Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2023 Nov 29.
Article in English | MEDLINE | ID: mdl-38077058

ABSTRACT

Hematopoietic stem cell (HSC) transplantation using umbilical cord blood (UCB) is a potentially life-saving treatment for leukemia and bone marrow failure but is limited by the low number of HSCs in UCB. The loss of HSCs after ex vivo manipulation is also a major obstacle to gene editing for inherited blood disorders. HSCs require a low rate of translation to maintain their capacity for self-renewal, but hematopoietic cytokines used to expand HSCs stimulate protein synthesis and impair long-term self-renewal. We previously described cytokine-free conditions that maintain but do not expand human and mouse HSCs ex vivo. Here we performed a high throughput screen and identified translation inhibitors that allow ex vivo expansion of human HSCs while minimizing cytokine exposure. Transplantation assays show a ~5-fold expansion of long-term HSCs from UCB after one week of culture in low cytokine conditions. Single cell transcriptomic analysis demonstrates maintenance of HSCs expressing mediators of the unfolded protein stress response, further supporting the importance of regulated proteostasis in HSC maintenance and expansion. This expansion method maintains and expands human HSCs after CRISPR/Cas9 editing of the BCL11A+58 enhancer, overcoming a major obstacle to ex vivo gene correction for human hemoglobinopathies.

2.
Blood Adv ; 3(4): 575-587, 2019 02 26.
Article in English | MEDLINE | ID: mdl-30787021

ABSTRACT

Phosphatidylinositol 3-kinase is an important signaling molecule that, once activated, leads to the generation of phosphatidylinositol (3,4,5)-trisphosphate (PIP3). We performed a proteomic screen to identify PIP3-interacting proteins in human platelets. Among these proteins, we found engulfment and cell motility 1 (ELMO1), a scaffold protein with no catalytic activity. ELMO1 is expressed in platelets and interacts with active RhoG. However, the function of ELMO1 in platelets is not known. The focus of this study was to determine the function of ELMO1 in platelets utilizing ELMO1-/- mice. Platelet aggregation, granule secretion, integrin αIIbß3 activation, and thromboxane generation were enhanced in ELMO1-/- platelets in response to glycoprotein VI (GPVI) agonists but unaltered when a protease-activated receptor 4 agonist was used. The kinetics of spreading on immobilized fibrinogen was enhanced in ELMO1-/- platelets compared with wild-type (WT) littermate controls. This suggests that ELMO1 plays a role downstream of the GPVI and integrin αIIbß3 pathway. Furthermore, whole blood from ELMO1-/- mice perfused over collagen exhibited enhanced thrombus formation compared with WT littermate controls. ELMO1-/- mice showed reduced survival compared with control following pulmonary embolism. ELMO1-/- mice also exhibited a shorter time to occlusion using the ferric-chloride injury model and reduced bleeding times compared with WT littermate controls. These results indicate that ELMO1 plays an important role in hemostasis and thrombosis in vivo. RhoG activity was enhanced in ELMO1-/- murine platelets compared with WT littermate controls in response to GPVI agonist. Together, these data suggest that ELMO1 negatively regulates GPVI-mediated thrombus formation via RhoG.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Blood Platelets/metabolism , Platelet Aggregation , Adaptor Proteins, Signal Transducing/genetics , Animals , Blood Platelets/cytology , Gene Deletion , Hemostasis , Humans , Mice , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Thrombosis/genetics , Thrombosis/metabolism , Thromboxanes/metabolism
3.
Stem Cell Res Ther ; 9(1): 16, 2018 01 23.
Article in English | MEDLINE | ID: mdl-29361987

ABSTRACT

BACKGROUND: Deciphering molecular mechanisms underlying the division of hematopoietic stem cells (HSCs) and malignant precursors would improve our understanding of the basis of stem cell-fate decisions and oncogenic transformation. METHODS: Using a novel reporter of hematopoietic precursor, Evi1-GFP, we tracked the division of hematopoietic precursors in culture in real time. RESULTS: First, we confirmed that Evi1-GFP is a faithful reporter of HSC activity and identified three dividing patterns of HSCs: symmetric renewal, symmetric differentiation, and asymmetric division. Moreover, we found that the cytokine and growth factor combination (STIF) promotes symmetric renewal, whereas OP9 stromal cells balance symmetric renewal and differentiation of HSCs ex vivo. Interestingly, we found that Tet2 knockout HSCs underwent more symmetric differentiation in culture compared with the wild-type control. Intriguingly, OP9 stromal cells reverse the phenotype of Tet2 knockout HSCs ex vivo. Furthermore, we demonstrated that Tet2 -/- ;Flt3ITD acute myeloid leukemia (AML) precursors primarily underwent symmetric renewal divisions in culture. Mechanistically, we demonstrated that inhibiting DNA methylation can reverse the aberrant division phenotypes of Tet2 -/- and Tet2 -/- ;FLT3ITD precursors, suggesting that abnormal DNA methylation plays an important role in controlling (pre-)leukemic precursor fate decision ex vivo. CONCLUSIONS: Our study exploited a new system to explore the molecular mechanisms of the regulation of benign and malignant hematopoietic precursor division ex vivo. The knowledge learned from these studies will provide new insights into the molecular mechanisms of HSC fate decision and leukemogenesis.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , DNA Methylation/genetics , DNA-Binding Proteins/genetics , Hematopoietic Stem Cells/cytology , Leukemia, Myeloid, Acute/pathology , Proto-Oncogene Proteins/genetics , Animals , Cell Differentiation/genetics , Cell Division/genetics , Dioxygenases , MDS1 and EVI1 Complex Locus Protein/genetics , MDS1 and EVI1 Complex Locus Protein/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Time-Lapse Imaging , fms-Like Tyrosine Kinase 3/genetics
5.
Cancer Res ; 77(16): 4402-4413, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28625976

ABSTRACT

Acute myeloid leukemia (AML) is a malignant hematopoietic disease and the most common type of acute leukemia in adults. The mechanisms underlying drug resistance in AML are poorly understood. Activating mutations in FMS-like tyrosine kinase 3 (FLT3) are the most common molecular abnormality in AML. Quizartinib (AC220) is a potent and selective second-generation inhibitor of FLT3. It is in clinical trials for the treatment of relapsed or refractory FLT3-ITD-positive and -negative AML patients and as maintenance therapy. To understand the mechanisms of drug resistance to AC220, we undertook an unbiased approach with a novel CRISPR-pooled library to screen new genes whose loss of function confers resistance to AC220. We identified SPRY3, an intracellular inhibitor of FGF signaling, and GSK3, a canonical Wnt signaling antagonist, and demonstrated reactivation of downstream FGF/Ras/ERK and Wnt signaling as major mechanisms of resistance to AC220. We confirmed these findings in primary AML patient samples. Expression of SPRY3 and GSK3A was dramatically reduced in AC220-resistant AML samples, and SPRY3-deleted primary AML cells were resistant to AC220. Intriguingly, expression of SPRY3 was greatly reduced in GSK3 knockout AML cells, which positioned SPRY3 downstream of GSK3 in the resistance pathway. Taken together, our study identified novel genes whose loss of function conferred resistance to a selective FLT3 inhibitor, providing new insight into signaling pathways that contribute to acquired resistance in AML. Cancer Res; 77(16); 4402-13. ©2017 AACR.


Subject(s)
Benzothiazoles/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Phenylurea Compounds/pharmacology , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , Apoptosis , Cell Line, Tumor , Cell Proliferation , Child , Clustered Regularly Interspaced Short Palindromic Repeats , Drug Resistance, Neoplasm , Humans , Leukemia, Myeloid, Acute/pathology , Male , Mutation , Protein Kinase Inhibitors/pharmacology , Signal Transduction , Transfection
6.
Blood Adv ; 1(14): 887-898, 2017 Jun 13.
Article in English | MEDLINE | ID: mdl-29296732

ABSTRACT

ATAC-seq provides genome-wide chromatin state in 3 cell types of hematopoietic stem/progenitor cells.Transcription factor cohorts are associated with dynamic changes of open chromatin during the differentiation of LT/ST-HSCs to MPPs.

8.
J Clin Invest ; 126(7): 2419-21, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27322056

ABSTRACT

The secretory protein Dickkopf-1 (DKK-1) is a known Wnt antagonist and has been shown to suppress tumorigenesis in some cancer cells; however, it is also upregulated in many types of cancer and associated with poor prognosis. Wnt-independent mechanisms by which DKK-1 promotes cancer cell proliferation are not well understood. In this issue of the JCI, Kimura and colleagues demonstrate that DKK-1 interacts with cytoskeleton-associated protein 4 (CKAP4) to promote activation of AKT. They show that both DKK-1 and CKAP4 are frequently upregulated in pancreatic and lung cancers. Importantly, targeting this interaction with an anti-CKAP4 antibody prevented tumor formation in murine xenograft models. These results identify a previously unrecognized DKK-1-mediated pathway and suggest CKAP4 as a potential therapeutic target for certain cancers.


Subject(s)
Gene Expression Regulation, Neoplastic , Intercellular Signaling Peptides and Proteins , Animals , Cell Line, Tumor , Cell Proliferation , Cytoskeletal Proteins , Mice
9.
Curr Stem Cell Rep ; 2(4): 379-387, 2016 Dec.
Article in English | MEDLINE | ID: mdl-28503404

ABSTRACT

Wnt signaling plays important roles in stem cell self-renewal and differentiation in adults as well as in embryonic development. Mutations that activate canonical Wnt/ß-catenin signaling also initiate and maintain several cancer states, including colorectal cancer and leukemia, and hence Wnt inhibitors are currently being explored as therapeutic options. In this review, we summarize previous studies and update recent findings on canonical Wnt signaling and its components, as well as their roles in somatic stem cell homeostasis and maintenance of cancer initiating cells.

10.
Thromb Haemost ; 114(3): 558-68, 2015 Aug 31.
Article in English | MEDLINE | ID: mdl-25947062

ABSTRACT

ADP-induced thromboxane generation depends on Src family kinases (SFKs) and is enhanced with pan-protein kinase C (PKC) inhibitors, but it is not clear how these two events are linked. The aim of the current study is to investigate the role of Y311 phosphorylated PKCδ in regulating ADP-induced platelet activation. In the current study, we employed various inhibitors and murine platelets from mice deficient in specific molecules to evaluate the role of PKCδ in ADP-induced platelet responses. We show that, upon stimulation of platelets with 2MeSADP, Y311 on PKCδ is phosphorylated in a P2Y1/Gq and Lyn-dependent manner. By using PKCδ and Lyn knockout murine platelets, we also show that tyrosine phosphorylated PKCδ plays a functional role in mediating 2MeSADP-induced thromboxane generation. 2MeSADP-induced PKCδ Y311 phosphorylation and thromboxane generation were potentiated in human platelets pre-treated with either a pan-PKC inhibitor, GF109203X or a PKC α/ß inhibitor and in PKC α or ß knockout murine platelets compared to controls. Furthermore, we show that PKC α/ß inhibition potentiates the activity of SFK, which further hyper-phosphorylates PKCδ and potentiates thromboxane generation. These results show for the first time that tyrosine phosphorylated PKCδ regulates ADP-induced thromboxane generation independent of its catalytic activity and that classical PKC isoforms α/ß regulate the tyrosine phosphorylation on PKCδ and subsequent thromboxane generation through tyrosine kinase, Lyn, in platelets.


Subject(s)
Adenosine Diphosphate/pharmacology , Blood Platelets/drug effects , Platelet Activation/drug effects , Protein Kinase C/blood , Purinergic P2Y Receptor Agonists/pharmacology , Thromboxane A2/blood , Animals , Blood Platelets/enzymology , Humans , Isoenzymes , Mice, Knockout , Phosphorylation , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/deficiency , Protein Kinase C/genetics , Protein Kinase Inhibitors/pharmacology , Receptors, Purinergic P2Y1/drug effects , Receptors, Purinergic P2Y1/metabolism , Signal Transduction/drug effects , src-Family Kinases/blood , src-Family Kinases/genetics
11.
Platelets ; 26(1): 2-9, 2015.
Article in English | MEDLINE | ID: mdl-24433221

ABSTRACT

Platelets upon activation change their shape, aggregate and secrete alpha and dense granule contents among which ADP acts as a feedback activator. Different Protein Kinase C (PKC) isoforms have specific non-redundant roles in mediating platelet responses including secretion and thrombus formation. Murine platelets lacking specific PKC isoforms have been used to evaluate the isoform specific functions. Novel PKC isoform δ has been shown to play an important role in some pathological processes. Lack of specific inhibitors for PKCδ has restricted analysis of its role in various cells. The current study was carried out to evaluate a novel small molecule PKCδ inhibitor, CGX1037 in platelets. Platelet aggregation, dense granule secretion and western blotting experiments were performed to evaluate CGX1037. In human platelets, CGX1037 inhibited PAR4-mediated phosphorylation on PKD2, a PKCδ-specific substrate. Pre-treatment of human or murine platelets with CGX1037 inhibited PAR4-mediated dense granule secretion whereas it potentiated GPVI-mediated dense granule secretion similar to the responses observed in murine platelets lacking PKCδ· Furthermore, pre-treatment of platelets from PKCδ(-/-) mice with CGX1037 had no significant additive effect on platelet responses suggesting the specificity of CGX1037. Hence, we show that CGX1037 is a selective small molecule inhibitor of PKCδ in platelets.


Subject(s)
Blood Platelets/drug effects , Blood Platelets/metabolism , Protein Kinase C-delta/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Animals , Humans , Inhibitory Concentration 50 , Mice , Mice, Knockout , Phosphorylation/drug effects , Platelet Aggregation/drug effects , Platelet Membrane Glycoproteins/metabolism , Protein Kinase C-delta/genetics , Protein Kinase C-delta/metabolism , Protein Kinase D2 , Protein Kinases/metabolism , Protein Transport , Secretory Vesicles/metabolism
12.
Arterioscler Thromb Vasc Biol ; 34(12): 2579-85, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25359855

ABSTRACT

OBJECTIVE: We previously determined that protein kinase C δ (PKCδ) regulates platelet function. However, the function of PKCδ in megakaryopoiesis is unknown. APPROACH AND RESULTS: Using PKCδ(-/-) and wild-type littermate mice, we found that deficiency of PKCδ caused an increase in white blood cells and platelet counts, as well as in bone marrow and splenic megakaryocytes (P<0.05). Additionally, the megakaryocyte number and DNA content were enhanced in PKCδ(-/-) mouse bone marrow after culturing with exogenous thrombopoietin compared with wild-type (P<0.05). Importantly, thrombopoietin-induced signaling was also altered with PKCδ deletion because both extracellular signal-regulated kinase and Akt308 phosphorylation were heightened in PKCδ(-/-) megakaryocytes compared with wild-type. Finally, PKCδ(-/-) mice recovered faster and had a heightened rebound thrombocytosis after thrombocytopenic challenge. CONCLUSIONS: These data suggest that PKCδ is an important megakaryopoietic protein, which regulates signaling induced by thrombopoietin and represents a potential therapeutic target.


Subject(s)
Megakaryocytes/cytology , Megakaryocytes/enzymology , Protein Kinase C-delta/deficiency , Thrombocytopenia/blood , Thrombocytopenia/enzymology , Thrombopoiesis/physiology , Animals , Bone Marrow Cells/cytology , Extracellular Signal-Regulated MAP Kinases/blood , Leukocyte Count , Mice , Mice, Inbred C57BL , Mice, Knockout , Platelet Count , Protein Kinase C-delta/blood , Protein Kinase C-delta/genetics , Proto-Oncogene Proteins c-akt/blood , RNA, Messenger/blood , RNA, Messenger/genetics , Signal Transduction , Spleen/cytology , Thrombocytopenia/immunology , Thrombopoiesis/genetics , Thrombopoietin/blood , Up-Regulation
13.
J Biol Chem ; 288(47): 34230-34238, 2013 Nov 22.
Article in English | MEDLINE | ID: mdl-24106269

ABSTRACT

We investigated the mechanism of activation and functional role of a hitherto uncharacterized signaling molecule, RhoG, in platelets. We demonstrate for the first time the expression and activation of RhoG in platelets. Platelet aggregation, integrin αIIbß3 activation, and α-granule and dense granule secretion in response to the glycoprotein VI (GPVI) agonists collagen-related peptide (CRP) and convulxin were significantly inhibited in RhoG-deficient platelets. In contrast, 2-MeSADP- and AYPGKF-induced platelet aggregation and secretion were minimally affected in RhoG-deficient platelets, indicating that the function of RhoG in platelets is GPVI-specific. CRP-induced phosphorylation of Syk, Akt, and ERK, but not SFK (Src family kinase), was significantly reduced in RhoG-deficient platelets. CRP-induced RhoG activation was consistently abolished by a pan-SFK inhibitor but not by Syk or PI3K inhibitors. Interestingly, unlike CRP, platelet aggregation and Syk phosphorylation induced by fucoidan, a CLEC-2 agonist, were unaffected in RhoG-deficient platelets. Finally, RhoG(-/-) mice had a significant delay in time to thrombotic occlusion in cremaster arterioles compared with wild-type littermates, indicating the important in vivo functional role of RhoG in platelets. Our data demonstrate that RhoG is expressed and activated in platelets, plays an important role in GPVI-Fc receptor γ-chain complex-mediated platelet activation, and is critical for thrombus formation in vivo.


Subject(s)
GTP Phosphohydrolases/metabolism , Platelet Aggregation , Platelet Membrane Glycoproteins/metabolism , Receptors, Fc/metabolism , Thrombosis/metabolism , rho GTP-Binding Proteins/metabolism , Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/pharmacology , Animals , Anticoagulants/pharmacology , Carrier Proteins/pharmacology , Crotalid Venoms/pharmacology , Female , GTP Phosphohydrolases/genetics , Humans , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Knockout , Oligopeptides/pharmacology , Peptides/pharmacology , Phosphorylation/drug effects , Phosphorylation/genetics , Platelet Membrane Glycoproteins/genetics , Polysaccharides/pharmacology , Protein Kinases , Receptors, Fc/genetics , Thionucleotides/pharmacology , Thrombosis/genetics , Thrombosis/pathology , rho GTP-Binding Proteins/genetics
14.
J Biol Chem ; 288(40): 29160-9, 2013 Oct 04.
Article in English | MEDLINE | ID: mdl-23960082

ABSTRACT

Protein kinase C (PKC) isoforms differentially regulate platelet functional responses downstream of glycoprotein VI (GPVI) signaling, but the role of PKCs regulating upstream effectors such as Syk is not known. We investigated the role of PKC on Syk tyrosine phosphorylation using the pan-PKC inhibitor GF109203X (GFX). GPVI-mediated phosphorylation on Syk Tyr-323, Tyr-352, and Tyr-525/526 was rapidly dephosphorylated, but GFX treatment inhibited this dephosphorylation on Tyr-525/526 in human platelets but not in wild type murine platelets. GFX treatment did not affect tyrosine phosphorylation on FcRγ chain or Src family kinases. Phosphorylation of Lat Tyr-191 and PLCγ2 Tyr-759 was also increased upon treatment with GFX. We evaluated whether secreted ADP is required for such dephosphorylation. Exogenous addition of ADP to GFX-treated platelets did not affect tyrosine phosphorylation on Syk. FcγRIIA- or CLEC-2-mediated Syk tyrosine phosphorylation was also potentiated with GFX in human platelets. Because potentiation of Syk phosphorylation is not observed in murine platelets, PKC-deficient mice cannot be used to identify the PKC isoform regulating Syk phosphorylation. We therefore used selective inhibitors of PKC isoforms. Only PKCß inhibition resulted in Syk hyperphosphorylation similar to that in platelets treated with GFX. This result indicates that PKCß is the isoform responsible for Syk negative regulation in human platelets. In conclusion, we have elucidated a novel pathway of Syk regulation by PKCß in human platelets.


Subject(s)
Blood Platelets/enzymology , Intracellular Signaling Peptides and Proteins/metabolism , Phosphotyrosine/metabolism , Protein-Tyrosine Kinases/metabolism , Adenosine Diphosphate/pharmacology , Animals , Blood Platelets/drug effects , Enzyme Activation/drug effects , Feedback, Physiological/drug effects , Humans , Isoenzymes/metabolism , Lectins, C-Type/metabolism , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Phospholipase C gamma/metabolism , Phosphorylation/drug effects , Platelet Aggregation/drug effects , Protein Kinase C beta/antagonists & inhibitors , Protein Kinase C beta/metabolism , Protein Kinase Inhibitors/pharmacology , Receptors, Fc/metabolism , Syk Kinase
15.
Biochem Pharmacol ; 82(7): 720-7, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21736870

ABSTRACT

Protein kinase D (PKD) is a subfamily of serine/threonine specific family of kinases, comprised of PKD1, PKD2 and PKD3 (PKCµ, PKD2 and PKCv in humans). It is known that PKCs activate PKD, but the relative expression of isoforms of PKD or the specific PKC isoform/s responsible for its activation in platelets is not known. This study is aimed at investigating the pathway involved in activation of PKD in platelets. We show that PKD2 is the major isoform of PKD that is expressed in human as well as murine platelets but not PKD1 or PKD3. PKD2 activation induced by AYPGKF was abolished with a G(q) inhibitor YM-254890, but was not affected by Y-27632, a RhoA/p160ROCK inhibitor, indicating that PKD2 activation is G(q)-, but not G12/13-mediated Rho-kinase dependent. Calcium-mediated signals are also required for activation of PKD2 as dimethyl BAPTA inhibited its phosphorylation. GF109203X, a pan PKC inhibitor abolished PKD2 phosphorylation but Go6976, a classical PKC inhibitor had no effect suggesting that novel PKC isoforms are involved in PKD2 activation. Importantly, Rottlerin, a non-selective PKCδ inhibitor, inhibited AYPGKF-induced PKD2 activation in human platelets. Similarly, AYPGKF- and Convulxin-induced PKD2 phosphorylation was dramatically inhibited in PKCδ-deficient platelets, but not in PKCθ- or PKCɛ-deficient murine platelets compared to that of wild type platelets. Hence, we conclude that PKD2 is a common signaling target downstream of various agonist receptors in platelets and G(q)-mediated signals along with calcium and novel PKC isoforms, in particular, PKCδ activate PKD2 in platelets.


Subject(s)
Blood Platelets/enzymology , Protein Kinase C-delta/physiology , Protein Kinases/metabolism , Animals , CD36 Antigens/physiology , Calcium/blood , Enzyme Activation , GTP-Binding Protein alpha Subunits, Gq-G11/physiology , Humans , In Vitro Techniques , Isoenzymes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphatidylinositol 3-Kinases/physiology , Phosphorylation , Protein Kinase C-delta/genetics , Protein Kinase D2 , Receptors, Thrombin/physiology , Species Specificity , src-Family Kinases/physiology
16.
Biochem Pharmacol ; 82(5): 505-13, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21645497

ABSTRACT

Protein kinase C-zeta (PKCζ), an atypical isoform of the PKC family of protein serine/threonine kinases, is expressed in human platelets. However, the mechanisms of its activation and the regulation of its activity in platelets are not known. We have found that under basal resting conditions, PKCζ has a high phosphorylation status at the activation loop threonine 410 (T410) and the turn motif (autophosphorylation site) threonine 560 (T560), both of which have been shown to be important for its catalytic activity. After stimulation with agonist under stirring conditions, the T410 residue was dephosphorylated in a time- and concentration-dependent manner, while the T560 phosphorylation remained unaffected. The T410 dephosphorylation could be significantly prevented by blocking the binding of fibrinogen to integrin αIIbß3 with an antagonist, SC-57101; or by okadaic acid used at concentrations that inhibits protein serine/threonine phosphatases PP1 and PP2A in vitro. The dephosphorylation of T410 residue on PKCζ was also observed in PP1cγ null murine platelets after agonist stimulation, suggesting that other isoforms of PP1c or another phosphatase could be responsible for this dephosphorylation event. We conclude that human platelets express PKCζ, and it may be constitutively phosphorylated at the activation loop threonine 410 and the turn motif threonine 560 under basal resting conditions, which are differentially dephosphorylated by outside-in signaling. This differential dephosphorylation of PKCζ might be an important regulatory mechanism for platelet functional responses.


Subject(s)
Blood Platelets/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/physiology , Protein Kinase C/metabolism , Animals , Blood Platelets/drug effects , Humans , Mice , Mice, Inbred BALB C , Oligopeptides/pharmacology , Phosphorylation , Platelet Aggregation , Protein Phosphatase 2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...