Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters











Database
Language
Publication year range
1.
Biochem J ; 476(13): 1943-1954, 2019 07 09.
Article in English | MEDLINE | ID: mdl-31208986

ABSTRACT

Metabolic remodeling plays an essential role in the pathophysiology of heart failure (HF). Many studies have shown that the disruption of phosphoinositide-dependent protein kinase-1 (PDK1) caused severe and lethal HF; however, the metabolic pattern of PDK1 deletion remains ambiguous. 1H nuclear magnetic resonance-based metabolomics was applied to explore the altered metabolic pattern in Pdk1-deficient mice. Principle component analysis showed significant separation as early as 4 weeks of age, and dysfunction of metabolism precedes a morphological change in Pdk1-deficient mice. A time trajectory plot indicated that disturbed metabolic patterns were related to the pathological process of the HF in Pdk1-deficient mice, rather than the age of mice. Metabolic profiles demonstrated significantly increased levels of acetate, glutamate, glutamine, and O-phosphocholine in Pdk1 deletion mice. Levels of lactate, alanine, glycine, taurine, choline, fumarate, IMP, AMP, and ATP were significantly decreased compared with controls. Furthermore, PDK1 knockdown decreased the oxygen consumption rate in H9C2 cells as determined using a Seahorse XF96 Analyzer. These findings imply that the disruption of metabolism and impaired mitochondrial activity might be involved in the pathogenesis of HF with PDK1 deletion.


Subject(s)
3-Phosphoinositide-Dependent Protein Kinases/deficiency , Heart Failure/metabolism , Mitochondria, Heart/metabolism , Myocytes, Cardiac/metabolism , Oxygen Consumption , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/deficiency , Animals , Heart Failure/genetics , Heart Failure/pathology , Mice , Mice, Knockout , Mitochondria, Heart/genetics , Mitochondria, Heart/pathology , Myocytes, Cardiac/pathology
2.
J Thromb Haemost ; 16(6): 1211-1225, 2018 06.
Article in English | MEDLINE | ID: mdl-29575487

ABSTRACT

Essentials Phosphoinositide 3-kinase and MAPK pathways crosstalk via PDK1. PDK1 is required for adenosine diphosphate-induced platelet activation and thromboxane generation. PDK1 regulates RAF proto-oncogene Ser/Thr kinase (Raf1) activation in the MAPK pathway. Genetic ablation of PDK1 protects against platelet-dependent thrombosis in vivo. SUMMARY: Background Platelets are dynamic effector cells with functions that span hemostatic, thrombotic and inflammatory continua. Phosphoinositide-dependent protein kinase 1 (PDK1) regulates protease-activated receptor 4-induced platelet activation and thrombus formation through glycogen synthase kinase3ß. However, whether PDK1 also signals through the ADP receptor and its functional importance in vivo remain unknown. Objective To establish the mechanism of PDK1 in ADP-induced platelet activation and thrombosis. Methods We assessed the role of PDK1 on 2MeSADP-induced platelet activation by measuring aggregation, thromboxane generation and phosphorylation events in the presence of BX-795, which inhibits PDK1, or by using platelet-specific PDK1 knockout mice and performing western blot analysis. PDK1 function in thrombus formation was assessed with an in vivo pulmonary embolism model. Results PDK1 inhibition with BX-795 reduced 2-methylthio-ADP (2MeSADP)-induced aggregation of human and murine platelets by abolishing thromboxane generation. Similar results were observed in pdk1-/- mice. PDK1 was also necessary for the phosphorylation of mitogen-activated protein kinase kinase 1/2 (MEK1/2), extracellular signal-regulated kinase 1/2, and cytosolic phospholipase A2, indicating that PDK1 regulates an upstream kinase in the mitogen-activated protein kinase (MAPK) pathway. We next determined that this upstream kinase is Raf-1, a serine/threonine kinase that is necessary for the phosphorylation of MEK1/2, as pharmacological inhibition and genetic ablation of PDK1 were sufficient to prevent Raf1 phosphorylation. Furthermore, in vivo inhibition or genetic ablation of PDK1 protected mice from collagen/epinephrine-induced pulmonary embolism. Conclusion PDK1 governs thromboxane generation and thrombosis in platelets that are stimulated with 2MeSADP by regulating activation of the MAPK pathway.


Subject(s)
3-Phosphoinositide-Dependent Protein Kinases/metabolism , Blood Platelets/enzymology , Mitogen-Activated Protein Kinases/blood , Platelet Aggregation/drug effects , Proto-Oncogene Proteins c-raf/blood , Pulmonary Embolism/enzymology , Thrombosis/enzymology , Thromboxanes/blood , 3-Phosphoinositide-Dependent Protein Kinases/antagonists & inhibitors , 3-Phosphoinositide-Dependent Protein Kinases/blood , 3-Phosphoinositide-Dependent Protein Kinases/deficiency , 3-Phosphoinositide-Dependent Protein Kinases/genetics , Animals , Blood Platelets/drug effects , Disease Models, Animal , Humans , Mice, Knockout , Phosphorylation , Platelet Aggregation Inhibitors/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Mas , Pulmonary Embolism/blood , Pulmonary Embolism/genetics , Pulmonary Embolism/prevention & control , Pyrimidines/pharmacology , Signal Transduction , Thiophenes/pharmacology , Thrombosis/blood , Thrombosis/genetics , Thrombosis/prevention & control
3.
Arterioscler Thromb Vasc Biol ; 36(8): 1507-16, 2016 08.
Article in English | MEDLINE | ID: mdl-27339458

ABSTRACT

OBJECTIVE: Activation of platelets by subendothelial collagen results in an increase of cytosolic Ca(2+) concentration ([Ca(2+)]i) and is followed by platelet activation and thrombus formation that may lead to vascular occlusion. The present study determined the role of phosphoinositide-dependent protein kinase 1 (PDK1) in collagen-dependent platelet Ca(2+) signaling and ischemic stroke in vivo. APPROACH AND RESULTS: Platelet activation with collagen receptor glycoprotein VI agonists collagen-related peptide or convulxin resulted in a significant increase in PDK1 activity independent of second-wave signaling. PDK1 deficiency was associated with reduced platelet phospholipase Cγ2-dependent inositol-1,4,5-trisphosphate production and intracellular [Ca(2+)]i in response to stimulation with collagen-related peptide or convulxin. The defective increase of [Ca(2+)]i resulted in a substantial defect in activation-dependent platelet secretion and aggregation on collagen-related peptide stimulation. Furthermore, Rac1 activation and spreading, adhesion to collagen, and thrombus formation under high arterial shear rates were significantly diminished in PDK1-deficient platelets. Mice with PDK1-deficient platelets were protected against arterial thrombotic occlusion after FeCl3-induced mesenteric arterioles injury and ischemic stroke in vivo. These mice had significantly reduced brain infarct volumes, with a significantly increased survival of 7 days after transient middle cerebral artery occlusion without increase of intracerebral hemorrhage. Tail bleeding time was prolonged in pdk1(-/-) mice, reflecting an important role of PDK1 in primary hemostasis. CONCLUSIONS: PDK1 is required for Ca(2+)-dependent platelet activation on stimulation of collagen receptor glycoprotein VI, arterial thrombotic occlusion, and ischemic stroke in vivo.


Subject(s)
3-Phosphoinositide-Dependent Protein Kinases/metabolism , Blood Platelets/enzymology , Calcium Signaling , Collagen/metabolism , Infarction, Middle Cerebral Artery/enzymology , Platelet Activation , Thrombosis/enzymology , 3-Phosphoinositide-Dependent Protein Kinases/deficiency , 3-Phosphoinositide-Dependent Protein Kinases/genetics , Animals , Disease Models, Animal , Genetic Predisposition to Disease , Infarction, Middle Cerebral Artery/blood , Infarction, Middle Cerebral Artery/pathology , Inositol 1,4,5-Trisphosphate/blood , Mice, Knockout , Neuropeptides/blood , Phenotype , Phospholipase C gamma/blood , Platelet Adhesiveness , Platelet Aggregation , Platelet Membrane Glycoproteins/deficiency , Platelet Membrane Glycoproteins/genetics , Thrombosis/blood , Thrombosis/pathology , Time Factors , rac1 GTP-Binding Protein/blood
4.
J Exp Med ; 212(2): 253-65, 2015 Feb 09.
Article in English | MEDLINE | ID: mdl-25624444

ABSTRACT

E4BP4, a circadian protein, is indispensable for NK cell development. It remains largely unknown which signal is required to induce E4BP4 expression and what effects it has during NK cell differentiation. Here, we reveal that PDK1, a kinase upstream of mTOR, connects IL-15 signaling to E4BP4. Early deletion of PDK1 caused a severe loss of NK cells and compromised antitumor activity in vivo. PDK1-deficient NK cells displayed much weaker IL-15-induced mTOR activation and E4BP4 induction, as well as remarkable reduction in CD122, a receptor subunit specifying NK cell responsiveness to IL-15. The phenotypes were partially reversible by ectopic expression of E4BP4 or bypassed activation of mTOR. We also determined that PDK1-mediated metabolic signaling was dispensable for NK cell terminal maturation and survival. Thus, we identify a role for PDK1 signaling as a key mediator in regulating E4BP4 expression during early NK cell development. Our findings underscore the importance of IL-15 self-responsiveness through a positive feedback loop that involves PDK1-mTOR-E4BP4-CD122 signaling.


Subject(s)
3-Phosphoinositide-Dependent Protein Kinases/metabolism , Basic-Leucine Zipper Transcription Factors/genetics , Gene Expression Regulation , Interleukin-15/metabolism , Killer Cells, Natural/metabolism , 3-Phosphoinositide-Dependent Protein Kinases/deficiency , 3-Phosphoinositide-Dependent Protein Kinases/genetics , Animals , Basic-Leucine Zipper Transcription Factors/metabolism , Cell Survival/drug effects , Gene Deletion , Gene Expression , Gene Expression Regulation/drug effects , Interleukin-15/pharmacology , Interleukin-2 Receptor beta Subunit/metabolism , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Mice, Transgenic , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL