Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Adv Mater ; 36(8): e2304615, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37934471

RESUMEN

The spleen is an important mediator of both adaptive and innate immunity. As such, attempts to modulate the immune response provided by the spleen may be conducive to improved outcomes for numerous diseases throughout the body. Here, biomimicry is used to rationally design nanomaterials capable of splenic retention and immunomodulation for the treatment of disease in a distant organ, the postinfarct heart. Engineered senescent erythrocyte-derived nanotheranostic (eSENTs) are generated, demonstrating significant uptake by the immune cells of the spleen including T and B cells, as well as monocytes and macrophages. When loaded with suberoylanilide hydroxamic acid (SAHA), the nanoagents exhibit a potent therapeutic effect, reducing infarct size by 14% at 72 h postmyocardial infarction when given as a single intravenous dose 2 h after injury. These results are supportive of the hypothesis that RBC-derived biomimicry may provide new approaches for the targeted modulation of the pathological processes involved in myocardial infarction, thus further experiments to decisively confirm the mechanisms of action are currently underway. This novel concept may have far-reaching applicability for the treatment of a number of both acute and chronic conditions where the immune responses are either stimulated or suppressed by the splenic (auto)immune milieu.


Asunto(s)
Biomimética , Infarto del Miocardio , Humanos , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/patología , Corazón , Inmunidad Innata , Inmunomodulación
2.
J Mol Cell Cardiol ; 175: 62-66, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36584478

RESUMEN

Myh6-Cre transgenic mouse line was known to express Cre recombinase only in the heart. Nevertheless, during breeding Myh6-Cre to Rosa26fstdTom reporter (tdTom) mouse line, we observed that a significant part of their F2 tdTom/+ offspring had tdTom reporter gene universally activated. Our results show that Myh6-Cre transgenic mice have Cre recombinase activity in a subpopulation of the male germline cells, and that Myh6 gene transcripts are enriched in the interstitial Leydig cells and the undifferentiated spermatogonia stem cells. In summary, the current study confirms that the previously known "heart-specific" Myh6 promoter drives Cre expression in the testis.


Asunto(s)
Células Germinativas , Integrasas , Masculino , Ratones , Animales , Regiones Promotoras Genéticas/genética , Ratones Transgénicos , Integrasas/genética , Integrasas/metabolismo , Células Germinativas/metabolismo
3.
ACS Sens ; 6(6): 2225-2232, 2021 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-34056903

RESUMEN

Platelets play a prominent role in multiple diseases, in particular arterial and venous thrombosis and also in atherosclerosis and cancer. To advance the in vivo study of the biological activity of this cell type from a basic experimental focus to a clinical focus, new translatable platelet-specific molecular imaging agents are required. Herein, we report the development of a near-infrared fluorescence probe based upon tirofiban, a clinically approved small-molecule glycoprotein IIb/IIIa inhibitor (GPIIb/IIIa). Through in vitro experiments with human platelets and in vivo ones in a murine model of deep-vein thrombosis, we demonstrate the avidity of the generated probe for activated platelets, with the added benefit of a short blood half-life, thereby enabling rapid in vivo visualization within the vasculature.


Asunto(s)
Plaquetas , Inhibidores de Agregación Plaquetaria , Animales , Humanos , Ratones , Imagen Óptica , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria , Tirofibán
4.
Circ Cardiovasc Imaging ; 14(3): e011898, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33724049

RESUMEN

BACKGROUND: The postthrombotic syndrome is a common, often morbid sequela of venous thrombosis (VT) that arises from thrombus persistence and inflammatory scarring of juxtaposed vein walls and valves. Noninvasive 18F-fluorodeoxyglucose (FDG) positron emission tomography/computed tomography (PET/CT) imaging can measure neutrophil inflammation in VT. Here, we hypothesized (1) early fluorodeoxyglucose positron emission tomography/computed tomography (FDG-PET/CT) VT inflammation can predict subsequent vein wall scarring (VWS) and (2) statin therapy can reduce FDG-PET VT inflammation and subsequent VWS. METHODS: C57BL/6J mice (n=75) underwent induction of stasis-induced VT of the inferior vena cava or jugular vein. Inferior vena cava VT mice (n=44) were randomized to daily oral rosuvastatin 5 mg/kg or saline starting at day -1. Subgroups of mice then underwent FDG-PET/CT 2 days after VT induction. On day 14, a subset of mice was euthanized, and VWS was assessed via histology. In vitro studies were further performed on bone marrow-derived neutrophils. RESULTS: Statin therapy reduced early day 2 FDG-PET VT inflammation, thrombus neutrophil influx, and plasma IL (interleukin)-6 levels. At day 14, statin therapy reduced VWS but did not affect day 2 thrombus mass, cholesterol, or white blood counts, nor reduce day 2 glucose transporter 1 or myeloperoxidase expression in thrombus or in isolated neutrophils. In survival studies, the day 2 FDG-PET VT inflammation signal as measured by mean and maximum standardized uptake values predicted the extent of day 14 VWS (area under the receiver operating characteristic curve =0.82) with a strong correlation coefficient (r) of r=0.73 and r=0.74, respectively. Mediation analyses revealed that 40% of the statin-induced VWS reduction was mediated by reductions in VT inflammation as quantified by FDG-PET. CONCLUSIONS: Early noninvasive FDG-PET/CT imaging of VT inflammation predicts the magnitude of subsequent VWS and may provide a new translatable approach to identify individuals at risk for postthrombotic syndrome and to assess anti-inflammatory postthrombotic syndrome therapies, such as statins.


Asunto(s)
Cicatriz/diagnóstico , Fluorodesoxiglucosa F18/farmacología , Tomografía Computarizada por Tomografía de Emisión de Positrones/métodos , Venas/diagnóstico por imagen , Trombosis de la Vena/diagnóstico , Animales , Cicatriz/etiología , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Radiofármacos/farmacología , Trombosis de la Vena/etiología
5.
Circulation ; 143(12): 1224-1238, 2021 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-33445952

RESUMEN

BACKGROUND: Up to 50% of patients with proximal deep vein thrombosis (DVT) will develop the postthrombotic syndrome characterized by limb swelling and discomfort, hyperpigmentation, skin ulcers, and impaired quality of life. Although catheter-based interventions enabling the restoration of blood flow (RBF) have demonstrated little benefit on postthrombotic syndrome, the impact on the acuity of the thrombus and mechanisms underlying this finding remain obscure. In experimental and clinical studies, we examined whether RBF has a restricted time window for improving DVT resolution. METHODS: First, experimental stasis DVT was generated in C57/BL6 mice (n=291) by inferior vena cava ligation. To promote RBF, mice underwent mechanical deligation with or without intravenous recombinant tissue plasminogen activator administered 2 days after deligation. RBF was assessed over time by ultrasonography and intravital microscopy. Resected thrombosed inferior vena cava specimens underwent thrombus and vein wall histological and gene expression assays. Next, in a clinical study, we conducted a post hoc analysis of the ATTRACT (Acute Venous Thrombosis: Thrombus Removal with Adjunctive Catheter-Directed Thrombolysis) pharmacomechanical catheter-directed thrombolysis (PCDT) trial (NCT00790335) to assess the effects of PCDT on Venous Insufficiency Epidemiological and Economic Study quality-of-life and Villalta scores for specific symptom-onset-to-randomization timeframes. RESULTS: Mice that developed RBF by day 4, but not later, exhibited reduced day 8 thrombus burden parameters and reduced day 8 vein wall fibrosis and inflammation, compared with controls. In mice without RBF, recombinant tissue plasminogen activator administered at day 4, but not later, reduced day 8 thrombus burden and vein wall fibrosis. It is notable that, in mice already exhibiting RBF by day 4, recombinant tissue plasminogen activator administration did not further reduce thrombus burden or vein wall fibrosis. In the ATTRACT trial, patients receiving PCDT in an intermediate symptom-onset-to-randomization timeframe of 4 to 8 days demonstrated maximal benefits in Venous Insufficiency Epidemiological and Economic Study quality-of-life and Villalta scores (between-group difference=8.41 and 1.68, respectively, P<0.001 versus patients not receiving PCDT). PCDT did not improve postthrombotic syndrome scores for patients having a symptom-onset-to-randomization time of <4 days or >8 days. CONCLUSIONS: Taken together, these data illustrate that, within a restricted therapeutic window, RBF improves DVT resolution, and PCDT may improve clinical outcomes. Further studies are warranted to examine the value of time-restricted RBF strategies to reduce postthrombotic syndrome in patients with DVT.


Asunto(s)
Circulación Sanguínea/fisiología , Endotelio Vascular/patología , Venas/patología , Trombosis de la Vena/fisiopatología , Animales , Femenino , Humanos , Masculino , Ratones , Calidad de Vida , Resultado del Tratamiento
6.
JACC Basic Transl Sci ; 5(7): 685-695, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32760856

RESUMEN

Paclitaxel drug-coated balloons (DCBs) reduce restenosis, but their overall safety has recently raised concerns. This study hypothesized that DCBs could lessen inflammation and reduce plaque progression. Using 25 rabbits with cholesterol feeding- and balloon injury-induced lesions, DCB-percutaneous transluminal angioplasty (PTA), plain PTA, or sham-PTA (balloon insertion without inflation) was investigated using serial intravascular near-infrared fluorescence-optical coherence tomography and serial intravascular ultrasound. In these experiments, DCB-PTA reduced inflammation and plaque burden in nonobstructive lesions compared with PTA or sham-PTA. These findings indicated the potential for DCBs to serve safely as regional anti-atherosclerosis therapy.

7.
J Am Soc Nephrol ; 31(5): 931-945, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32152232

RESUMEN

BACKGROUND: Arteriovenous fistulas placed surgically for dialysis vascular access have a high primary failure rate resulting from excessive inward remodeling, medial fibrosis, and thrombosis. No clinically established pharmacologic or perisurgical therapies currently address this unmet need. Statins' induction of multiple anti-inflammatory and antithrombotic effects suggests that these drugs might reduce arteriovenous fistula failure. Yet, the in vivo physiologic and molecular effects of statins on fistula patency and maturation remain poorly understood. METHODS: We randomized 108 C57Bl/6J mice to receive daily atorvastatin 1.14 mg/kg or PBS (control) starting 7 days before end-to-side carotid artery-jugular vein fistula creation and for up to 42 days after fistula creation. We then assessed longitudinally the effects of statin therapy on primary murine fistula patency and maturation. We concomitantly analyzed the in vivo arteriovenous fistula thrombogenic and inflammatory macrophage response to statin therapy, using the fibrin-targeted, near-infrared fluorescence molecular imaging agent FTP11-CyAm7 and dextranated, macrophage-avid nanoparticles CLIO-VT680. RESULTS: In vivo molecular-structural imaging demonstrated that atorvastatin significantly reduced fibrin deposition at day 7 and macrophage accumulation at days 7 and 14, findings supported by histopathologic and gene-expression analyses. Structurally, atorvastatin promoted favorable venous limb outward remodeling, preserved arteriovenous fistula blood flow, and prolonged primary arteriovenous fistula patency through day 42 (P<0.05 versus control for all measures). CONCLUSIONS: These findings provide new in vivo evidence that statins improve experimental arteriovenous fistula patency and maturation, indicating that additional clinical evaluation of statin therapy in patients on dialysis undergoing arteriovenous fistula placement is warranted.


Asunto(s)
Derivación Arteriovenosa Quirúrgica , Atorvastatina/uso terapéutico , Fibrina/metabolismo , Macrófagos/efectos de los fármacos , Grado de Desobstrucción Vascular/efectos de los fármacos , Animales , Atorvastatina/farmacología , Arteria Carótida Interna , Colágeno/metabolismo , Femenino , Fibrosis/prevención & control , Hemorreología , Inflamación/prevención & control , Venas Yugulares/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Imagen Molecular , Nanopartículas , ARN Mensajero/biosíntesis , Distribución Aleatoria , Trombosis/prevención & control , Transcripción Genética , Dispositivos de Acceso Vascular
8.
JCI Insight ; 4(2)2019 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-30674723

RESUMEN

Patients with heterozygous missense mutations in the ACTA2 or MYH11 gene are known to exhibit thoracic aortic aneurysm and a risk of early-onset aortic dissection. However, less common phenotypes involving arterial obstruction are also observed, including coronary and cerebrovascular stenotic disease. Herein we implicate the HDAC9 complex in transcriptional silencing of contractile protein-associated genes, known to undergo downregulation in stenotic lesions. Furthermore, neointimal formation was inhibited in HDAC9- or MALAT1-deficient mice with preservation of contractile protein expression. Pharmacologic targeting of the HDAC9 complex through either MALAT1 antisense oligonucleotides or inhibition of the methyltransferase EZH2 (catalytic mediator recruited by the HDAC9 complex) reduced neointimal formation. In conclusion, we report the implication of the HDAC9 complex in stenotic disease and demonstrate that pharmacologic therapy targeting epigenetic complexes can ameliorate arterial obstruction in an experimental system.

9.
JCI Insight ; 3(5)2018 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-29515022

RESUMEN

Loss-of-function mutations in genes encoding contractile proteins have been observed in thoracic aortic aneurysms (TAA). To gain insight into the contribution of contractile protein deficiency in the pathogenesis of TAA, we examined human aneurysm samples. We found multiple contractile gene products deficient in TAA samples, and in particular, expression of SM22α was inversely correlated with aneurysm size. SM22α-deficient mice demonstrated pregnancy-induced aortic dissection, and SM22α deficiency worsened aortic aneurysm in Fbn1C1039G/+ (Marfan) mice, validating this gene product as a TAA effector. We found that repression of SM22α was enforced by increased activity of the methyltransferase EZH2. TGF-ß effectors such as SMAD3 were excluded from binding SM22α-encoding chromatin (TAGLN) in TAA samples, while treatment with the EZH2 inhibitor GSK343 improved cytoskeletal architecture and restored SM22α expression. Finally, inhibition of EZH2 improved aortic performance in Fbn1C1039G/+ mice, in association with restoration of contractile protein expression (including SM22α). Together, these data inform our understanding of contractile protein deficiency in TAA and support the pursuit of chromatin modifying factors as therapeutic targets in aortic disease.


Asunto(s)
Aneurisma de la Aorta Torácica/tratamiento farmacológico , Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Indazoles/administración & dosificación , Músculo Liso Vascular/patología , Piridonas/administración & dosificación , Animales , Aorta Torácica/patología , Aorta Torácica/fisiopatología , Aneurisma de la Aorta Torácica/etiología , Aneurisma de la Aorta Torácica/patología , Aneurisma de la Aorta Torácica/fisiopatología , Metilación de ADN/efectos de los fármacos , Modelos Animales de Enfermedad , Proteína Potenciadora del Homólogo Zeste 2/genética , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Epigénesis Genética/efectos de los fármacos , Fibrilina-1/genética , Técnicas de Inactivación de Genes , Histonas/metabolismo , Humanos , Losartán/administración & dosificación , Síndrome de Marfan/complicaciones , Síndrome de Marfan/genética , Ratones , Ratones Noqueados , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Contracción Muscular/efectos de los fármacos , Contracción Muscular/fisiología , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/patología , ARN Interferente Pequeño/metabolismo
10.
Nat Commun ; 9(1): 1009, 2018 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-29520069

RESUMEN

Thoracic aortic aneurysm (TAA) has been associated with mutations affecting members of the TGF-ß signaling pathway, or components and regulators of the vascular smooth muscle cell (VSMC) actomyosin cytoskeleton. Although both clinical groups present similar phenotypes, the existence of potential common mechanisms of pathogenesis remain obscure. Here we show that mutations affecting TGF-ß signaling and VSMC cytoskeleton both lead to the formation of a ternary complex comprising the histone deacetylase HDAC9, the chromatin-remodeling enzyme BRG1, and the long noncoding RNA MALAT1. The HDAC9-MALAT1-BRG1 complex binds chromatin and represses contractile protein gene expression in association with gain of histone H3-lysine 27 trimethylation modifications. Disruption of Malat1 or Hdac9 restores contractile protein expression, improves aortic mural architecture, and inhibits experimental aneurysm growth. Thus, we highlight a shared epigenetic pathway responsible for VSMC dysfunction in both forms of TAA, with potential therapeutic implication for other known HDAC9-associated vascular diseases.


Asunto(s)
Aneurisma de la Aorta Torácica/genética , ADN Helicasas/genética , Histona Desacetilasas/genética , Músculo Liso Vascular/patología , Proteínas Nucleares/genética , ARN Largo no Codificante/genética , Proteínas Represoras/genética , Factores de Transcripción/genética , Factor de Crecimiento Transformador beta/metabolismo , Actomiosina/genética , Actomiosina/metabolismo , Animales , Aorta/patología , Aneurisma de la Aorta Torácica/patología , Línea Celular , Núcleo Celular/metabolismo , Cromatina/metabolismo , ADN Helicasas/metabolismo , Metilación de ADN , Modelos Animales de Enfermedad , Femenino , Técnica del Anticuerpo Fluorescente , Histona Desacetilasas/metabolismo , Histonas/metabolismo , Humanos , Masculino , Ratones , Ratones Noqueados , Músculo Liso Vascular/citología , Mutación , Miocitos del Músculo Liso , Proteínas Nucleares/metabolismo , Fenotipo , Cultivo Primario de Células , Interferencia de ARN , ARN Largo no Codificante/metabolismo , ARN Interferente Pequeño/metabolismo , Proteínas Represoras/metabolismo , Transducción de Señal/genética , Factores de Transcripción/metabolismo
11.
JCI Insight ; 2(11)2017 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-28570273

RESUMEN

Using transcriptional profiling of platelets from patients presenting with acute myocardial infarction, we identified myeloid-related protein-14 (MRP-14, also known as S100A9) as an acute myocardial infarction gene and reported that platelet MRP-14 binding to platelet CD36 regulates arterial thrombosis. However, whether MRP-14 plays a role in venous thrombosis is unknown. We subjected WT and Mrp-14-deficient (Mrp-14-/-) mice to experimental models of deep vein thrombosis (DVT) by stasis ligation or partial flow restriction (stenosis) of the inferior vena cava. Thrombus weight in response to stasis ligation or stenosis was reduced significantly in Mrp-14-/- mice compared with WT mice. The adoptive transfer of WT neutrophils or platelets, or the infusion of recombinant MRP-8/14, into Mrp-14-/- mice rescued the venous thrombosis defect in Mrp-14-/- mice, indicating that neutrophil- and platelet-derived MRP-14 directly regulate venous thrombogenesis. Stimulation of neutrophils with MRP-14 induced neutrophil extracellular trap (NET) formation, and NETs were reduced in venous thrombi harvested from Mrp-14-/- mice and in Mrp-14-/- neutrophils stimulated with ionomycin. Given prior evidence that MRP-14 also regulates arterial thrombosis, but not hemostasis (i.e., reduced bleeding risk), MRP-14 appears to be a particularly attractive molecular target for treating thrombotic cardiovascular diseases, including myocardial infarction, stroke, and venous thromboembolism.

12.
J Nucl Med ; 58(6): 871-877, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28450558

RESUMEN

Metabolic and molecular imaging continues to advance our understanding of vascular disease pathophysiology. At present, 18F-FDG PET imaging is the most widely used clinical tool for metabolic and molecular imaging of atherosclerosis. However, novel nuclear tracers and intravascular optical near-infrared fluorescence imaging catheters are emerging to assess new biologic targets in vivo and in coronary arteries. This review highlights current metabolic and molecular imaging clinical and near-clinical applications within atherosclerosis and venous thromboembolism, and explores the potential for metabolic and molecular imaging to affect patient-level risk prediction and disease treatment.


Asunto(s)
Aterosclerosis/diagnóstico por imagen , Aterosclerosis/metabolismo , Imagen Molecular/métodos , Tomografía de Emisión de Positrones/métodos , Tromboembolia Venosa/diagnóstico por imagen , Tromboembolia Venosa/metabolismo , Aterosclerosis/patología , Biomarcadores/metabolismo , Humanos , Radiofármacos/farmacocinética , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Tromboembolia Venosa/patología
13.
Thromb Haemost ; 117(2): 339-348, 2017 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-27975098

RESUMEN

Venous thromboembolism is a major cause of death during and immediately post-sepsis. Venous thrombosis (VT) is mediated by cell adhesion molecules and leukocytes, including neutrophil extracellular traps (NETs). Sepsis, or experimentally, endotoxaemia, shares similar characteristics and is modulated via toll like receptor 4 (TLR4). This study was undertaken to determine if endotoxaemia potentiates early stasis thrombogenesis, and secondarily to determine the role of VT TLR4, ICAM-1 and neutrophils (PMNs). Wild-type (WT), ICAM-1-/- and TLR4-/- mice underwent treatment with saline or LPS (10 mg/kg i. p.) alone, or followed by inferior vena cava (IVC) ligation to generate stasis VT. In vivo microscopy of leukocyte trafficking was performed in non-thrombosed mice, and tissue and plasma were harvested during early VT formation. Pre-thrombosis, circulating ICAM-1 was elevated and increased leukocyte adhesion and rolling occurred on the IVC of LPS-treated mice. Post-thrombosis, endotoxaemic mice formed larger, platelet-poor thrombi. Endotoxaemic TLR4-/- mice did not have an augmented thrombotic response and exhibited significantly decreased circulating ICAM-1 compared to endotoxaemic WT controls. Endotoxaemic ICAM-1-/- mice had significantly smaller thrombi compared to controls. Hypothesising that PMNs localised to the inflamed endothelium were promoting thrombosis, PMN depletion using anti-Ly6G antibody was performed. Paradoxically, VT formed without PMNs was amplified, potentially related to endotoxaemia induced elevation of PAI-1 and circulating FXIII, and decreased uPA. Endotoxaemia enhanced early VT occurs in a TLR-4 and ICAM-1 dependent fashion, and is potentiated by neutropenia. ICAM-1 and/or TLR-4 inhibition may be a unique strategy to prevent sepsis-associated VT.


Asunto(s)
Coagulación Sanguínea , Endotoxemia/complicaciones , Molécula 1 de Adhesión Intercelular/metabolismo , Neutropenia/complicaciones , Neutrófilos/metabolismo , Receptor Toll-Like 4/metabolismo , Trombosis de la Vena/etiología , Animales , Plaquetas/metabolismo , Adhesión Celular , Modelos Animales de Enfermedad , Endotoxemia/sangre , Factor VIII/metabolismo , Fibrinógeno/metabolismo , Molécula 1 de Adhesión Intercelular/genética , Rodamiento de Leucocito , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Neutropenia/sangre , Transducción de Señal , Factores de Tiempo , Receptor Toll-Like 4/deficiencia , Receptor Toll-Like 4/genética , Trombosis de la Vena/sangre
14.
Theranostics ; 5(12): 1317-27, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26516370

RESUMEN

Fibrinolytic therapy of venous thromboembolism (VTE) is increasingly utilized, yet limited knowledge is available regarding in vivo mechanisms that govern fibrinolytic efficacy. In particular, it is unknown how age-dependent thrombus organization limits direct blood contact with fibrin, the target of blood-based fibrinolytic agents. Utilizing high-resolution in vivo optical molecular imaging with FTP11, a near-infrared fluorescence (NIRF) fibrin-specific reporter, here we investigated the in vivo interrelationships of blood accessibility to fibrin, thrombus age, thrombus neoendothelialization, and fibrinolysis in murine venous thrombosis (VT). In both stasis VT and non-stasis VT, NIRF microscopy showed that FTP11 fibrin binding was thrombus age-dependent. FTP11 localized to the luminal surface of early-stage VT, but only minimally to subacute VT (p<0.001). Transmission electron microscopy of early stage VT revealed direct blood cell contact with luminal fibrin-rich surfaces. In contrast, subacute VT exhibited an encasing CD31+ neoendothelial layer that limited blood cell contact with thrombus fibrin in both VT models. Next we developed a theranostic strategy to predict fibrinolytic efficacy based on the in vivo fibrin accessibility to blood NIRF signal. Mice with variably aged VT underwent FTP11 injection and intravital microscopy (IVM), followed by tissue plasminogen activator infusion to induce VT fibrinolysis. Fibrin molecular IVM revealed that early stage VT, but not subacute VT, bound FTP11 (p<0.05), and experienced higher rates of fibrinolysis and total fibrinolysis (p<0.05 vs. subacute VT). Before fibrinolysis, the baseline FTP11 NIRF signal predicted the net fibrinolysis at 60 minutes (p<0.001). Taken together, these data provide novel insights into the temporal evolution of VT and its susceptibility to therapeutic fibrinolysis. Fibrin molecular imaging may provide a theranostic strategy to identify venous thrombi amenable to fibrinolytic therapies.


Asunto(s)
Fibrina/análisis , Fibrinolíticos/administración & dosificación , Imagen Molecular/métodos , Trombosis/patología , Trombosis de la Vena/tratamiento farmacológico , Trombosis de la Vena/patología , Animales , Modelos Animales de Enfermedad , Indoles/metabolismo , Masculino , Ratones Endogámicos C57BL , Oligopéptidos/metabolismo , Coloración y Etiquetado/métodos
16.
PLoS One ; 10(2): e0116621, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25680183

RESUMEN

Despite anticoagulation therapy, up to one-half of patients with deep vein thrombosis (DVT) will develop the post-thrombotic syndrome (PTS). Improving the long-term outcome of DVT patients at risk for PTS will therefore require new approaches. Here we investigate the effects of statins--lipid-lowering agents with anti-thrombotic and anti-inflammatory properties--in decreasing thrombus burden and decreasing vein wall injury, mediators of PTS, in established murine stasis and non-stasis chemical-induced venous thrombosis (N = 282 mice). Treatment of mice with daily atorvastatin or rosuvastatin significantly reduced stasis venous thrombus burden by 25% without affecting lipid levels, blood coagulation parameters, or blood cell counts. Statin-driven reductions in VT burden (thrombus mass for stasis thrombi, intravital microscopy thrombus area for non-stasis thrombi) compared similarly to the therapeutic anticoagulant effects of low molecular weight heparin. Blood from statin-treated mice showed significant reductions in platelet aggregation and clot stability. Statins additionally reduced thrombus plasminogen activator inhibitor-1 (PAI-1), tissue factor, neutrophils, myeloperoxidase, neutrophil extracellular traps (NETs), and macrophages, and these effects were most notable in the earlier timepoints after DVT formation. In addition, statins reduced DVT-induced vein wall scarring by 50% durably up to day 21 in stasis VT, as shown by polarized light microscopy of picrosirius red-stained vein wall collagen. The overall results demonstrate that statins improve VT resolution via profibrinolytic, anticoagulant, antiplatelet, and anti-vein wall scarring effects. Statins may therefore offer a new pharmacotherapeutic approach to improve DVT resolution and to reduce the post-thrombotic syndrome, particularly in subjects who are ineligible for anticoagulation therapy.


Asunto(s)
Antiinflamatorios/farmacología , Anticoagulantes/farmacología , Cicatriz/tratamiento farmacológico , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Inhibidores de Agregación Plaquetaria/farmacología , Venas/efectos de los fármacos , Trombosis de la Vena/tratamiento farmacológico , Animales , Antiinflamatorios/uso terapéutico , Anticoagulantes/uso terapéutico , Cicatriz/complicaciones , Fibrinólisis/efectos de los fármacos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Masculino , Ratones , Inhibidores de Agregación Plaquetaria/uso terapéutico , Trombosis de la Vena/complicaciones , Trombosis de la Vena/fisiopatología
17.
Arterioscler Thromb Vasc Biol ; 35(1): 189-96, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25395614

RESUMEN

OBJECTIVE: In vivo assessment of pathological endothelium within arteriovenous fistula (AVF) could provide new insights into inflow stenosis, a common cause of AVF primary failure in end-stage renal disease patients. Here we developed nanoparticle-based imaging strategies to assess pathological endothelium in vivo and elucidate its relationship to neointimal hyperplasia formation in AVF. APPROACH AND RESULTS: Jugular-carotid AVFs were created in C57BL/6 mice (n=38). Pathological endothelium in the AVF was visualized and quantified in vivo using dextranated magnetofluorescent nanoparticles (CLIO-VT680 [cross-linked iron oxide-VivoTag680]). At day 14, CLIO-VT680 was deposited in AVF, but only minimally in sham-operated arteries. Transmission electron microscopy revealed that CLIO-VT680 resided within endothelial cells and in the intimal extracellular space. Endothelial cells of AVF, but not control arteries, expressed vascular cell adhesion molecule-1 and showed augmented endothelial permeability near the anastomosis. Intravital microscopy demonstrated that CLIO-VT680 deposited most intensely near the AVF anastomosis (P<0.0001). The day 14 intravital microscopy CLIO-VT680 signal predicted the subsequent site and magnitude of AVF neointimal hyperplasia at day 42 (r=0.58, P<0.05). CLIO-VT680 deposition in AVF was further visualized by ex vivo MRI. CONCLUSIONS: AVF develop a pathological endothelial response that can be assessed in vivo via nanoparticle-enhanced imaging. AVF endothelium is activated and exhibits augmented permeability, offering a targeting mechanism for nanoparticle deposition and retention in pathological endothelium. The in vivo AVF nanoparticle signal identified and predicted subsequent inflow neointimal hyperplasia. This approach could be used to test therapeutic interventions aiming to restore endothelial health and to decrease early AVF failure caused by inflow stenosis.


Asunto(s)
Fístula Arteriovenosa/patología , Arterias Carótidas/patología , Dextranos , Endotelio Vascular/patología , Colorantes Fluorescentes , Venas Yugulares/patología , Imagen por Resonancia Magnética , Nanopartículas de Magnetita , Microscopía Fluorescente , Animales , Fístula Arteriovenosa/metabolismo , Fístula Arteriovenosa/fisiopatología , Velocidad del Flujo Sanguíneo , Permeabilidad Capilar , Arterias Carótidas/metabolismo , Arterias Carótidas/fisiopatología , Arterias Carótidas/cirugía , Arterias Carótidas/ultraestructura , Proliferación Celular , Constricción Patológica , Modelos Animales de Enfermedad , Endotelio Vascular/metabolismo , Endotelio Vascular/fisiopatología , Endotelio Vascular/cirugía , Endotelio Vascular/ultraestructura , Hiperplasia , Venas Yugulares/metabolismo , Venas Yugulares/fisiopatología , Venas Yugulares/cirugía , Venas Yugulares/ultraestructura , Masculino , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , Neointima , Valor Predictivo de las Pruebas , Flujo Sanguíneo Regional , Factores de Tiempo , Molécula 1 de Adhesión Celular Vascular/metabolismo
18.
Sci Signal ; 7(348): ra100, 2014 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-25336613

RESUMEN

The Ras-related guanosine triphosphatase RhoA mediates pathological cardiac hypertrophy, but also promotes cell survival and is cardioprotective after ischemia/reperfusion injury. To understand how RhoA mediates these opposing roles in the myocardium, we generated mice with a cardiomyocyte-specific deletion of RhoA. Under normal conditions, the hearts from these mice showed functional, structural, and growth parameters similar to control mice. Additionally, the hearts of the cardiomyocyte-specific, RhoA-deficient mice subjected to transverse aortic constriction (TAC)-a procedure that induces pressure overload and, if prolonged, heart failure-exhibited a similar amount of hypertrophy as those of the wild-type mice subjected to TAC. Thus, neither normal cardiac homeostasis nor the initiation of compensatory hypertrophy required RhoA in cardiomyocytes. However, in response to chronic TAC, hearts from mice with cardiomyocyte-specific deletion of RhoA showed greater dilation, with thinner ventricular walls and larger chamber dimensions, and more impaired contractile function than those from control mice subjected to chronic TAC. These effects were associated with aberrant calcium signaling, as well as decreased activity of extracellular signal-regulated kinases 1 and 2 (ERK1/2) and AKT. In addition, hearts from mice with cardiomyocyte-specific RhoA deficiency also showed less fibrosis in response to chronic TAC, with decreased transcriptional activation of genes involved in fibrosis, including myocardin response transcription factor (MRTF) and serum response factor (SRF), suggesting that the fibrotic response to stress in the heart depends on cardiomyocyte-specific RhoA signaling. Our data indicated that RhoA regulates multiple pathways in cardiomyocytes, mediating both cardioprotective (hypertrophy without dilation) and cardio-deleterious effects (fibrosis).


Asunto(s)
Fibrosis Endomiocárdica/enzimología , Insuficiencia Cardíaca/enzimología , Sistema de Señalización de MAP Quinasas , Miocitos Cardíacos/enzimología , Estrés Fisiológico , Proteínas de Unión al GTP rho/metabolismo , Animales , Enfermedades de la Aorta/enzimología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Fibrosis Endomiocárdica/genética , Fibrosis Endomiocárdica/patología , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/patología , Ratones , Ratones Transgénicos , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Miocitos Cardíacos/patología , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas de Unión al GTP rho/genética , Proteína de Unión al GTP rhoA
19.
Circulation ; 130(13): 1044-52, 2014 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-25070665

RESUMEN

BACKGROUND: Accurate detection of recurrent same-site deep vein thrombosis (DVT) is a challenging clinical problem. Because DVT formation and resolution are associated with a preponderance of inflammatory cells, we investigated whether noninvasive (18)F-fluorodeoxyglucose (FDG)-positron emission tomography (PET) imaging could identify inflamed, recently formed thrombi and thereby improve the diagnosis of recurrent DVT. METHODS AND RESULTS: We established a stasis-induced DVT model in murine jugular veins and also a novel model of recurrent stasis DVT in mice. C57BL/6 mice (n=35) underwent ligation of the jugular vein to induce stasis DVT. FDG-PET/computed tomography (CT) was performed at DVT time points of day 2, 4, 7, 14, or 2+16 (same-site recurrent DVT at day 2 overlying a primary DVT at day 16). Antibody-based neutrophil depletion was performed in a subset of mice before DVT formation and FDG-PET/CT. In a clinical study, 38 patients with lower extremity DVT or controls undergoing FDG-PET were analyzed. Stasis DVT demonstrated that the highest FDG signal occurred at day 2, followed by a time-dependent decrease (P<0.05). Histological analyses demonstrated that thrombus neutrophils (P<0.01), but not macrophages, correlated with thrombus PET signal intensity. Neutrophil depletion decreased FDG signals in day 2 DVT in comparison with controls (P=0.03). Recurrent DVT demonstrated significantly higher FDG uptake than organized day 14 DVT (P=0.03). The FDG DVT signal in patients also exhibited a time-dependent decrease (P<0.01). CONCLUSIONS: Noninvasive FDG-PET/CT identifies neutrophil-dependent thrombus inflammation in murine DVT, and demonstrates a time-dependent signal decrease in both murine and clinical DVT. FDG-PET/CT may offer a molecular imaging strategy to accurately diagnose recurrent DVT.


Asunto(s)
Neutrófilos/diagnóstico por imagen , Tomografía de Emisión de Positrones , Trombosis/diagnóstico por imagen , Tomografía Computarizada por Rayos X , Trombosis de la Vena/diagnóstico por imagen , Animales , Estudios de Casos y Controles , Estudios de Cohortes , Modelos Animales de Enfermedad , Femenino , Fluorodesoxiglucosa F18/farmacocinética , Humanos , Ligadura , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Imagen Multimodal , Neutropenia/diagnóstico por imagen , Recurrencia , Estudios Retrospectivos , Sensibilidad y Especificidad , Trombosis/metabolismo , Factores de Tiempo , Trombosis de la Vena/metabolismo
20.
Cell Metab ; 19(2): 246-58, 2014 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-24506866

RESUMEN

Endothelial dysfunction is a central hallmark of diabetes. The transcriptional coactivator PGC-1α is a powerful regulator of metabolism, but its role in endothelial cells remains poorly understood. We show here that endothelial PGC-1α expression is high in diabetic rodents and humans and that PGC-1α powerfully blocks endothelial migration in cell culture and vasculogenesis in vivo. Mechanistically, PGC-1α induces Notch signaling, blunts activation of Rac/Akt/eNOS signaling, and renders endothelial cells unresponsive to established angiogenic factors. Transgenic overexpression of PGC-1α in the endothelium mimics multiple diabetic phenotypes, including aberrant re-endothelialization after carotid injury, blunted wound healing, and reduced blood flow recovery after hindlimb ischemia. Conversely, deletion of endothelial PGC-1α rescues the blunted wound healing and recovery from hindlimb ischemia seen in type 1 and type 2 diabetes. Endothelial PGC-1α thus potently inhibits endothelial function and angiogenesis, and induction of endothelial PGC-1α contributes to multiple aspects of vascular dysfunction in diabetes.


Asunto(s)
Diabetes Mellitus/metabolismo , Células Endoteliales/metabolismo , Factores de Transcripción/metabolismo , Animales , Movimiento Celular/fisiología , Células Cultivadas , Células Endoteliales/citología , Regulación de la Expresión Génica , Miembro Posterior/patología , Humanos , Ratones , Ratones Noqueados , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA