Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 210
Filter
Add more filters










Publication year range
1.
Immunology ; 2024 May 26.
Article in English | MEDLINE | ID: mdl-38798068

ABSTRACT

Members of the Protein kinases D (PKD) family are described as regulators of T cell responses. From the two T cell-expressed isoforms PKD2 and PKD3, so far mainly the former was thoroughly investigated and is well understood. Recently, we have investigated also PKD3 using conventional as well as conditional T cell-specific knockout models. These studies suggested PKD3 to be a T cell-extrinsic regulator of the cells' fate. However, these former model systems did not take into account possible redundancies with the highly homologous PKD2. To overcome this issue and thus properly unravel PKD3's T cell-intrinsic functions, here we additionally used a mouse model overexpressing a constitutively active isoform of PKD3 specifically in the T cell compartment. These transgenic mice showed a slightly higher proportion of central memory T cells in secondary lymphoid organs and blood. This effect could not be explained via differences upon polyclonal stimulation in vitro, however, may be connected to the observed developmental aberrances in the CD8 single positive compartment during thymic development. Lastly, the observed alterations in the CD8+ T cell compartment did not impact proper immune response upon immunization with ovalbumin or in a subcutaneous tumour model suggesting only a small to absent biological relevance. Taking together the knowledge of all our published studies on PKD3 in the T cell compartment, we now conclude that T cell-intrinsic PKD3 is a fine-tuner of central memory T cell as well as CD8 single positive thymocyte development.

2.
Front Immunol ; 13: 1049033, 2022.
Article in English | MEDLINE | ID: mdl-36466811

ABSTRACT

Protein kinases D (PKDs) are implicated in T cell receptor (TCR) signaling. Of the two T cell-expressed isoforms PKD2 and PKD3, however, only the former one is rather well understood in this immune cell type. Recently, we have observed a putative hyper-phenotype of T cells from conventional PKD3-knockout mice, which we explained as a secondary effect due to a skewed T cell compartment from naïve towards effector/memory T cells already under steady state conditions. Nonetheless, to this end it is not clear whether these aberrations are mediated by a T cell-intrinsic or -extrinsic function of PKD3. To address this question, we have investigated mice lacking PKD3 specifically in the T cell compartment. We could show that T cells from CD4-Cre-driven conditional knockout mice did not phenocopy the ones from conventional PKD3-knockout mice. In brief, no skewing in the T cell compartment of peripheral lymphoid organs, no hyper-activation upon stimulation in vitro or in vivo as well as no aberrations in follicular helper T cells (TFH) upon immunization were observed. Hence, although PKD3 is strongly regulated upon TCR stimulation, in T cells this kinase seems to be dispensable for their activation. The described skewing in the T cell compartment of conventional PKD3-deficient mice seems to be mediated by T cell-extrinsic mechanisms, thus once more emphasizing the importance of cell type-specific mouse models.


Subject(s)
Germinal Center , Receptors, Antigen, T-Cell , Mice , Animals , Lymphocyte Count , Mice, Knockout , Receptors, Antigen, T-Cell/genetics
3.
Cell Commun Signal ; 20(1): 54, 2022 04 19.
Article in English | MEDLINE | ID: mdl-35440091

ABSTRACT

BACKGROUND: The Protein kinase D3 (PKD3) has been implicated in signal transduction downstream of the T cell receptor (TCR). However, its role for the activation of primary T lymphocytes has not been elucidated so far. METHODS: Expression of PKD isoforms in primary murine T cells was determined by RT-PCR and SDS-Page. A germline PKD3-knockout mouse line was analyzed for its immune response to OVA/alum intraperitoneal immunization. Phenotyping of the T cell compartment ex vivo as well as upon stimulation in vitro was performed by flow cytometry. Additionally, cytokine expression was assessed by flow cytometry, RT-PCR and Luminex technology. RESULTS: PKD expression in T cells is modulated by TCR stimulation, leading to a rapid down-regulation on mRNA and on protein level. PKD3-deficient mice respond to immunization with enhanced T follicular helper cell generation. Furthermore, peripheral PKD3-deficient CD4+ T cells express more interleukin-2 than wild type CD4+ T cells upon TCR stimulation ex vivo. However, purified naïve CD4+ T cells do not differ in their phenotype upon differentiation in vitro from wild type T cells. Moreover, we observed a shift towards an effector/memory phenotype of splenic T cells at steady state, which might explain the contradictory results obtained with pan-T cells ex vivo and naïve-sorted T cells. CONCLUSION: While PKD3-deficiency in vivo in mice leads to a skewing of the T cell compartment towards a more activated phenotype, this kinase seems to be dispensable for naïve CD4+ T cell differentiation in vitro. Video Abstract.


Subject(s)
DNA-Activated Protein Kinase/metabolism , DNA-Binding Proteins/metabolism , T-Lymphocytes , Animals , CD4-Positive T-Lymphocytes , Mice , Mice, Knockout , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/metabolism
4.
Cancers (Basel) ; 14(3)2022 Feb 04.
Article in English | MEDLINE | ID: mdl-35159064

ABSTRACT

Protein kinase C iota (PKCι) functions as a bonafide human oncogene in lung and ovarian cancer and is required for KrasG12D-mediated lung cancer initiation and progression. PKCι expression is required for pancreatic cancer cell growth and maintenance of the transformed phenotype; however, nothing is known about the role of PKCι in pancreas development or pancreatic tumorigenesis. In this study, we investigated the effect of pancreas-specific ablation of PKCι expression on pancreatic cellular homeostasis, susceptibility to pancreatitis, and KrasG12D-mediated pancreatic cancer development. Knockout of pancreatic Prkci significantly increased pancreatic immune cell infiltration, acinar cell DNA damage, and apoptosis, but reduced sensitivity to caerulein-induced pancreatitis. Prkci-ablated pancreatic acinar cells exhibited P62 aggregation and a loss of autophagic vesicles. Loss of pancreatic Prkci promoted KrasG12D-mediated pancreatic intraepithelial neoplasia formation but blocked progression to adenocarcinoma, consistent with disruption of autophagy. Our results reveal a novel promotive role for PKCι in pancreatic epithelial cell autophagy and pancreatic cancer progression.

5.
Curr Alzheimer Res ; 18(12): 941-955, 2021.
Article in English | MEDLINE | ID: mdl-34951366

ABSTRACT

Βackground: ß-Amyloid precursor protein-cleaving enzyme-1 (BACE1) initiates the production of Aß-peptides that form Aß-plaque in Alzheimer's disease. METHODS: Reportedly, acute insulin treatment in normal mice, and hyperinsulinemia in high-fat-fed (HFF) obese/diabetic mice, increase BACE1 activity and levels of Aß-peptides and phospho- -thr-231-tau in the brain; moreover, these effects are blocked by PKC-λ/ι inhibitors. However, as chemical inhibitors may affect unsuspected targets, we presently used knockout methodology to further examine PKC-λ/ι requirements. We found that total-body heterozygous PKC-λ knockout reduced acute stimulatory effects of insulin and chronic effects of hyperinsulinemia in HFF/obese/diabetic mice, on brain PKC-λ activity and production of Aß1-40/42 and phospho-thr-231-tau. This protection in HFF mice may reflect that hepatic PKC-λ haploinsufficiency prevents the development of glucose intolerance and hyperinsulinemia. RESULTS: On the other hand, heterozygous knockout of PKC-λ markedly reduced brain levels of BACE1 protein and mRNA, and this may reflect diminished activation of nuclear factor kappa-B (NFκB), which is activated by PKC-λ and increases BACE1 and proinflammatory cytokine transcription. Accordingly, whereas intravenous administration of aPKC inhibitor diminished aPKC activity and BACE1 levels by 50% in the brain and 90% in the liver, nasally-administered inhibitor reduced aPKC activity and BACE1 mRNA and protein levels by 50-70% in the brain while sparing the liver. Additionally, 24-hour insulin treatment in cultured human-derived neurons increased NFκB activity and BACE1 levels, and these effects were blocked by various PKC-λ/ι inhibitors. CONCLUSION: PKC-λ/ι controls NFκB activity and BACE1 expression; PKC-λ/ι inhibitors may be used nasally to target brain PKC-λ/ι or systemically to block both liver and brain PKC-λ/ι, to regulate NFκB-dependent BACE1 and proinflammatory cytokine expression.


Subject(s)
Alzheimer Disease , Amyloid Precursor Protein Secretases , Aspartic Acid Endopeptidases , Diabetes Mellitus, Experimental , NF-kappa B , Protein Kinase C , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Mice , NF-kappa B/metabolism , Protein Kinase C/genetics
6.
Cell Rep ; 37(8): 110054, 2021 11 23.
Article in English | MEDLINE | ID: mdl-34818553

ABSTRACT

We report that atypical protein kinase Cι (PKCι) is an oncogenic driver of glioblastoma (GBM). Deletion or inhibition of PKCι significantly impairs tumor growth and prolongs survival in murine GBM models. GBM cells expressing elevated PKCι signaling are sensitive to PKCι inhibitors, whereas those expressing low PKCι signaling exhibit active SRC signaling and sensitivity to SRC inhibitors. Resistance to the PKCι inhibitor auranofin is associated with activated SRC signaling and response to a SRC inhibitor, whereas resistance to a SRC inhibitor is associated with activated PKCι signaling and sensitivity to auranofin. Interestingly, PKCι- and SRC-dependent cells often co-exist in individual GBM tumors, and treatment of GBM-bearing mice with combined auranofin and SRC inhibitor prolongs survival beyond either drug alone. Thus, we identify PKCι and SRC signaling as distinct therapeutic vulnerabilities that are directly translatable into an improved treatment for GBM.


Subject(s)
Glioblastoma/genetics , Glioblastoma/metabolism , Isoenzymes/metabolism , Protein Kinase C/metabolism , Animals , Carcinogenesis/genetics , Cell Line, Tumor , Disease Models, Animal , Gene Expression/genetics , Gene Expression Regulation, Neoplastic/genetics , Glioblastoma/classification , Humans , Isoenzymes/genetics , Mice , Oncogenes/genetics , Protein Kinase C/genetics , Protein Kinase C/physiology , Signal Transduction/physiology
7.
J Clin Invest ; 131(16)2021 08 16.
Article in English | MEDLINE | ID: mdl-34255743

ABSTRACT

In view of emerging drug-resistant tuberculosis (TB), host-directed adjunct therapies are urgently needed to improve treatment outcomes with currently available anti-TB therapies. One approach is to interfere with the formation of lipid-laden "foamy" macrophages in the host, as they provide a nutrient-rich host cell environment for Mycobacterium tuberculosis (Mtb). Here, we provide evidence that Wnt family member 6 (WNT6), a ligand of the evolutionarily conserved Wingless/Integrase 1 (WNT) signaling pathway, promotes foam cell formation by regulating key lipid metabolic genes including acetyl-CoA carboxylase 2 (ACC2) during pulmonary TB. Using genetic and pharmacological approaches, we demonstrated that lack of functional WNT6 or ACC2 significantly reduced intracellular triacylglycerol (TAG) levels and Mtb survival in macrophages. Moreover, treatment of Mtb-infected mice with a combination of a pharmacological ACC2 inhibitor and the anti-TB drug isoniazid (INH) reduced lung TAG and cytokine levels, as well as lung weights, compared with treatment with INH alone. This combination also reduced Mtb bacterial numbers and the size of mononuclear cell infiltrates in livers of infected mice. In summary, our findings demonstrate that Mtb exploits WNT6/ACC2-induced storage of TAGs in macrophages to facilitate its intracellular survival, a finding that opens new perspectives for host-directed adjunctive treatment of pulmonary TB.


Subject(s)
Acetyl-CoA Carboxylase/metabolism , Macrophages/metabolism , Macrophages/microbiology , Mycobacterium tuberculosis/metabolism , Mycobacterium tuberculosis/pathogenicity , Proto-Oncogene Proteins/metabolism , Triglycerides/metabolism , Wnt Proteins/metabolism , Acetyl-CoA Carboxylase/antagonists & inhibitors , Animals , Antitubercular Agents/administration & dosage , Enzyme Inhibitors/administration & dosage , Foam Cells/metabolism , Host Microbial Interactions/drug effects , Host Microbial Interactions/physiology , Humans , Isoniazid/administration & dosage , Lung/drug effects , Lung/metabolism , Lung/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mycobacterium tuberculosis/drug effects , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Signal Transduction/drug effects , Tuberculosis, Pulmonary/drug therapy , Tuberculosis, Pulmonary/metabolism , Tuberculosis, Pulmonary/microbiology , Wnt Proteins/deficiency , Wnt Proteins/genetics
8.
Life Sci Alliance ; 4(8)2021 08.
Article in English | MEDLINE | ID: mdl-34145024

ABSTRACT

Members of the protein kinase D (PKD) family (PKD1, 2, and 3) integrate hormonal and nutritional inputs to regulate complex cellular metabolism. Despite the fact that a number of functions have been annotated to particular PKDs, their molecular targets are relatively poorly explored. PKD3 promotes insulin sensitivity and suppresses lipogenesis in the liver of animals fed a high-fat diet. However, its substrates are largely unknown. Here we applied proteomic approaches to determine PKD3 targets. We identified more than 300 putative targets of PKD3. Furthermore, biochemical analysis revealed that PKD3 regulates cAMP-dependent PKA activity, a master regulator of the hepatic response to glucagon and fasting. PKA regulates glucose, lipid, and amino acid metabolism in the liver, by targeting key enzymes in the respective processes. Among them the PKA targets phenylalanine hydroxylase (PAH) catalyzes the conversion of phenylalanine to tyrosine. Consistently, we showed that PKD3 is activated by glucagon and promotes glucose and tyrosine levels in hepatocytes. Therefore, our data indicate that PKD3 might play a role in the hepatic response to glucagon.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Glucagon/pharmacology , Hepatocytes/cytology , Protein Kinase C/metabolism , Proteomics/methods , Animals , Cells, Cultured , Fasting , Glucose/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Mice , Phenylalanine Hydroxylase/metabolism , Phosphorylation , Primary Cell Culture , Protein Interaction Maps , Tyrosine/metabolism
9.
iScience ; 24(1): 102019, 2021 Jan 22.
Article in English | MEDLINE | ID: mdl-33521594

ABSTRACT

Doublecortin-like kinase 1 (DCLK1)-positive pancreatic cancer stem cells develop at a precancerous stage and may contribute to the lack of efficacy of pancreatic cancer therapy. Although PanIN cells express oncogenic KRas and have an increased activity of epidermal growth factor receptor (EGFR), we demonstrate that, in DCLK1+ PanIN cells, EGFR signaling is not propagated to the nucleus. Mimicking blockage of EGFR with erlotinib in PanIN organoid culture or in p48cre;KrasG12D mice led to a significant increase in DCLK1+ PanIN cells. As a mechanism of how EGFR inhibition leads to formation of DCLK1+ cells, we identify an increase in hydrogen peroxide contributing to activation of Protein Kinase D1 (PKD1). Active PKD1 then drives stemness and abundance of DCLK1+ cells in lesions. Our data suggest a signaling mechanism that leads to the development of DCLK1+ pancreatic cancer stem cells, which can be exploited to target this population in potential therapeutic approaches.

10.
Stem Cells ; 39(6): 819-830, 2021 06.
Article in English | MEDLINE | ID: mdl-33539629

ABSTRACT

Survival of chronic lymphocytic leukemia (CLL) cells critically depends on the support of an adapted and therefore appropriate tumor microenvironment. Increasing evidence suggests that B-cell receptor-associated kinases such as protein kinase C-ß (PKCß) or Lyn kinase are essential for the formation of a microenvironment supporting leukemic growth. Here, we describe the impact of PKCß on the glucose metabolism in bone marrow stromal cells (BMSC) upon CLL contact. BMSC get activated by CLL contact expressing stromal PKCß that diminishes mitochondrial stress and apoptosis in CLL cells by stimulating glucose uptake. In BMSC, the upregulation of PKCß results in increased mitochondrial depolarization and leads to a metabolic switch toward oxidative phosphorylation. In addition, PKCß-deficient BMSC regulates the expression of Hnf1 promoting stromal insulin signaling after CLL contact. Our data suggest that targeting PKCß and the glucose metabolism of the leukemic niche could be a potential therapeutic strategy to overcome stroma-mediated drug resistance.


Subject(s)
Bone Marrow Cells/metabolism , Glucose/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Protein Kinase C beta/metabolism , Bone Marrow Cells/drug effects , Cell Communication/drug effects , Cell Survival/drug effects , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Protein Kinase C beta/drug effects , Protein Kinase Inhibitors/pharmacology , Tumor Microenvironment/drug effects
11.
Adv Biol Regul ; 78: 100756, 2020 12.
Article in English | MEDLINE | ID: mdl-32992232

ABSTRACT

PKC isozymes have been put in place as oncoproteins since the discovery that they can function as receptors for potent tumor-promoting phorbol esters in the 1980s. Despite nearly two decades of research, a clear in vivo proof of that concept was missing. The availability of so-called knock out mouse lines of individual PKC genes provided a tool to investigate isozyme specific in vivo functions in the context of tumor initiation, development and progression. This review aims to provide a limited overview of how the application of these mouse lines in combination with a cancer mouse model helped to understand PKC's in vivo function during tumorigenesis. The focus of this review will be on skin, colon and lung cancer.


Subject(s)
Carcinogenesis , Isoenzymes/metabolism , Protein Kinase C/metabolism , Animals , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Disease Models, Animal , Isoenzymes/genetics , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Mice, Knockout , Protein Kinase C/genetics
12.
Sci Transl Med ; 12(526)2020 01 15.
Article in English | MEDLINE | ID: mdl-31941829

ABSTRACT

Overcoming drug resistance remains a key challenge to cure patients with acute and chronic B cell malignancies. Here, we describe a stromal cell-autonomous signaling pathway, which contributes to drug resistance of malignant B cells. We show that protein kinase C (PKC)-ß-dependent signals from bone marrow-derived stromal cells markedly decrease the efficacy of cytotoxic therapies. Conversely, small-molecule PKC-ß inhibitors antagonize prosurvival signals from stromal cells and sensitize tumor cells to targeted and nontargeted chemotherapy, resulting in enhanced cytotoxicity and prolonged survival in vivo. Mechanistically, stromal PKC-ß controls the expression of adhesion and matrix proteins, required for activation of phosphoinositide 3-kinases (PI3Ks) and the extracellular signal-regulated kinase (ERK)-mediated stabilization of B cell lymphoma-extra large (BCL-XL) in tumor cells. Central to the stroma-mediated drug resistance is the PKC-ß-dependent activation of transcription factor EB, regulating lysosome biogenesis and plasma membrane integrity. Stroma-directed therapies, enabled by direct inhibition of PKC-ß, enhance the effectiveness of many antileukemic therapies.


Subject(s)
Protein Kinase C beta/metabolism , Apoptosis/drug effects , Drug Resistance, Neoplasm/genetics , Humans , Phosphorylation/drug effects , Signal Transduction/genetics , Signal Transduction/physiology , Stromal Cells/drug effects , Stromal Cells/metabolism , Tumor Cells, Cultured
14.
Cell Commun Signal ; 17(1): 141, 2019 11 06.
Article in English | MEDLINE | ID: mdl-31694643

ABSTRACT

BACKGROUND: Protein kinase C θ has been established as an important signaling intermediate in T-effector-cell activation and survival pathways by controlling activity of the key transcription factors NF-κB and NFAT. Previous studies identified an activation-induced auto-phosphorylation site at Thr-219, located between the tandem C1 domains of the regulatory fragment in PKCθ, as a structural requirement for its correct membrane translocation and the subsequent transactivation of downstream signals leading to IL-2 production in a human T cell line. METHODS: The present work aimed to define the role of this phosphorylation switch on PKCθ in a physiological context through a homozygous T219A knockin mouse strain. T cell activation was analyzed by H3-thymidine uptake (proliferative response), qRT-PCR and luminex measurements (cytokine production). NFAT and NF-κB transactivation responses were estimated by Gel mobility shift and Alpha Screen assays. Frequencies of T cell subsets were analyzed by flow cytometry. RESULTS: Despite a normal T cell development, in vitro activated effector T cells clearly revealed a requirement of Thr-219 phosphorylation site on PKCθ for a transactivation of NF-κB and NFAT transcription factors and, subsequently, robust IL-2 and IFN-γ expression. CONCLUSION: This phenotype is reminiscent of the PKCθ knockout T cells, physiologically validating that this (p) Thr-219 auto-phosphorylation site indeed critically regulates PKCθ function in primary mouse T cells.


Subject(s)
Gene Knock-In Techniques , Phenotype , Protein Kinase C-theta/genetics , Protein Kinase C-theta/metabolism , Animals , Cytokines/metabolism , Mice , T-Lymphocytes/cytology , T-Lymphocytes/metabolism
15.
Sci Signal ; 12(593)2019 08 06.
Article in English | MEDLINE | ID: mdl-31387939

ABSTRACT

Hepatic activation of protein kinase C (PKC) isoforms by diacylglycerol (DAG) promotes insulin resistance and contributes to the development of type 2 diabetes (T2D). The closely related protein kinase D (PKD) isoforms act as effectors for DAG and PKC. Here, we showed that PKD3 was the predominant PKD isoform expressed in hepatocytes and was activated by lipid overload. PKD3 suppressed the activity of downstream insulin effectors including the kinase AKT and mechanistic target of rapamycin complex 1 and 2 (mTORC1 and mTORC2). Hepatic deletion of PKD3 in mice improved insulin-induced glucose tolerance. However, increased insulin signaling in the absence of PKD3 promoted lipogenesis mediated by SREBP (sterol regulatory element-binding protein) and consequently increased triglyceride and cholesterol content in the livers of PKD3-deficient mice fed a high-fat diet. Conversely, hepatic-specific overexpression of a constitutively active PKD3 mutant suppressed insulin-induced signaling and caused insulin resistance. Our results indicate that PKD3 provides feedback on hepatic lipid production and suppresses insulin signaling. Therefore, manipulation of PKD3 activity could be used to decrease hepatic lipid content or improve hepatic insulin sensitivity.


Subject(s)
Cholesterol/biosynthesis , Hepatocytes/metabolism , Insulin/metabolism , Protein Kinase C/metabolism , Signal Transduction , Triglycerides/biosynthesis , Animals , Cholesterol/genetics , Insulin/genetics , Lipogenesis/genetics , Mice , Mice, Transgenic , Protein Kinase C/genetics , Triglycerides/genetics
16.
Cancer Cell ; 36(2): 156-167.e7, 2019 08 12.
Article in English | MEDLINE | ID: mdl-31378680

ABSTRACT

We report that mouse LSL-KrasG12D;Trp53fl/fl (KP)-mediated lung adenocarcinoma (LADC) tumorigenesis can proceed through both PKCι-dependent and PKCι-independent pathways. The predominant pathway involves PKCι-dependent transformation of bronchoalveolar stem cells (BASCs). However, KP mice harboring conditional knock out Prkci alleles (KPI mice) develop LADC tumors through PKCι-independent transformation of Axin2+ alveolar type 2 (AT2) stem cells. Transformed growth of KPI, but not KP, tumors is blocked by Wnt pathway inhibition in vitro and in vivo. Furthermore, a KPI-derived genomic signature predicts sensitivity of human LADC cells to Wnt inhibition, and identifies a distinct subset of primary LADC tumors exhibiting a KPI-like genotype. Thus, LADC can develop through both PKCι-dependent and PKCι-independent pathways, resulting in tumors exhibiting distinct oncogenic signaling and pharmacologic vulnerabilities.


Subject(s)
Adenocarcinoma of Lung/enzymology , Cell Transformation, Neoplastic/metabolism , Genes, ras , Isoenzymes/metabolism , Lung Neoplasms/enzymology , Protein Kinase C/metabolism , Tumor Suppressor Protein p53/metabolism , Wnt Signaling Pathway , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Isoenzymes/deficiency , Isoenzymes/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Protein Kinase C/deficiency , Protein Kinase C/genetics , Protein Kinase Inhibitors/pharmacology , Tumor Burden , Tumor Cells, Cultured , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , beta Catenin/genetics , beta Catenin/metabolism
17.
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
18.
Nat Neurosci ; 21(8): 1027-1037, 2018 08.
Article in English | MEDLINE | ID: mdl-30013171

ABSTRACT

The protein kinase C (PKC) enzymes have long been established as critical for synaptic plasticity. However, it is unknown whether Ca2+-dependent PKC isozymes are activated in dendritic spines during plasticity and, if so, how this synaptic activity is encoded by PKC. Here, using newly developed, isozyme-specific sensors, we demonstrate that classical isozymes are activated to varying degrees and with distinct kinetics. PKCα is activated robustly and rapidly in stimulated spines and is the only isozyme required for structural plasticity. This specificity depends on a PDZ-binding motif present only in PKCα. The activation of PKCα during plasticity requires both NMDA receptor Ca2+ flux and autocrine brain-derived neurotrophic factor (BDNF)-TrkB signaling, two pathways that differ vastly in their spatiotemporal scales of signaling. Our results suggest that, by integrating these signals, PKCα combines a measure of recent, nearby synaptic plasticity with local synaptic input, enabling complex cellular computations such as heterosynaptic facilitation of plasticity necessary for efficient hippocampus-dependent learning.


Subject(s)
Autocrine Communication/physiology , Brain-Derived Neurotrophic Factor/physiology , Calcium Signaling/physiology , Neuronal Plasticity/physiology , Protein Kinase C-alpha/physiology , Animals , Autocrine Communication/genetics , Brain-Derived Neurotrophic Factor/genetics , Calcium Signaling/genetics , Dendritic Spines , Enzyme Activation , Hippocampus/physiology , Isoenzymes , Kinetics , Learning/physiology , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Kinase C-alpha/genetics , Receptors, N-Methyl-D-Aspartate/metabolism
19.
Immunity ; 48(6): 1144-1159.e5, 2018 06 19.
Article in English | MEDLINE | ID: mdl-29884460

ABSTRACT

PKCß-null (Prkcb-/-) mice are severely immunodeficient. Here we show that mice whose B cells lack PKCß failed to form germinal centers and plasma cells, which undermined affinity maturation and antibody production in response to immunization. Moreover, these mice failed to develop plasma cells in response to viral infection. At the cellular level, we have shown that Prkcb-/- B cells exhibited defective antigen polarization and mTORC1 signaling. While altered antigen polarization impaired antigen presentation and likely restricted the potential of GC development, defective mTORC1 signaling impaired metabolic reprogramming, mitochondrial remodeling, and heme biosynthesis in these cells, which altogether overwhelmingly opposed plasma cell differentiation. Taken together, our study reveals mechanistic insights into the function of PKCß as a key regulator of B cell polarity and metabolic reprogramming that instructs B cell fate.


Subject(s)
B-Lymphocytes/immunology , Cell Differentiation/immunology , Lymphocyte Activation/immunology , Plasma Cells/immunology , Protein Kinase C beta/immunology , Animals , Heme/biosynthesis , Mice , Mice, Knockout , Mitochondria/immunology , Mitochondria/metabolism , Plasma Cells/cytology
20.
Oncogene ; 37(37): 5136-5146, 2018 09.
Article in English | MEDLINE | ID: mdl-29789715

ABSTRACT

The polarity proteins Par3 and aPKC are key regulators of processes altered in cancer. Par3/aPKC are thought to dynamically interact with Par6 but increasing evidence suggests that aPKC and Par3 also exert complex-independent functions. Whereas aPKCλ serves as tumor promotor, Par3 can either promote or suppress tumorigenesis. Here we asked whether and how Par3 and aPKCλ genetically interact to control two-stage skin carcinogenesis. Epidermal loss of Par3, aPKCλ, or both, strongly reduced tumor multiplicity and increased latency but inhibited invasion to similar extents, indicating that Par3 and aPKCλ function as a complex to promote tumorigenesis. Molecularly, Par3/aPKCλ cooperate to promote Akt, ERK and NF-κB signaling during tumor initiation to sustain growth, whereas aPKCλ dominates in promoting survival. In the inflammatory tumorigenesis phase Par3/aPKCλ cooperate to drive Stat3 activation and hyperproliferation. Unexpectedly, the reduced inflammatory signaling did not alter carcinogen-induced immune cell numbers but reduced IL-4 Receptor-positive stromal macrophage numbers in all mutant mice, suggesting that epidermal aPKCλ and Par3 promote a tumor-permissive environment. Importantly, aPKCλ also serves a distinct, carcinogen-independent role in controlling skin immune cell homeostasis. Collectively, our data demonstrates that Par3 and aPKCλ cooperate to promote skin tumor initiation and progression, likely through sustaining growth, survival, and inflammatory signaling.


Subject(s)
Carcinogenesis/genetics , Cell Adhesion Molecules/genetics , Protein Kinase C/genetics , Skin Neoplasms/genetics , Skin/pathology , Adaptor Proteins, Signal Transducing , Animals , Carcinogenesis/pathology , Cell Cycle Proteins , Cell Polarity/genetics , Cell Proliferation/genetics , Disease Models, Animal , Inflammation/pathology , Macrophages/pathology , Mice , Mice, Knockout , NF-kappa B/genetics , Receptors, Interleukin-4/genetics , STAT3 Transcription Factor/genetics , Signal Transduction/genetics , Skin Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...