Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters











Database
Language
Publication year range
1.
Blood Adv ; 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38968150

ABSTRACT

Platelet CLEC-2 is a hemITAM-containing receptor which has a critical role in venous thrombosis, but minimal involvement in haemostasis. CLEC-2 can be blocked by Btk inhibitors. Treatment with ibrutinib is associated with increased bleeding due to off-target inhibition of Src family kinases (SFKs). Patients with X-linked agammaglobulinemia (XLA) who lack Btk however do not bleed, suggesting selective Btk inhibition is a viable antithrombotic strategy. We assessed the effects of selective Btk inhibitors PRN1008 (rilzabrutinib) and PRN473 on platelet signalling and function mediated by CLEC-2 and GPVI. We used healthy donor and XLA platelets to determine off-target inhibitor effects. Inferior vena cava (IVC) stenosis and Salmonella infection mouse models were used to assess antithrombotic effects of PRN473 in vivo. PRN1008 and PRN473 potently inhibited CLEC-2-mediated platelet activation to rhodocytin. No off-target inhibition of SFKs was seen. PRN1008 treatment of Btk-deficient platelets resulted in minor additional inhibition of aggregation and tyrosine phosphorylation, likely reflecting inhibition of Tec. No effect on GPCR-mediated platelet function was observed. PRN473 significantly reduced the number of thrombi in podoplanin positive vessels following Salmonella infection and the presence of IVC thrombosis following vein stenosis. The potent inhibition of human platelet CLEC-2, and reduced thrombosis in in vivo models, together with the lack of off-target SFK inhibition and absence of bleeding reported in rilzabrutinib treated immune thrombocytopenia patients, suggest Btk inhibition as a promising antithrombotic strategy.

2.
Dev Cell ; 52(4): 446-460.e5, 2020 02 24.
Article in English | MEDLINE | ID: mdl-32032546

ABSTRACT

Hematopoietic stem and progenitor cells (HSPCs), first specified from hemogenic endothelium (HE) in the ventral dorsal aorta (VDA), support lifelong hematopoiesis. Their de novo production promises significant therapeutic value; however, current in vitro approaches cannot efficiently generate multipotent long-lived HSPCs. Presuming this reflects a lack of extrinsic cues normally impacting the VDA, we devised a human dorsal aorta-on-a-chip platform that identified Yes-activated protein (YAP) as a cyclic stretch-induced regulator of HSPC formation. In the zebrafish VDA, inducible Yap overexpression significantly increased runx1 expression in vivo and the number of CD41+ HSPCs downstream of HE specification. Endogenous Yap activation by lats1/2 knockdown or Rho-GTPase stimulation mimicked Yap overexpression and induced HSPCs in embryos lacking blood flow. Notably, in static human induced pluripotent stem cell (iPSC)-derived HE culture, compound-mediated YAP activation enhanced RUNX1 levels and hematopoietic colony-forming potential. Together, our findings reveal a potent impact of hemodynamic Rho-YAP mechanotransduction on HE fate, relevant to de novo human HSPC production.


Subject(s)
Cell Cycle Proteins/metabolism , Core Binding Factor Alpha 2 Subunit/metabolism , Endothelium, Vascular/cytology , Hematopoiesis , Hematopoietic Stem Cells/cytology , Induced Pluripotent Stem Cells/cytology , Mechanotransduction, Cellular , Transcription Factors/metabolism , Animals , Aorta/cytology , Aorta/embryology , Cell Cycle Proteins/genetics , Cell Differentiation , Core Binding Factor Alpha 2 Subunit/genetics , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Endothelium, Vascular/metabolism , Hematopoietic Stem Cells/physiology , Hemodynamics , Humans , Induced Pluripotent Stem Cells/physiology , Transcription Factors/genetics , Zebrafish , rho GTP-Binding Proteins/metabolism
3.
Stem Cell Reports ; 8(5): 1226-1241, 2017 05 09.
Article in English | MEDLINE | ID: mdl-28416284

ABSTRACT

Hematopoietic stem/progenitor cells (HSPCs) are formed during ontogeny from hemogenic endothelium in the ventral wall of the dorsal aorta (VDA). Critically, the cellular mechanism(s) allowing HSPC egress and migration to secondary niches are incompletely understood. Matrix metalloproteinases (MMPs) are inflammation-responsive proteins that regulate extracellular matrix (ECM) remodeling, cellular interactions, and signaling. Here, inhibition of vascular-associated Mmp2 function caused accumulation of fibronectin-rich ECM, retention of runx1/cmyb+ HSPCs in the VDA, and delayed caudal hematopoietic tissue (CHT) colonization; these defects were absent in fibronectin mutants, indicating that Mmp2 facilitates endothelial-to-hematopoietic transition via ECM remodeling. In contrast, Mmp9 was dispensable for HSPC budding, being instead required for proper colonization of secondary niches. Significantly, these migration defects were mimicked by overexpression and blocked by knockdown of C-X-C motif chemokine-12 (cxcl12), suggesting that Mmp9 controls CHT homeostasis through chemokine regulation. Our findings indicate Mmp2 and Mmp9 play distinct but complementary roles in developmental HSPC production and migration.


Subject(s)
Cell Movement , Embryonic Stem Cells/cytology , Hematopoietic Stem Cells/cytology , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Stem Cell Niche , Animals , Cell Proliferation , Chemokine CXCL12/metabolism , Core Binding Factor Alpha 2 Subunit/metabolism , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/physiology , Extracellular Matrix/metabolism , Fibronectins/metabolism , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/physiology , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/genetics , Zebrafish/embryology , Zebrafish/metabolism , Zebrafish Proteins/metabolism
4.
Exp Hematol ; 46: 83-95.e6, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27751871

ABSTRACT

Hematopoietic stem cells (HSCs) have the ability to both self-renew and differentiate each of the mature blood cell lineages and thereby reconstitute the entire blood system. Therefore, HSCs are therapeutically valuable for treatment of hematological malignances and bone marrow failure. We showed recently that transient glucose elevation elicited dose-dependent effects on HSCs through elevated metabolic activity and subsequent reactive oxygen species-mediated induction of Hypoxia-Inducible Factor 1α (Hif1α). Platelet-Derived Growth Factor B (pdgfb), a Hif1α-target, and its receptor, pdgfrb, were significantly upregulated in response to metabolic stimulation. Although the function of PDGF signaling is well established in vascular development, its role in hematopoiesis is less understood. Exposure to either a pan-PDGF inhibitor or a PDGFRß-selective antagonist in the context of Hif1α stimulation blocked elevations in hematopoietic stem and progenitor cell (HSPC) formation as determined by runx1;cmyb whole-mount in situ hybridization (WISH) and HSPC-reporter flow cytometry analysis. Similar results were observed for morpholino (MO) knockdown of pdgfrb or dominant-negative pdgfrb expression, indicating that PDGFRß signaling is a key downstream mediator of Hif1α-mediated induction of HSPCs. Notably, overexpression of Pdgfb ligand enhanced HSPC numbers in the aorta-gonado-mesonephros (AGM) at 36 hours postfertilization (hpf) and in the caudal hematopoietic tissue at 48 hpf. A survey of known PDGF-B/PDGFRß regulatory targets by expression analysis revealed a significant increase in inflammatory intermediates, including Interleukin 6 (IL-6) and its receptor (IL-6R). MO-mediated knockdown of il6 or chemical inhibition of IL-6R antagonized the effect of Pdgfb overexpression. Furthermore, epistatic analysis of IL-6/IL-6R function confirmed activity downstream of Hif1α. Together, these findings define a Hif1α-regulated signaling axis mediated through PBFGB/PDGFRß and IL-6/IL-6R that acts to control embryonic HSPC production.


Subject(s)
Hematopoietic Stem Cells/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit , Interleukin-6/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Signal Transduction , Animals , Cell Differentiation , Cell Proliferation , Gene Expression , Hematopoietic Stem Cells/cytology , Immunohistochemistry , Protein Stability , Receptor, Platelet-Derived Growth Factor beta/genetics , Zebrafish
5.
Cell Rep ; 17(2): 458-468, 2016 10 04.
Article in English | MEDLINE | ID: mdl-27705794

ABSTRACT

Vitamin D insufficiency is a worldwide epidemic affecting billions of individuals, including pregnant women and children. Despite its high incidence, the impact of active vitamin D3 (1,25(OH)D3) on embryonic development beyond osteo-regulation remains largely undefined. Here, we demonstrate that 1,25(OH)D3 availability modulates zebrafish hematopoietic stem and progenitor cell (HSPC) production. Loss of Cyp27b1-mediated biosynthesis or vitamin D receptor (VDR) function by gene knockdown resulted in significantly reduced runx1 expression and Flk1+cMyb+ HSPC numbers. Selective modulation in vivo and in vitro in zebrafish indicated that vitamin D3 acts directly on HSPCs, independent of calcium regulation, to increase proliferation. Notably, ex vivo treatment of human HSPCs with 1,25(OH)D3 also enhanced hematopoietic colony numbers, illustrating conservation across species. Finally, gene expression and epistasis analysis indicated that CXCL8(IL-8) was a functional target of vitamin D3-mediated HSPC regulation. Together, these findings highlight the relevance of developmental 1,25(OH)D3 availability for definitive hematopoiesis and suggest potential therapeutic utility in HSPC expansion.


Subject(s)
Cytochrome P-450 Enzyme System/genetics , Hematopoietic Stem Cells/metabolism , Interleukin-8/genetics , Receptors, Calcitriol/genetics , Vitamin D/genetics , Zebrafish Proteins/genetics , Animals , Biological Availability , Calcium Signaling/genetics , Core Binding Factor Alpha 2 Subunit/genetics , Embryonic Development/genetics , Female , Gene Expression Regulation, Developmental , Hematopoiesis/genetics , Humans , Interleukin-8/metabolism , Pregnancy , Vascular Endothelial Growth Factor Receptor-2/genetics , Vitamin D/metabolism , Vitamin D Deficiency/genetics , Vitamin D Deficiency/metabolism , Zebrafish/genetics , Zebrafish/growth & development
6.
Cell Stem Cell ; 19(3): 370-82, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27424782

ABSTRACT

Hematopoietic stem and progenitor cell (HSPC) specification is regulated by numerous defined factors acting locally within the hemogenic niche; however, it is unclear whether production can adapt to fluctuating systemic needs. Here we show that the CNS controls embryonic HSPC numbers via the hypothalamic-pituitary-adrenal/interrenal (HPA/I) stress response axis. Exposure to serotonin or the reuptake inhibitor fluoxetine increased runx1 expression and Flk1(+)/cMyb(+) HSPCs independent of peripheral innervation. Inhibition of neuronal, but not peripheral, tryptophan hydroxlyase (Tph) persistently reduced HSPC number. Consistent with central HPA/I axis induction and glucocorticoid receptor (GR) activation, GR agonists enhanced, whereas GR loss diminished, HSPC formation. Significantly, developmental hypoxia, as indicated by Hif1α function, induced the HPA/I axis and cortisol production. Furthermore, Hif1α-stimulated HSPC enhancement was attenuated by neuronal tph or GR loss. Our data establish that embryonic HSC production responds to physiologic stress via CNS-derived serotonin synthesis and central feedback regulation to control HSC numbers.


Subject(s)
Central Nervous System/metabolism , Embryonic Stem Cells/metabolism , Hematopoietic Stem Cells/metabolism , Receptors, Glucocorticoid/metabolism , Signal Transduction , Animals , Cell Count , Cell Hypoxia/drug effects , Cell Proliferation/drug effects , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Embryonic Stem Cells/drug effects , Fluoxetine/pharmacology , Hematopoietic Stem Cells/drug effects , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney/drug effects , Kidney/metabolism , Serotonergic Neurons/drug effects , Serotonergic Neurons/metabolism , Serotonin/pharmacology , Signal Transduction/drug effects , Stress, Physiological/drug effects , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/metabolism , Tryptophan Hydroxylase/metabolism , Zebrafish/embryology
SELECTION OF CITATIONS
SEARCH DETAIL