the target regions. These amplicons covered all KIV 2 copies of an individual allele simultaneously. In addition, cloned amplicons from genomic DNA of an African individual were sequenced. Our data suggest that sequence variation in this genomic region may be higher than previously appreciated. Detection probability of variants appeared to depend on the KIV 2 copy number of the analyzed DNA and on the proportion of copies carrying the variant. Asians had a high frequency of so-called KIV 2 type B and type C (together 70% of alleles), which differ by three or two synonymous substitutions respectively from the reference type A. This is most likely explained by the strong bottleneck suggested to have occurred when modern humans migrated to East Asia. A higher frequency of variable sites was detected in the Africans. In particular, two previously unreported splice site variants were found. One was associated with non-detectable Lp(a). The other was observed at high population frequencies (10% to 40%). Like the KIV 2 type B and C variants, this latter variant was also found in a high proportion of KIV 2 repeats in the affected alleles and in alleles differing in copy numbers. Our findings may have implications for the interpretation of SNP analyses in other repetitive loci of the human genome.


Subject(s)
DNA Copy Number Variations , Genetic Variation , Lipoprotein(a)/genetics , Asian People/genetics , Base Sequence , Black People/genetics , Cloning, Molecular , Ethnicity/genetics , Exons , Female , Gene Frequency , Humans , Introns , Kringles/genetics , Male , White People/genetics
11.
J Biol Chem ; 289(28): 19420-34, 2014 Jul 11.
Article in English | MEDLINE | ID: mdl-24872409

ABSTRACT

Rab GTPases control membrane traffic and receptor-mediated endocytosis. Within this context, Rab5a plays an important role in the spatial regulation of intracellular transport and signal transduction processes. Here, we report a previously uncharacterized role for Rab5a in the regulation of T-cell motility. We show that Rab5a physically associates with protein kinase Cϵ (PKCϵ) in migrating T-cells. After stimulation of T-cells through the integrin LFA-1 or the chemokine receptor CXCR4, Rab5a is phosphorylated on an N-terminal Thr-7 site by PKCϵ. Both Rab5a and PKCϵ dynamically interact at the centrosomal region of migrating cells, and PKCϵ-mediated phosphorylation on Thr-7 regulates Rab5a trafficking to the cell leading edge. Furthermore, we demonstrate that Rab5a Thr-7 phosphorylation is functionally necessary for Rac1 activation, actin rearrangement, and T-cell motility. We present a novel mechanism by which a PKCϵ-Rab5a-Rac1 axis regulates cytoskeleton remodeling and T-cell migration, both of which are central for the adaptive immune response.


Subject(s)
Adaptive Immunity/physiology , Cell Movement/physiology , Protein Kinase C-epsilon/metabolism , T-Lymphocytes/metabolism , rab5 GTP-Binding Proteins/metabolism , Cell Line , Cytoskeleton/genetics , Cytoskeleton/metabolism , Female , Humans , Lymphocyte Function-Associated Antigen-1/genetics , Lymphocyte Function-Associated Antigen-1/metabolism , Male , Phosphorylation/physiology , Protein Kinase C-epsilon/genetics , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , T-Lymphocytes/cytology , rab5 GTP-Binding Proteins/genetics
12.
Immunity ; 38(1): 41-52, 2013 Jan 24.
Article in English | MEDLINE | ID: mdl-23290522

ABSTRACT

Transforming growth-factor ß (TGFß) has been implicated in T helper 17 (Th17) cell biology and in triggering expression of interleukin-17A (IL-17A), which is a key Th17 cell cytokine. Deregulated TGFß receptor (TGFßR) signaling has been implicated in Th17-cell-mediated autoimmune pathogenesis. Nevertheless, the full molecular mechanisms involved in the activation of the TGFßR pathway in driving IL-17A expression remain unknown. Here, we identified protein kinase C α (PKCα) as a signaling intermediate specific to the Th17 cell subset in the activation of TGFßRI. We have shown that PKCα physically interacts and functionally cooperates with TGFßRI to promote robust SMAD2-3 activation. Furthermore, PKCα-deficient (Prkca(-/-)) cells demonstrated a defect in SMAD-dependent IL-2 suppression, as well as decreased STAT3 DNA binding within the Il17a promoter. Consistently, Prkca(-/-) cells failed to mount appropriate IL-17A, but not IL-17F, responses in vitro and were resistant to induction of Th17-cell-dependent experimental autoimmune encephalomyelitis in vivo.


Subject(s)
Interleukin-17/metabolism , Protein Kinase C-alpha/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Animals , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Gene Expression Regulation , Interleukin-17/immunology , Mice , Mice, Knockout , Myelin-Oligodendrocyte Glycoprotein/adverse effects , Peptide Fragments/adverse effects , Protein Kinase C-alpha/genetics , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Smad Proteins/metabolism , Substrate Specificity
13.
PLoS One ; 8(1): e53709, 2013.
Article in English | MEDLINE | ID: mdl-23335970

ABSTRACT

In T cells PKCθ mediates the activation of critical signals downstream of TCR/CD28 stimulation. We investigated the molecular mechanisms by which PKCθ regulates NFκB transactivation by examining PKCθ/ß single and double knockout mice and observed a redundant involvement of PKCθ and PKCß in this signaling pathway. Mechanistically, we define a PKCθ-CYLD protein complex and an interaction between the positive PKCθ/ß and the negative CYLD signaling pathways that both converge at the level of TAK1/IKK/I-κBα/NFκB and NFAT transactivation. In Jurkat leukemic T cells, CYLD is endoproteolytically processed in the initial minutes of stimulation by the paracaspase MALT1 in a PKC-dependent fashion, which is required for robust IL-2 transcription. However, in primary T cells, CYLD processing occurs with different kinetics and an altered dependence on PKC. The formation of a direct PKCθ/CYLD complex appears to regulate the short-term spatial distribution of CYLD, subsequently affecting NFκB and NFAT repressional activity of CYLD prior to its MALT1-dependent inactivation. Taken together, our study establishes CYLD as a new and critical PKCθ interactor in T cells and reveals that antagonistic PKCθ/ß-CYLD crosstalk is crucial for the adjustment of immune thresholds in primary mouse CD3(+) T cells.


Subject(s)
CD3 Complex , Cysteine Endopeptidases/metabolism , NF-kappa B/metabolism , NFATC Transcription Factors/metabolism , Protein Kinase C/metabolism , Signal Transduction , T-Lymphocytes/metabolism , Animals , CD3 Complex/metabolism , Caspases/metabolism , Cell Line , Cysteine Endopeptidases/genetics , Deubiquitinating Enzyme CYLD , Enzyme Activation , Gene Expression Regulation , Humans , Jurkat Cells , Lymphocyte Activation , MAP Kinase Kinase 4/metabolism , MAP Kinase Kinase Kinases/metabolism , Mice , Mice, Knockout , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein , Neoplasm Proteins/metabolism , Phenotype , Protein Binding , Protein Kinase C/genetics , Proteolysis , T-Lymphocytes/immunology , Transcriptional Activation
14.
Immunol Invest ; 42(2): 164-77, 2013.
Article in English | MEDLINE | ID: mdl-23323525

ABSTRACT

One of the shortcomings of vaccinia virus (VACV) as immunization vector is the down-regulation of HLA and costimulatory molecules in antigen presenting cells. To overcome this problem we investigated the use of protein kinase C (PKC) as immune stimulatory agent. Thus several classical and atypical PKCs were inserted into wild-type or attenuated VACV using recombination into the hemagglutinin gene and the expression driven by the VACV 7,5K-IE gene promoter. Recombinant constructs expressing PKC-alpha, -beta, -theta as well as wild-type, constitutive active or dominant negative PKC-zeta constructs were generated. Additional constructs expressing PKB/Akt1 and ICAM-1 were used for comparison. Immature and mature peripheral blood derived-dendritic cells (DC) as well as lymphoid cell lines capable of obtaining a DC-like phenotype upon mitogen stimulation were infected. Disappointingly, VACV-driven PKC overexpression did not significantly enhance expression of various activation markers or costimulatory molecules tested. Neither CD86 nor HLA-DR expression was upregulated and also no influence on the maturation of DC, as measured by DC-SIGN and CD83, was observed. However, VACV did not interfere with LPS induced up-regulation of CD83 and did not lead to substantial apoptosis of infected DC within the first 24 hours.


Subject(s)
B7-2 Antigen/metabolism , Dendritic Cells/immunology , Dendritic Cells/virology , HLA-DR Antigens/metabolism , Protein Kinase C/metabolism , Up-Regulation , Animals , B7-2 Antigen/genetics , B7-2 Antigen/immunology , Cell Differentiation , Cell Line , Coculture Techniques , Dendritic Cells/metabolism , Genetic Vectors , HLA-DR Antigens/genetics , HLA-DR Antigens/immunology , Humans , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Protein Kinase C/genetics , Protein Kinase C/immunology , Recombination, Genetic , Vaccinia virus/enzymology , Vaccinia virus/genetics
15.
J Autoimmun ; 39(4): 428-40, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22921335

ABSTRACT

Interleukin-17A (IL-17A) is the signature cytokine produced by Th17 CD4(+) T cells and has been tightly linked to autoimmune pathogenesis. In particular, the transcription factors NFAT and RORγt are known to activate Il17a transcription, although the detailed mechanism of action remains incompletely understood. Here, we show that the nuclear orphan receptor NR2F6 can attenuate the capacity of NFAT to bind to critical regions of the Il17a gene promoter. In addition, because NR2F6 binds to defined hormone response elements (HREs) within the Il17a locus, it interferes with the ability of RORγt to access the DNA. Consistently, NFAT and RORγt binding within the Il17a locus were enhanced in Nr2f6-deficient CD4(+) Th17 cells but decreased in Nr2f6-overexpressing transgenic CD4(+) Th17 cells. Taken together, our findings uncover an example of antagonistic regulation of Il17a transcription through the direct reciprocal actions of NR2F6 versus NFAT and RORγt.


Subject(s)
COUP Transcription Factors/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Interleukin-17/immunology , NFATC Transcription Factors/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Th17 Cells/immunology , Animals , Binding Sites , Binding, Competitive , COUP Transcription Factors/deficiency , COUP Transcription Factors/genetics , DNA/immunology , DNA/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Gene Expression Regulation/immunology , Interleukin-17/genetics , Interleukin-17/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Protein Binding , Repressor Proteins , Response Elements/immunology , Signal Transduction , Th17 Cells/metabolism , Th17 Cells/pathology , Transcription, Genetic
16.
J Autoimmun ; 37(3): 198-208, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21700422

ABSTRACT

Transforming growth factor ß (TGFß) plays a central role in maintaining immune homeostasis by regulating the initiation and termination of immune responses and thus preventing the development of autoimmune diseases. In this study, we describe an essential mechanism by which the actin regulatory protein Coronin 1A (Coro1A) ensures the proper response of Th17 CD4(+) T cells to TGFß. Coro1A has been established as a key player in T cell survival, migration, activation, and Ca(2+) regulation in naive T cells. We show that mice lacking Coro1a developed less severe experimental autoimmune encephalomyelitis (EAE). Unexpectedly, upon the re-induction of EAE, Coro1a(-/-) mice exhibited enhanced EAE signs that correlated with increased numbers of IL-17 producing CD4(+) cells in the central nervous system (CNS) compared to wild-type mice. In vitro differentiated Coro1a(-/-) Th17 CD4(+) T cells consistently produced more IL-17 than wild-type cells and displayed a Th17/Th1-like phenotype in regard to the expression of the Th1 markers T-bet and IFNγ. Mechanistically, the Coro1a(-/-) Th17 cell phenotype correlated with a severe defect in TGFßR-mediated SMAD3 activation. Taken together, these data provide experimental evidence of a non-redundant role of Coro1A in the regulation of Th17 CD4(+) cell effector functions and, subsequently, in the development of autoimmunity.


Subject(s)
Gene Expression/immunology , Lupus Erythematosus, Systemic/immunology , Microfilament Proteins/immunology , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/immunology , Smad3 Protein/metabolism , Transforming Growth Factor beta/metabolism , Animals , Cell Differentiation/immunology , Cell Movement/immunology , Disease Models, Animal , Female , Flow Cytometry , Interferon-gamma/biosynthesis , Interleukin-17/biosynthesis , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/pathology , Mice , Mice, Knockout , Microfilament Proteins/deficiency , Microfilament Proteins/genetics , Real-Time Polymerase Chain Reaction , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/immunology , Signal Transduction/genetics , Smad3 Protein/genetics , Smad3 Protein/immunology , Th1 Cells/immunology , Th1 Cells/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Transforming Growth Factor beta/immunology
17.
Am J Med Genet A ; 149A(11): 2522-6, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19842202

ABSTRACT

Partial duplication 3q is a well defined clinical entity characterized by growth retardation, cryptorchism, microcephaly, and characteristic dysmorphisms. Most patients present with large duplications or are associated with a second chromosomal imbalance, which makes the definition of the phenotype difficult. Here, we report on a 4-year and 8-month-old girl with pre- and postnatal measurements in the high normal range, developmental delay, minor dysmorphic features, and a de novo unbalanced 3/4 translocation with trisomy 3q27 --> qter and monosomy of the subtelomeric region of 4p. Conventional karyotyping, FISH with probes from the Wolf-Hirschhorn syndrome critical region and chromosome 4p locus-specific probes, microsatellite marker-based haplotyping, and SNP microarray copy number analysis revealed a terminal 4p deletion of less than 500 kb with a breakpoint distal to the Wolf-Hirschhorn syndrome critical region, a chromosome 3q duplication of around 15.3 Mb, with origin of the rearrangement in paternal meiosis. Thus, our case clearly characterizes the phenotype of pure partial duplication 3q more exactly, and moreover, indicates that small chromosome rearrangements might lead to growth in the upper normal range or even cause overgrowth.


Subject(s)
Chromosomes, Human, Pair 3/genetics , Trisomy/genetics , Adult , Child, Preschool , Chromosome Banding , Female , Gene Duplication , Humans , In Situ Hybridization, Fluorescence , Infant, Newborn , Phenotype
18.
Sci Signal ; 2(76): ra30, 2009 Jun 23.
Article in English | MEDLINE | ID: mdl-19549985

ABSTRACT

The E3 ubiquitin ligase Casitas B-lineage lymphoma (Cbl-b) is central to antigen-induced immune tolerance and regulates the CD28 dependence of T cell activation. Cbl-b undergoes ubiquitination and proteasomal degradation after adequate costimulation of T cells; however, the mechanism involved is unknown. Here, we identified protein kinase C-theta (PKC-theta) as the critical intermediary for the inactivation of Cbl-b in response to costimulation of T cells through CD28. PKC-theta associated with Cbl-b on stimulation of the T cell receptor. After costimulation of T cells through CD28, Cbl-b was ubiquitinated and degraded through a mechanism that depended on the kinase activity of PKC-theta. Consistent with this mechanism, the impaired responses of PKCtheta-deficient T cells were at least partially restored by the concomitant genetic loss of cblb. Thus, our data establish a nonredundant antagonism between PKC-theta and Cbl-b that regulates T cell activation responses.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Isoenzymes/immunology , Lymphocyte Activation/immunology , Protein Kinase C/immunology , Proto-Oncogene Proteins c-cbl/immunology , T-Lymphocytes/immunology , Ubiquitin-Protein Ligases/immunology , Ubiquitination/immunology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , CD28 Antigens/genetics , CD28 Antigens/immunology , CD28 Antigens/metabolism , Isoenzymes/genetics , Isoenzymes/metabolism , Lymphocyte Activation/genetics , Mice , Mice, Knockout , Protein Kinase C/genetics , Protein Kinase C/metabolism , Protein Kinase C-theta , Proto-Oncogene Proteins c-cbl/genetics , Proto-Oncogene Proteins c-cbl/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/enzymology , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/genetics
19.
Blood ; 112(12): 4617-27, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-18796635

ABSTRACT

The antigen-specific interaction of a T cell with an antigen-presenting cell (APC) results in the formation of an immunologic synapse (IS) between the membranes of the 2 cells. beta(2) integrins on the T cell, namely, leukocyte function-associated antigen 1 (LFA-1) and its counter ligand, namely, immunoglobulin-like cell adhesion molecule 1 (ICAM-1) on the APC, critically stabilize this intercellular interaction. The small GTPase Rap1 controls T-cell adhesion through modulating the affinity and/or spatial organization of LFA-1; however, the upstream regulatory components triggered by the T-cell receptor (TCR) have not been resolved. In the present study, we identified a previously unknown function of a protein kinase C- theta (PKC-theta)/RapGEF2 complex in LFA-1 avidity regulation in T lymphocytes. After T-cell activation, the direct phosphorylation of RapGEF2 at Ser960 by PKC- theta regulates Rap1 activation as well as LFA-1 adhesiveness to ICAM-1. In OT-II TCR-transgenic CD4(+) T cells, clustering of LFA-1 after antigen activation was impaired in the absence of PKC- theta. These data define that, among other pathways acting on LFA-1 regulation, PKC- theta and its effector RapGEF2 are critical factors in TCR signaling to Rap1. Taken together, PKC- theta sets the threshold for T-cell activation by positively regulating both the cytokine responses and the adhesive capacities of T lymphocytes.


Subject(s)
Cell Adhesion/genetics , Isoenzymes/physiology , Protein Kinase C/physiology , T-Lymphocytes/physiology , rap1 GTP-Binding Proteins/metabolism , Animals , Cell Adhesion/drug effects , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Jurkat Cells , Lymphocyte Function-Associated Antigen-1/physiology , Mice , Mice, Knockout , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Phosphorylation , Protein Binding , Protein Kinase C/genetics , Protein Kinase C/metabolism , Protein Kinase C-theta , RNA, Small Interfering/pharmacology , Substrate Specificity , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Up-Regulation/genetics , Up-Regulation/physiology
20.
Immunity ; 29(2): 205-16, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18701084

ABSTRACT

The protein kinase C (PKC) family of serine-threonine kinases plays a central role in T lymphocyte activation. Here, we identify NR2F6, a nuclear zinc-finger orphan receptor, as a critical PKC substrate and essential regulator of CD4(+) T cell activation responses. NR2F6 potently antagonized the ability of T helper 0 (Th0) and Th17 CD4(+) T cells to induce expression of key cytokine genes such as interleukin-2 (IL-2) and IL-17. Mechanistically, NR2F6 directly interfered with the DNA binding of nuclear factor of activated T cells (NF-AT):activator protein 1 (AP-1) but not nuclear factor kappaB (NF-kappa B) and, subsequently, transcriptional activity of the NF-AT-dependent IL-17A cytokine promoter. Consistent with our model, Nr2f6-deficient mice had hyperreactive lymphocytes, developed a late-onset immunopathology, and were hypersusceptible to Th17-dependent experimental autoimmune encephalomyelitis. Our study establishes NR2F6 as a transcriptional repressor of IL-17 expression in Th17-differentiated CD4(+) T cells in vitro and in vivo.


Subject(s)
Autoimmune Diseases/immunology , CD4-Positive T-Lymphocytes/immunology , DNA-Binding Proteins/metabolism , Interleukin-17/metabolism , Lymphocyte Activation , Protein Kinase C/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , T-Lymphocytes, Helper-Inducer/immunology , Animals , Autoimmune Diseases/metabolism , Autoimmunity/immunology , CD4-Positive T-Lymphocytes/metabolism , COUP Transcription Factors , DNA-Binding Proteins/deficiency , Interleukin-17/immunology , Interleukin-2/immunology , Interleukin-2/metabolism , Mice , Mice, Knockout , Receptors, Cytoplasmic and Nuclear/deficiency , Repressor Proteins , T-Lymphocytes, Helper-Inducer/metabolism , Transcription Factor AP-1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...