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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Circ Res ; 133(6): 463-480, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37555328

RESUMEN

BACKGROUND: Cardiac valve disease is observed in 2.5% of the general population and 10% of the elderly people. Effective pharmacological treatments are currently not available, and patients with severe cardiac valve disease require surgery. PROX1 (prospero-related homeobox transcription factor 1) and FOXC2 (Forkhead box C2 transcription factor) are transcription factors that are required for the development of lymphatic and venous valves. We found that PROX1 and FOXC2 are expressed in a subset of valvular endothelial cells (VECs) that are located on the downstream (fibrosa) side of cardiac valves. Whether PROX1 and FOXC2 regulate cardiac valve development and disease is not known. METHODS: We used histology, electron microscopy, and echocardiography to investigate the structure and functioning of heart valves from Prox1ΔVEC mice in which Prox1 was conditionally deleted from VECs. Isolated valve endothelial cells and valve interstitial cells were used to identify the molecular mechanisms in vitro, which were tested in vivo by RNAScope, additional mouse models, and pharmacological approaches. The significance of our findings was tested by evaluation of human samples of mitral valve prolapse and aortic valve insufficiency. RESULTS: Histological analysis revealed that the aortic and mitral valves of Prox1ΔVEC mice become progressively thick and myxomatous. Echocardiography revealed that the aortic valves of Prox1ΔVEC mice are stenotic. FOXC2 was downregulated and PDGF-B (platelet-derived growth factor-B) was upregulated in the VECs of Prox1ΔVEC mice. Conditional knockdown of FOXC2 and conditional overexpression of PDGF-B in VECs recapitulated the phenotype of Prox1ΔVEC mice. PDGF-B was also increased in mice lacking FOXC2 and in human mitral valve prolapse and insufficient aortic valve samples. Pharmacological inhibition of PDGF-B signaling with imatinib partially ameliorated the valve defects of Prox1ΔVEC mice. CONCLUSIONS: PROX1 antagonizes PDGF-B signaling partially via FOXC2 to maintain the extracellular matrix composition and prevent myxomatous degeneration of cardiac valves.


Asunto(s)
Enfermedades de las Válvulas Cardíacas , Prolapso de la Válvula Mitral , Animales , Humanos , Ratones , Células Endoteliales/metabolismo , Enfermedades de las Válvulas Cardíacas/genética , Enfermedades de las Válvulas Cardíacas/prevención & control , Enfermedades de las Válvulas Cardíacas/metabolismo , Válvula Mitral/metabolismo , Prolapso de la Válvula Mitral/metabolismo , Factores de Transcripción/metabolismo , Proteínas Proto-Oncogénicas c-sis/metabolismo
2.
Proc Natl Acad Sci U S A ; 119(34): e2207592119, 2022 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-35969769

RESUMEN

Vaso-occlusive episode (VOE) is a common and critical complication of sickle cell disease (SCD). Its pathogenesis is incompletely understood. von Willebrand factor (VWF), a multimeric plasma hemostatic protein synthesized and secreted by endothelial cells and platelets, is increased during a VOE. However, whether and how VWF contributes to the pathogenesis of VOE is not fully understood. In this study, we found increased VWF levels during tumor necrosis factor (TNF)-induced VOE in a humanized mouse model of SCD. Deletion of endothelial VWF decreased hemolysis, vascular occlusion, and organ damage caused by TNF-induced VOE in SCD mice. Moreover, administering ADAMTS13, the VWF-cleaving plasma protease, reduced plasma VWF levels, decreased inflammation and vaso-occlusion, and alleviated organ damage during VOE. These data suggest that promoting VWF cleavage via ADAMTS13 may be an effective treatment for reducing hemolysis, inflammation, and vaso-occlusion during VOE.


Asunto(s)
Anemia de Células Falciformes , Enfermedades Vasculares , Factor de von Willebrand , Proteína ADAMTS13/metabolismo , Proteína ADAMTS13/farmacología , Proteína ADAMTS13/uso terapéutico , Animales , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Eliminación de Gen , Hemólisis/efectos de los fármacos , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Ratones , Enfermedades Vasculares/tratamiento farmacológico , Enfermedades Vasculares/etiología , Factor de von Willebrand/genética , Factor de von Willebrand/metabolismo
3.
Am J Pathol ; 192(9): 1282-1294, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35640675

RESUMEN

Apart from autopsy, tissue correlates of coronavirus disease 2019 (COVID-19) clinical stage are lacking. In the current study, cutaneous punch biopsy specimens of 15 individuals with severe/critical COVID-19 and six with mild/moderate COVID-19 were examined. Evidence for arterial and venous microthrombi, deposition of C5b-9 and MASP2 (representative of alternative and lectin complement pathways, respectively), and differential expression of interferon type I-driven antiviral protein MxA (myxovirus resistance A) versus SIN3A, a promoter of interferon type I-based proinflammatory signaling, were assessed. Control subjects included nine patients with sepsis-related acute respiratory distress syndrome (ARDS) and/or acute kidney injury (AKI) pre-COVID-19. Microthrombi were detected in 13 (87%) of 15 patients with severe/critical COVID-19 versus zero of six patients with mild/moderate COVID-19 (P < 0.001) and none of the nine patients with pre-COVID-19 ARDS/AKI (P < 0.001). Cells lining the microvasculature staining for spike protein of severe acute respiratory syndrome coronavirus 2, the etiologic agent of COVID-19, also expressed tissue factor. C5b-9 deposition occurred in 13 (87%) of 15 patients with severe/critical COVID-19 versus zero of six patients with mild/moderate COVID-19 (P < 0.001) and none of the nine patients with pre-COVID-19 ARDS/AKI (P < 0.001). MASP2 deposition was also restricted to severe/critical COVID-19 cases. MxA expression occurred in all six mild/moderate versus two (15%) of 13 severe/critical cases (P < 0.001) of COVID-19. In contrast, SIN3A was restricted to severe/critical COVID-19 cases co-localizing with severe acute respiratory syndrome coronavirus 2 spike protein. SIN3A was also elevated in plasma of patients with severe/critical COVID-19 versus control subjects (P ≤ 0.02). In conclusion, the study identified premortem tissue correlates of COVID-19 clinical stage using skin. If validated in a longitudinal cohort, this approach could identify individuals at risk for disease progression and enable targeted interventions.


Asunto(s)
Lesión Renal Aguda , COVID-19 , Interferón Tipo I , Síndrome de Dificultad Respiratoria , Trombosis , Antivirales , Biopsia , Complejo de Ataque a Membrana del Sistema Complemento , Humanos , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa , Glicoproteína de la Espiga del Coronavirus
4.
Blood ; 132(13): 1426-1437, 2018 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-30068506

RESUMEN

Inflammation is a major contributor to deep vein thrombosis (DVT). Flow restriction of the inferior vena cava (IVC) in mice induces DVT like that in humans. In this model, P-selectin-dependent adhesion of neutrophils and monocytes leads to release of neutrophil extracellular traps (NETs) and expression of tissue factor. However, it is not known what signals cause myeloid cells to generate these procoagulant effectors. Using ultrasonography and spinning-disk intravital microscopy in genetically engineered mice, we found that engagement of P-selectin glycoprotein ligand-1 (PSGL-1) and the chemokine receptor CXCR2 on rolling neutrophils propagated signals that cooperated to induce ß2 integrin-dependent arrest in flow-restricted IVCs. Unlike previous reports, PSGL-1 signaling in neutrophils did not require L-selectin, and it used tyrosine 145 rather than tyrosines 112 and 128 on the adaptor Src homology domain-containing leukocyte phosphoprotein of 76 kDa. PSGL-1 and CXCR2 signaling cooperated to increase the frequency and size of thrombi, in part by stimulating release of NETs. Unlike in neutrophils, blocking PSGL-1 or CXCR2 signaling in monocytes did not affect their recruitment into thrombi or their expression of tissue factor. Our results demonstrate that neutrophils cooperatively signal through PSGL-1 and CXCR2 to promote DVT.


Asunto(s)
Glicoproteínas de Membrana/metabolismo , Neutrófilos/metabolismo , Receptores de Interleucina-8B/metabolismo , Transducción de Señal , Trombosis de la Vena/metabolismo , Animales , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Neutrófilos/patología , Trombosis de la Vena/patología
5.
Arterioscler Thromb Vasc Biol ; 34(9): 1924-32, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24903096

RESUMEN

OBJECTIVE: Aortic valve stenosis (AS) is characterized by fibrosis and calcification of valves leading to aortic valve narrowing, resulting in high wall shear stress across the valves. We previously demonstrated that high shear stress can activate platelet-derived transforming growth factor-ß1 (TGF-ß1), a cytokine that induces fibrosis and calcification. The aim of this study was to investigate the role of shear-induced platelet release of TGF-ß1 and its activation in AS. APPROACH AND RESULTS: We studied hypercholesterolemic Ldlr(-/-)Apob(100/100)/Mttp(fl/fl)/Mx1Cre(+/+) (Reversa) mice that develop AS on Western diet and a surgical ascending aortic constriction mouse model that acutely simulates the hemodynamics of AS to study shear-induced platelet TGF-ß1 release and activation. Reversa mice on Western diet for 6 months had thickening of the aortic valves, increased wall shear stress, and increased plasma TGF-ß1 levels. There were weak and moderate correlations between wall shear stress and TGF-ß1 levels in the progression and reversed Reversa groups and a stronger correlation in the ascending aortic constriction model in wild-type mice but not in mice with a targeted deletion of megakaryocyte and platelet TGF-ß1 (Tgfb1(flox)). Plasma total TGF-ß1 levels correlated with collagen deposition in the stenotic valves in Reversa mice. Although active TGF-ß1 levels were too low to be measured directly, we found (1) canonical TGF-ß1 (phosphorylated small mothers against decapentaplegic 2/3) signaling in the leukocytes and canonical and noncanonical (phosphorylated extracellular signal-regulated kinases 1/2) TGF-ß1 signaling in aortic valves of Reversa mice on a Western diet, and (2) TGF-ß1 signaling of both pathways in the ascending aortic constriction stenotic area in wild-type but not Tgfb1(flox) mice. CONCLUSIONS: Shear-induced, platelet-derived TGF-ß1 activation may contribute to AS.


Asunto(s)
Estenosis de la Válvula Aórtica/etiología , Plaquetas/metabolismo , Hemorreología , Estrés Mecánico , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Válvula Aórtica/patología , Estenosis de la Válvula Aórtica/sangre , Estenosis de la Válvula Aórtica/fisiopatología , Apolipoproteína B-100/genética , Calcinosis/sangre , Calcinosis/etiología , Calcinosis/fisiopatología , Colágeno/metabolismo , Dieta Aterogénica , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Fibrosis , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Hipercolesterolemia/etiología , Hipercolesterolemia/genética , Hipercolesterolemia/metabolismo , Leucocitos/metabolismo , Sistema de Señalización de MAP Quinasas , Ratones , Receptores de LDL/deficiencia , Transducción de Señal , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta1/farmacología
6.
J Biol Chem ; 288(15): 10628-39, 2013 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-23463512

RESUMEN

TGF-ß1 is a disulfide-bonded homodimeric protein produced by platelets and other cells that plays a role in many physiologic and pathologic processes. TGF-ß1 is secreted as an inactive large latent complex (LLC) comprised of TGF-ß1, latency-associated peptide, and latent TGF-ß binding protein 1. We previously demonstrated that shear force can activate LLC and that thiol-disulfide exchange contributes to the process. We have now investigated the role of thiol isomerases in the activation of LLC in platelet releasates (PR) and recombinant LLC. The wasp venom peptide mastoparan, which inhibits the chaperone activity of PDI, inhibited stirring- and shear-induced activation of latent TGF-ß1 by 90 and 75% respectively. To identify the proteins that bind to mastoparan either directly or indirectly, PR were chromatographed on a mastoparan affinity column. Latent TGF-ß binding protein 1, latency-associated peptide, TGF-ß1, clusterin, von Willebrand factor, multimerin-1, protein disulfide isomerase (PDI), ERp5, ERp57, and ERp72 eluted specifically from the column. Anti-PDI RL90 attenuated the inhibitory effect of mastoparan on LLC activation. Furthermore, reduced PDI inhibited activation of PR LLC, whereas oxidized PDI had no effect. We conclude that thiol isomerases and thiol-disulfide exchange contribute to TGF-ß1 activation and identify a number of molecules that may participate in the process.


Asunto(s)
Plaquetas/metabolismo , Proteínas de Unión a TGF-beta Latente/metabolismo , Péptidos/farmacología , Proteína Disulfuro Isomerasas/antagonistas & inhibidores , Factor de Crecimiento Transformador beta1/metabolismo , Venenos de Avispas/farmacología , Proteínas Sanguíneas/metabolismo , Línea Celular , Clusterina/metabolismo , Disulfuros/metabolismo , Femenino , Humanos , Péptidos y Proteínas de Señalización Intercelular , Masculino , Unión Proteica/efectos de los fármacos , Proteína Disulfuro Isomerasas/metabolismo , Factor de von Willebrand/metabolismo
7.
Am J Physiol Heart Circ Physiol ; 307(10): H1529-38, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25239803

RESUMEN

Bone marrow-derived inflammatory cells, including platelets, may contribute to the progression of pressure overload-induced left ventricular hypertrophy (LVH). However, the underlying mechanisms for this are still unclear. One potential mechanism is through release of granule cargo. Unc13-d(Jinx) (Jinx) mice, which lack Munc13-4, a limiting factor in vesicular priming and fusion, have granule secretion defects in a variety of hematopoietic cells, including platelets. In the current study, we investigated the role of granule secretion in the development of LVH and cardiac remodeling using chimeric mice specifically lacking Munc13-4 in marrow-derived cells. Pressure overload was elicited by transverse aortic constriction (TAC). Chimeric mice were created by bone marrow transplantation. Echocardiography, histology staining, immunohistochemistry, real-time polymerase chain reaction, enzyme-linked immunosorbent assay, and mass spectrometry were used to study LVH progression and inflammatory responses. Wild-type (WT) mice that were transplanted with WT bone marrow (WT→WT) and WT mice that received Jinx bone marrow (Jinx→WT) developed LVH and a classic fetal reprogramming response early (7 days) after TAC. However, at late times (5 wk), mice lacking Munc13-4 in bone marrow-derived cells (Jinx→WT) failed to sustain the cardiac hypertrophy observed in WT chimeric mice. No difference in cardiac fibrosis was observed at early or late time points. Reinjection of WT platelets or platelet releasate partially restored cardiac hypertrophy in Jinx chimeric mice. These results suggest that sustained LVH in the setting of pressure overload depends on one or more factors secreted from bone marrow-derived cells, possibly from platelets. Inhibiting granule cargo release may represent a novel target for preventing sustained LVH.


Asunto(s)
Plaquetas/metabolismo , Células de la Médula Ósea/metabolismo , Hipertrofia Ventricular Izquierda/metabolismo , Proteínas de la Membrana/metabolismo , Miocardio/metabolismo , Vesículas Secretoras/metabolismo , Animales , Trasplante de Médula Ósea , Modelos Animales de Enfermedad , Hipertensión/complicaciones , Hipertrofia Ventricular Izquierda/etiología , Hipertrofia Ventricular Izquierda/patología , Hipertrofia Ventricular Izquierda/prevención & control , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/patología , Transfusión de Plaquetas , Factores de Tiempo , Función Ventricular Izquierda , Remodelación Ventricular
8.
Blood ; 119(4): 1064-74, 2012 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-22134166

RESUMEN

Circulating platelets contain high concentrations of TGF-ß1 in their α-granules and release it on platelet adhesion/activation. We hypothesized that uncontrolled in vitro release of platelet TGF-ß1 may confound measurement of plasma TGF-ß1 in mice and that in vivo release and activation may contribute to cardiac pathology in response to constriction of the transverse aorta, which produces both high shear and cardiac pressure overload. Plasma TGF-ß1 levels in blood collected from C57Bl/6 mice by the standard retro-bulbar technique were much higher than those obtained when prostaglandin E1 was added to inhibit release or when blood was collected percutaneously from the left ventricle under ultrasound guidance. Even with optimal blood drawing, plasma TGF-ß1 was lower in mice rendered profoundly thrombocytopenic or mice with selectively low levels of platelet TGF-ß1 because of megakaryocyte-specific disruption of their TGF-ß1 gene (Tgfb1(flox)). Tgfb1(flox) mice were also partially protected from developing cardiac hypertrophy, fibrosis, and systolic dysfunction in response to transverse aortic constriction. These studies demonstrate that plasma TGF-ß1 levels can be assessed accurately, but it requires special precautions; that platelet TGF-ß1 contributes to plasma levels of TGF-ß1; and that platelet TGF-ß1 contributes to the pathologic cardiac changes that occur in response to aortic constriction.


Asunto(s)
Plaquetas/metabolismo , Corazón/fisiopatología , Hipertensión/fisiopatología , Miocardio/patología , Factor de Crecimiento Transformador beta1/metabolismo , Alprostadil/metabolismo , Animales , Plaquetas/patología , Vasos Sanguíneos/patología , Cruzamientos Genéticos , Modelos Animales de Enfermedad , Fibrosis/etiología , Fibrosis/patología , Hemorragia/etiología , Hipertensión/sangre , Hipertensión/metabolismo , Hipertensión/patología , Integrasas/genética , Megacariocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Agregación Plaquetaria , Anomalías Cutáneas/etiología , Trombocitopenia/etiología , Factor de Crecimiento Transformador beta1/sangre , Factor de Crecimiento Transformador beta1/genética
9.
Matrix Biol Plus ; 22: 100149, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38831847

RESUMEN

Although the mechanism for activation of latent TGFß1 and TGFß3 is understood to involve the binding of the TGFß propeptide (LAP) to both an integrin and an insoluble substrate, the activation of latent TGFß2 has been unclear because the TGFß2 LAP does not have the classical integrin binding sequence found in the other two TGFß isoform LAPs. To assess the potential requirement for covalent linkage with a matrix or cell surface protein for the activation of latent TGFß2, we generated mice in which the TGFß2 Cys residue predicted to be involved in binding was mutated to Ser (Tgfb2C24S). We reasoned that, if covalent interaction with a second molecule is required for latent TGFß2 activation, mutant mice should display a Tgfb2 null (Tgfb2-/-)-like phenotype. Tgfb2C24S mice closely phenocopy Tgfb2-/- mice with death in utero between E18 and P1 and with congenital heart and kidney defects similar to those described for Tgfb2-/- mice. The mutant latent TGFß2 is secreted at levels similar to WT, yet TGFß signaling monitored as nuclear pSmad2 is suppressed. We conclude that, like latent TGFß1, latent TGFß2 activation requires binding to an immobilized matrix or plasma membrane molecule.

10.
JACC Basic Transl Sci ; 9(2): 185-199, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38510715

RESUMEN

The severity of aortic stenosis (AS) is associated with acquired von Willebrand syndrome (AVWS) and gastrointestinal bleeding, leading to anemia (Heyde's syndrome). We investigated how anemia is linked with AS and AVWS using the LA100 mouse model and patients with AS. Induction of anemia in LA100 mice increased transforming growth factor (TGF)-ß1 activation, AVWS, and AS progression. Patients age >75 years with severe AS had higher plasma TGF-ß1 levels and more severe anemia than AS patients age <75 years, and there was a correlation between TGF-ß1 and anemia. These data are compatible with the hypothesis that the blood loss anemia of Heyde's syndrome contributes to AS progression via WSS-induced activation of platelet TGF-ß1 and additional gastrointestinal bleeding via WSS-induced AVWS.

11.
Thromb Res ; 225: 47-56, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37001283

RESUMEN

BACKGROUND AND OBJECTIVES: COVID-19 progression is characterized by systemic small vessel arterial and venous thrombosis. Microvascular endothelial cell (MVEC) activation and injury, platelet activation, and histopathologic features characteristic of acute COVID-19 also describe certain thrombotic microangiopathies, including atypical hemolytic-uremic syndrome (aHUS), thrombotic thrombocytopenic purpura (TTP), and hematopoietic stem cell transplant (HSCT)-associated veno-occlusive disease (VOD). We explored the effect of clinically relevant doses of defibrotide, approved for HSCT-associated VOD, on MVEC activation/injury. METHODS: Human dermal MVEC were exposed to plasmas from patients with acute TMAs or acute COVID-19 in the presence and absence of defibrotide (5µg/ml) and caspase 8, a marker of EC activation and apoptosis, was assessed. RNAseq was used to explore potential mechanisms of defibrotide activity. RESULTS: Defibrotide suppressed TMA plasma-induced caspase 8 activation in MVEC (mean 60.2 % inhibition for COVID-19; p = 0.0008). RNAseq identified six major cellular pathways associated with defibrotide's alteration of COVID-19-associated MVEC changes: TNF-α signaling; IL-17 signaling; extracellular matrix (ECM)-EC receptor and platelet receptor interactions; ECM formation; endothelin activity; and fibrosis. Communications across these pathways were revealed by STRING analyses. Forty transcripts showing the greatest changes induced by defibrotide in COVID-19 plasma/MVEC cultures included: claudin 14 and F11R (JAM), important in maintaining EC tight junctions; SOCS3 and TNFRSF18, involved in suppression of inflammation; RAMP3 and transgelin, which promote angiogenesis; and RGS5, which regulates caspase activation and apoptosis. CONCLUSION: Our data, in the context of a recent clinical trial in severe COVID-19, suggest benefits to further exploration of defibrotide and these pathways in COVID-19 and related endotheliopathies.


Asunto(s)
COVID-19 , Trasplante de Células Madre Hematopoyéticas , Enfermedades Vasculares , Humanos , Caspasa 8 , COVID-19/complicaciones , Células Endoteliales , Anticoagulantes
12.
Geroscience ; 45(2): 983-999, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36460774

RESUMEN

SIRT3 is a longevity factor that acts as the primary deacetylase in mitochondria. Although ubiquitously expressed, previous global SIRT3 knockout studies have shown primarily a cardiac-specific phenotype. Here, we sought to determine how specifically knocking out SIRT3 in cardiomyocytes (SIRTcKO mice) temporally affects cardiac function and metabolism. Mice displayed an age-dependent increase in cardiac pathology, with 10-month-old mice exhibiting significant loss of systolic function, hypertrophy, and fibrosis. While mitochondrial function was maintained at 10 months, proteomics and metabolic phenotyping indicated SIRT3 hearts had increased reliance on glucose as an energy substrate. Additionally, there was a significant increase in branched-chain amino acids in SIRT3cKO hearts without concurrent increases in mTOR activity. Heavy water labeling experiments demonstrated that, by 3 months of age, there was an increase in protein synthesis that promoted hypertrophic growth with a potential loss of proteostasis in SIRT3cKO hearts. Cumulatively, these data show that the cardiomyocyte-specific loss of SIRT3 results in severe pathology with an accelerated aging phenotype.


Asunto(s)
Sirtuina 3 , Ratones , Animales , Sirtuina 3/genética , Sirtuina 3/metabolismo , Proteostasis , Ratones Noqueados , Miocitos Cardíacos , Mitocondrias/metabolismo
13.
J Clin Invest ; 132(15)2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35912863

RESUMEN

SARS-CoV-2-infected individuals may suffer a multi-organ system disorder known as "long COVID" or post-acute sequelae of SARS-CoV-2 infection (PASC). There are no standard treatments, the pathophysiology is unknown, and incidence varies by clinical phenotype. Acute COVID-19 correlates with biomarkers of systemic inflammation, hypercoagulability, and comorbidities that are less prominent in PASC. Macrovessel thrombosis, a hallmark of acute COVID-19, is less frequent in PASC. Female sex at birth is associated with reduced risk for acute COVID-19 progression, but with increased risk of PASC. Persistent microvascular endotheliopathy associated with cryptic SARS-CoV-2 tissue reservoirs has been implicated in PASC pathology. Autoantibodies, localized inflammation, and reactivation of latent pathogens may also be involved, potentially leading to microvascular thrombosis, as documented in multiple PASC tissues. Diagnostic assays illuminating possible therapeutic targets are discussed.


Asunto(s)
COVID-19 , Trombosis , COVID-19/complicaciones , COVID-19/terapia , Femenino , Humanos , Inflamación , SARS-CoV-2 , Trombosis/etiología , Trombosis/terapia , Síndrome Post Agudo de COVID-19
14.
Blood Adv ; 6(11): 3321-3328, 2022 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-35358295

RESUMEN

Transforming growth factor ß1 (TGF-ß1) regulates a wide variety of events in adult bone marrow (BM), including quiescence of hematopoietic stem cells, via undefined mechanisms. Because megakaryocytes (MKs)/platelets are a rich source of TGF-ß1, we assessed whether TGF-ß1 might inhibit its own production by comparing mice with conditional inactivation of Tgfb1 in MKs (PF4Cre;Tgfb1flox/flox) and control mice. PF4Cre;Tgfb1flox/flox mice had ∼30% more MKs in BM and ∼15% more circulating platelets than control mice (P < .001). Thrombopoietin (TPO) levels in plasma and TPO expression in liver were approximately twofold higher in PF4Cre;Tgfb1flox/flox than in control mice (P < .01), whereas TPO expression in BM cells was similar between these mice. In BM cell culture, TPO treatment increased the number of MKs from wild-type mice by approximately threefold, which increased approximately twofold further in the presence of a TGF-ß1-neutralizing antibody and increased the number of MKs from PF4Cre;Tgfb1flox/flox mice approximately fourfold. Our data reveal a new role for TGF-ß1 produced by MKs/platelets in regulating its own production in BM via increased TPO production in the liver. Additional studies are required to determine the mechanism.


Asunto(s)
Médula Ósea/metabolismo , Megacariocitos , Trombopoyetina , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Plaquetas/metabolismo , Hígado/metabolismo , Megacariocitos/citología , Megacariocitos/metabolismo , Ratones , Trombopoyetina/metabolismo
15.
Nat Med ; 10(5): 502-9, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15098027

RESUMEN

Hemostasis initiates angiogenesis-dependent wound healing, and thrombosis is frequently associated with advanced cancer. Although activation of coagulation generates potent regulators of angiogenesis, little is known about how this pathway supports angiogenesis in vivo. Here we show that the tissue factor (TF)-VIIa protease complex, independent of triggering coagulation, can promote tumor and developmental angiogenesis through protease-activated receptor-2 (PAR-2) signaling. In this context, the TF cytoplasmic domain negatively regulates PAR-2 signaling. Mice from which the TF cytoplasmic domain has been deleted (TF Delta CT mice) show enhanced PAR-2-dependent angiogenesis, in synergy with platelet-derived growth factor BB (PDGF-BB). Ocular tissue from diabetic patients shows PAR-2 colocalization with phosphorylated TF specifically on neovasculature, suggesting that phosphorylation of the TF cytoplasmic domain releases its negative regulatory control of PAR-2 signaling in angiogenesis. Targeting the TF-VIIa signaling pathway may thus enhance the efficacy of angiostatic treatments for cancer and neovascular eye diseases.


Asunto(s)
Neovascularización Patológica , Neovascularización Fisiológica , Tromboplastina/fisiología , Animales , Aorta/patología , Oftalmopatías/patología , Oftalmopatías/fisiopatología , Humanos , Técnicas In Vitro , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Estructura Terciaria de Proteína , Receptor PAR-2/fisiología , Transducción de Señal , Tromboplastina/química , Tromboplastina/genética
16.
Artículo en Inglés | MEDLINE | ID: mdl-34619980

RESUMEN

Objective: Aortic stenosis (AS) is characterized by narrowing of the aortic valve opening, resulting in peak blood flow velocity that induces high wall shear stress (WSS) across the valve. Severe AS leads to heart failure and death. There is no treatment available for AS other than valve replacement. Platelet-derived transforming growth factor beta 1 (TGF-ß1) partially contributes to AS progression in mice, and WSS is a potent activator of latent TGF-ß1. N-acetylcysteine (NAC) inhibits WSS-induced TGF-ß1 activation in vitro. We hypothesize that NAC will inhibit AS progression by inhibiting WSS-induced TGF-ß1 activation. Approach: We treated a cohort of Ldlr(-/-)Apob(100/100) low density lipoprotein receptor (LDLR) mice fed a high-fat diet with NAC (2% in drinking water) at different stages of disease progression and measured its effect on AS progression and TGF-ß1 activation. Results: Short-term NAC treatment inhibited AS progression in mice with moderate and severe AS relative to controls, but not in LDLR mice lacking platelet-derived TGF-ß1 (TGF-ß1platlet-KO-LDLR). NAC treatment reduced TGF-ß signaling, p-Smad2 and collagen levels, and mesenchymal transition from isolectin B4 and CD45-positive cells in LDLR mice. Mechanistically, NAC treatment resulted in plasma NAC concentrations ranging from 75.5 to 449.2 ng/mL, which were sufficient to block free thiol labeling of plasma proteins and reduce active TGF-ß1 levels without substantially affecting reactive oxygen species-modified products in valvular cells. Conclusions: Short-term treatment with NAC inhibits AS progression by inhibiting WSS-induced TGF-ß1 activation in the LDLR mouse model of AS, motivating a clinical trial of NAC and/or other thiol-reactive agent(s) as a potential therapy for AS.

17.
J Thromb Haemost ; 19(9): 2268-2274, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34236752

RESUMEN

BACKGROUND: A substantial proportion of patients infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) develop severe/critical coronavirus disease 2019 (COVID-19) characterized by acute respiratory distress syndrome (ARDS) with thrombosis. OBJECTIVES: We tested the hypothesis that SARS-CoV-2--induced upregulation of tissue factor (TF) expression may be responsible for thrombus formation in COVID-19. METHODS: We compared autopsy lung tissues from 11 patients with COVID-19--associated ARDS with samples from 6 patients with ARDS from other causes (non-COVID-19 ARDS) and 11 normal control lungs. RESULTS: Dual RNA in situ hybridization for SARS-CoV-2 and TF identified sporadic clustered SARS-CoV-2 with prominent co-localization of SARS-CoV-2 and TF RNA. TF expression was 2-fold higher in COVID-19 than in non-COVID-19 ARDS lungs (P = .017) and correlated with the intensity of SARS-CoV-2 staining (R2  = .36, P = .04). By immunofluorescence, TF protein expression was 2.1-fold higher in COVID-19 versus non-COVID-19 ARDS lungs (P = .0048) and 11-fold (P < .001) higher than control lungs. Fibrin thrombi and thrombi positive for platelet factor 4 (PF4) were found in close proximity to regions expressing TF in COVID-19 ARDS lung, and correlated with TF expression (fibrin, R2  = .52, P < .001; PF4, R2  = .59, P < .001). CONCLUSIONS: These data suggest that upregulation of TF expression is associated with thrombus formation in COVID-19 lungs and could be a key therapeutic target. Correlation of TF expression with SARS-CoV-2 in lungs of COVID-19 patients also raises the possibility of direct TF induction by the virus.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , Pulmón , Tromboplastina , Regulación hacia Arriba
18.
JCI Insight ; 6(14)2021 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-34291736

RESUMEN

Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), remains a pandemic. Severe disease is associated with dysfunction of multiple organs, but some infected cells do not express ACE2, the canonical entry receptor for SARS-CoV-2. Here, we report that the C-type lectin receptor L-SIGN interacted in a Ca2+-dependent manner with high-mannose-type N-glycans on the SARS-CoV-2 spike protein. We found that L-SIGN was highly expressed on human liver sinusoidal endothelial cells (LSECs) and lymph node lymphatic endothelial cells but not on blood endothelial cells. Using high-resolution confocal microscopy imaging, we detected SARS-CoV-2 viral proteins within the LSECs from liver autopsy samples from patients with COVID-19. We found that both pseudo-typed virus enveloped with SARS-CoV-2 spike protein and authentic SARS-CoV-2 virus infected L-SIGN-expressing cells relative to control cells. Moreover, blocking L-SIGN function reduced CoV-2-type infection. These results indicate that L-SIGN is a receptor for SARS-CoV-2 infection. LSECs are major sources of the clotting factors vWF and factor VIII (FVIII). LSECs from liver autopsy samples from patients with COVID-19 expressed substantially higher levels of vWF and FVIII than LSECs from uninfected liver samples. Our data demonstrate that L-SIGN is an endothelial cell receptor for SARS-CoV-2 that may contribute to COVID-19-associated coagulopathy.


Asunto(s)
COVID-19 , Capilares , Moléculas de Adhesión Celular/metabolismo , Células Endoteliales , Lectinas Tipo C/metabolismo , Hígado/irrigación sanguínea , Vasos Linfáticos , Receptores de Superficie Celular/metabolismo , SARS-CoV-2/fisiología , COVID-19/metabolismo , COVID-19/patología , COVID-19/virología , Capilares/metabolismo , Capilares/patología , Capilares/virología , Células Endoteliales/metabolismo , Células Endoteliales/patología , Células Endoteliales/virología , Perfilación de la Expresión Génica/métodos , Humanos , Hígado/patología , Vasos Linfáticos/metabolismo , Vasos Linfáticos/patología , Vasos Linfáticos/virología , Glicoproteína de la Espiga del Coronavirus , Internalización del Virus
19.
Blood ; 112(9): 3650-60, 2008 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-18544680

RESUMEN

Transforming growth factor-beta1 (TGF-beta1) has potent physiologic and pathologic effects on a variety of cell types at subnanomolar concentrations. Platelets contain 40 times as much TGF-beta1 as other cells and secrete it as an inactive (latent) form in complex with latency-associated peptide (LAP), which is disulfide bonded via Cys33 to latent TGF-beta binding protein 1 (LTBP-1). Little is known about how latent TGF-beta1 becomes activated in vivo. Here we show that TGF-beta1 released from platelets or fibroblasts undergoes dramatic activation when subjected to stirring or shear forces, providing a potential mechanism for physiologic control. Thiol-disulfide exchange appears to contribute to the process based on the effects of thiol-reactive reagents and differences in thiol labeling of TGF-beta1 before and after stirring or shear. Activation required the presence of LTBP, as TGF-beta1 contained in complex with only LAP could not be activated by stirring when studied as either a recombinant purified protein complex or in the platelet releasates or sera of mice engineered to contain an LAP C33S mutation. Release and activation of latent TGF-beta1 in vivo was demonstrated in a mouse model 5 minutes after thrombus formation. These data potentially provide a novel mechanism for in vivo activation of TGF-beta1.


Asunto(s)
Plaquetas/metabolismo , Factor de Crecimiento Transformador beta1/sangre , Animales , Fenómenos Biofísicos , Biofisica , Proteínas Sanguíneas/metabolismo , Trombosis de las Arterias Carótidas/sangre , Línea Celular , Medios de Cultivo Condicionados , Fibroblastos/metabolismo , Humanos , Técnicas In Vitro , Proteínas de Unión a TGF-beta Latente/sangre , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Estrés Mecánico , Compuestos de Sulfhidrilo/sangre , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo
20.
Circ Res ; 102(4): 457-64, 2008 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-18174463

RESUMEN

The serine protease zymogen factor X is converted to its catalytically active form factor Xa by the binary complex of factor VIIa bound to its cell surface receptor tissue factor (TF) or by the intrinsic Xase complex, which consists of active factors VIII (VIIIa), IX (IXa), factor X, and Ca2+. Factor Xa has procoagulant activity by conversion of prothrombin to thrombin and also induces signal transduction, either alone or in the ternary TF:VIIa:factor Xa coagulation initiation complex. Factor Xa cleaves and activates protease activated receptor (PAR)1 or -2, but factor Xa signaling efficiency varies among cell types. We show here that annexin 2 acts as a receptor for factor Xa on the surface of human umbilical vein endothelial cells and that annexin 2 facilitates factor Xa activation of PAR-1 but does not enhance coagulant function of factor Xa. Overexpression of TF abolishes annexin 2 dependence on factor Xa signaling and diminishes binding to cell surface annexin 2, whereas selectively abolishing TF promotes the annexin 2/factor Xa interaction. We propose that annexin 2 serves to regulate factor Xa signaling specifically in the absence of cell surface TF and may thus play physiological or pathological roles when factor Xa is generated in a TF-depleted environment.


Asunto(s)
Anexina A2/metabolismo , Células Endoteliales/metabolismo , Factor Xa/metabolismo , Receptor PAR-1/metabolismo , Transducción de Señal/fisiología , Animales , Coagulación Sanguínea/fisiología , Células Cultivadas , Células Endoteliales/citología , Factor Xa/química , Humanos , Inmunoprecipitación , Lisina/metabolismo , Ratones , Ratones Mutantes , Estructura Terciaria de Proteína , Tromboplastina/metabolismo , Venas Umbilicales/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA