Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters








Publication year range
1.
Biomolecules ; 11(2)2021 02 05.
Article in English | MEDLINE | ID: mdl-33562506

ABSTRACT

Protein Kinase C theta (PKCθ) is a serine/threonine kinase that belongs to the novel PKC subfamily. In normal tissue, its expression is restricted to skeletal muscle cells, platelets and T lymphocytes in which PKCθ controls several essential cellular processes such as survival, proliferation and differentiation. Particularly, PKCθ has been extensively studied for its role in the immune system where its translocation to the immunological synapse plays a critical role in T cell activation. Beyond its physiological role in immune responses, increasing evidence implicates PKCθ in the pathology of various diseases, especially autoimmune disorders and cancers. In this review, we discuss the implication of PKCθ in various types of cancers and the PKCθ-mediated signaling events controlling cancer initiation and progression. In these types of cancers, the high PKCθ expression leads to aberrant cell proliferation, migration and invasion resulting in malignant phenotype. The recent development and application of PKCθ inhibitors in the context of autoimmune diseases could benefit the emergence of treatment for cancers in which PKCθ has been implicated.


Subject(s)
Autoimmune Diseases/genetics , Gene Expression Regulation, Neoplastic , Neoplasms/genetics , Protein Kinase C-theta/genetics , Protein Subunits/genetics , Signal Transduction/genetics , Animals , Autoimmune Diseases/enzymology , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Cell Movement , Cell Proliferation , Humans , NF-kappa B/genetics , NF-kappa B/metabolism , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasms/enzymology , Neoplasms/immunology , Neoplasms/pathology , Protein Binding , Protein Interaction Domains and Motifs , Protein Kinase C-theta/chemistry , Protein Kinase C-theta/immunology , Protein Subunits/chemistry , Protein Subunits/immunology , Th17 Cells/immunology , Th17 Cells/pathology , Th2 Cells/immunology , Th2 Cells/pathology
2.
J Biol Chem ; 295(41): 14214-14221, 2020 10 09.
Article in English | MEDLINE | ID: mdl-32796034

ABSTRACT

T-cell activation is a critical part of the adaptive immune system, enabling responses to foreign cells and external stimulus. In this process, T-cell antigen receptor (TCR) activation stimulates translocation of the downstream kinase PKCθ to the membrane, leading to NF-κB activation and thus transcription of relevant genes. However, the details of how PKCθ is recruited to the membrane remain enigmatic. It is known that annexin A5 (ANXA5), a calcium-dependent membrane-binding protein, has been reported to mediate PKCδ activation by interaction with PKCδ, a homologue of PKCθ, which implicates a potential role of ANXA5 involved in PKCθ signaling. Here we demonstrate that ANXA5 does play a critical role in the recruitment of PKCθ to the membrane during T-cell activation. ANXA5 knockout in Jurkat T cells substantially inhibited the membrane translocation of PKCθ upon TCR engagement and blocked the recruitment of CARMA1-BCL10-MALT1 signalosome, which provides a platform for the catalytic activation of IKKs and subsequent activation of canonical NF-κB signaling in activated T cells. As a result, NF-κB activation was impaired in ANXA5-KO T cells. T-cell activation was also suppressed by ANAX5 knockdown in primary T cells. These results demonstrated a novel role of ANXA5 in PKC translocation and PKC signaling during T-cell activation.


Subject(s)
Annexin A5/immunology , Lymphocyte Activation , Protein Kinase C-theta/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Animals , Annexin A5/genetics , Humans , Jurkat Cells , Mice , Protein Kinase C-theta/genetics , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology
3.
Mol Ther ; 28(9): 1987-2006, 2020 09 02.
Article in English | MEDLINE | ID: mdl-32492367

ABSTRACT

Regulatory T cells maintain immunological tolerance and dampen inflammatory responses. Administering regulatory T cells can prevent the immune-mediated tissue destruction of graft-versus-host disease, which frequently accompanies hematopoietic stem cell transfer. Neutralizing the T cell-specific kinase, protein kinase C theta, which promotes T cell effector functions and represses regulatory T cell differentiation, augments regulatory T cell immunosuppression and stability. We used a synthetic, cell-penetrating peptide mimic to deliver antibodies recognizing protein kinase C theta into primary human CD4 T cells. When differentiated ex vivo into induced regulatory T cells, treated cells expressed elevated levels of the regulatory T cell transcriptional regulator forkhead box P3, the surface-bound immune checkpoint receptor programmed death receptor-1, and pro-inflammatory interferon gamma, previously ascribed to a specific population of stable, highly suppressive human induced regulatory T cells. The in vitro suppressive capacity of these induced regulatory T cells was 10-fold greater than that of T cells differentiated without antibody delivery. When administered at the time of graft-versus-host disease induction, using a humanized mouse model, antibody-treated regulatory T cells were superior to non-treated T cells in attenuating lethal outcomes. This antibody delivery approach may overcome obstacles currently encountered using patient-derived regulatory T cells as a cell-based therapy for immune modulation.


Subject(s)
Adoptive Transfer/methods , Antibodies/immunology , Antibodies/pharmacology , Cell-Penetrating Peptides , Graft vs Host Disease/therapy , Immune Tolerance/drug effects , Intracellular Fluid/immunology , Protein Kinase C-theta/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cells, Cultured , Disease Models, Animal , Female , Forkhead Transcription Factors/metabolism , Graft vs Host Disease/immunology , Humans , Immune Tolerance/immunology , Interferon-gamma/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Programmed Cell Death 1 Receptor/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , Treatment Outcome
4.
Fish Shellfish Immunol ; 87: 788-795, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30716520

ABSTRACT

Haemorrhagic disease caused by grass carp reovirus (GCRV) can result in large-scale death of young grass carp, leading to irreparable economic losses that seriously affect large-scale breeding. Protein kinase C (PKC, also known as PRKC) represents a family of serine/threonine protein kinases that includes multiple isozymes in many species. Among these, PKC-θ (PKC theta, also written as PRKCQ) is a novel isoform, mainly expressed in T cells, that is known to be involved in immune system function in mammals. To date, no research on immunological functions of fish Pkc-θ has been reported. To address this issue, we cloned the grass carp pkc-θ gene. Phylogenetic and syntenic analysis showed that this gene is the most evolutionarily conserved relative to zebrafish. Real-time quantitative PCR (RT-qPCR) indicated that pkc-θ was expressed at high levels in the gills and spleen of healthy grass carp. Infection with GCRV down regulated pkc-θ expression in the gills and spleen. Gene products that function upstream and downstream of pkc-θ were up regulated in the gill, but were down-regulated in the spleen. These results suggest that direct or indirect targeting of pkc-θ by GCRV may help the virus evade host immune defences in the spleen. Phorbol ester (PMA) treatment of Jurkat T cells induced translocation of grass carp Pkc-θ from the cytoplasm to the plasma membrane. This response to PMA suggests evolutionary conservation of an immune response function in fish Pkc-θ, as well as conservation of its sequence and structural domains. This study expanded our knowledge of the fish PKC gene family, and explored the role of pkc-θ in function of the grass carp immune system, providing new insights which may facilitate further studies of its biological functions.


Subject(s)
Carps/genetics , Carps/immunology , Fish Diseases/immunology , Gene Expression Regulation/immunology , Immunity, Innate/genetics , Protein Kinase C-theta/genetics , Protein Kinase C-theta/immunology , Amino Acid Sequence , Animals , Base Sequence , Fish Proteins/chemistry , Fish Proteins/genetics , Fish Proteins/immunology , Gene Expression Profiling/veterinary , Protein Kinase C-theta/chemistry , Random Allocation , Reoviridae/physiology , Reoviridae Infections/immunology , Reoviridae Infections/veterinary , Sequence Alignment/veterinary
5.
Fish Shellfish Immunol ; 84: 1100-1107, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30408601

ABSTRACT

A primitive adaptive immune system has recently been suggested to be present in a basal chordate amphioxus (Branchiostoma belcheri, Bb), making it an ideal model for studying the origin of adaptive immune. The novel protein kinase C isoform PKC-θ, but not its closest isoform PKC-δ, plays a critical role for mammalian T-cell activation via translocation to immunological synapse (IS) mediated by a unique PKC-θ V3 domain containing one PxxP motif. To understand the evolution of this unique PKC-θ V3 domain and the primitive adaptive immune system in amphioxus, we comparatively studied the orthologs of PKC-δ and -θ from amphioxus and other species. Phylogenetic analysis showed BbPKC-δ/θ to be the common ancestor of vertebrate PKC-δ and PKC-θ, with a V3 domain containing two PxxP motifs. One motif is conserved in both zebrafish and mammalian PKC-θ but is absent in PKC-δ V3 domain of these species, and has already emerged in drosophila PKC-δ. The other non-conserved motif emerged in BbPKC-δ/θ, and only retained in Danio rerio PKC-δ (DrPKC-δ) but lost in mammalian PKC-δ and -θ. Comparative analyses of the sequence and function of BbPKC-δ/θ, DrPKC-δ, DrPKC-θ and Homo sapiens PKC-θ (HsPKC-θ) in IS translocation and T-cell receptor (TCR)-induced NF-κB activation revealed that retention of the conserved PxxP motif and loss of the non-conserved PxxP motif in mammalian PKC-θ and loss of both PxxP motifs in mammalian PKC-δ accomplish the unique function of PKC-θ in T cells. Together, this study suggests an evolutionary mechanism for PKC-θ unique V3 and reveals BbPKC-δ/θ is the common ancestor of PKC-δ and -θ with a functional proto-V3 domain, supplying new evidence for the existence of primitive adaptive immune system in amphioxus.


Subject(s)
Adaptive Immunity/genetics , Fish Diseases/immunology , Gene Expression Regulation/immunology , Lancelets/genetics , Lancelets/immunology , Protein Kinase C-delta/genetics , Protein Kinase C-delta/immunology , Protein Kinase C-theta/genetics , Protein Kinase C-theta/immunology , Amino Acid Sequence , Animals , Gene Expression Profiling/veterinary , Lancelets/enzymology , Phylogeny , Protein Kinase C-delta/chemistry , Protein Kinase C-theta/chemistry , Sequence Alignment/veterinary
6.
J Clin Invest ; 128(10): 4604-4621, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30106752

ABSTRACT

Regulatory T cells (Tregs) are critical for maintaining immune homeostasis. However, current Treg immunotherapies do not optimally treat inflammatory diseases in patients. Understanding the cellular processes that control Treg function may allow for the augmentation of therapeutic efficacy. In contrast to activated conventional T cells, in which protein kinase C-θ (PKC-θ) localizes to the contact point between T cells and antigen-presenting cells, in human and mouse Tregs, PKC-θ localizes to the opposite end of the cell in the distal pole complex (DPC). Here, using a phosphoproteomic screen, we identified the intermediate filament vimentin as a PKC-θ phospho target and show that vimentin forms a DPC superstructure on which PKC-θ accumulates. Treatment of mouse Tregs with either a clinically relevant PKC-θ inhibitor or vimentin siRNA disrupted vimentin and enhanced Treg metabolic and suppressive activity. Moreover, vimentin-disrupted mouse Tregs were significantly better than controls at suppressing alloreactive T cell priming in graft-versus-host disease (GVHD) and GVHD lethality, using a complete MHC-mismatch mouse model of acute GVHD (C57BL/6 donor into BALB/c host). Interestingly, vimentin disruption augmented the suppressor function of PKC-θ-deficient mouse Tregs. This suggests that enhanced Treg activity after PKC-θ inhibition is secondary to effects on vimentin, not just PKC-θ kinase activity inhibition. Our data demonstrate that vimentin is a key metabolic and functional controller of Treg activity and provide proof of principle that disruption of vimentin is a feasible, translationally relevant method to enhance Treg potency.


Subject(s)
Antigen-Presenting Cells/immunology , Graft vs Host Disease/immunology , Intermediate Filaments/immunology , Lymphocyte Activation , T-Lymphocytes, Regulatory/immunology , Vimentin/immunology , Animals , Antigen-Presenting Cells/pathology , Disease Models, Animal , Graft vs Host Disease/genetics , Graft vs Host Disease/pathology , Humans , Intermediate Filaments/genetics , Intermediate Filaments/pathology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Protein Kinase C-theta/genetics , Protein Kinase C-theta/immunology , T-Lymphocytes, Regulatory/pathology , Vimentin/genetics
7.
Cell Immunol ; 331: 137-145, 2018 09.
Article in English | MEDLINE | ID: mdl-29954581

ABSTRACT

Platelets modulate the immune system following injury by interacting with CD4+ T regulatory cells (CD4+ Tregs). The underlying mechanisms remain unsolved. We hypothesize paracrine interactions via Tumor necrosis factor-alpha (TNFα)-, Toll like receptor-4 (TLR4)-, and Interleukin-10 (IL-10). In the murine burn injury model, CD4+ Treg activation pathways were selectively addressed using TNFR2-, TLR4- and IL-10-deficient mice. The CD4+ Treg signalling molecule PKC-θ was analyzed using phospho-flow cytometry to detect rapid cell activation. Thromboelastometry (ROTEM®) was used to assess platelet activation. Injury induced significant early activation of CD4+ Tregs, disruption of TNFR2 and TLR4 activation pathways resulted in lower activity. The disruption of IL-10 crosstalk had no significant impact. Selective disruption of paracrine interactions is associated with changes in posttraumatic hemostasis parameters. TNFR2- and TLR4-dependent pathways modulate the activation of CD4+ Tregs following trauma. In contrast, we did not observe a role of IL-10 in the posttraumatic activation of CD4+ Tregs. ONE SENTENCE SUMMARY: TLR4- and TNFR2-dependent mechanisms, but not IL-10-dependent pathways, modulate the anti-inflammatory response of CD4+ Tregs following trauma.


Subject(s)
Burns/immunology , Interleukin-10/immunology , Lymphocyte Activation/immunology , Receptors, Tumor Necrosis Factor, Type II/immunology , T-Lymphocytes, Regulatory/immunology , Toll-Like Receptor 4/immunology , Animals , Burns/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Interleukin-10/genetics , Interleukin-10/metabolism , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Platelet Activation/immunology , Protein Kinase C-theta/immunology , Protein Kinase C-theta/metabolism , Receptors, Tumor Necrosis Factor, Type II/genetics , Receptors, Tumor Necrosis Factor, Type II/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes, Regulatory/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
8.
Parasite Immunol ; 40(2)2018 02.
Article in English | MEDLINE | ID: mdl-29117626

ABSTRACT

Cerebral malaria (CM) is one complication of Plasmodium parasite infection that can lead to strong inflammatory immune responses in the central nervous system (CNS), accompanied by lung inflammation and anaemia. Here, we focus on the role of the innate immune response in experimental cerebral malaria (ECM) caused by blood-stage murine Plasmodium berghei ANKA infection. While T cells are important for ECM pathogenesis, the role of innate lymphoid cells (ILCs) is only emerging. The role of ILCs and non-lymphoid cells, such as neutrophils and platelets, contributing to the host immune response and leading to ECM and human cerebral malaria (HCM) is reviewed.


Subject(s)
Cytokines/immunology , Immunity, Innate/immunology , Lymphocytes/immunology , Malaria, Cerebral/immunology , Plasmodium berghei/immunology , Animals , Humans , Killer Cells, Natural/immunology , Macrophages/immunology , Malaria, Cerebral/parasitology , Mice , Mice, Inbred C57BL , Neutrophils/immunology , Protein Kinase C-theta/immunology
9.
EBioMedicine ; 25: 165-174, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29050947

ABSTRACT

The molecular mechanisms that govern differential T cell development into pro-inflammatory Th17 vs. regulatory T (Treg) cells remain unclear. Here, we show that selective deletion of CREB in T cells or Th17 cells impaired Th17 cell differentiation in vitro and in vivo, and led to resistance to autoimmune diseases. Mechanistically, CREB, activated by CD3-PKC-Ï´ signaling, plays a key role in regulating Th17 cell differentiation, at least in part through directly binding to the Il17-Il17f gene locus. Unexpectedly, although dispensable for FOXP3 expression and for the homeostasis and suppressive function of thymus-derived Treg cells, CREB negatively regulates the survival of TGF-ß-induced Treg cells, and deletion of CREB resulted in increased FOXP3+ Treg cells in the intestine and protection in a colitis model. Thus, CREB is critical in autoimmune diseases by promoting Th17 cell and inhibiting de novo Treg cell generation.


Subject(s)
Autoimmune Diseases/genetics , Cyclic AMP Response Element-Binding Protein/genetics , Forkhead Transcription Factors/genetics , Interleukin-17/genetics , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , CD3 Complex/genetics , CD3 Complex/immunology , Cell Differentiation/genetics , Cell Differentiation/immunology , Cyclic AMP Response Element-Binding Protein/immunology , Forkhead Transcription Factors/immunology , Humans , Interleukin-17/immunology , Lymphocyte Activation , Mice , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Protein Kinase C-theta/genetics , Protein Kinase C-theta/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/immunology
10.
Sci Rep ; 7(1): 4258, 2017 06 26.
Article in English | MEDLINE | ID: mdl-28652577

ABSTRACT

In vitro induced human regulatory T cells (iTregs) have demonstrated in vivo therapeutic utility, but pathways regulating their function have not been elucidated. Here, we report that human iTregs generated in vitro from naïve cord blood cells preferentially recruit Disc large homolog 1 (Dlgh1) and exclude protein kinase C (PKC)-θ from immunological synapses formed on supported lipid bilayers with laterally mobile ICAM-1 and anti-CD3 mAb. Also, iTregs display elevated Dlgh1 overall and Dlgh1-dependent p38 phosphorylation, higher levels of phosphatase and tensin homolog (PTEN), and diminished Akt phosphorylation. Pharmacological interruption of PKC-θ increases and Dlgh1 silencing decreases the ability of iTregs to suppress interferon-γ production by CD4+CD25- effector T cells (Teff). Comparison with expanded cord blood-derived CD4+CD25hi tTreg and expanded Teffs from the same donors indicate that iTreg are intermediate between expanded CD4+CD25hi tTregs and Teffs, whereas modulation of suppressive activities by PKC-θ and Dlgh1 signaling pathways are shared.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Immunological Synapses/genetics , Membrane Proteins/genetics , Protein Kinase C-theta/genetics , T-Lymphocytes, Regulatory/metabolism , Adaptor Proteins, Signal Transducing/immunology , CD4 Antigens/genetics , Cell Differentiation/genetics , Discs Large Homolog 1 Protein , Fetal Blood/cytology , Fetal Blood/metabolism , Humans , Immunological Synapses/metabolism , Intercellular Adhesion Molecule-1/genetics , Interleukin-2 Receptor alpha Subunit/genetics , Lipid Bilayers/immunology , Lipid Bilayers/metabolism , Lymphocyte Activation , Membrane Proteins/immunology , Phosphorylation , Protein Kinase C-theta/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , p38 Mitogen-Activated Protein Kinases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL