Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
JCI Insight ; 8(19)2023 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-37651191

RESUMEN

Thrombosis is a common complication of advanced cancer, yet the cellular mechanisms linking malignancy to thrombosis are poorly understood. The unfolded protein response (UPR) is an ER stress response associated with advanced cancers. A proteomic evaluation of plasma from patients with gastric and non-small cell lung cancer who were monitored prospectively for venous thromboembolism demonstrated increased levels of UPR-related markers in plasma of patients who developed clots compared with those who did not. Release of procoagulant activity into supernatants of gastric, lung, and pancreatic cancer cells was enhanced by UPR induction and blocked by antagonists of the UPR receptors inositol-requiring enzyme 1α (IRE1α) and protein kinase RNA-like endoplasmic reticulum kinase (PERK). Release of extracellular vesicles bearing tissue factor (EVTFs) from pancreatic cancer cells was inhibited by siRNA-mediated knockdown of IRE1α/XBP1 or PERK pathways. Induction of UPR did not increase tissue factor (TF) synthesis, but rather stimulated localization of TF to the cell surface. UPR-induced TF delivery to EVTFs was inhibited by ADP-ribosylation factor 1 knockdown or GBF1 antagonism, verifying the role of vesicular trafficking. Our findings show that UPR activation resulted in increased vesicular trafficking leading to release of prothrombotic EVTFs, thus providing a mechanistic link between ER stress and cancer-associated thrombosis.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Neoplasias Pancreáticas , Humanos , Proteínas Serina-Treonina Quinasas/metabolismo , Endorribonucleasas/genética , Proteómica , Tromboplastina/metabolismo , Respuesta de Proteína Desplegada , Neoplasias Pancreáticas/complicaciones , Factores de Intercambio de Guanina Nucleótido/metabolismo
2.
Blood ; 139(1): 104-117, 2022 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-34329392

RESUMEN

Tyrosine phosphorylation of extracellular proteins is observed in cell cultures and in vivo, but little is known about the functional roles of tyrosine phosphorylation of extracellular proteins. Vertebrate lonesome kinase (VLK) is a broadly expressed secretory pathway tyrosine kinase present in platelet α-granules. It is released from platelets upon activation and phosphorylates substrates extracellularly. Its role in platelet function, however, has not been previously studied. In human platelets, we identified phosphorylated tyrosines mapped to luminal or extracellular domains of transmembrane and secreted proteins implicated in the regulation of platelet activation. To determine the role of VLK in extracellular tyrosine phosphorylation and platelet function, we generated mice with a megakaryocyte/platelet-specific deficiency of VLK. Platelets from these mice are normal in abundance and morphology but have significant changes in function both in vitro and in vivo. Resting and thrombin-stimulated VLK-deficient platelets exhibit a significant decrease in several tyrosine phosphobands. Results of functional testing of VLK-deficient platelets show decreased protease-activated receptor 4-mediated and collagen-mediated platelet aggregation but normal responses to adenosine 5'-diphosphate. Dense granule and α-granule release are reduced in these platelets. Furthermore, VLK-deficient platelets exhibit decreased protease-activated receptor 4-mediated Akt (S473) and Erk1/2 (T202/Y204) phosphorylation, indicating altered proximal signaling. In vivo, mice lacking VLK in megakaryocytes/platelets display strongly reduced platelet accumulation and fibrin formation after laser-induced injury of cremaster arterioles compared with control mice but with normal bleeding times. These studies show that the secretory pathway tyrosine kinase VLK is critical for stimulus-dependent platelet activation and thrombus formation, providing the first evidence that a secreted protein kinase is required for normal platelet function.


Asunto(s)
Plaquetas/metabolismo , Activación Plaquetaria , Proteínas Tirosina Quinasas/metabolismo , Trombosis/metabolismo , Animales , Plaquetas/patología , Eliminación de Gen , Células HEK293 , Humanos , Ratones Transgénicos , Proteínas Tirosina Quinasas/genética , Trombosis/patología
3.
Blood Adv ; 3(20): 3092-3098, 2019 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-31648331

RESUMEN

In addition to their primary roles in hemostasis and thrombosis, platelets participate in many other physiological and pathological processes, including, but not limited to inflammation, wound healing, tumor metastasis, and angiogenesis. Among their most interesting properties is the large number of bioactive proteins stored in their α-granules, the major storage granule of platelets. We previously showed that platelets differentially package pro- and antiangiogenic proteins in distinct α-granules that undergo differential release upon platelet activation. Nevertheless, how megakaryocytes achieve differential packaging is not fully understood. In this study, we use a mouse megakaryocyte culture system and endocytosis assay to establish when and where differential packaging occurs during platelet production. Live cell microscopy of primary mouse megakaryocytes incubated with fluorescently conjugated fibrinogen and endostatin showed differential endocytosis and packaging of the labeled proteins into distinct α-granule subpopulations. Super-resolution microscopy of mouse proplatelets and human whole-blood platelet α-granules simultaneously probed for 2 different membrane proteins (VAMP-3 and VAMP-8), and multiple granular content proteins (bFGF, ENDO, TSP, VEGF) confirmed differential packaging of protein contents into α-granules. These data suggest that megakaryocytes differentially sort and package α-granule contents, which are preserved as α-granule subpopulations during proplatelet extension and platelet production.


Asunto(s)
Plaquetas/metabolismo , Gránulos Citoplasmáticos/metabolismo , Megacariocitos/metabolismo , Animales , Transporte Biológico , Biomarcadores , Diferenciación Celular , Técnica del Anticuerpo Fluorescente , Humanos , Megacariocitos/citología , Ratones , Trombopoyesis
4.
Proc Natl Acad Sci U S A ; 115(5): E982-E991, 2018 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-29343648

RESUMEN

Stimulation of protease-activated receptor 1 (PAR1) on endothelium by activated protein C (APC) is protective in several animal models of disease, and APC has been used clinically in severe sepsis and wound healing. Clinical use of APC, however, is limited by its immunogenicity and its anticoagulant activity. We show that a class of small molecules termed "parmodulins" that act at the cytosolic face of PAR1 stimulates APC-like cytoprotective signaling in endothelium. Parmodulins block thrombin generation in response to inflammatory mediators and inhibit platelet accumulation on endothelium cultured under flow. Evaluation of the antithrombotic mechanism showed that parmodulins induce cytoprotective signaling through Gßγ, activating a PI3K/Akt pathway and eliciting a genetic program that includes suppression of NF-κB-mediated transcriptional activation and up-regulation of select cytoprotective transcripts. STC1 is among the up-regulated transcripts, and knockdown of stanniocalin-1 blocks the protective effects of both parmodulins and APC. Induction of this signaling pathway in vivo protects against thromboinflammatory injury in blood vessels. Small-molecule activation of endothelial cytoprotection through PAR1 represents an approach for treatment of thromboinflammatory disease and provides proof-of-principle for the strategy of targeting the cytoplasmic surface of GPCRs to achieve pathway selective signaling.


Asunto(s)
Células Endoteliales/metabolismo , Inflamación/metabolismo , Receptor PAR-1/agonistas , Trombosis/metabolismo , Animales , Apoptosis , Factor Xa/metabolismo , Técnicas de Silenciamiento del Gen , Glicoproteínas/genética , Glicoproteínas/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Lipopolisacáridos , Masculino , Ratones , Ratones Endogámicos C57BL , Microcirculación , Péptido Hidrolasas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Transcripción Genética , Regulación hacia Arriba
5.
Cancer Res ; 77(1): 112-122, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-27784746

RESUMEN

Novel agents are needed to improve chemoradiotherapy for locally advanced rectal cancer. In this study, we assessed the ability of CRLX101, an investigational nanoparticle-drug conjugate containing the payload camptothecin (CPT), to improve therapeutic responses as compared with standard chemotherapy. CRLX101 was evaluated as a radiosensitizer in colorectal cancer cell lines and murine xenograft models. CRLX101 was as potent as CPT in vitro in its ability to radiosensitize cancer cells. Evaluations in vivo demonstrated that the addition of CRLX101 to standard chemoradiotherapy significantly increased therapeutic efficacy by inhibiting DNA repair and HIF1α pathway activation in tumor cells. Notably, CRLX101 was more effective than oxaliplatin at enhancing the efficacy of chemoradiotherapy, with CRLX101 and 5-fluorouracil producing the highest therapeutic efficacy. Gastrointestinal toxicity was also significantly lower for CRLX101 compared with CPT when combined with radiotherapy. Our results offer a preclinical proof of concept for CRLX101 as a modality to improve the outcome of neoadjuvant chemoradiotherapy for rectal cancer treatment, in support of ongoing clinical evaluation of this agent (LCC1315 NCT02010567). Cancer Res; 77(1); 112-22. ©2016 AACR.


Asunto(s)
Camptotecina/farmacología , Ciclodextrinas/farmacología , Reparación del ADN/efectos de los fármacos , Subunidad alfa del Factor 1 Inducible por Hipoxia/biosíntesis , Fármacos Sensibilizantes a Radiaciones/farmacología , Neoplasias del Recto/patología , Animales , Western Blotting , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Quimioradioterapia/métodos , Técnica del Anticuerpo Fluorescente , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Nanoconjugados , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Cancer Res ; 76(15): 4493-503, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27325647

RESUMEN

VEGF pathway-targeting antiangiogenic drugs, such as bevacizumab, when combined with chemotherapy have changed clinical practice for the treatment of a broad spectrum of human cancers. However, adaptive resistance often develops, and one major mechanism is elevated tumor hypoxia and upregulated hypoxia-inducible factor-1α (HIF1α) caused by antiangiogenic treatment. Reduced tumor vessel numbers and function following antiangiogenic therapy may also affect intratumoral delivery of concurrently administered chemotherapy. Nonetheless, combining chemotherapy and bevacizumab can lead to improved response rates, progression-free survival, and sometimes, overall survival, the extent of which can partly depend on the chemotherapy backbone. A rational, complementing chemotherapy partner for combination with bevacizumab would not only reduce HIF1α to overcome hypoxia-induced resistance, but also improve tumor perfusion to maintain intratumoral drug delivery. Here, we evaluated bevacizumab and CRLX101, an investigational nanoparticle-drug conjugate containing camptothecin, in preclinical mouse models of orthotopic primary triple-negative breast tumor xenografts, including a patient-derived xenograft. We also evaluated long-term efficacy of CRLX101 and bevacizumab to treat postsurgical, advanced metastatic breast cancer in mice. CRLX101 alone and combined with bevacizumab was highly efficacious, leading to complete tumor regressions, reduced metastasis, and greatly extended survival of mice with metastatic disease. Moreover, CRLX101 led to improved tumor perfusion and reduced hypoxia, as measured by contrast-enhanced ultrasound and photoacoustic imaging. CRLX101 durably suppressed HIF1α, thus potentially counteracting undesirable effects of elevated tumor hypoxia caused by bevacizumab. Our preclinical results show pairing a potent cytotoxic nanoparticle chemotherapeutic that complements and improves concurrent antiangiogenic therapy may be a promising treatment strategy for metastatic breast cancer. Cancer Res; 76(15); 4493-503. ©2016 AACR.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Bevacizumab/uso terapéutico , Camptotecina/uso terapéutico , Ciclodextrinas/uso terapéutico , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Inhibidores de la Angiogénesis/administración & dosificación , Inhibidores de la Angiogénesis/farmacología , Animales , Bevacizumab/administración & dosificación , Bevacizumab/farmacología , Camptotecina/administración & dosificación , Camptotecina/farmacología , Línea Celular Tumoral , Ciclodextrinas/administración & dosificación , Ciclodextrinas/farmacología , Femenino , Humanos , Ratones , Ratones SCID , Nanopartículas , Neoplasias de la Mama Triple Negativas/patología
7.
Blood ; 126(5): 651-60, 2015 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-25999457

RESUMEN

Platelet activation results in profound morphologic changes accompanied by release of granule contents. Recent evidence indicates that fusion of granules with the plasma membrane during activation provides auxiliary membrane to cover growing actin structures. Yet little is known about how membrane fusion is coupled with actin reorganization. Vesicle-associated membrane protein (VAMP)-7 is found on platelet vesicles and possesses an N-terminal longin domain capable of linking exocytosis to cytoskeletal remodeling. We have evaluated platelets from VAMP-7(-/-) mice to determine whether this VAMP isoform contributes to granule release and platelet spreading. VAMP-7(-/-) platelets demonstrated a partial defect in dense granule exocytosis and impaired aggregation. α Granule exocytosis from VAMP-7(-/-) platelets was diminished both in vitro and in vivo during thrombus formation. Consistent with a role of VAMP-7 in cytoskeletal remodeling, spreading on matrices was decreased in VAMP-7(-/-) platelets compared to wild-type controls. Immunoprecipitation of VAMP-7 revealed an association with VPS9-domain ankyrin repeat protein (VARP), an adaptor protein that interacts with both membrane-bound and cytoskeleton proteins and with Arp2/3. VAMP-7, VARP, and Arp2/3 localized to the platelet periphery during spreading. These studies demonstrate that VAMP-7 participates in both platelet granule secretion and spreading and suggest a mechanism whereby VAMP-7 links granule exocytosis with actin reorganization.


Asunto(s)
Activación Plaquetaria/fisiología , Proteínas R-SNARE/sangre , Citoesqueleto de Actina/fisiología , Complejo 2-3 Proteico Relacionado con la Actina/sangre , Animales , Plaquetas/fisiología , Plaquetas/ultraestructura , Gránulos Citoplasmáticos/fisiología , Exocitosis/fisiología , Factores de Intercambio de Guanina Nucleótido/sangre , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Selectina-P/sangre , Agregación Plaquetaria/fisiología , Proteínas R-SNARE/deficiencia , Proteínas R-SNARE/genética
8.
Breast Cancer Res Treat ; 150(3): 559-67, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25833208

RESUMEN

Antiangiogenic therapies inhibit the development of new tumor blood vessels, thereby blocking tumor growth. Despite the advances in developing antiangiogenic agents, clinical data indicate that these drugs have limited efficacy in breast cancer patients. Tumors inevitably develop resistance to antiangiogenics, which is attributed in part to the induction of intra-tumoral hypoxia and stabilization of hypoxia-inducible factor 1α (HIF-1α), a transcription factor that promotes tumor angiogenesis, invasion, metastasis, and cancer stem cell (CSC) self-renewal. Here, we tested whether inhibiting HIF-1α can reverse the stimulatory effects of antiangiogenic-induced hypoxia on breast CSCs. Breast cancer cells grown under hypoxic conditions were treated with the dual topoisomerase-1 (TOPO-1) and HIF-1α inhibitor camptothecin and assessed for their CSC content. In a preclinical model of breast cancer, treatment with bevacizumab was compared to the combination treatment of bevacizumab with CRLX101, an investigational nanoparticle-drug conjugate with a camptothecin payload or CRLX101 monotherapy. While exposure to hypoxia increased the number of breast CSCs, treatment with CPT blocked this effect. In preclinical mouse models, concurrent administration of CRLX101 impeded the induction of both HIF-1α and CSCs in breast tumors induced by bevacizumab treatment. Greater tumor regression and delayed tumor recurrence were observed with the combination of these agents compared to bevacizumab alone. Tumor reimplantation experiments demonstrated that the combination therapy effectively targets the CSC populations. The results from these studies support the combined administration of dual TOPO-1- and HIF-1α-targeted agents like CRLX101 with antiangiogenic agents to increase the efficacy of these treatments.


Asunto(s)
Camptotecina/administración & dosificación , Ciclodextrinas/administración & dosificación , Resistencia a Antineoplásicos/efectos de los fármacos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Células Madre Neoplásicas/efectos de los fármacos , Inhibidores de la Angiogénesis/administración & dosificación , Inhibidores de la Angiogénesis/farmacología , Animales , Protocolos de Quimioterapia Combinada Antineoplásica , Bevacizumab/administración & dosificación , Bevacizumab/farmacología , Camptotecina/farmacología , Línea Celular Tumoral , Ciclodextrinas/farmacología , Femenino , Humanos , Células MCF-7 , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Ratones , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Blood ; 125(12): 1976-85, 2015 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-25587041

RESUMEN

Protease-activated receptor-1 (PAR1) couples the coagulation cascade to platelet activation during myocardial infarction and to endothelial inflammation during sepsis. This receptor demonstrates marked signaling bias. Its activation by thrombin stimulates prothrombotic and proinflammatory signaling, whereas its activation by activated protein C (APC) stimulates cytoprotective and antiinflammatory signaling. A challenge in developing PAR1-targeted therapies is to inhibit detrimental signaling while sparing beneficial pathways. We now characterize a novel class of structurally unrelated small-molecule PAR1 antagonists, termed parmodulins, and compare the activity of these compounds to previously characterized compounds that act at the PAR1 ligand-binding site. We find that parmodulins target the cytoplasmic face of PAR1 without modifying the ligand-binding site, blocking signaling through Gαq but not Gα13 in vitro and thrombus formation in vivo. In endothelium, parmodulins inhibit prothrombotic and proinflammatory signaling without blocking APC-mediated pathways or inducing endothelial injury. In contrast, orthosteric PAR1 antagonists such as vorapaxar inhibit all signaling downstream of PAR1. Furthermore, exposure of endothelial cells to nanomolar concentrations of vorapaxar induces endothelial cell barrier dysfunction and apoptosis. These studies demonstrate how functionally selective antagonism can be achieved by targeting the cytoplasmic face of a G-protein-coupled receptor to selectively block pathologic signaling while preserving cytoprotective pathways.


Asunto(s)
Endotelio Vascular/lesiones , Lactonas/efectos adversos , Piridinas/efectos adversos , Receptor PAR-1/antagonistas & inhibidores , Trombosis/tratamiento farmacológico , Trombosis/prevención & control , Animales , Apoptosis , Sitios de Unión , Células COS , Chlorocebus aethiops , Citoplasma/metabolismo , Endotelio Vascular/efectos de los fármacos , Exocitosis , Células Endoteliales de la Vena Umbilical Humana , Humanos , Inflamación , Ligandos , Inhibidores de Agregación Plaquetaria/química , Proteína C/química , ARN Interferente Pequeño/metabolismo , Transducción de Señal
10.
Clin Cancer Res ; 21(4): 808-18, 2015 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-25524310

RESUMEN

PURPOSE: Increased tumor hypoxia and hence elevated hypoxia-inducible factor-1α (HIF1α) is thought to limit the efficacy of vascular endothelial growth factor (VEGF) pathway-targeting drugs by upregulating adaptive resistance genes. One strategy to counteract this is to combine antiangiogenic drugs with agents able to suppress HIF1α. One such possibility is the investigational drug CRLX101, a nanoparticle-drug conjugate (NDC) containing the payload camptothecin, a known topoisomerase-I poison. EXPERIMENTAL DESIGN: CRLX101 was evaluated both as a monotherapy and combination with bevacizumab in a preclinical mouse model of advanced metastatic ovarian cancer. These preclinical studies contributed to the rationale for undertaking a phase II clinical study to evaluate CRLX101 monotherapy in patients with advanced platinum-resistant ovarian cancer. RESULTS: Preclinically, CRLX101 is highly efficacious as a monotherapy when administered at maximum-tolerated doses. Furthermore, chronic low-dose CRLX101 with bevacizumab reduced bevacizumab-induced HIF1α upregulation and resulted in synergistic efficacy, with minimal toxicity in mice. In parallel, initial data reported here from an ongoing phase II clinical study of CRLX101 monotherapy shows measurable tumor reductions in 74% of patients and a 16% RECIST response rate to date. CONCLUSIONS: Given these preclinical and initial clinical results, further clinical studies are currently evaluating CRLX101 in combination with bevacizumab in ovarian cancer and warrant the evaluation of this therapy combination in other cancer types where HIF1α is implicated in pathogenesis, as it may potentially be able to improve the efficacy of antiangiogenic drugs.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Bevacizumab/administración & dosificación , Camptotecina/administración & dosificación , Ciclodextrinas/administración & dosificación , Nanopartículas/administración & dosificación , Neoplasias Ováricas/patología , Animales , Bevacizumab/efectos adversos , Camptotecina/efectos adversos , Ciclodextrinas/efectos adversos , Sinergismo Farmacológico , Femenino , Humanos , Ratones , Ratones SCID , Nanopartículas/efectos adversos , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Proc Natl Acad Sci U S A ; 110(37): 15127-32, 2013 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-23980155

RESUMEN

Nanoparticles are currently being investigated in a number of human clinical trials. As information on how nanoparticles function in humans is difficult to obtain, animal studies that can be correlative to human behavior are needed to provide guidance for human clinical trials. Here, we report correlative studies on animals and humans for CRLX101, a 20- to 30-nm-diameter, multifunctional, polymeric nanoparticle containing camptothecin (CPT). CRLX101 is currently in phase 2 clinical trials, and human data from several of the clinical investigations are compared with results from multispecies animal studies. The pharmacokinetics of polymer-conjugated CPT (indicative of the CRLX101 nanoparticles) in mice, rats, dogs, and humans reveal that the area under the curve scales linearly with milligrams of CPT per square meter for all species. Plasma concentrations of unconjugated CPT released from CRLX101 in animals and humans are consistent with each other after accounting for differences in serum albumin binding of CPT. Urinary excretion of polymer-conjugated CPT occurs primarily within the initial 24 h after dosing in animals and humans. The urinary excretion dynamics of polymer-conjugated and unconjugated CPT appear similar between animals and humans. CRLX101 accumulates into solid tumors and releases CPT over a period of several days to give inhibition of its target in animal xenograft models of cancer and in the tumors of humans. Taken in total, the evidence provided from animal models on the CRLX101 mechanism of action suggests that the behavior of CRLX101 in animals is translatable to humans.


Asunto(s)
Camptotecina/administración & dosificación , Ciclodextrinas/administración & dosificación , Nanoconjugados/administración & dosificación , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/metabolismo , Animales , Antineoplásicos Fitogénicos/administración & dosificación , Antineoplásicos Fitogénicos/farmacocinética , Antineoplásicos Fitogénicos/uso terapéutico , Área Bajo la Curva , Camptotecina/farmacocinética , Camptotecina/uso terapéutico , Línea Celular Tumoral , Ensayos Clínicos como Asunto , Ciclodextrinas/farmacocinética , Ciclodextrinas/uso terapéutico , Perros , Sistemas de Liberación de Medicamentos , Femenino , Humanos , Ratones , Ratones Desnudos , Nanoconjugados/química , Nanoconjugados/uso terapéutico , Ratas , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/metabolismo , Investigación Biomédica Traslacional
12.
Arterioscler Thromb Vasc Biol ; 33(3): 481-8, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23288151

RESUMEN

OBJECTIVE: Platelet granule exocytosis serves a central role in hemostasis and thrombosis. Recently, single-cell amperometry has shown that platelet membrane fusion during granule exocytosis results in the formation of a fusion pore that subsequently expands to enable the extrusion of granule contents. However, the molecular mechanisms that control platelet fusion pore expansion and collapse are not known. METHODS AND RESULTS: We identified dynamin-related protein-1 (Drp1) in platelets and found that an inhibitor of Drp1, mdivi-1, blocked exocytosis of both platelet dense and α-granules. We used single-cell amperometry to monitor serotonin release from individual dense granules and, thereby, measured the effect of Drp1 inhibition on fusion pore dynamics. Inhibition of Drp1 increased spike width and decreased prespike foot events, indicating that Drp1 influences fusion pore formation and expansion. Platelet-mediated thrombus formation in vivo after laser-induced injury of mouse cremaster arterioles was impaired after infusion of mdivi-1. CONCLUSIONS: These results demonstrate that inhibition of Drp1 disrupts platelet fusion pore dynamics and indicate that Drp1 can be targeted to control thrombus formation in vivo.


Asunto(s)
Plaquetas/metabolismo , Dinaminas/sangre , Exocitosis , GTP Fosfohidrolasas/sangre , Fusión de Membrana , Proteínas Asociadas a Microtúbulos/sangre , Proteínas Mitocondriales/sangre , Vesículas Secretoras/metabolismo , Trombosis/sangre , Lesiones del Sistema Vascular/sangre , Animales , Arteriolas/lesiones , Plaquetas/efectos de los fármacos , Modelos Animales de Enfermedad , Dinaminas/antagonistas & inhibidores , Exocitosis/efectos de los fármacos , Fibrinolíticos/farmacología , GTP Fosfohidrolasas/antagonistas & inhibidores , Humanos , Rayos Láser , Fusión de Membrana/efectos de los fármacos , Ratones , Proteínas Asociadas a Microtúbulos/antagonistas & inhibidores , Proteínas Mitocondriales/antagonistas & inhibidores , Selectina-P/sangre , Quinazolinonas/farmacología , Conejos , Vesículas Secretoras/efectos de los fármacos , Serotonina/sangre , Trombosis/etiología , Trombosis/prevención & control , Factores de Tiempo , Lesiones del Sistema Vascular/etiología
13.
Blood ; 120(1): 199-206, 2012 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-22589474

RESUMEN

There has been recent controversy as to whether platelet α-granules represent a single granule population or are composed of different subpopulations that serve discrete functions. To address this question, we evaluated the localization of vesicle-associated membrane proteins (VAMPs) in spread platelets to determine whether platelets actively sort a specific subpopulation of α-granules to the periphery during spreading. Immunofluorescence microscopy demonstrated that granules expressing VAMP-3 and VAMP-8 localized to the central granulomere of spread platelets along with the granule cargos von Willebrand factor and serotonin. In contrast, α-granules expressing VAMP-7 translocated to the periphery of spread platelets along with the granule cargos TIMP2 and VEFG. Time-lapse microscopy demonstrated that α-granules expressing VAMP-7 actively moved from the granulomere to the periphery during spreading. Platelets from a patient with gray platelet syndrome lacked α-granules and demonstrated only minimal spreading. Similarly, spreading was impaired in platelets obtained from Unc13d(Jinx) mice, which are deficient in Munc13-4 and have an exocytosis defect. These studies identify a new α-granule subtype expressing VAMP-7 that moves to the periphery during spreading, supporting the premise that α-granules are heterogeneous and demonstrating that granule exocytosis is required for platelet spreading.


Asunto(s)
Plaquetas/metabolismo , Gránulos Citoplasmáticos/metabolismo , Exocitosis/fisiología , Síndrome de Plaquetas Grises/metabolismo , Proteínas R-SNARE/metabolismo , Animales , Preescolar , Femenino , Humanos , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Persona de Mediana Edad , Transporte de Proteínas/fisiología , Seudópodos/metabolismo , Proteína 3 de Membrana Asociada a Vesículas/metabolismo
14.
Biochemistry ; 49(21): 4533-42, 2010 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-20429610

RESUMEN

Following platelet activation, platelets undergo a dramatic shape change mediated by the actin cytoskeleton and accompanied by secretion of granule contents. While the actin cytoskeleton is thought to influence platelet granule secretion, the mechanism for this putative regulation is not known. We found that disruption of the actin cytoskeleton by latrunculin A inhibited alpha-granule secretion induced by several different platelet agonists without significantly affecting activation-induced platelet aggregation. In a cell-free secretory system, platelet cytosol was required for alpha-granule secretion. Inhibition of actin polymerization prevented alpha-granule secretion in this system, and purified platelet actin could substitute for platelet cytosol to support alpha-granule secretion. To determine whether SNAREs physically associate with the actin cytoskeleton, we isolated the Triton X-100 insoluble actin cytoskeleton from platelets. VAMP-8 and syntaxin-2 associated only with actin cytoskeletons of activated platelets. Syntaxin-4 and SNAP-23 associated with cytoskeletons isolated from either resting or activated platelets. When syntaxin-4 and SNAP-23 were tested for actin binding in a purified protein system, only syntaxin-4 associated directly with polymerized platelet actin. These data show that the platelet cytoskeleton interacts with select SNAREs and that actin polymerization facilitates alpha-granule release.


Asunto(s)
Plaquetas/metabolismo , Gránulos Citoplasmáticos/metabolismo , Citoesqueleto/metabolismo , Activación Plaquetaria/fisiología , Proteínas SNARE/metabolismo , Actinas/inmunología , Actinas/metabolismo , Plaquetas/fisiología , Compuestos Bicíclicos Heterocíclicos con Puentes , Gránulos Citoplasmáticos/inmunología , Citoesqueleto/inmunología , Citoesqueleto/fisiología , Humanos , Octoxinol/metabolismo , Activación Plaquetaria/efectos de los fármacos , Activación Plaquetaria/inmunología , Proteínas Qa-SNARE/inmunología , Proteínas Qa-SNARE/metabolismo , Sintaxina 1/metabolismo , Tiazolidinas , beta-Tromboglobulina
15.
Am J Physiol Gastrointest Liver Physiol ; 295(3): G629-40, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18669626

RESUMEN

An organotypic slice preparation of the adult mouse parotid salivary gland amenable to a variety of optical assessments of fluid and protein secretion dynamics is described. The semi-intact preparation rendered without the use of enzymatic treatment permitted live-cell imaging and multiphoton analysis of cellular and supracellular signals. Toward this end we demonstrated that the parotid slice is a significant addition to the repertoire of tools available to investigators to probe exocrine structure and function since there is currently no cell culture system that fully recapitulates parotid acinar cell biology. Importantly, we show that a subpopulation of the acinar cells of parotid slices can be maintained in short-term culture and retain their morphology and function for up to 2 days. This in vitro model system is a significant step forward compared with enzymatically dispersed acini that rapidly lose their morphological and functional characteristics over several hours, and it was shown to be long enough for the expression and trafficking of exogenous protein following adenoviral infection. This system is compatible with a variety of genetic and physiological approaches used to study secretory function.


Asunto(s)
Señalización del Calcio , Exocitosis , Glándula Parótida/citología , Glándula Parótida/metabolismo , Actinas/metabolismo , Adenoviridae/genética , Agonistas Adrenérgicos/farmacología , Animales , Apoptosis , Señalización del Calcio/efectos de los fármacos , Polaridad Celular , Proliferación Celular , Forma de la Célula , Supervivencia Celular , Agonistas Colinérgicos/farmacología , Estimulación Eléctrica , Exocitosis/efectos de los fármacos , Vectores Genéticos , Masculino , Ratones , Microscopía Confocal , Microscopía de Fluorescencia por Excitación Multifotónica , Glándula Parótida/efectos de los fármacos , Glándula Parótida/ultraestructura , Transporte de Proteínas , Proteínas Recombinantes de Fusión/metabolismo , Vesículas Secretoras/metabolismo , Factores de Tiempo , Técnicas de Cultivo de Tejidos , Transducción Genética
16.
J Mol Cell Cardiol ; 40(3): 361-74, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16458920

RESUMEN

Atrial cardiac myocytes secrete the vasoactive hormone atrial natriuretic peptide (ANP) by both constitutive and regulated exocytotic fusion of ANP-containing large dense core vesicles (LDCV) with the sarcolemma. Detailed information, however, regarding the identity and function of specific membrane fusion proteins (SNARE proteins) involved in exocytosis in the endocrine heart is lacking. In the current study, we identified SNARE proteins and determined their association with ANP-containing secretory granules using primary cultures of neonatal and adult rat atrial cardiac myocytes. Using RT-PCR, cardiac myocytes were screened for SNARE and SNARE-associated transcripts. Identified SNARE proteins that have been implicated in exocytosis in neuroendocrine cells were further characterized by Western blot analysis. Functional interaction between SNARE proteins was demonstrated using immunoprecipitation. Using cell fractionation and immunocytochemical methods, it was revealed that VAMP-1, VAMP-2 and synaptotagmin-1 (the putative Ca(2+) sensor) localized to subpopulations of ANP-containing secretory granules in atrial myocytes. Currently, there is conflicting data regarding the role of Ca(2+) in ANP exocytosis. To judge whether secretory activity could be evoked by intracellular Ca(2+) elevation, time-resolved membrane capacitance measurements were used in combination with the flash photolysis of caged compounds to follow the exocytotic activity of single neonatal atrial myocytes. These studies demonstrated that multiple SNARE proteins are present in neonatal and adult cardiac myocytes and suggest the importance of Ca(2+) in exocytosis of ANP from neonatal atrial cardiac myocytes.


Asunto(s)
Miocitos Cardíacos/metabolismo , Proteínas SNARE/metabolismo , Animales , Animales Recién Nacidos , Factor Natriurético Atrial/metabolismo , Western Blotting , Células Cultivadas , Atrios Cardíacos/citología , Inmunohistoquímica , Masculino , Microscopía Confocal , Técnicas de Placa-Clamp , Pruebas de Precipitina , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Q-SNARE/genética , Proteínas Q-SNARE/metabolismo , Proteínas Qa-SNARE/genética , Proteínas Qa-SNARE/metabolismo , Proteínas R-SNARE/genética , Proteínas R-SNARE/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas SNARE/genética , Fracciones Subcelulares , Sinaptotagmina I/genética , Sinaptotagmina I/metabolismo , Proteína 1 de Membrana Asociada a Vesículas/genética , Proteína 1 de Membrana Asociada a Vesículas/metabolismo , Proteína 2 de Membrana Asociada a Vesículas/genética , Proteína 2 de Membrana Asociada a Vesículas/metabolismo , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...