Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 127
Filtrar
1.
EBioMedicine ; 75: 103812, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-35033854

RESUMEN

BACKGROUND: Thromboembolism is a life-threatening manifestation of coronavirus disease 2019 (COVID-19). We investigated a dysfunctional phenotype of vascular endothelial cells in the lungs during COVID-19. METHODS: We obtained the lung specimens from the patients who died of COVID-19. The phenotype of endothelial cells and immune cells was examined by flow cytometry and immunohistochemistry (IHC) analysis. We tested the presence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in the endothelium using IHC and electron microscopy. FINDINGS: The autopsy lungs of COVID-19 patients exhibited severe coagulation abnormalities, immune cell infiltration, and platelet activation. Pulmonary endothelial cells of COVID-19 patients showed increased expression of procoagulant von Willebrand factor (VWF) and decreased expression of anticoagulants thrombomodulin and endothelial protein C receptor (EPCR). In the autopsy lungs of COVID-19 patients, the number of macrophages, monocytes, and T cells was increased, showing an activated phenotype. Despite increased immune cells, adhesion molecules such as ICAM-1, VCAM-1, E-selectin, and P-selectin were downregulated in pulmonary endothelial cells of COVID-19 patients. Notably, decreased thrombomodulin expression in endothelial cells was associated with increased immune cell infiltration in the COVID-19 patient lungs. There were no SARS-CoV-2 particles detected in the lung endothelium of COVID-19 patients despite their dysfunctional phenotype. Meanwhile, the autopsy lungs of COVID-19 patients showed SARS-CoV-2 virions in damaged alveolar epithelium and evidence of hypoxic injury. INTERPRETATION: Pulmonary endothelial cells become dysfunctional during COVID-19, showing a loss of thrombomodulin expression related to severe thrombosis and infiltration, and endothelial cell dysfunction might be caused by a pathologic condition in COVID-19 patient lungs rather than a direct infection with SARS-CoV-2. FUNDING: This work was supported by the Johns Hopkins University, the American Heart Association, and the National Institutes of Health.


Asunto(s)
Trastornos de la Coagulación Sanguínea/metabolismo , COVID-19/metabolismo , Regulación hacia Abajo , Endotelio Vascular/metabolismo , Hipoxia/metabolismo , Pulmón/metabolismo , SARS-CoV-2/metabolismo , Trombomodulina/biosíntesis , Anciano , Anciano de 80 o más Años , Trastornos de la Coagulación Sanguínea/patología , COVID-19/patología , Células Endoteliales/metabolismo , Células Endoteliales/ultraestructura , Endotelio Vascular/ultraestructura , Femenino , Humanos , Hipoxia/patología , Pulmón/ultraestructura , Masculino , Persona de Mediana Edad
2.
Blood ; 137(7): 977-982, 2021 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-32870264

RESUMEN

Excess platelet activation by extracellular vesicles (EVs) results in trophoblast inflammasome activation, interleukin 1ß (IL-1ß) activation, preeclampsia (PE), and partial embryonic lethality. Embryonic thrombomodulin (TM) deficiency, which causes embryonic lethality hallmarked by impaired trophoblast proliferation, has been linked with maternal platelet activation. We hypothesized that placental TM loss, platelet activation, and embryonic lethality are mechanistically linked to trophoblast inflammasome activation. Here, we uncover unidirectional interaction of placental inflammasome activation and reduced placental TM expression: although inflammasome inhibition did not rescue TM-null embryos from lethality, the inflammasome-dependent cytokine IL-1ß reduced trophoblast TM expression and impaired pregnancy outcome. EVs, known to induce placental inflammasome activation, reduced trophoblast TM expression and proliferation. Trophoblast TM expression correlated negatively with IL-1ß expression and positively with platelet numbers and trophoblast proliferation in human PE placentae, implying translational relevance. Soluble TM treatment or placental TM restoration ameliorated the EV-induced PE-like phenotype in mice, preventing placental thromboinflammation and embryonic death. The lethality of TM-null embryos is not a consequence of placental NLRP3 inflammasome activation. Conversely, EV-induced placental inflammasome activation reduces placental TM expression, promoting placental and embryonic demise. These data identify a new function of placental TM in PE and suggest that soluble TM limits thromboinflammatory pregnancy complications.


Asunto(s)
Muerte Fetal/etiología , Inflamasomas/metabolismo , Placenta/metabolismo , Preeclampsia/metabolismo , Trombomodulina/deficiencia , Animales , División Celular , Regulación hacia Abajo , Vesículas Extracelulares , Femenino , Genes Letales , Humanos , Proteína Antagonista del Receptor de Interleucina 1/farmacología , Interleucina-1beta/biosíntesis , Interleucina-1beta/genética , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR/fisiología , Placenta/irrigación sanguínea , Activación Plaquetaria , Plasma Rico en Plaquetas , Embarazo , Resultado del Embarazo , Receptores de Trombina , Proteínas Recombinantes/farmacología , Trombomodulina/antagonistas & inhibidores , Trombomodulina/biosíntesis , Trombomodulina/genética , Trofoblastos/metabolismo
3.
Mol Biol Rep ; 47(10): 7575-7582, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32930934

RESUMEN

Thrombomodulin (THBD) is an endothelial surface glycoprotein receptor, having a pivotal role in maintaining laminar blood flow. It functions to protect endothelial integrity by exhibiting anti-coagulation and anti-inflammatory properties thereby playing a key role in cardiovascular disease (CVD) pathology. Cholesterol lowering drugs have shown to alter the anti-inflammatory effects of cytokines. Understanding the molecular aspects of THBD gene and its relation to inflammatory cytokines is important to identify new prognostic and therapeutic targets for the CVD treatments. The present study was conducted to measure the expression of THBD, TNF-α and NF-kB genes in coronary artery disease patients (CAD) in Pakistani population. Lipid profile and BMI was compared both on fifty CAD patients and fifty healthy individuals. Expression analysis for THBD, TNF-α and NF-kB was carried out using real time PCR. The effect of lipid lowering drugs on cardiometabolic risk variables especially gene expression was analyzed. Our results indicated that the difference in BMI was marginal; however LDL-cholesterol and triglycerides levels in CAD patients were significantly higher than healthy individuals. THBD gene was significantly up-regulated whereas TNF-α and NF-kB were significantly down regulated in CAD individuals. Further exploration revealed that these variations were accounted to the use of statins by the patients. The use of statins by CAD patients up-regulated the mRNA expression of THBD by down-regulation of inflammatory mediators. The enhanced expression of endothelial THBD in response to cholesterol lowering drugs establishes a novel pleiotropic target that can be of clinical significance in thromboembolic and inflammatory disorders.


Asunto(s)
Enfermedad de la Arteria Coronaria/metabolismo , Regulación de la Expresión Génica , FN-kappa B/biosíntesis , Trombomodulina/biosíntesis , Factor de Necrosis Tumoral alfa/biosíntesis , Adulto , Anciano , Enfermedad de la Arteria Coronaria/tratamiento farmacológico , Enfermedad de la Arteria Coronaria/patología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Pakistán
4.
Atherosclerosis ; 287: 54-63, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31212235

RESUMEN

BACKGROUND AND AIMS: Thrombomodulin (TM) is an endothelial cell membrane-bound anticoagulant protein expressed in normal arteries. After vascular injury, medial and neointimal smooth muscle cells (SMCs) exhibit large amounts of TM. The purpose of this study was to investigate the physiological significance of vascular SMC-bound TM. METHODS: The morphology, expression of phenotype markers and cell behaviors of cultured aortic SMCs after knockdown of TM were observed. Transgenic mice with SMC-specific TM deletion were generated, and carotid neointima formation was induced by carotid ligation. RESULTS: Cultured human aortic SMCs displayed a synthetic phenotype with a rhomboid-shaped morphology and expressed TM. TM knockdown induced a spindle-shaped change in morphology with an increased expression of contractile phenotype marker and decreased expression of synthetic phenotype marker. TM knockdown not only attenuated the proliferation of SMCs but also reduced tumor necrosis factor-α-induced nuclear factor-κB activation and interlukin-6 production. In a carotid artery ligation model, transgenic mice with SMC-specific TM deletion (SM22-cretg/TMflox/flox) had significantly less cellular proliferation in arterial walls compared with wild type mice (SM22-cretg/TM+/+). The neointima area and neointima/media area ratio were smaller in SM22-cretg/TMflox/flox mice at 4 weeks after ligation. CONCLUSIONS: Our results indicate that vascular SMC-bound TM plays a role in changes of the SMC phenotype. It also influences SMC cell behavior and injury-induced neointima formation.


Asunto(s)
Traumatismos de las Arterias Carótidas/genética , Regulación de la Expresión Génica , Músculo Liso Vascular/patología , Neointima/patología , Trombomodulina/genética , Animales , Arterias Carótidas/metabolismo , Arterias Carótidas/patología , Traumatismos de las Arterias Carótidas/metabolismo , Traumatismos de las Arterias Carótidas/patología , Membrana Celular/metabolismo , Movimiento Celular , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Transgénicos , Músculo Liso Vascular/metabolismo , Neointima/metabolismo , Fenotipo , ARN/genética , Trombomodulina/biosíntesis
5.
Shock ; 51(3): 364-371, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-29608549

RESUMEN

Severe sepsis is critical to health and can result in acute renal failure (ARF). Tissue factor (TF) and thrombomodulin (TM) play key roles in vascular endothelial functions by helping maintain microcirculation in the kidney. Budding uninhibited by benzimidazole-1 (Bub1) plays a role in Akt and JNK signaling, which control TF and TM, respectively. We hypothesized that Bub1 could control vascular endothelial function in sepsis. The aim of this study was to determine the role of Bub1 in septic ARF. We used Mouse cecum ligation and puncture (CLP) using low Bub1 expressing (Bub1) and wild-type (Bub1) mice in vivo and lipopolysaccharide (LPS) stimulation of human aortic endothelial cell (HAEC) in vitro. Bub1 mice had a higher survival rate after CLP than Bub1. Bub1 mice had more severe ARF after CLP than Bub1 with blood biochemical and pathological analyses. TF expression in Bub1 mice and control HAEC (control) significantly increased in the septic model compared with Bub1 and Bub1 silenced HAEC (siBub1). TM expression in the control significantly decreased after LPS stimulation compared with siBub1. Akt and JNK phosphorylation of siBub1 were attenuated after LPS stimulation. Associations of Bub1 with Akt or JNK after LPS stimulation of HAEC were detected using immunoprecipitation, suggesting that Bub1 is involved in the phosphorylation of Akt and JNK after LPS stimulation. Bub1 insufficiency attenuates TF expression and reduces TM suppression by blocking Akt and JNK phosphorylation, respectively, thus leading to the prevention of ARF and death caused by sepsis.


Asunto(s)
Lesión Renal Aguda/metabolismo , Células Endoteliales/metabolismo , Proteínas Serina-Treonina Quinasas/deficiencia , Sepsis/metabolismo , Trombomodulina/biosíntesis , Tromboplastina/biosíntesis , Lesión Renal Aguda/genética , Lesión Renal Aguda/patología , Animales , Modelos Animales de Enfermedad , Células Endoteliales/patología , Humanos , Ratones , Ratones Mutantes , Proteínas Serina-Treonina Quinasas/metabolismo , Sepsis/genética , Sepsis/patología , Trombomodulina/genética , Tromboplastina/genética
6.
Biochim Biophys Acta Gene Regul Mech ; 1861(8): 752-761, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29940355

RESUMEN

Thrombomodulin (TM, encoded by the THBD gene) expressed in vascular endothelial cells plays pivotal roles maintaining the equilibrium of coagulation and anti-coagulation. TM levels can be regulated at the transcriptional level although the epigenetic mechanism is underexplored. Here we report that transcriptional activation of TM in both immortalized vascular endothelial cells (EAhy926) and primary human aortic endothelial cells (HAEC) by all-trans retinoic acid (RA) paralleled accumulation of trimethylated histone H3K4, a prominent marker for active chromatin, surrounding the THBD promoter. RA treatment up-regulated the expression of SETD1A (SET1), a dedicated H3K4 methyltransferase, and augmented SETD1A occupancies on the THBD promoter. Further analysis revealed that the sequence-specific transcription factor Kruppel-like factor 4 (KLF4) interacted with and recruited SETD1A to the THBD promoter. Interestingly, SETD1A was recruited to the KLF4 promoter by retinoic acid receptor (RAR) and mediated the up-regulation of KLF4 expression by RA stimulation. In summary, our data illustrate a previously unrecognized pathway in which SETD1A contributes to RA-induced TM expression in vascular endothelial cells by modulating the activity and expression of KLF4.


Asunto(s)
Endotelio Vascular/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Trombomodulina/biosíntesis , Activación Transcripcional , Células Cultivadas , Células Endoteliales/metabolismo , Humanos , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/biosíntesis , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Receptores de Ácido Retinoico/metabolismo , Trombomodulina/genética , Tretinoina/fisiología
7.
Interact Cardiovasc Thorac Surg ; 27(2): 177-181, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29547930

RESUMEN

OBJECTIVES: We retrospectively analysed data from non-valvular atrial fibrillation (AF) patients who underwent minimally invasive surgical AF ablation at our centre. Our purpose was to explore the atrial endocardial expression of von Willebrand factor (vWF) and thrombomodulin (TM) and their association with rhythm results after the procedure. METHODS: From January 2014 to May 2015, 60 patients underwent minimally invasive surgical AF ablation at our centre. Left atrial appendage samples were obtained during the procedure and immunohistochemistry for endocardial markers including vWF and TM was performed and semi-quantitatively graded. All patients underwent postoperative rhythm documentation at 3, 6, 12 and 24 months. RESULTS: At the 2-year interval, 49 (82%) patients maintained sinus rhythm, and all patients were asymptomatic. Univariate analysis shows that patients with AF recurrence have higher vWF score 2/3 and longer AF duration (P < 0.05). In the multivariate analysis, AF duration, vWF score, TM score, left atrial diameter (LAD) and non-paroxysmal AF are included. The result suggests that higher vWF score 2/3, lower TM score 0/1 and non-paroxysmal AF are statistically significant (P < 0.05). In addition, higher vWF score 2/3 is associated with larger LAD (45.2 ± 5.6 mm vs 41.2 ± 7.6 mm, P = 0.032), while higher TM score 2/3, on the other hand, is associated with smaller LAD (44.6 ± 6.1 mm vs 39.9 ± 7.8 mm, P = 0.032). The Kaplan-Meier analysis shows that higher vWF score 2/3 and lower TM score 0/1 appear to be accompanied with higher recurrence rate (vWF: P = 0.021; TM: P = 0.036). CONCLUSIONS: Atrial endocardial expression of vWF and TM might be associated with recurrence after minimally invasive surgical AF ablation. Patients with AF recurrence seem to have elevated vWF expression and decreased TM expression.


Asunto(s)
Apéndice Atrial/metabolismo , Fibrilación Atrial/cirugía , Ablación por Catéter/efectos adversos , Endocardio/metabolismo , Trombomodulina/biosíntesis , Factor de von Willebrand/biosíntesis , Anciano , Fibrilación Atrial/metabolismo , Biomarcadores/metabolismo , Ablación por Catéter/métodos , Femenino , Atrios Cardíacos/metabolismo , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Procedimientos Quirúrgicos Mínimamente Invasivos/efectos adversos , Recurrencia , Estudios Retrospectivos , Factores de Tiempo
8.
J Biosci Bioeng ; 124(4): 445-451, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28694021

RESUMEN

Thrombomodulin (TM) is an endothelial cell membrane protein that acts as a major cofactor in the protein C anticoagulant pathway. The EGF-like domains 4-6 of TM (TM456) are essential for PC activation. In this study, we proposed a liposomal recombinant TM conjugate to mimic the membrane TM structure and its anticoagulant activity. First, a DSPE-PEG2000-TM456 was successfully synthesized by site-specific conjugation of azido-TM456 with DSPE-PEG2000-DBCO via copper-free click chemistry quantitatively. Then, liposome-TM456 was fabricated via direct liposome formation with the DSPE-PEG2000-TM456 and other lipids. This liposomal formulation of TM456 retained protein C activation activity as that of TM456. Also, liposome-TM456 was much more stable and had a longer plasma half-life than TM456 and DSPE-PEG2000-TM456, respectively. Moreover, liposome-TM456 showed in vivo anticoagulant effect by decreasing the mortality from 80% to 20% in a thrombin-induced thromboembolism mouse model. The reported liposome-TM456 conjugate mimics the endothelial TM anticoagulation activity and may serve as an effective anticoagulant agent candidate for future development.


Asunto(s)
Trombomodulina/biosíntesis , Trombomodulina/uso terapéutico , Trombosis/tratamiento farmacológico , Animales , Anticoagulantes/síntesis química , Anticoagulantes/química , Anticoagulantes/uso terapéutico , Pruebas de Coagulación Sanguínea , Química Clic , Humanos , Liposomas , Masculino , Ratones , Fosfatidiletanolaminas , Polietilenglicoles , Proteína C/metabolismo , Trombina/antagonistas & inhibidores , Trombina/metabolismo , Trombomodulina/química , Trombomodulina/genética , Trombomodulina/metabolismo
9.
Atherosclerosis ; 262: 62-70, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28525804

RESUMEN

BACKGROUND AND AIMS: Thrombomodulin (TM), through its lectin-like domain (TMD1), sequesters proinflammatory high-mobility group box 1 (HMGB1) to prevent it from engaging the receptor for advanced glycation end product (RAGE) that sustains inflammation and tissue damage. Our previous study demonstrated that short-term treatment with recombinant TM containing all the extracellular domains (i.e., rTMD123) inhibits HMGB1-RAGE signaling and confers protection against CaCl2-induced AAA formation. In this study, we attempted to further optimize TM domains, as a potential therapeutic agent for AAA, using the recombinant adeno-associated virus (AAV) vector. METHODS: The therapeutic effects of recombinant TMD1 (rTMD1) and recombinant AAV vectors carrying the lectin-like domain of TM (rAAV-TMD1) were evaluated in the CaCl2-induced AAA model and angiotensin II-infused AAA model, respectively. RESULTS: In the CaCl2-induced model, treatment with rTMD1 suppressed the tissue levels of HMGB1 and RAGE, macrophage accumulation, elastin destruction and AAA formation, and the effects were comparable to a mole-equivalent dosage of rTMD123. In the angiotensin II-infused model, a single intravenous injection of rAAV-TMD1 (1011 genome copies), which resulted in a persistently high serum level of TMD1 for at least 12 weeks, effectively attenuated AAA formation with suppression of HMGB1 and RAGE levels and inhibition of proinflammatory cytokine production, macrophage accumulation, matrix metalloproteinase activities and oxidative stress in the aortic wall. CONCLUSIONS: These findings corroborate the therapeutic potential of the TM lectin-like domain in AAA. The attenuation of angiotensin II-infused AAA by one-time delivery of rAAV-TMD1 provides a proof-of-concept validation of its application as potential gene therapy for aneurysm development.


Asunto(s)
Aneurisma de la Aorta Abdominal/prevención & control , Dependovirus/genética , Terapia Genética/métodos , Vectores Genéticos , Trombomodulina/genética , Angiotensina II , Animales , Aorta Abdominal/metabolismo , Aorta Abdominal/patología , Aneurisma de la Aorta Abdominal/inducido químicamente , Aneurisma de la Aorta Abdominal/genética , Aneurisma de la Aorta Abdominal/metabolismo , Cloruro de Calcio , Citocinas/metabolismo , Modelos Animales de Enfermedad , Elastina/metabolismo , Proteína HMGB1/metabolismo , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Metaloproteinasas de la Matriz/metabolismo , Ratones Noqueados para ApoE , Estrés Oxidativo , Dominios Proteicos , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Trombomodulina/biosíntesis , Trombomodulina/metabolismo , Remodelación Vascular
10.
J Immunol ; 195(6): 2743-53, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26259584

RESUMEN

The mechanisms leading to the life-threatening dengue hemorrhagic fever (DHF) remain elusive. DHF preferentially occurs during secondary dengue infections, suggesting that aberrant immune responses are involved in its development. We previously demonstrated that the autoantibodies elicited by dengue virus (DENV) nonstructural protein 1 (NS1; anti-NS1 Igs) induce plasma leakage and mortality in mice with warfarinized anticoagulant suppression. However, the involved pathogenic Ig fractions of anti-NS1 Igs remain unclear. In this study, the autoreactive Igs in patients with DHF and in NS1-immunized rabbits crossreacted with TNF-related apoptosis-inducing ligand receptor 1 (death receptor [DR]4). Challenges with the DENV in a subcytotoxic dose sensitized endothelial cells to apoptosis. Treatments with the autoantibodies induced proapoptotic activities and suppressed the surface expression of endothelial anticoagulant thrombomodulin. Combined treatments comprising the DENV and DR4 affinity-purified fractions of anti-NS1 IgGs (anti-NS1-DR4 Ig), but not preimmune control IgGs, in subcytotoxic doses led to apoptosis in endothelial cells. Treatments with the anti-NS1-DR4 Ig led to plasma leakage, coagulopathy, and morality in mice with warfarinized anticoagulant suppression. These results suggest that DR4-induced endothelial cell sensitization through NS1-elicited autoantibodies exacerbates anticoagulant suppression, vascular injury, and plasma leakage. Detecting and blocking anti-DR Igs in patients may be novel strategies for managing severe DENV infection.


Asunto(s)
Autoanticuerpos/inmunología , Virus del Dengue/inmunología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/inmunología , Dengue Grave/patología , Proteínas no Estructurales Virales/inmunología , Animales , Anticuerpos/inmunología , Anticuerpos/farmacología , Anticoagulantes , Apoptosis/inmunología , Coagulación Sanguínea , Línea Celular , Supervivencia Celular , Embrión de Pollo , Culicidae , Células Endoteliales/inmunología , Células Endoteliales/patología , Humanos , Inmunoglobulina G/inmunología , Ratones , Interferencia de ARN , ARN Interferente Pequeño , Conejos , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Dengue Grave/inmunología , Trombomodulina/biosíntesis
11.
Thromb Res ; 135(6): 1154-9, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25840743

RESUMEN

BACKGROUND: Thalidomide (Thal) treatment of patients with multiple myeloma (MM) is associated with vascular thrombosis, but the underlying mechanism is unknown. OBJECTIVES: To evaluate the hypothesis that Thal, dexamethasone (Dex) and MM serum perturb the hemostatic balance on human umbilical vein endothelial cells (HUVECs). METHODS: Drugs with or without the serum of MM patients or healthy controls were incubated with HUVECs. Analyses of phosphatidylserine (PS), tissue factor (TF), endothelial protein C receptor (EPCR) and thrombomodulin (TM) were performed using flow cytometry. The production of thrombin and activated protein C (APC) were measured by chromogenic assay. The roles of IL-6 and TNFα in regulating these indicators were also investigated. RESULTS: We found that Thal or Dex alone could not increase TF and PS expression in HUVECs. However, when pretreated with MM serum, their expression was significantly increased by either Thal or Dex. Concurrent changes were also detected in thrombin generation. In contrast, Thal and Dex had a direct inhibitory effect on the expression of EPCR and TM, and this inhibitory effect was especially significant when MM serum was added. The generation of APC paralleled the expression of EPCR and TM. All of the above outcomes were reversed to a variable extent by anti-IL-6R and anti-TNFα antibodies. CONCLUSIONS: These findings suggest Thal may act as a procoagulant by altering the balance between procoagulant and anticoagulant proteins on the surface of HUVECs, thereby contributing to thrombogenesis. MM serum plays a synergistic role in this process.


Asunto(s)
Células Endoteliales/metabolismo , Mieloma Múltiple/sangre , Mieloma Múltiple/tratamiento farmacológico , Talidomida/efectos adversos , Adulto , Inhibidores de la Angiogénesis/efectos adversos , Antígenos CD/biosíntesis , Coagulantes/química , Dexametasona/efectos adversos , Receptor de Proteína C Endotelial , Femenino , Hemostasis/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Interleucina-6/sangre , Masculino , Persona de Mediana Edad , Fosfatidilserinas/química , Receptores de Superficie Celular/biosíntesis , Trombomodulina/biosíntesis , Tromboplastina/biosíntesis , Factor de Necrosis Tumoral alfa/biosíntesis , Trombosis de la Vena/inducido químicamente
12.
PLoS One ; 10(3): e0122491, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25816372

RESUMEN

PURPOSE: To determine the role of thrombomodulin (TM) in corneal epithelial wound healing, and to investigate whether recombinant TM epidermal growth factor-like domain plus serine/threonine-rich domain (rTMD23) has therapeutic potential in corneal epithelial wound healing. METHODS: TM localization and expression in the murine cornea were examined by immunofluorescence staining. TM expression after injury was also studied. The effect of rTMD23 on corneal wound healing was evaluated by in vitro and in vivo assays. RESULTS: TM was expressed in the cornea in normal adult mice. TM expression increased in the early phase of wound healing and decreased after wound recovery. In the in vitro study, platelet-derived growth factor-BB (PDGF-BB) induced TM expression in murine corneal epithelial cells by mediating E26 transformation-specific sequence-1 (Ets-1) via the mammalian target of rapamycin (mTOR) signaling pathway. The administration of rTMD23 increased the rate of corneal epithelial wound healing. CONCLUSIONS: TM expression in corneal epithelium was modulated during the corneal wound healing process, and may be regulated by PDGF-BB. In addition, rTMD23 has therapeutic potential in corneal injury.


Asunto(s)
Lesiones de la Cornea/genética , Proteínas Proto-Oncogénicas c-sis/genética , Trombomodulina/genética , Cicatrización de Heridas/genética , Animales , Becaplermina , Lesiones de la Cornea/patología , Lesiones de la Cornea/terapia , Epitelio Corneal/metabolismo , Epitelio Corneal/patología , Ratones , Proteínas Proto-Oncogénicas c-sis/metabolismo , Proteínas Recombinantes/administración & dosificación , Transducción de Señal/efectos de los fármacos , Trombomodulina/administración & dosificación , Trombomodulina/biosíntesis , Cicatrización de Heridas/efectos de los fármacos
13.
BMC Cancer ; 14: 375, 2014 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-24886404

RESUMEN

BACKGROUND: The identification of potential tumor markers will help improve therapeutic planning and patient management. Thrombomodulin (TM) is a sensitive urothelial marker. TM was reported to be one of the endogenous anti-metastatic factors and has diagnostic and prognostic values for the progression of carcinoma. In the present study, we examine the role of TM in bladder cancer. METHODS: We studied the role of TM in tumor behavior and related signaling pathways in vitro using the human bladder cancer cell lines HT1376, HT1197, J82 and T24, and in vivo using animal models. We also selected clinical specimens from 100 patients with bladder cancer for immunohistochemical staining to evaluate the predictive capacity of TM in tumor invasiveness. RESULTS: The data revealed that positive immunoreactivity for TM was inversely correlated with clinical stage and DNA methyltransferase 1 immunoreactivity. Decreased TM expression could predict the aggressive tumor growth and advanced clinical stage in bladder cancer. When TM was inhibited, tumor growth rate and invasion ability were augmented in vitro and in vivo. The underlying changes included increased cell proliferation, enhanced epithelial-mesenchymal transition (EMT) and angiogenesis. Moreover, inhibition of NF-κB activation significantly increased TM expression and attenuated tumor aggressiveness in bladder cancer. CONCLUSIONS: TM plays an important role in bladder cancer tumor aggressiveness in vitro and in vivo and is a clinically significant predictor that may represent a suitable therapeutic target for bladder cancer.


Asunto(s)
Carcinogénesis , Transformación Celular Neoplásica/genética , Trombomodulina/biosíntesis , Neoplasias de la Vejiga Urinaria/genética , Animales , Biomarcadores de Tumor , Línea Celular Tumoral , Transición Epitelial-Mesenquimal/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , FN-kappa B/genética , Pronóstico , Trombomodulina/genética , Neoplasias de la Vejiga Urinaria/patología , Ensayos Antitumor por Modelo de Xenoinjerto
14.
PLoS One ; 9(5): e96005, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24797675

RESUMEN

Statins exert atheroprotective effects through the induction of specific transcriptional factors in multiple organs. In endothelial cells, statin-dependent atheroprotective gene up-regulation is mediated by Kruppel-like factor (KLF) family transcription factors. To dissect the mechanism of gene regulation, we sought to determine molecular targets by performing microarray analyses of human umbilical vein endothelial cells (HUVECs) treated with pitavastatin, and KLF4 was determined to be the most highly induced gene. In addition, it was revealed that the atheroprotective genes induced with pitavastatin, such as nitric oxide synthase 3 (NOS3) and thrombomodulin (THBD), were suppressed by KLF4 knockdown. Myocyte enhancer factor-2 (MEF2) family activation is reported to be involved in pitavastatin-dependent KLF4 induction. We focused on MEF2C among the MEF2 family members and identified a novel functional MEF2C binding site 148 kb upstream of the KLF4 gene by chromatin immunoprecipitation along with deep sequencing (ChIP-seq) followed by luciferase assay. By applying whole genome and quantitative chromatin conformation analysis {chromatin interaction analysis with paired end tag sequencing (ChIA-PET), and real time chromosome conformation capture (3C) assay}, we observed that the MEF2C-bound enhancer and transcription start site (TSS) of KLF4 came into closer spatial proximity by pitavastatin treatment. 3D-Fluorescence in situ hybridization (FISH) imaging supported the conformational change in individual cells. Taken together, dynamic chromatin conformation change was shown to mediate pitavastatin-responsive gene induction in endothelial cells.


Asunto(s)
Ensamble y Desensamble de Cromatina/efectos de los fármacos , Cromatina/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Factores de Transcripción de Tipo Kruppel/biosíntesis , Quinolinas/farmacología , Cromatina/genética , Regulación de la Expresión Génica/genética , Técnicas de Silenciamiento del Gen , Células Endoteliales de la Vena Umbilical Humana/citología , Humanos , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción MEF2/genética , Factores de Transcripción MEF2/metabolismo , Óxido Nítrico Sintasa de Tipo III/biosíntesis , Óxido Nítrico Sintasa de Tipo III/genética , Elementos de Respuesta , Trombomodulina/biosíntesis , Trombomodulina/genética
15.
J R Soc Interface ; 11(94): 20131079, 2014 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-24554575

RESUMEN

Drug eluting stents are associated with late stent thrombosis (LST), delayed healing and prolonged exposure of stent struts to blood flow. Using macroscale disturbed and undisturbed fluid flow waveforms, we numerically and experimentally determined the effects of microscale model strut geometries upon the generation of prothrombotic conditions that are mediated by flow perturbations. Rectangular cross-sectional stent strut geometries of varying heights and corresponding streamlined versions were studied in the presence of disturbed and undisturbed bulk fluid flow. Numerical simulations and particle flow visualization experiments demonstrated that the interaction of bulk fluid flow and stent struts regulated the generation, size and dynamics of the peristrut flow recirculation zones. In the absence of endothelial cells, deposition of thrombin-generated fibrin occurred primarily in the recirculation zones. When endothelium was present, peristrut expression of anticoagulant thrombomodulin (TM) was dependent on strut height and geometry. Thinner and streamlined strut geometries reduced peristrut flow recirculation zones decreasing prothrombotic fibrin deposition and increasing endothelial anticoagulant TM expression. The studies define physical and functional consequences of macro- and microscale variables that relate to thrombogenicity associated with the most current stent designs, and particularly to LST.


Asunto(s)
Fibrina/metabolismo , Regulación de la Expresión Génica , Hemodinámica , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Modelos Cardiovasculares , Stents , Trombomodulina/biosíntesis , Trombosis/metabolismo , Células Cultivadas , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Trombosis/etiología , Trombosis/patología
16.
Inflammation ; 37(1): 27-34, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23912649

RESUMEN

Animal models have shown that mesenteric lymph plays important roles in the pathogenesis of endothelium injury in many critical ill states. Gut-derived septicemia and endothelium injury are the two key pathogenesis of heat stroke (HS); however, it is unclear whether mesenteric lymph is cytotoxic to endothelium in HS. HS rat models were prepared in a prewarmed incubator. Mesenteric lymph, collected pre-, during, and post-HS, was analyzed for biological activity on human umbilical vein endothelial cell (HUVEC) in vitro. The effect of HS lymph on the production of von Willebrand factor (vWF), thrombomodulin (TM), and IL-6 by HUVEC was investigated. In vivo, vascular endothelium injury biomarkers, circulating endothelial cell (CEC), as well as serum soluble vWF and TM were tested in rats of HS and HS with mesenteric lymph duct ligation (HS-LDL). HS but not heat stroke sham mesenteric lymph-injured endothelial cells showed significantly increased HUVEC cytotoxicity and enhanced HUVEC monolayer permeability as well as elevated levels of vWF and TM production by HUVEC. IL-6 production by HUVEC was augmented by HS lymph in vitro. The effects of HS lymph on IL-6 production had a time course resembling that of the toxic effects of HS lymph on HUVEC. In vivo, when compared with HS rats, decreased CEC counts as well as lower serum vWF and TM concentrations were detected in HS-LDL rats. HS mesenteric lymph is probably harmful to vascular endothelium, which indicates that the modulation of mesenteric lymph may have some potential benefits to HS.


Asunto(s)
Golpe de Calor/sangre , Interleucina-6/biosíntesis , Linfa/metabolismo , Trombomodulina/sangre , Factor de von Willebrand/biosíntesis , Animales , Biomarcadores/sangre , Células Cultivadas , Modelos Animales de Enfermedad , Endotelio Vascular/lesiones , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Lipoproteínas LDL/metabolismo , Masculino , Ratas , Ratas Wistar , Trombomodulina/biosíntesis
17.
J Thromb Thrombolysis ; 34(4): 468-74, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22903729

RESUMEN

Metabolic syndrome is associated with significant hypercoagulable prothrombotic tendency; however, the mechanism for the prothrombotic state is not completely understood. We hypothesize that higher circulating plasma free fatty acids (FFAs) in metabolic syndrome inhibit the endothelial thrombomodulin (TM)-endothelial protein C receptor (EPCR) pathway, thereby promoting thrombus formation. Human umbilical vein endothelial cells were cultured in media supplemented with various doses of palmitic acid (PA), in the presence or absence of JNK inhibitor, and the expression of TM and EPCR was measured by western blot. The thrombotic state of high fat fed C57BL/6J mice was examined by tail bleeding time and deep venous thrombosis (DVT) model. As a result, PA inhibited the expression of TM and EPCR in endothelial cells, and this effect was blunted by inhibiting JNK signaling. High fat diet fed mice had higher level of circulating FFAs and exhibited prothrombotic state, evidenced by increased tail bleeding time and enlarged thrombotic size in DVT model, compared to the control diet fed mice. Hence, FFAs inhibit TM-EPCR-Protein C system in endothelial cells through activating JNK signaling, which may be a mechanism for the prothrombotic state in metabolic syndrome.


Asunto(s)
Antígenos CD/biosíntesis , Ácidos Grasos no Esterificados/metabolismo , Regulación de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana/metabolismo , MAP Quinasa Quinasa 4/metabolismo , Síndrome Metabólico/metabolismo , Receptores de Superficie Celular/biosíntesis , Transducción de Señal , Trombomodulina/biosíntesis , Trombosis de la Vena/metabolismo , Animales , Tiempo de Sangría , Células Cultivadas , Grasas de la Dieta/efectos adversos , Grasas de la Dieta/farmacología , Receptor de Proteína C Endotelial , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Masculino , Síndrome Metabólico/complicaciones , Síndrome Metabólico/patología , Ratones , Trombosis de la Vena/etiología , Trombosis de la Vena/patología
18.
Haematologica ; 97(11): 1674-7, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22689672

RESUMEN

Steroid-refractory graft-versus-host disease causes significant morbidity and mortality after allogeneic stem cell transplantation. The pathomechanism of steroid resistance is currently not understood, but it has been suggested that endothelial cell dysfunction plays a role. Endothelial thrombomodulin was quantified along with histological markers of epithelial damage and cytotoxic T cells in colon biopsies from 51 allografted patients, and retrospectively correlated with response to steroids and survival. Loss of endothelial thrombomodulin was the strongest predictor of response to steroids (P=0.02) and nonrelapse mortality (P=0.01) in multivariate analyses adjusting for T-cell infiltrates, histological grading, vessel density, disease status, donor type, and conditioning therapy. Our data provide evidence that at disease onset, loss of endothelial thrombomodulin expression rather than excessive T-cell infiltration associates with steroid-refractory graft-versus-host disease and mortality. Prospective histological investigations are now warranted to improve diagnosis and prognostication of this core complication of stem cell transplantation.


Asunto(s)
Endotelio Vascular/metabolismo , Enfermedad Injerto contra Huésped , Enfermedades Intestinales , Esteroides/administración & dosificación , Trombomodulina/biosíntesis , Supervivencia sin Enfermedad , Endotelio Vascular/patología , Femenino , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Enfermedad Injerto contra Huésped/metabolismo , Enfermedad Injerto contra Huésped/mortalidad , Enfermedad Injerto contra Huésped/patología , Trasplante de Células Madre Hematopoyéticas , Humanos , Enfermedades Intestinales/tratamiento farmacológico , Enfermedades Intestinales/metabolismo , Enfermedades Intestinales/mortalidad , Enfermedades Intestinales/patología , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Tasa de Supervivencia , Trasplante Homólogo
19.
Am J Physiol Heart Circ Physiol ; 303(2): H189-96, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22661506

RESUMEN

Hypercholesterolemia characterized by elevation of low-density lipoprotein (LDL) cholesterol is a major risk factor for atherosclerotic vascular disease. p66shc mediates hypercholesterolemia-induced endothelial dysfunction and atheromatous plaque formation. We asked if LDL upregulates endothelial p66shc via changes in the epigenome and examined the role of p66shc in LDL-stimulated endothelial cell dysfunction. Human LDL stimulates human p66shc promoter activity and p66shc expression in human endothelial cells. LDL leads to hypomethylation of two CpG dinucleotides and acetylation of histone 3 in the human p66shc promoter. These two CpG dinucleotides mediate LDL-stimulated p66shc promoter activity. Inhibition or knock down of DNA methyltransferases negates LDL-induced endothelial p66shc expression. p66shc mediates LDL-stimulated increase in expression of endothelial intercellular adhesion molecule-1 (ICAM1) and decrease in expression of thrombomodulin (TM). Mirroring these changes in ICAM1 and TM expression, p66shc mediates LDL-stimulated adhesion of monocytes to endothelial cells and plasma coagulation on endothelial cells. These findings indicate that LDL cholesterol upregulates human endothelial p66shc expression via hypomethylation of CpG dinucleotides in the p66shc promoter. Moreover, they show that LDL-stimulated p66shc expression mediates a dysfunctional endothelial cell surface, with proadhesive and procoagulant features.


Asunto(s)
LDL-Colesterol/fisiología , Células Endoteliales/fisiología , Epigénesis Genética , Proteínas Adaptadoras de la Señalización Shc/genética , Acetilación , Coagulación Sanguínea/fisiología , Adhesión Celular/fisiología , Línea Celular , LDL-Colesterol/farmacología , Metilasas de Modificación del ADN/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Histonas/metabolismo , Humanos , Molécula 1 de Adhesión Intercelular/biosíntesis , Monocitos/fisiología , Regiones Promotoras Genéticas , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src , Trombomodulina/biosíntesis , Regulación hacia Arriba
20.
Xenotransplantation ; 19(2): 82-91, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22497510

RESUMEN

For long-term xenograft survival, coagulation control is one of the remaining critical issues. Our attention has been directed toward human thrombomodulin (hTM), because it is expected to exhibit the following beneficial effects on coagulation control and cytoprotection: (i) to solve the problem of molecular incompatibility in protein C activation; (ii) to exert a role as a physiological regulator, only when thrombin is formed; (iii) to suppress direct prothrombinase activity; and (iv) to have anti-inflammatory properties. hTM gene was transfected into pig (Landrace/Yorkshire) fibroblasts using pCAGGS expression vector and pPGK-puro vector. After puromycin selection, only fibroblasts expressing a high level of hTM were collected by cell sorting and then applied to nuclear transfer. Following electroactivation and subsequent culture, a total of 1547 cleaved embryos were transferred to seven surrogate mother pigs. Two healthy cloned piglets expressing hTM were born, successfully grew to maturity and produced normal progeny. Immunohistochemical staining of organs from F1 generation pigs demonstrated hTM expression in endothelial cells as well as parenchymal cells. High expression was observed particularly in endothelial cells of kidney and liver. Aortic endothelial cells from cloned pigs were found to express hTM levels similar to human umbilical vein endothelial cells (HUVEC) and to make it possible to convert protein C into activated protein C. The blockade of human endothelial cell protein C receptor (hEPCR) significantly reduced APC production in HUVEC, but not in hTM-PAEC. Although no bleeding tendency was observed in hTM-cloned pigs, activated partial thromboplastin time (APTT) was slightly prolonged and soluble hTM was detected in pig plasma. hTM was expressed in platelets and mononuclear cells, but not in RBC. Cloned pigs expressing hTM in endothelial cells at a comparable level to HUVEC were produced. As complete suppression of antigen-antibody reaction in the graft is essential for accurate assessment of transgene related to coagulation control, production of genetically engineered pigs expressing hTM and complement regulatory protein based on galactosyltransferase knockout is desired.


Asunto(s)
Clonación de Organismos/métodos , Sus scrofa/genética , Trombomodulina/biosíntesis , Trombomodulina/genética , Animales , Animales Modificados Genéticamente , Secuencia de Bases , Células Sanguíneas/metabolismo , Coagulación Sanguínea , Cartilla de ADN/genética , Células Endoteliales/metabolismo , Femenino , Expresión Génica , Ingeniería Genética , Supervivencia de Injerto , Células Endoteliales de la Vena Umbilical Humana , Humanos , Hibridación Genética , Inmunohistoquímica , Masculino , Tiempo de Tromboplastina Parcial , Embarazo , Proteína C/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/sangre , Proteínas Recombinantes/genética , Sus scrofa/sangre , Sus scrofa/metabolismo , Trombomodulina/sangre , Distribución Tisular , Trasplante Heterólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA