Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Metab Brain Dis ; 38(7): 2383-2391, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37341855

RESUMEN

Multiple sclerosis (MS) is the most common causes of non-traumatic disability in young adults worldwide. MS pathophysiologies include the formation of inflammatory lesions, axonal damage and demyelination, and blood brain barrier (BBB) disruption. Coagulation proteins, including factor (F)XII, can serve as important mediators of the adaptive immune response during neuroinflammation. Indeed, plasma FXII levels are increased during relapse in relapsing-remitting MS patients, and previous studies showed that reducing FXII levels was protective in a murine model of MS, experimental autoimmune encephalomyelitis (EAE). Our objective was to determine if pharmacological targeting of FXI, a major substrate of activated FXII (FXIIa), improves neurological function and attenuates CNS damage in the setting of EAE. EAE was induced in male mice using murine myelin oligodendrocyte glycoprotein peptides combined with heat-inactivated Mycobacterium tuberculosis and pertussis toxin. Upon onset of symptoms, mice were treated every other day intravenously with anti-FXI antibody, 14E11, or saline. Disease scores were recorded daily until euthanasia for ex vivo analyses of inflammation. Compared to the vehicle control, 14E11 treatment reduced the clinical severity of EAE and total mononuclear cells, including CD11b+CD45high macrophage/microglia and CD4+ T cell numbers in brain. Following pharmacological targeting of FXI, BBB disruption was reduced, as measured by decreased axonal damage and fibrin(ogen) accumulation in the spinal cord. These data demonstrate that pharmacological inhibition of FXI reduces disease severity, immune cell migration, axonal damage, and BBB disruption in mice with EAE. Thus, therapeutic agents targeting FXI and FXII may provide a useful approach for treating autoimmune and neurologic disorders.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Esclerosis Múltiple , Animales , Masculino , Ratones , Encéfalo/metabolismo , Encefalomielitis Autoinmune Experimental/metabolismo , Factor XI/antagonistas & inhibidores , Factor XI/metabolismo , Ratones Endogámicos C57BL , Esclerosis Múltiple/patología , Glicoproteína Mielina-Oligodendrócito , Médula Espinal/metabolismo
2.
Platelets ; 30(1): 126-135, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30560697

RESUMEN

Human platelets express two protease-activated receptors (PARs), PAR1 (F2R) and PAR4 (F2RL3), which are activated by a number of serine proteases that are generated during pathological events and cause platelet activation. Recent interest has focused on PAR4 as a therapeutic target, given PAR4 seems to promote experimental thrombosis and procoagulant microparticle formation, without a broadly apparent role in hemostasis. However, it is not yet known whether PAR4 activity plays a role in platelet-leukocyte interactions, which are thought to contribute to both thrombosis and acute or chronic thrombo-inflammatory processes. We sought to determine whether PAR4 activity contributes to granule secretion from activated platelets and platelet-leukocyte interactions. We performed in vitro and ex vivo studies of platelet granule release and platelet-leukocyte interactions in the presence of PAR4 agonists including PAR4 activating peptide, thrombin, cathepsin G, and plasmin in combination with small-molecule PAR4 antagonists. Activation of human platelets with thrombin, cathepsin G, or plasmin potentiated platelet dense granule secretion that was specifically impaired by PAR4 inhibitors. Platelet-leukocyte interactions and platelet P-selectin exposure the following stimulation with PAR4 agonists were also impaired by activated PAR4 inhibition in either a purified system or in whole blood. These results indicate PAR4-specific promotion of platelet granule release and platelet-leukocyte aggregate formation and suggest that pharmacological control of PAR4 activity could potentially attenuate platelet granule release or platelet-leukocyte interaction-mediated pathological processes.


Asunto(s)
Plaquetas/metabolismo , Comunicación Celular , Gránulos Citoplasmáticos/metabolismo , Leucocitos/metabolismo , Receptores de Trombina/metabolismo , Animales , Biomarcadores , Citometría de Flujo , Humanos , Masculino , Papio , Activación Plaquetaria , Agregación Plaquetaria
3.
J Biol Chem ; 292(21): 8616-8629, 2017 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-28408624

RESUMEN

Activated protein C (APC) is a multifunctional serine protease with anticoagulant, cytoprotective, and anti-inflammatory activities. In addition to the cytoprotective effects of APC on endothelial cells, podocytes, and neurons, APC cleaves and detoxifies extracellular histones, a major component of neutrophil extracellular traps (NETs). NETs promote pathogen clearance but also can lead to thrombosis; the pathways that negatively regulate NETosis are largely unknown. Thus, we studied whether APC is capable of directly inhibiting NETosis via receptor-mediated cell signaling mechanisms. Here, by quantifying extracellular DNA or myeloperoxidase, we demonstrate that APC binds human leukocytes and prevents activated platelet supernatant or phorbol 12-myristate 13-acetate (PMA) from inducing NETosis. Of note, APC proteolytic activity was required for inhibiting NETosis. Moreover, antibodies against the neutrophil receptors endothelial protein C receptor (EPCR), protease-activated receptor 3 (PAR3), and macrophage-1 antigen (Mac-1) blocked APC inhibition of NETosis. Select mutations in the Gla and protease domains of recombinant APC caused a loss of NETosis. Interestingly, pretreatment of neutrophils with APC prior to induction of NETosis inhibited platelet adhesion to NETs. Lastly, in a nonhuman primate model of Escherichia coli-induced sepsis, pretreatment of animals with APC abrogated release of myeloperoxidase from neutrophils, a marker of neutrophil activation. These findings suggest that the anti-inflammatory function of APC at therapeutic concentrations may include the inhibition of NETosis in an EPCR-, PAR3-, and Mac-1-dependent manner, providing additional mechanistic insight into the diverse functions of neutrophils and APC in disease states including sepsis.


Asunto(s)
Trampas Extracelulares/inmunología , Activación Neutrófila/inmunología , Neutrófilos/inmunología , Proteína C/inmunología , Animales , Antígenos CD/inmunología , Antígenos CD/metabolismo , Modelos Animales de Enfermedad , Receptor de Proteína C Endotelial , Escherichia coli , Infecciones por Escherichia coli/sangre , Infecciones por Escherichia coli/inmunología , Trampas Extracelulares/metabolismo , Femenino , Humanos , Antígeno de Macrófago-1/inmunología , Antígeno de Macrófago-1/metabolismo , Masculino , Activación Neutrófila/efectos de los fármacos , Neutrófilos/metabolismo , Papio anubis , Proteína C/metabolismo , Receptores de Superficie Celular/inmunología , Receptores de Superficie Celular/metabolismo , Sepsis/sangre , Sepsis/inmunología , Acetato de Tetradecanoilforbol/farmacología
4.
Am J Physiol Cell Physiol ; 310(7): C568-75, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26764050

RESUMEN

Molecular chaperones that support protein quality control, including heat shock protein 70 (Hsp70), participate in diverse aspects of cellular and physiological function. Recent studies have reported roles for specific chaperone activities in blood platelets in maintaining hemostasis; however, the functions of Hsp70 in platelet physiology remain uninvestigated. Here we characterize roles for Hsp70 activity in platelet activation and function. In vitro biochemical, microscopy, flow cytometry, and aggregometry assays of platelet function, as well as ex vivo analyses of platelet aggregate formation in whole blood under shear, were carried out under Hsp70-inhibited conditions. Inhibition of platelet Hsp70 blocked platelet aggregation and granule secretion in response to collagen-related peptide (CRP), which engages the immunoreceptor tyrosine-based activation motif-bearing collagen receptor glycoprotein VI (GPVI)-Fc receptor-γ chain complex. Hsp70 inhibition also reduced platelet integrin-αIIbß3 activation downstream of GPVI, as Hsp70-inhibited platelets showed reduced PAC-1 and fibrinogen binding. Ex vivo, pharmacological inhibition of Hsp70 in human whole blood prevented the formation of platelet aggregates on collagen under shear. Biochemical studies supported a role for Hsp70 in maintaining the assembly of the linker for activation of T cells signalosome, which couples GPVI-initiated signaling to integrin activation, secretion, and platelet function. Together, our results suggest that Hsp70 regulates platelet activation and function by supporting linker for activation of T cells-associated signaling events downstream of platelet GPVI engagement, suggesting a role for Hsp70 in the intracellular organization of signaling systems that mediate platelet secretion, "inside-out" activation of platelet integrin-αIIbß3, platelet-platelet aggregation, and, ultimately, hemostatic plug and thrombus formation.


Asunto(s)
Plaquetas/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Integrinas/metabolismo , Activación Plaquetaria/fisiología , Agregación Plaquetaria/fisiología , Citometría de Flujo , Humanos , Transducción de Señal/fisiología
5.
Am J Physiol Cell Physiol ; 310(5): C373-80, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26659727

RESUMEN

The Tec family kinase Bruton's tyrosine kinase (Btk) plays an important signaling role downstream of immunoreceptor tyrosine-based activation motifs in hematopoietic cells. Mutations in Btk are involved in impaired B-cell maturation in X-linked agammaglobulinemia, and Btk has been investigated for its role in platelet activation via activation of the effector protein phospholipase Cγ2 downstream of the platelet membrane glycoprotein VI (GPVI). Because of its role in hematopoietic cell signaling, Btk has become a target in the treatment of chronic lymphocytic leukemia and mantle cell lymphoma; the covalent Btk inhibitor ibrutinib was recently approved by the US Food and Drug Administration for treatment of these conditions. Antihemostatic events have been reported in some patients taking ibrutinib, although the mechanism of these events remains unknown. We sought to determine the effects of Btk inhibition on platelet function in a series of in vitro studies of platelet activation, spreading, and aggregation. Our results show that irreversible inhibition of Btk with two ibrutinib analogs in vitro decreased human platelet activation, phosphorylation of Btk, P-selectin exposure, spreading on fibrinogen, and aggregation under shear flow conditions. Short-term studies of ibrutinib analogs administered in vivo also showed abrogation of platelet aggregation in vitro, but without measurable effects on plasma clotting times or on bleeding in vivo. Taken together, our results suggest that inhibition of Btk significantly decreased GPVI-mediated platelet activation, spreading, and aggregation in vitro; however, prolonged bleeding was not observed in a model of bleeding.


Asunto(s)
Plaquetas/efectos de los fármacos , Activación Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Glicoproteínas de Membrana Plaquetaria/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Administración Oral , Agammaglobulinemia Tirosina Quinasa , Animales , Plaquetas/metabolismo , Hemorragia/inducido químicamente , Humanos , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Papio , Activación Plaquetaria/fisiología , Transducción de Señal/efectos de los fármacos
6.
Blood ; 133(5): 385-386, 2019 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-30705047
7.
Metab Brain Dis ; 30(1): 57-65, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24810631

RESUMEN

Multiple sclerosis (MS) is a neuroinflammatory disease characterized by demyelination and axonal damage of the central nervous system. The pathogenesis of MS has also been linked to vascular inflammation and local activation of the coagulation system, resulting in perivascular fibrin deposition. Treatment of experimental autoimmune encephalomyelitis (EAE), a model of human MS, with antithrombotic and antiinflammatory activated protein C (APC) reduces disease severity. Since recombinant APC (Drotecogin alfa), originally approved for the treatment of severe sepsis, is not available for human MS studies, we tested the hypothesis that pharmacologic activation of endogenous protein C could likewise improve the outcome of EAE. Mice were immunized with murine myelin oligodendrocyte glycoprotein (MOG) peptides and at the onset of EAE symptoms, were treated every other day with either WE thrombin (25 µg/kg; i.v.), a selective recombinant protein C activator thrombin analog, or saline control. Mice were monitored for changes in disease score until euthanized for ex vivo analysis of inflammation. Administration of WE thrombin significantly ameliorated clinical severity of EAE, reduced inflammatory cell infiltration and demyelination, suppressed the activation of macrophages comprising the CD11b + population and reduced accumulation of fibrin (ogen) in the spinal cord. These data suggest that symptomatic MS may respond to a treatment strategy that involves temporal pharmacological enhancement of endogenous APC generation.


Asunto(s)
Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Proteína C/agonistas , Trombina/uso terapéutico , Animales , Evaluación Preclínica de Medicamentos , Encefalomielitis Autoinmune Experimental/etiología , Encefalomielitis Autoinmune Experimental/patología , Activación Enzimática , Fibrina/análisis , Fibrinógeno/análisis , Humanos , Molécula 1 de Adhesión Intercelular/biosíntesis , Activación de Macrófagos , Masculino , Ratones , Esclerosis Múltiple , Glicoproteína Mielina-Oligodendrócito/inmunología , Fragmentos de Péptidos/inmunología , Mutación Puntual , Proteína C/metabolismo , Médula Espinal/patología , Bazo/inmunología , Bazo/patología , Trombina/genética , Resultado del Tratamiento , Factor de Necrosis Tumoral alfa/biosíntesis , Sustancia Blanca/patología
8.
J Thromb Haemost ; 22(7): 2039-2051, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38670314

RESUMEN

BACKGROUND: Activated protein C (APC) has anticoagulant and cytoprotective cell-signaling activities, which often require protease-activated receptor (PAR) 1 and PAR3 and PAR cleavages at noncanonical sites (R46-N47 and R41-G42, respectively). Some PAR1-derived (P1) peptides and PAR3-derived (P3) peptides, eg, P1-47-66 and P3-42-65, mimic APC's cell signaling. In anti-inflammatory assays, these 2 peptides at low concentrations synergistically attenuate cellular inflammation. OBJECTIVES: To determine whether a P1 peptide covalently linked to a P3 peptide mimics APC's anti-inflammatory and endothelial barrier stabilization activities. METHODS: Anti-inflammatory assays employed stimulated THP-1 cells and caspase-1 measurements. Cultured human EA.hy926 or murine aortic endothelial cells (ECs) exposed to thrombin were monitored for transendothelial electrical resistance. Bivalent covalently linked P1:P3 peptides were studied for APC-like activities. RESULTS: In anti-inflammatory assays, P1-47-55 was as active as P1-47-66 and some P3 peptides (eg, P3-44-54 and P3-51-65) were as active as P3-42-65. The bivalent P1:P3 peptide comprising P1-47-55-(Gly[10 residues])-P3-51-65 (designated "G10 peptide") was more potently anti-inflammatory than the P1 or P3 peptide alone. In transendothelial electrical resistance studies of thrombin-challenged ECs, P1-47-55 and the G10 peptide mimicked APC's protective actions. In dose-response studies, the G10 peptide was more potent than the P1-47-55 peptide. In murine EC studies, the murine PAR-sequence-derived G10 peptide mimicked murine APC's activity. Anti-PAR1 and anti-PAR3 antibodies, but not anti-endothelial protein C receptor antibodies, abated G10's cytoprotection, showing that G10's actions involve PAR1:PAR3. G10 significantly increased survival in murine endotoxemia. CONCLUSION: The PAR-sequence-derived G10 peptide is a bivalent agonist that mimics APC's cytoprotective, anti-inflammatory, and endothelial barrier-stabilizing actions and APC's protection against endotoxemic mortality.


Asunto(s)
Células Endoteliales , Proteína C , Receptor PAR-1 , Proteína C/metabolismo , Proteína C/química , Humanos , Animales , Receptor PAR-1/agonistas , Receptor PAR-1/metabolismo , Células Endoteliales/metabolismo , Células Endoteliales/efectos de los fármacos , Ratones , Antiinflamatorios/farmacología , Antiinflamatorios/química , Ratones Endogámicos C57BL , Células THP-1 , Trombina/metabolismo , Receptor de Proteína C Endotelial/metabolismo , Receptores de Trombina/agonistas , Receptores de Trombina/metabolismo , Transducción de Señal , Receptores Proteinasa-Activados/agonistas , Receptores Proteinasa-Activados/metabolismo , Péptidos/farmacología , Péptidos/química , Endotoxemia/tratamiento farmacológico , Endotoxemia/metabolismo , Fragmentos de Péptidos/farmacología , Masculino , Modelos Animales de Enfermedad
9.
Am J Physiol Cell Physiol ; 305(12): C1230-9, 2013 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-24025866

RESUMEN

The tubulin cytoskeleton plays a key role in maintaining the characteristic quiescent discoid shape of resting platelets. Upon activation, platelets undergo a dramatic change in shape; however, little is known of how the microtubule system contributes to regulating platelet shape and function. Here we investigated the role of the covalent modification of α-tubulin by acetylation in the regulation of platelet physiology during activation. Superresolution microscopy analysis of the platelet tubulin cytoskeleton showed that the marginal band together with an interconnected web of finer tubulin structures collapsed upon platelet activation with the glycoprotein VI (GPVI)-agonist collagen-related peptide (CRP). Western blot analysis revealed that α-tubulin was acetylated in resting platelets and deacetylated during platelet activation. Tubacin, a specific inhibitor of the tubulin deacetylase HDAC6, prevented tubulin deacetylation upon platelet activation with CRP. Inhibition of HDAC6 upregulated tubulin acetylation and disrupted the organization of the platelet microtubule marginal band without significantly affecting platelet volume changes in response to CRP stimulation. HDAC6 inhibitors also inhibited platelet aggregation in response to CRP and blocked platelet signaling events upstream of platelet Rho GTPase activation. Together, these findings support a role for acetylation signaling in controlling the resting structure of the platelet tubulin marginal band as well as in the coordination of signaling systems that drive platelet cytoskeletal changes and aggregation.


Asunto(s)
Plaquetas/fisiología , Citoesqueleto/fisiología , Histona Desacetilasas/metabolismo , Activación Plaquetaria/fisiología , Transducción de Señal/fisiología , Tubulina (Proteína)/metabolismo , Plaquetas/citología , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Regulación Enzimológica de la Expresión Génica/fisiología , Histona Desacetilasa 6 , Histona Desacetilasas/genética , Humanos , Péptidos/genética , Péptidos/metabolismo
10.
Sci Rep ; 12(1): 15517, 2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-36109550

RESUMEN

Coronavirus disease 2019 (COVID-19) continues to significantly impact the global population, thus countermeasure platforms that enable rapid development of therapeutics against variants of SARS-CoV-2 are essential. We report use of a phage display human antibody library approach to rapidly identify neutralizing antibodies (nAbs) against SARS-CoV-2. We demonstrate the binding and neutralization capability of two nAbs, STI-2020 and STI-5041, against the SARS-CoV-2 WA-1 strain as well as the Alpha and Beta variants. STI-2020 and STI-5041 were protective when administered intravenously or intranasally in the golden (Syrian) hamster model of COVID-19 challenged with the WA-1 strain or Beta variant. The ability to administer nAbs intravenously and intranasally may have important therapeutic implications and Phase 1 healthy subjects clinical trials are ongoing.


Asunto(s)
COVID-19 , Animales , Anticuerpos Monoclonales , Anticuerpos Neutralizantes/uso terapéutico , Anticuerpos Antivirales/uso terapéutico , Cricetinae , Humanos , Mesocricetus , Pruebas de Neutralización , SARS-CoV-2
11.
J Thromb Haemost ; 19(1): 269-280, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33049092

RESUMEN

Essentials Activated protein C (APC) is a serine protease with anticoagulant and cytoprotective effects. We tested whether APC or non-canonical PAR-derived peptides suppress inflammasome activity. APC or PAR1- and PAR3-derived peptides restrict inflammasome-dependent caspase-1 activity. Combined PAR1-derived and PAR3-derived peptides synergistically suppress caspase-1 activity. ABSTRACT: Background Activated protein C (APC) has been shown to restrict murine inflammasome activity. However, whether APC can exert anti-inflammatory activity in part through suppression of inflammasome activation in human systems is unknown. Objectives Studies were made to determine whether either APC or protease activated receptor (PAR)-derived peptides can reduce NLRP3 inflammasome activity in differentiated human THP-1 macrophage-like cells or in primary human monocytes stimulated to activate the inflammasome. Methods Human THP-1 cells or primary human monocytes were differentiated, treated with APC or PAR-derived peptides, and then stimulated with lipopolysaccharide and ATP to induce caspase-1 activity, a product of inflammasome activation. Results Activated protein C or noncanonical PAR1-derived or PAR3-derived peptides significantly reduced caspase-1 activity, detection of fluorescent NLRP3, and IL-1ß release from THP-1 cells. At low concentrations where no effect was observed for each individual peptide, combinations of the PAR1-derived peptide and the PAR3-derived peptide resulted in a significant synergistic decrease in caspase-1 and IL-1ß release. Caspase-1 activity was also reduced in primary human monocytes. Studies using blocking antibodies and small molecule PAR1 inhibitors suggest that EPCR, PAR1, and PAR3 each play roles in the observed anti-inflammatory effects. Several shortened versions of the PAR1- and PAR3-derived peptide reduced caspase-1 activity and exhibited synergistic anti-inflammatory effects. Conclusions The results indicate that both APC and certain PAR1- and PAR3-derived peptides, which are biased agonists for PAR1 or PAR3, can reduce inflammasome activity in stimulated human monocytes as measured by caspase-1 activity and IL-1ß release and that PAR-derived biased peptide agonist combinations are synergistically anti-inflammatory.


Asunto(s)
Inflamasomas , Proteína C , Proteínas Adaptadoras Transductoras de Señales , Antiinflamatorios/farmacología , Caspasa 1/metabolismo , Proteínas de Ciclo Celular , Receptor de Proteína C Endotelial , Humanos , Interleucina-1beta , Macrófagos/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR , Péptidos/farmacología , Proteína C/metabolismo , Receptor PAR-1 , Transducción de Señal , Células THP-1
12.
Ann Thorac Surg ; 107(5): 1571-1581, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30458159

RESUMEN

BACKGROUND: The optimal antithrombotic regimen after bioprosthetic aortic valve replacement (bAVR) is unclear. We conducted a systematic review of various anticoagulation strategies following surgical or transcatheter bAVR (TAVR). METHODS: We searched Medline, PubMed, Embase, Evidence-Based Medicine Reviews, and gray literature through June 2017 for controlled clinical trials and cohort studies that directly compared different antithrombotic strategies in nonpregnant adults who had undergone bAVR. We assessed risk of bias and graded the strength of the evidence using established methods. RESULTS: Of 4,554 titles reviewed, 6 clinical trials and 13 cohort studies met inclusion criteria. We found moderate-strength evidence that mortality, thromboembolic events, and bleeding rates are similar between aspirin and warfarin after surgical bAVR. Observational data suggest lower mortality and thromboembolic events with aspirin combined with warfarin compared with aspirin alone after surgical bAVR, but the effect size is small and the combination is associated with a substantial increase in bleeding risk. We found insufficient evidence for all other treatment comparisons in surgical bAVR. In TAVR patients, we found moderate-strength evidence that mortality, stroke, and major cardiac events are similar between dual antiplatelet therapy and aspirin alone, though a nonsignificantly lower rate of bleeding occurred with aspirin alone. CONCLUSIONS: Treatment with warfarin or aspirin leads to similar outcomes after surgical bAVR. Combining aspirin with warfarin may lead to a small decrease in thromboembolism and mortality, but is accompanied by increased bleeding. For TAVR patients, aspirin is equivalent to dual antiplatelet therapy for reducing thromboembolism and mortality, with a possible decrease in bleeding.


Asunto(s)
Válvula Aórtica , Bioprótesis , Fibrinolíticos/uso terapéutico , Enfermedades de las Válvulas Cardíacas/cirugía , Implantación de Prótesis de Válvulas Cardíacas , Prótesis Valvulares Cardíacas , Humanos
13.
Blood Adv ; 3(20): 3080-3091, 2019 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-31648335

RESUMEN

In humans, platelet count within the normal range is required for physiological hemostasis, but, adversely, platelets also support pathological thrombosis. Moreover, by releasing growth factors, they may enhance neoplastic proliferation. We hypothesize that platelet count correlates with platelet-dependent pathologies, even within the range of hemostatic competence. Because platelet production is promoted by thrombopoietin signaling through the myeloproliferative leukemia virus oncogene (cMPL), a receptor expressed on megakaryocytes, we evaluated the feasibility of selective targeting of hepatic thrombopoietin production to test this hypothesis. We synthesized murine- and primate-specific antisense oligonucleotides (THPO-ASO) that silence hepatic thrombopoietin gene (THPO) expression without blocking extrahepatic THPO. Repeated doses of THPO-ASO were administered to mice and a baboon, causing a sustained 50% decline in plasma thrombopoietin levels and platelet count within 4 weeks in both species. To investigate whether reducing platelet count within the translationally relevant hemostatic range could alter a neoplastic process, we administered THPO-ASO to 6-week-old transgenic MMTV-PyMT mice that develop early ductal atypia that progresses into cMPL-negative fatal metastatic breast cancer within 2 to 3 months. THPO-ASO treatment increased the average time to euthanasia (primary humane endpoint) at 2 cm3 combined palpable tumor volume. Our results show that THPO-ASO reduced blood platelet count, plasma platelet factor 4, vascular endothelial growth factor, thrombopoietin levels, bone marrow megakaryocyte density, tumor growth rate, proliferation index, vascularization, platelet and macrophage content, and pulmonary metastases vs untreated controls. These findings confirm that sustained and moderate pharmacological platelet count reduction is feasible with THPO-ASO administration and can delay progression of certain platelet-dependent pathological processes within a safe hemostatic platelet count range.


Asunto(s)
Neoplasias de la Mama/sangre , Neoplasias de la Mama/etiología , Silenciador del Gen , Hígado/metabolismo , Recuento de Plaquetas , Trombopoyetina/genética , Animales , Neoplasias de la Mama/patología , Movimiento Celular , Transformación Celular Neoplásica , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Haplorrinos , Ratones , Ratones Transgénicos , Estadificación de Neoplasias , Microambiente Tumoral/genética
14.
J Leukoc Biol ; 2018 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-29601101

RESUMEN

Innate immune cells are an essential part of the host defense response, promoting inflammation through release of proinflammatory cytokines or formation of neutrophil extracellular traps. While these processes are important for defense against infectious agents or injury, aberrant activation potentiates pathologic inflammatory disease. Thus, understanding regulatory mechanisms that limit neutrophil extracellular traps formation and cytokine release is of therapeutic interest for targeting pathologic diseases. Activated protein C is an endogenous serine protease with anticoagulant activity as well as anti-inflammatory and cytoprotective functions, the latter of which are mediated through binding cell surface receptors and inducing intracellular signaling. In this review, we discuss certain leukocyte functions, namely neutrophil extracellular traps formation and cytokine release, and the inhibition of these processes by activated protein C.

15.
J Immunol Methods ; 435: 77-84, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27286714

RESUMEN

BACKGROUND: Neutrophils, the most populous innate immune cell type, are the first responders to sites of infection and inflammation. Neutrophils can release their DNA to form extracellular traps (NETs), webs of DNA and granular proteases that contribute to pathogen clearance and promote thrombus formation. At present, the study of NETs is in part limited to the qualitative analysis of fluorescence microscopy-based images, thus quantification of the interactions between NETs and coagulation factors remains ill-defined. AIM: Develop a quantitative method to measure the spatial distribution of DNA and colocalization of coagulation factor binding to neutrophils and NETs utilizing fluorescence-based microscopy. APPROACH: Human neutrophils were purified from peripheral blood, bound to fibronectin and treated with the PKC-activator phorbol myristate acetate (PMA) to induce neutrophil activation and NETs formation. Samples were incubated with purified coagulation factors or plasma before staining with a DNA-binding dye and coagulation factor-specific antibodies. The spatial distribution of DNA and coagulation factors was imaged via fluorescence microscopy and quantified via a custom-built MATLAB-based image analysis algorithm. The algorithm first established global thresholding parameters on a training set of fluorescence image data and then systematically quantified intensity profiles across treatment conditions. Quantitative comparison of treatment conditions was enabled through the normalization of fluorescent intensities using the number of cells per image to determine the percent and area of DNA and coagulation factor binding per cell. RESULTS: Upon stimulation with PMA, NETs formation resulted in an increase in the area of DNA per cell. The coagulation factor fibrinogen bound to both the neutrophil cell body as well as NETs, while prothrombin, FX and FVIIa binding was restricted to the neutrophil cell body. The Gla domain of FX was required to mediate FX-neutrophil binding. Activated protein C (APC), but not Gla-less APC, bound to neutrophil cell bodies and NETs in a punctate manner. Neither FXIIa nor FXIa were found to bind either neutrophil cell bodies or NETs. Fibrinogen binding was dependent on extracellular DNA, while FX and APC required phosphatidylserine exposure for binding to activated neutrophils. CONCLUSIONS: We have developed a quantitative measurement platform to define the spatial localization of fluorescently-labeled coagulation factor binding to neutrophils and extracellular DNA during NETosis.


Asunto(s)
ADN/análisis , Trampas Extracelulares , Microscopía Fluorescente/métodos , Neutrófilos/inmunología , Fenómenos Biofísicos , Factores de Coagulación Sanguínea , ADN/metabolismo , Trampas Extracelulares/inmunología , Técnica del Anticuerpo Fluorescente/métodos , Humanos , Microscopía Fluorescente/instrumentación , Activación Neutrófila , Neutrófilos/ultraestructura , Acetato de Tetradecanoilforbol/farmacología
16.
Cell Mol Bioeng ; 8(4): 543-552, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26640599

RESUMEN

Physical theories of active matter continue to provide a quantitative understanding of dynamic cellular phenomena, including cell locomotion. Although various investigations of the rheology of cells have identified important viscoelastic and traction force parameters for use in these theoretical approaches, a key variable has remained elusive both in theoretical and experimental approaches: the spatiotemporal behavior of the subcellular density. The evolution of the subcellular density has been qualitatively observed for decades as it provides the source of image contrast in label-free imaging modalities (e.g., differential interference contrast, phase contrast) used to investigate cellular specimens. While these modalities directly visualize cell structure, they do not provide quantitative access to the structures being visualized. We present an established quantitative imaging approach, non-interferometric quantitative phase microscopy, to elucidate the subcellular density dynamics in neutrophils undergoing chemokinesis following uniform bacterial peptide stimulation. Through this approach, we identify a power law dependence of the neutrophil mean density on time with a critical point, suggesting a critical density is required for motility on 2D substrates. Next we elucidate a continuum law relating mean cell density, area, and total mass that is conserved during neutrophil polarization and migration. Together, our approach and quantitative findings will enable investigators to define the physics coupling cytoskeletal dynamics with subcellular density dynamics during cell migration.

17.
Thromb Res ; 135(1): 155-60, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25527332

RESUMEN

BACKGROUND: Treatment of chronic myelogenous leukemia (CML) with the BCR-ABL tyrosine kinase inhibitor (TKI) imatinib significantly improves patient outcomes. As some patients are unresponsive to imatinib, next generation BCR-ABL inhibitors such as nilotinib have been developed to treat patients with imatinib-resistant CML. The use of some BCR-ABL inhibitors has been associated with bleeding diathesis, and these inhibitors have been shown to inhibit platelet functions, which may explain the hemostasis impairment. Surprisingly, a new TKI, ponatinib, has been associated with a high incidence of severe acute ischemic cardiovascular events. The mechanism of this unexpected adverse effect remains undefined. OBJECTIVE AND METHODS: This study used biochemical and functional assays to evaluate whether ponatinib was different from the other BCR-ABL inhibitors with respect to platelet activation, spreading, and aggregation. RESULTS AND CONCLUSIONS: Our results show that ponatinib, similar to other TKIs, acts as a platelet antagonist. Ponatinib inhibited platelet activation, spreading, granule secretion, and aggregation, likely through broad spectrum inhibition of platelet tyrosine kinase signaling, and also inhibited platelet aggregate formation in whole blood under shear. As our results indicate that pobatinib inhibits platelet function, the adverse cardiovascular events observed in patients taking ponatinib may be the result of the effect of ponatinib on other organs or cell types, or disease-specific processes, such as BCR-ABL+cells undergoing apoptosis in response to chemotherapy, or drug-induced adverse effects on the integrity of the vascular endothelium in ponatinib-treated patients.


Asunto(s)
Plaquetas/efectos de los fármacos , Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Imidazoles/antagonistas & inhibidores , Motivo de Activación del Inmunorreceptor Basado en Tirosina , Piridazinas/antagonistas & inhibidores , Secuencias de Aminoácidos , Apoptosis , Plaquetas/citología , Colágeno/química , Células Endoteliales/citología , Fibrinógeno/química , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Selectina-P/química , Fosfatidilserinas/química , Fosforilación , Activación Plaquetaria , Agregación Plaquetaria , Resistencia al Corte , Transducción de Señal , Tirosina/química
18.
Cell Mol Bioeng ; 7(1): 1-14, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24563678

RESUMEN

The dynamics of the cellular and molecular constituents of the circulatory system are regulated by the biophysical properties of the heart, vasculature and blood cells and proteins. In this review, we discuss measurement techniques that have been developed to characterize the physical and mechanical parameters of the circulatory system across length scales ranging from the tissue scale (centimeter) to the molecular scale (nanometer) and time scales of years to milliseconds. We compare the utility of measurement techniques as a function of spatial resolution and penetration depth from both a diagnostic and research perspective. Together, this review provides an overview of the utility of measurement science techniques to study the spatial systems of the circulatory system in health and disease.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA