Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Nat Biotechnol ; 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38689027

ABSTRACT

Drugs are administered at a dosing schedule set by their therapeutic index, and termination of action is achieved by clearance and metabolism of the drug. In some cases, such as anticoagulant drugs or immunotherapeutics, it is important to be able to quickly reverse the drug's action. Here, we report a general strategy to achieve on-demand reversibility by designing a supramolecular drug (a noncovalent assembly of two cooperatively interacting drug fragments held together by transient hybridization of peptide nucleic acid (PNA)) that can be reversed with a PNA antidote that outcompetes the hybridization between the fragments. We demonstrate the approach with thrombin-inhibiting anticoagulants, creating very potent and reversible bivalent direct thrombin inhibitors (Ki = 74 pM). The supramolecular inhibitor effectively inhibited thrombus formation in mice in a needle injury thrombosis model, and this activity could be reversed by administration of the PNA antidote. This design is applicable to therapeutic targets where two binding sites can be identified.

2.
Blood ; 143(4): 342-356, 2024 Jan 25.
Article in English | MEDLINE | ID: mdl-37922495

ABSTRACT

ABSTRACT: Glycoprotein Ibα (GPIbα) is expressed on the surface of platelets and megakaryocytes (MKs) and anchored to the membrane skeleton by filamin A (flnA). Although GPIb and flnA have fundamental roles in platelet biogenesis, the nature of this interaction in megakaryocyte biology remains ill-defined. We generated a mouse model expressing either human wild-type (WT) GPIbα (hGPIbαWT) or a flnA-binding mutant (hGPIbαFW) and lacking endogenous mouse GPIbα. Mice expressing the mutant GPIbα transgene exhibited macrothrombocytopenia with preserved GPIb surface expression. Platelet clearance was normal and differentiation of MKs to proplatelets was unimpaired in hGPIbαFW mice. The most striking abnormalities in hGPIbαFW MKs were the defective formation of the demarcation membrane system (DMS) and the redistribution of flnA from the cytoplasm to the peripheral margin of MKs. These abnormalities led to disorganized internal MK membranes and the generation of enlarged megakaryocyte membrane buds. The defective flnA-GPIbα interaction also resulted in misdirected release of buds away from the vasculature into bone marrow interstitium. Restoring the linkage between flnA and GPIbα corrected the flnA redistribution within MKs and DMS ultrastructural defects as well as restored normal bud size and release into sinusoids. These studies define a new mechanism of macrothrombocytopenia resulting from dysregulated MK budding. The link between flnA and GPIbα is not essential for the MK budding process, however, it plays a major role in regulating the structure of the DMS, bud morphogenesis, and the localized release of buds into the circulation.


Subject(s)
Megakaryocytes , Platelet Glycoprotein GPIb-IX Complex , Thrombocytopenia , Animals , Humans , Mice , Blood Platelets/metabolism , Cytoplasm/metabolism , Filamins/genetics , Filamins/metabolism , Megakaryocytes/metabolism , Morphogenesis , Platelet Glycoprotein GPIb-IX Complex/genetics , Platelet Glycoprotein GPIb-IX Complex/metabolism , Thrombocytopenia/genetics , Thrombocytopenia/metabolism
4.
J Biol Chem ; 298(11): 102536, 2022 11.
Article in English | MEDLINE | ID: mdl-36174675

ABSTRACT

The cellular response to hypoxia is regulated through enzymatic oxygen sensors, including the prolyl hydroxylases, which control degradation of the well-known hypoxia inducible factors (HIFs). Other enzymatic oxygen sensors have been recently identified, including members of the KDM histone demethylase family. Little is known about how different oxygen-sensing pathways interact and if this varies depending on the form of hypoxia, such as chronic or intermittent. In this study, we investigated how two proposed cellular oxygen-sensing systems, HIF-1 and KDM4A, KDM4B, and KDM4C, respond in cells exposed to rapid forms of intermittent hypoxia (minutes) and compared to chronic hypoxia (hours). We found that intermittent hypoxia increases HIF-1α protein through a pathway distinct from chronic hypoxia, involving the KDM4A, KDM4B, and KDM4C histone lysine demethylases. Intermittent hypoxia increases the quantity and activity of KDM4A, KDM4B, and KDM4C, resulting in a decrease in histone 3 lysine 9 (H3K9) trimethylation near the HIF1A locus. We demonstrate that this contrasts with chronic hypoxia, which decreases KDM4A, KDM4B, and KDM4C activity, leading to hypertrimethylation of H3K9 globally and at the HIF1A locus. Altogether, we found that demethylation of histones bound to the HIF1A gene in intermittent hypoxia increases HIF1A mRNA expression, which has the downstream effect of increasing overall HIF-1 activity and expression of HIF target genes. This study highlights how multiple oxygen-sensing pathways can interact to regulate and fine tune the cellular hypoxic response depending on the period and length of hypoxia.


Subject(s)
Histones , Hypoxia-Inducible Factor 1, alpha Subunit , Protein Processing, Post-Translational , Humans , Demethylation , Histone Demethylases/metabolism , Histones/genetics , Histones/metabolism , Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Oxygen/metabolism
5.
Blood Adv ; 6(18): 5449-5462, 2022 09 27.
Article in English | MEDLINE | ID: mdl-35767737

ABSTRACT

Recanalization with restored cerebral perfusion is the primary goal of thrombolytic therapy in acute ischemic stroke. The identification of adjunctive therapies that can be safely used to enhance thrombolysis in stroke remains an elusive goal. We report here the development of a mouse in situ carotid artery thrombolysis (iCAT) stroke model involving graded cerebral ischemia to induce unihemispheric infarction after thrombotic occlusion of the common carotid artery (CCA). Electrolytic-induced thrombotic occlusion of the left CCA enabled real-time assessment of recanalization and rethrombosis events after thrombolysis with recombinant tissue-type plasminogen activator (rtPA). Concurrent transient stenosis of the right CCA induced unihemispheric hypoperfusion and infarction in the left middle cerebral artery territory. Real-time assessment of thrombolysis revealed recanalization rates <30% in rtPA-treated animals with high rates of rethrombosis. Addition of the direct thrombin inhibitor argatroban increased recanalization rates to 50% and reduced rethrombosis. Paradoxically, this was associated with increased cerebral ischemia and stroke-related mortality (25%-42%). Serial analysis of carotid and cerebral blood flow showed that coadministration of argatroban with rtPA resulted in a marked increase in carotid artery embolization, leading to distal obstruction of the middle cerebral artery. Real-time imaging of carotid thrombi revealed that adjunctive anticoagulation destabilized platelet-rich thrombi at the vessel wall, leading to dislodgement of large platelet emboli. These studies confirm the benefits of anticoagulants in enhancing thrombolysis and large artery recanalization; however, at high levels of anticoagulation (∼3-fold prolongation of activated partial thromboplastin time), this effect is offset by increased incidence of carotid artery embolization and distal middle cerebral artery occlusion. The iCAT stroke model should provide important new insight into the effects of adjunctive antithrombotic agents on real-time thrombus dynamics during thrombolysis and their correlation with stroke outcomes.


Subject(s)
Brain Ischemia , Ischemic Stroke , Stroke , Thromboembolism , Animals , Anticoagulants/therapeutic use , Antithrombins/therapeutic use , Arginine/analogs & derivatives , Brain Ischemia/complications , Brain Ischemia/drug therapy , Carotid Artery, Common , Fibrinolytic Agents/pharmacology , Fibrinolytic Agents/therapeutic use , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/drug therapy , Mice , Pipecolic Acids , Stroke/drug therapy , Stroke/etiology , Sulfonamides , Thrombolytic Therapy/adverse effects , Thrombolytic Therapy/methods , Tissue Plasminogen Activator/pharmacology , Tissue Plasminogen Activator/therapeutic use , Treatment Outcome
6.
Analyst ; 147(6): 1222-1235, 2022 Mar 14.
Article in English | MEDLINE | ID: mdl-35212697

ABSTRACT

Microvascular thrombosis and inflammation (thromboinflammation) are major causes of morbidity and mortality in critically ill patients with limited therapeutic options. Platelets are central to thromboinflammation, and microvascular platelet thrombi are highly effective at recruiting and activating leukocytes at sites of endothelial injury. Whilst parallel-plate flow chambers, microslides and straight microchannel assays have been widely used to recapitulate leukocyte adhesive behavior on 2-dimensional (2D) surfaces, none of these methods achieve high fidelity 3-dimensional (3D) geometries emulating microvascular platelet thrombi. As a result, the role of hydrodynamic factors in regulating leukocyte interactions with platelet thrombi remains ill-defined. Here, we report a microfluidic post model that allows visualization and analysis of neutrophil-platelet interactions in a 3D flow field. We have utilized the unique mechanosensitive features of platelets to enable selective micropatterning of the 3D posts with human or mouse platelets. By modulating the activation status of platelets, our method enables precise control of platelet surface reactivity and neutrophil recruitment. In addition, our microfluidic post assay accurately recapitulated the rolling versus stationary adhesion behavior of single neutrophils and demonstrated the efficacy of the P-selectin and Mac-1 blocking antibodies to reduce neutrophil recruitment and stationary adhesion, respectively. Moreover, the geometry of posts had a major influence on the efficiency of neutrophil recruitment and adhesion stability. This new post method highlights the importance of platelet 3D geometries in facilitating efficient, localized neutrophil recruitment. These findings have potentially important implications for the potent proinflammatory function of microvascular platelet thrombi.


Subject(s)
Blood Platelets , Thrombosis , Animals , Cell Adhesion , Humans , Inflammation , Leukocytes , Mice , Microfluidics , Neutrophils
7.
Angew Chem Int Ed Engl ; 60(10): 5348-5356, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33345438

ABSTRACT

Blood feeding arthropods, such as leeches, ticks, flies and mosquitoes, provide a privileged source of peptidic anticoagulant molecules. These primarily operate through inhibition of the central coagulation protease thrombin by binding to the active site and either exosite I or exosite II. Herein, we describe the rational design of a novel class of trivalent thrombin inhibitors that simultaneously block both exosites as well as the active site. These engineered hybrids were synthesized using tandem diselenide-selenoester ligation (DSL) and native chemical ligation (NCL) reactions in one-pot. The most potent trivalent inhibitors possessed femtomolar inhibition constants against α-thrombin and were selective over related coagulation proteases. A lead hybrid inhibitor possessed potent anticoagulant activity, blockade of both thrombin generation and platelet aggregation in vitro and efficacy in a murine thrombosis model at 1 mg kg-1 . The rational engineering approach described here lays the foundation for the development of potent and selective inhibitors for a range of other enzymatic targets that possess multiple sites for the disruption of protein-protein interactions, in addition to an active site.


Subject(s)
Anticoagulants/therapeutic use , Platelet Aggregation Inhibitors/therapeutic use , Salivary Proteins and Peptides/therapeutic use , Thrombosis/drug therapy , Amblyomma/chemistry , Animals , Anopheles/chemistry , Anticoagulants/chemical synthesis , Anticoagulants/metabolism , Catalytic Domain , Humans , Male , Mice, Inbred C57BL , Platelet Aggregation Inhibitors/chemical synthesis , Platelet Aggregation Inhibitors/metabolism , Protein Binding , Protein Engineering , Salivary Proteins and Peptides/chemical synthesis , Salivary Proteins and Peptides/metabolism , Thrombin/chemistry , Thrombin/metabolism , Tsetse Flies/chemistry
8.
ACS Cent Sci ; 4(4): 468-476, 2018 Apr 25.
Article in English | MEDLINE | ID: mdl-29721529

ABSTRACT

The anophelins are small protein thrombin inhibitors that are produced in the salivary glands of the Anopheles mosquito to fulfill a vital role in blood feeding. A bioinformatic analysis of anophelin sequences revealed the presence of conserved tyrosine residues in an acidic environment that were predicted to be post-translationally sulfated in vivo. To test this prediction, insect cell expression of two anophelin proteins, from Anopheles albimanus and Anopheles gambiae, was performed, followed by analysis by mass spectrometry, which showed heterogeneous sulfation at the predicted sites. Homogeneously sulfated variants of the two proteins were subsequently generated by chemical synthesis via a one-pot ligation-desulfurization strategy. Tyrosine sulfation of the anophelins was shown to significantly enhance the thrombin inhibitory activity, with a doubly sulfated variant of the anophelin from A. albimanus exhibiting a 100-fold increase in potency compared with the unmodified homologue. Sulfated anophelins were also shown to exhibit potent in vivo anticoagulant and antithrombotic activity.

9.
Blood ; 132(2): 197-209, 2018 07 12.
Article in English | MEDLINE | ID: mdl-29784641

ABSTRACT

The circulating life span of blood platelets is regulated by the prosurvival protein BCL-XL It restrains the activity of BAK and BAX, the essential prodeath mediators of intrinsic apoptosis. Disabling the platelet intrinsic apoptotic pathway in mice by deleting BAK and BAX results in a doubling of platelet life span and concomitant thrombocytosis. Apoptotic platelets expose phosphatidylserine (PS) via a mechanism that is distinct from that driven by classical agonists. Whether there is any role for apoptotic PS in platelet function in vivo, however, is unclear. Apoptosis has also been associated with the platelet storage lesion (PSL), the constellation of biochemical deteriorations that occur during blood bank storage. In this study, we investigated the role of BAK/BAX-mediated apoptosis in hemostasis and thrombosis and in the development of the PSL. We show that although intrinsic apoptosis is rapidly induced during storage at 37°C, it is not detected when platelets are kept at the standard storage temperature of 22°C. Remarkably, loss of BAK and BAX did not prevent the development of the PSL at either temperature. BAK/BAX-deficient mice exhibited increased bleeding times and unstable thrombus formation. This phenotype was not caused by impaired PS exposure, but was associated with a defect in granule release from aged platelets. Strikingly, rejuvenation of BAK/BAX-deficient platelets in vivo completely rescued the observed hemostatic defects. Thus, apoptotic culling of old platelets from the bloodstream is essential to maintain a functional, hemostatically reactive platelet population. Inhibiting intrinsic apoptosis in blood banked platelets is unlikely to yield significant benefit.


Subject(s)
Apoptosis , Blood Platelets/metabolism , Disease Susceptibility , Animals , Apoptosis/genetics , Biomarkers , Bleeding Time , Blood Cell Count , Blood Coagulation , Caspases/metabolism , Cell Survival/genetics , Female , Genotype , Male , Mice , Mice, Knockout , Mitochondria/metabolism , Signal Transduction , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism , bcl-X Protein/genetics , bcl-X Protein/metabolism
10.
Nat Commun ; 9(1): 1087, 2018 03 14.
Article in English | MEDLINE | ID: mdl-29540687

ABSTRACT

Diabetes is associated with an exaggerated platelet thrombotic response at sites of vascular injury. Biomechanical forces regulate platelet activation, although the impact of diabetes on this process remains ill-defined. Using a biomembrane force probe (BFP), we demonstrate that compressive force activates integrin αIIbß3 on discoid diabetic platelets, increasing its association rate with immobilized fibrinogen. This compressive force-induced integrin activation is calcium and PI 3-kinase dependent, resulting in enhanced integrin affinity maturation and exaggerated shear-dependent platelet adhesion. Analysis of discoid platelet aggregation in the mesenteric circulation of mice confirmed that diabetes leads to a marked enhancement in the formation and stability of discoid platelet aggregates, via a mechanism that is not inhibited by therapeutic doses of aspirin and clopidogrel, but is eliminated by PI 3-kinase inhibition. These studies demonstrate the existence of a compression force sensing mechanism linked to αIIbß3 adhesive function that leads to a distinct prothrombotic phenotype in diabetes.


Subject(s)
Blood Platelets/metabolism , Diabetes Mellitus, Type 1/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Thrombosis/metabolism , Adult , Animals , Aspirin/pharmacology , Blood Platelets/drug effects , Clopidogrel , Female , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Platelet Adhesiveness/drug effects , Platelet Adhesiveness/physiology , Platelet Aggregation/drug effects , Platelet Aggregation/physiology , Ticlopidine/analogs & derivatives , Ticlopidine/pharmacology
11.
Semin Thromb Hemost ; 44(2): 102-113, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29294493

ABSTRACT

Ischemia-reperfusion (IR) injury is a common complication of a variety of cardiovascular diseases, including ischemic stroke and myocardial infarction (MI). While timely re-establishment of blood flow in a thrombosed artery is the primary goal of acute therapy in these diseases, paradoxically, reperfusion of ischemic tissue can cause widespread microvascular dysfunction that significantly exacerbates organ damage. Reperfusion injury is associated with activation of the humoral and cellular components of the hemostatic and innate immune systems and also with excessive reactive oxygen species production, endothelial dysfunction, thrombosis, and inflammation. Platelets are critical mediators of thromboinflammation during reperfusion injury and a hyperactive platelet phenotype may contribute to an exaggerated IR injury response. This is particularly relevant to diabetes which is characteristically associated with hyperactive platelets, significantly worse IR injury, increased organ damage, and increased risk of death. However, the mechanisms underlying vulnerability to IR injury in diabetic individuals is not well defined, nor the role of "diabetic platelets" in this process. This review summarizes recent progress in understanding the role of platelets in promoting microvascular dysfunction and inflammation in the context of IR injury. Furthermore, the authors discuss aspects of the thromboinflammatory function of platelets that are dysregulated in diabetes. They conclude that diabetes likely enhances the capacity of platelets to mediate microvascular thrombosis and inflammation during IR injury, which has potentially important implications for the future design of antiplatelet agents that can reduce microvascular dysfunction and inflammation.


Subject(s)
Blood Platelets/immunology , Diabetes Mellitus/blood , Inflammation/immunology , Reperfusion Injury/blood , Thrombosis/immunology , Diabetes Mellitus/immunology , Humans
12.
Blood ; 130(23): 2453-2462, 2017 12 07.
Article in English | MEDLINE | ID: mdl-29074499

ABSTRACT

Clot retraction refers to the process whereby activated platelets transduce contractile forces onto the fibrin network of a thrombus, which over time increases clot density and decreases clot size. This process is considered important for promoting clot stability and maintaining blood vessel patency. Insights into the mechanisms regulating clot retraction at sites of vascular injury have been hampered by a paucity of in vivo experimental models. By pairing localized vascular injury with thrombin microinjection in the mesenteric circulation of mice, we have demonstrated that the fibrin network of thrombi progressively compacts over a 2-hour period. This was a genuine retraction process, as treating thrombi with blebbistatin to inhibit myosin IIa-mediated platelet contractility prevented shrinkage of the fibrin network. Real-time confocal analysis of fibrinolysis after recombinant tissue-type plasminogen activator (tPA) administration revealed that incomplete proteolysis of fibrin polymers markedly facilitated clot retraction. Similarly, inhibiting endogenous fibrinolysis with tranexamic acid reduced retraction of fibrin polymers in vivo. In vitro clot retraction experiments indicated that subthreshold doses of tPA facilitated clot retraction through a plasmin-dependent mechanism. These effects correlated with changes in the elastic modulus of fibrin clots. These findings define the endogenous fibrinolytic system as an important regulator of clot retraction, and show that promoting clot retraction is a novel and complementary means by which fibrinolytic enzymes can reduce thrombus size.


Subject(s)
Clot Retraction , Fibrinolysis , Actomyosin/metabolism , Animals , Blood Platelets/drug effects , Blood Platelets/metabolism , Fibrin/metabolism , Fibrinolysis/drug effects , Humans , Male , Mice , Nonmuscle Myosin Type IIA/metabolism , Thrombosis/diagnostic imaging , Thrombosis/metabolism , Thrombosis/pathology , Tissue Plasminogen Activator/metabolism , Tissue Plasminogen Activator/pharmacology , Tranexamic Acid/pharmacology
13.
Sci Transl Med ; 9(409)2017 Sep 27.
Article in English | MEDLINE | ID: mdl-28954929

ABSTRACT

Gut ischemia is common in critically ill patients, promoting thrombosis and inflammation in distant organs. The mechanisms linking hemodynamic changes in the gut to remote organ thrombosis remain ill-defined. We demonstrate that gut ischemia in the mouse induces a distinct pulmonary thrombotic disorder triggered by neutrophil macroaggregates. These neutrophil aggregates lead to widespread occlusion of pulmonary arteries, veins, and the microvasculature. A similar pulmonary neutrophil-rich thrombotic response occurred in humans with the acute respiratory distress syndrome. Intravital microscopy during gut ischemia-reperfusion injury revealed that rolling neutrophils extract large membrane fragments from remnant dying platelets in multiple organs. These platelet fragments bridge adjacent neutrophils to facilitate macroaggregation. Platelet-specific deletion of cyclophilin D, a mitochondrial regulator of cell necrosis, prevented neutrophil macroaggregation and pulmonary thrombosis. Our studies demonstrate the existence of a distinct pulmonary thrombotic disorder triggered by dying platelets and neutrophil macroaggregates. Therapeutic targeting of platelet death pathways may reduce pulmonary thrombosis in critically ill patients.


Subject(s)
Gastrointestinal Tract/blood supply , Gastrointestinal Tract/pathology , Ischemia/complications , Lung/pathology , Neutrophils/pathology , Thrombosis/etiology , Thrombosis/pathology , Animals , Blood Platelets/metabolism , Cell Aggregation , Cell Membrane/metabolism , Peptidyl-Prolyl Isomerase F , Cyclophilins/deficiency , Cyclophilins/metabolism , Gastrointestinal Tract/physiopathology , Humans , Ischemia/physiopathology , Lung/blood supply , Lung/physiopathology , Mice, Inbred C57BL , Phosphatidylserines/metabolism , Respiratory Distress Syndrome/pathology , Respiratory Distress Syndrome/physiopathology , Splanchnic Circulation
14.
Nat Commun ; 6: 7835, 2015 Jul 23.
Article in English | MEDLINE | ID: mdl-26204458

ABSTRACT

Thrombin is a central regulator of leukocyte recruitment and inflammation at sites of vascular injury, a function thought to involve primarily endothelial PAR cleavage. Here we demonstrate the existence of a distinct leukocyte-trafficking mechanism regulated by components of the haemostatic system, including platelet PAR4, GPIbα and fibrin. Utilizing a mouse endothelial injury model we show that thrombin cleavage of platelet PAR4 promotes leukocyte recruitment to sites of vascular injury. This process is negatively regulated by GPIbα, as seen in mice with abrogated thrombin-platelet GPIbα binding (hGPIbα(D277N)). In addition, we demonstrate that fibrin limits leukocyte trafficking by forming a physical barrier to intravascular leukocyte migration. These studies demonstrate a distinct 'checkpoint' mechanism of leukocyte trafficking involving balanced thrombin interactions with PAR4, GPIbα and fibrin. Dysregulation of this checkpoint mechanism is likely to contribute to the development of thromboinflammatory disorders.


Subject(s)
Leukocytes/physiology , Platelet Activation , Platelet Glycoprotein GPIb-IX Complex/metabolism , Receptors, Thrombin/metabolism , Thrombin/metabolism , Animals , Cell Movement , Endothelial Cells/physiology , Fibrinolysis , Humans , Male , Mice , Mice, Inbred C57BL , Models, Animal
15.
Reprod Toxicol ; 50: 68-86, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25242112

ABSTRACT

Neonatal treatment of hamsters with diethylstilbestrol (DES) induces uterine hyperplasia/dysplasia/neoplasia (endometrial adenocarcinoma) in adult animals. We subsequently determined that the neonatal DES exposure event directly and permanently disrupts the developing hamster uterus (initiation stage) so that it responds abnormally when it is stimulated with estrogen in adulthood (promotion stage). To identify candidate molecular elements involved in progression of the disruption/neoplastic process, we performed: (1) immunoblot analyses and (2) microarray profiling (Affymetrix Gene Chip System) on sets of uterine protein and RNA extracts, respectively, and (3) immunohistochemical analysis on uterine sections; all from both initiation stage and promotion stage groups of animals. Here we report that: (1) progression of the neonatal DES-induced hyperplasia/dysplasia/neoplasia phenomenon in the hamster uterus involves a wide spectrum of specific gene expression alterations and (2) the gene products involved and their manner of altered expression differ dramatically during the initiation vs. promotion stages of the phenomenon.


Subject(s)
Diethylstilbestrol/toxicity , Uterine Neoplasms/chemically induced , Uterus/drug effects , Animals , Animals, Newborn , Cadherins/physiology , Female , Gene Expression Profiling , Guinea Pigs , Hyperplasia , Insulin Receptor Substrate Proteins/analysis , Mesocricetus , Oncogenes , Pregnancy , Proliferating Cell Nuclear Antigen/analysis , Receptors, Androgen/analysis , Uterine Neoplasms/metabolism , Uterus/metabolism , Uterus/pathology
16.
J Biol Chem ; 289(8): 5051-60, 2014 Feb 21.
Article in English | MEDLINE | ID: mdl-24385425

ABSTRACT

The Dok proteins are a family of adaptor molecules that have a well defined role in regulating cellular migration, immune responses, and tumor progression. Previous studies have demonstrated that Doks-1 to 3 are expressed in platelets and that Dok-2 is tyrosine-phosphorylated downstream of integrin αIIbß3, raising the possibility that it participates in integrin αIIbß3 outside-in signaling. We demonstrate that Dok-2 in platelets is primarily phosphorylated by Lyn kinase. Moreover, deficiency of Dok-2 leads to dysregulated integrin αIIbß3-dependent cytosolic calcium flux and phosphatidylinositol(3,4)P2 accumulation. Although agonist-induced integrin αIIbß3 affinity regulation was unaltered in Dok-2(-/-) platelets, Dok-2 deficiency was associated with a shear-dependent increase in integrin αIIbß3 adhesive function, resulting in enhanced platelet-fibrinogen and platelet-platelet adhesive interactions under flow. This increase in adhesion was restricted to discoid platelets and involved the shear-dependent regulation of membrane tethers. Dok-2 deficiency was associated with an increased rate of platelet aggregate formation on thrombogenic surfaces, leading to accelerated thrombus growth in vivo. Overall, this study defines an important role for Dok-2 in regulating biomechanical adhesive function of discoid platelets. Moreover, they define a previously unrecognized prothrombotic mechanism that is not detected by conventional platelet function assays.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Phosphoproteins/metabolism , Platelet Adhesiveness/physiology , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Shear Strength , Adaptor Proteins, Signal Transducing/deficiency , Animals , Blood Platelets/drug effects , Blood Platelets/metabolism , Blood Platelets/ultrastructure , Calcium/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Fibrinogen/pharmacology , Hemorheology/drug effects , Humans , Immobilized Proteins/pharmacology , Mice , Mice, Inbred C57BL , Phosphatidylinositol Phosphates/metabolism , Phosphoproteins/deficiency , Platelet Adhesiveness/drug effects , Shear Strength/drug effects , Thrombosis/metabolism , Thrombosis/pathology , Thrombosis/physiopathology , Time Factors
17.
Blood ; 121(22): 4555-66, 2013 May 30.
Article in English | MEDLINE | ID: mdl-23550035

ABSTRACT

Thrombosis promotes leukocyte infiltration into inflamed tissues, leading to organ injury in a broad range of diseases; however, the mechanisms by which thrombi guide leukocytes to sites of vascular injury remain ill-defined. Using mouse models of endothelial injury (traumatic or ischemia reperfusion), we demonstrate a distinct process of leukocyte recruitment, termed "directed intravascular migration," specifically mediated by platelet thrombi. Single adherent platelets and platelet aggregates stimulated leukocyte shape change at sites of endothelial injury; however, only thrombi were capable of inducing directed intravascular leukocyte migration. Leukocyte recruitment and migration induced by platelet thrombi occurred most prominently in veins but could also occur in arteries following ischemia-reperfusion injury. In vitro studies demonstrated a major role for platelet-derived NAP-2 (CXCL-7) and its CXCR1/2 receptor in regulating leukocyte polarization and motility. In vivo studies demonstrated the presence of an NAP-2 chemotactic gradient within the thrombus body. Pharmacologic blockade of CXCR1/2 as well as genetic deletion of NAP-2 markedly reduced leukocyte shape change and intrathrombus migration. These studies define a distinct process of leukocyte migration that is initiated by homotypic adhesive interactions between platelets, leading to the development of an NAP-2 chemotactic gradient within the thrombus body that guides leukocytes to sites of vascular injury.


Subject(s)
Blood Platelets/cytology , Chemokines, CXC/metabolism , Leukocytes/cytology , Receptors, Interleukin-8A/metabolism , Receptors, Interleukin-8B/metabolism , Thrombosis/immunology , Animals , Blood Platelets/immunology , Blood Platelets/metabolism , Cell Adhesion/immunology , Cell Movement/immunology , Cell Polarity/immunology , Green Fluorescent Proteins/genetics , Leukocytes/immunology , Mesenteric Arteries/immunology , Mesenteric Arteries/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Needlestick Injuries/immunology , Needlestick Injuries/pathology , Neutrophils/cytology , Neutrophils/immunology , Reperfusion Injury/immunology , Reperfusion Injury/pathology
18.
Reprod Toxicol ; 32(4): 472-83, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21963885

ABSTRACT

We assessed neonatal diethylstilbestrol (DES)-induced disruption at various endocrine levels in the hamster. In particular, we used organ transplantation into the hamster cheek pouch to determine whether abnormalities observed in the post-pubertal ovary are due to: (a) a direct (early) mechanism or (b) an indirect (late) mechanism that involves altered development and function of the hypothalamus and/or pituitary. Of the various disruption endpoints and attributes assessed: (1) some were consistent with the direct mechanism (altered uterine and cervical dimensions/organization, ovarian polyovular follicles, vaginal hypospadius, endometrial hyperplasia/dysplasia); (2) some were consistent with the indirect mechanism (ovarian/oviductal salpingitis, cystic ovarian follicles); (3) some were consistent with a combination of the direct and indirect mechanisms (altered endocrine status); and (4) the mechanism(s) for one (lack of corpora lutea) was uncertain. This study also generated some surprising observations regarding vaginal estrous assessments as a means to monitor periodicity of ovarian function in the hamster.


Subject(s)
Animals, Newborn , Diethylstilbestrol/toxicity , Estrogens, Non-Steroidal/toxicity , Genitalia, Female/drug effects , Animals , Cervix Uteri/anatomy & histology , Cricetinae , Estrous Cycle , Fallopian Tubes/anatomy & histology , Fallopian Tubes/drug effects , Fallopian Tubes/physiology , Female , Genitalia, Female/anatomy & histology , Genitalia, Female/physiology , Hormones/blood , Hypothalamus/physiology , Mesocricetus , Ovariectomy , Ovary/anatomy & histology , Ovary/physiology , Ovary/transplantation , Pituitary Gland/physiology , Pregnancy , Sexual Maturation , Uterus/anatomy & histology , Vagina/physiology
19.
Blood ; 118(6): 1663-74, 2011 Aug 11.
Article in English | MEDLINE | ID: mdl-21673344

ABSTRACT

BH3 mimetics are a new class of proapo-ptotic anticancer agents that have shown considerable promise in preclinical animal models and early-stage human trials. These agents act by inhibiting the pro-survival function of one or more Bcl-2-related proteins. Agents that inhibit Bcl-x(L) induce rapid platelet death that leads to thrombocytopenia; however, their impact on the function of residual circulating platelets remains unclear. In this study, we demonstrate that the BH3 mimetics, ABT-737 or ABT-263, induce a time- and dose-dependent decrease in platelet adhesive function that correlates with ectodomain shedding of the major platelet adhesion receptors, glycoprotein Ibα and glycoprotein VI, and functional down-regulation of integrin α(IIb)ß(3). Analysis of platelets from mice treated with higher doses of BH3 mimetics revealed the presence of a subpopulation of circulating platelets undergoing cell death that have impaired activation responses to soluble agonists. Functional analysis of platelets by intravital microscopy revealed a time-dependent defect in platelet aggregation at sites of vascular injury that correlated with an increase in tail bleeding time. Overall, these studies demonstrate that Bcl-x(L)-inhibitory BH3 mimetics not only induce thrombocytopenia but also a transient thrombocytopathy that can undermine the hemostatic function of platelets.


Subject(s)
Blood Platelets/physiology , Hemostasis/physiology , Thrombocytopenia/physiopathology , bcl-X Protein/metabolism , Aniline Compounds/pharmacology , Animals , Apoptosis/drug effects , Biphenyl Compounds/pharmacology , Blood Platelets/metabolism , Blood Platelets/ultrastructure , Blotting, Western , Collagen/metabolism , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Hemostasis/drug effects , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Electron, Scanning , Nitrophenols/pharmacology , Phosphatidylserines/metabolism , Piperazines/pharmacology , Platelet Adhesiveness/drug effects , Platelet Aggregation/drug effects , Platelet Count , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Glycoprotein GPIb-IX Complex , Platelet Membrane Glycoproteins/metabolism , Sulfonamides/pharmacology , Thrombocytopenia/chemically induced , Time Factors , bcl-X Protein/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...