Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Res Pract Thromb Haemost ; 8(4): 102472, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-39036672

RESUMEN

Background: Five secreted platelet protein disulfide isomerases (PDIs) and 1 transmembrane PDI regulate platelet function and thrombosis. Thioredoxin-related transmembrane protein 1 (TMX1) was the first member of the PDI family found to negatively regulate platelet aggregation and platelet accumulation in vivo. The effect of TMX1 on coagulation is unknown. Objectives: To determine the effect of TMX1 on coagulation. Methods: TMX1-/- mice were used to study platelet accumulation and fibrin deposition in vivo in the laser-induced thrombosis injury model. Annexin V deposition at the site of vascular injury was studied using conditional TMX1 knockout mice. Annexin V binding to platelets was studied using human platelets, anti-TMX1 antibodies, and TMX1-deficient platelets. Results: TMX1-/- mice had increased fibrin deposition that was reversed with infusion of recombinant TMX1. Infusion of recombinant TMX1 inhibited platelet accumulation and fibrin deposition in wild-type mice and inhibited fibrin deposition in ß3-null mice. Platelet accumulation is absent in ß3-null mice, suggesting that TMX1 inhibits coagulation independently of platelets. Annexin V binding was increased in activated human platelets incubated with an anti-TMX1 antibody and mouse platelets lacking TMX1. Addition of recombinant TMX1 decreased annexin V binding to platelets. Annexin V binding was increased at the site of vascular injury in Tie2-Cre/TMX1fl/fl mice deficient in endothelial cell TMX1. Conclusion: TMX1 decreases coagulation at the site of vascular injury and negatively regulates phosphatidylserine exposure on endothelial cells and platelets.

2.
Biomolecules ; 13(9)2023 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-37759735

RESUMEN

Extracellular histones, part of the protein group known as damage-associated molecular patterns (DAMPs), are released from damaged or dying cells and can instigate cellular toxicity. Within the context of chronic obstructive pulmonary disease (COPD), there is an observed abundance of extracellular histone H3.3, indicating potential pathogenic implications. Notably, histone H3.3 is often found hyperacetylated (AcH3.3) in the lungs of COPD patients. Despite these observations, the specific role of these acetylated histones in inducing pulmonary tissue damage in COPD remains unclear. To investigate AcH3.3's impact on lung tissue, we administered recombinant histones (rH2A, rH3.3, and rAcH3.3) or vehicle solution to mice via intratracheal instillation. After 48 h, we evaluated the lung toxicity damage and found that the rAcH3.3 treated animals exhibited more severe lung tissue damage compared to those treated with non-acetylated H3.3 and controls. The rAcH3.3 instillation resulted in significant histological changes, including alveolar wall rupture, epithelial cell damage, and immune cell infiltration. Micro-CT analysis confirmed macroscopic structural changes. The rAcH3.3 instillation also increased apoptotic activity (cleavage of caspase 3 and 9) and triggered acute systemic inflammatory marker activation (TNF-α, IL-6, MCP-3, or CXCL-1) in plasma, accompanied by leukocytosis and lymphocytosis. Confocal imaging analysis confirmed lymphocytic and monocytic/macrophage lung infiltration in response to H3.3 and AcH3.3 administration. Taken together, our findings implicate extracellular AcH3.3 in inducing cytotoxicity and acute inflammatory responses, suggesting its potential role in promoting COPD-related lung damage progression.

3.
Sci Transl Med ; 15(715): eade2966, 2023 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-37756380

RESUMEN

Hepatic fibrosis is the primary determinant of mortality in patients with metabolic dysfunction-associated steatohepatitis (MASH). Transforming growth factor-ß (TGFß), a master profibrogenic cytokine, is a promising therapeutic target that has not yet been translated into an effective therapy in part because of liabilities associated with systemic TGFß antagonism. We have identified that soluble folate receptor γ (FOLR3), which is expressed in humans but not in rodents, is a secreted protein that is elevated in the livers of patients with MASH but not in those with metabolic dysfunction-associated steatotic liver disease, those with type II diabetes, or healthy individuals. Global proteomics showed that FOLR3 was the most highly significant MASH-specific protein and was positively correlated with increasing fibrosis stage, consistent with stimulation of activated hepatic stellate cells (HSCs), which are the key fibrogenic cells in the liver. Exposure of HSCs to exogenous FOLR3 led to elevated extracellular matrix (ECM) protein production, an effect synergistically potentiated by TGFß1. We found that FOLR3 interacts with the serine protease HTRA1, a known regulator of TGFBR, and activates TGFß signaling. Administration of human FOLR3 to mice induced severe bridging fibrosis and an ECM pattern resembling human MASH. Our study thus uncovers a role of FOLR3 in enhancing fibrosis.


Asunto(s)
Diabetes Mellitus Tipo 2 , Hígado Graso , Humanos , Animales , Ratones , Factor de Crecimiento Transformador beta , Células Estrelladas Hepáticas , Ácido Fólico
4.
Int J Mol Sci ; 23(4)2022 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-35216100

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) is the number one cause of chronic liver disease worldwide, with 25% of these patients developing nonalcoholic steatohepatitis (NASH). NASH significantly increases the risk of cirrhosis and decompensated liver failure. Past studies in rodent models have shown that glycine-N-methyltransferase (GNMT) knockout results in rapid steatosis, fibrosis, and hepatocellular carcinoma progression. However, the attenuation of GNMT in subjects with NASH and the molecular basis for its impact on the disease process is still unclear. To address this knowledge gap, we show the reduction of GNMT protein levels in the liver of NASH subjects compared to healthy controls. To gain insight into the impact of decreased GNMT in the disease process, we performed global label-free proteome studies on the livers from a murine modified amylin diet-based model of NASH. Histological and molecular characterization of the animal model demonstrate a high resemblance to human disease. We found that a reduction of GNMT leads to a significant increase in S-adenosylmethionine (AdoMet), an essential metabolite for transmethylation reactions and a substrate for polyamine synthesis. Further targeted proteomic and metabolomic studies demonstrated a decrease in GNMT transmethylation, increased flux through the polyamine pathway, and increased oxidative stress production contributing to NASH pathogenesis.


Asunto(s)
Cirrosis Hepática/metabolismo , Hígado/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Estrés Oxidativo/fisiología , Poliaminas/metabolismo , S-Adenosilmetionina/metabolismo , Adulto , Animales , Carcinoma Hepatocelular/metabolismo , Modelos Animales de Enfermedad , Femenino , Glicina N-Metiltransferasa/metabolismo , Humanos , Neoplasias Hepáticas/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Oxidación-Reducción , Proteómica/métodos
5.
CRISPR J ; 4(6): 854-871, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34847745

RESUMEN

The lack of efficient tools to label multiple endogenous targets in cell lines without staining or fixation has limited our ability to track physiological and pathological changes in cells over time via live-cell studies. Here, we outline the FAST-HDR vector system to be used in combination with CRISPR-Cas9 to allow visual live-cell studies of up to three endogenous proteins within the same cell line. Our approach utilizes a novel set of advanced donor plasmids for homology-directed repair and a streamlined workflow optimized for microscopy-based cell screening to create genetically modified cell lines that do not require staining or fixation to accommodate microscopy-based studies. We validated this new methodology by developing two advanced cell lines with three fluorescent-labeled endogenous proteins that support high-content imaging without using antibodies or exogenous staining. We applied this technology to study seven severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2/COVID-19) viral proteins to understand better their effects on autophagy, mitochondrial dynamics, and cell growth. Using these two cell lines, we were able to identify the protein ORF3a successfully as a potent inhibitor of autophagy, inducer of mitochondrial relocalization, and a growth inhibitor, which highlights the effectiveness of live-cell studies using this technology.


Asunto(s)
Autofagia , COVID-19 , Sistemas CRISPR-Cas , Marcación de Gen , Dinámicas Mitocondriales , SARS-CoV-2 , Proteínas Viroporinas , COVID-19/genética , COVID-19/metabolismo , Células HCT116 , Células HEK293 , Células HeLa , Humanos , Microscopía , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , Proteínas Viroporinas/genética , Proteínas Viroporinas/metabolismo
6.
ACS Med Chem Lett ; 11(11): 2156-2164, 2020 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-33214824

RESUMEN

The dual PI3Kα/ m TOR inhibitors represent a promising molecularly targeted therapy for cancer. Here, we documented the discovery of new 2,4-disubstituted quinazoline analogs as potent dual PI3Kα/sm TOR inhibitors. Our structure based chemistry endeavor yielded six excellent compounds 9e, 9f, 9g, 9k, 9m, and 9o with single/double digit nanomolar IC50 values against both enzymes and acceptable aqueous solubility and stability to oxidative metabolism. One of those analogs, 9m, possessed a sulfonamide substituent, which has not been described for this chemical scaffold before. The short direct synthetic routes, structure-activity relationship, in vitro 2D cell culture viability assays against normal fibroblasts and 3 breast cancer cell lines, and in vitro 3D culture viability assay against MCF7 cells for this series are described.

7.
Bioorg Med Chem Lett ; 27(14): 3117-3122, 2017 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-28571824

RESUMEN

PI3Kα/mTOR ATP-competitive inhibitors are considered as one of the promising molecularly targeted cancer therapeutics. Based on lead compound A from the literature, two similar series of 2-substituted-4-morpholino-pyrido[3,2-d]pyrimidine and pyrido[2,3-d]pyrimidine analogs were designed and synthesized as PI3Kα/mTOR dual inhibitors. Interestingly, most of the series gave excellent inhibition for both enzymes with IC50 values ranging from single to double digit nM. Unlike many PI3Kα/mTOR dual inhibitors, our compounds displayed selectivity for PI3Kα. Based on its potent enzyme inhibitory activity, selectivity for PI3Kα and good therapeutic index in 2D cell culture viability assays, compound 4h was chosen to be evaluated in 3D culture for its IC50 against MCF7 breast cancer cells as well as for docking studies with both enzymes.


Asunto(s)
Antineoplásicos/síntesis química , Diseño de Fármacos , Inhibidores de las Quinasa Fosfoinosítidos-3 , Pirimidinas/química , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Adenosina Trifosfato/química , Adenosina Trifosfato/metabolismo , Antineoplásicos/química , Antineoplásicos/farmacología , Sitios de Unión , Unión Competitiva , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Fosfatidilinositol 3-Quinasa Clase I , Humanos , Concentración 50 Inhibidora , Células MCF-7 , Simulación del Acoplamiento Molecular , Fosfatidilinositol 3-Quinasas/metabolismo , Estructura Terciaria de Proteína , Pirimidinas/síntesis química , Pirimidinas/farmacología , Relación Estructura-Actividad , Serina-Treonina Quinasas TOR/metabolismo
8.
Arterioscler Thromb Vasc Biol ; 36(5): 961-71, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27055904

RESUMEN

OBJECTIVE: Platelets modulate hemostasis and immune responses via interactions with immune cells through secretion of immunemodulators and cell-cell interactions. The P2Y12 receptor mediates ADP-induced aggregation and secretion in platelets. APPROACH AND RESULTS: Using a mouse model of intra-abdominal sepsis and acute lung injury, we investigated the role of the P2Y12 receptor in neutrophil migration and lung inflammation in P2Y12 null mice and in mice pretreated with the P2Y12 antagonist clopidogrel. Our data show a decrease in circulating white blood cells and a decrease in platelet activation and platelet-leukocyte interactions in treated mice compared with untreated mice. Additionally, lung injury and platelet sequestration were diminished in clopidogrel-treated mice compared with their untreated septic littermates. Similar results were observed in P2Y12 null mice: platelet activation and platelet-leukocyte aggregates were decreased in septic P2Y12 null mice compared with wild-type mice. P2Y12 null mice were refractory to lung injury compared with wild-type mice. Finally, to evaluate P2Y12-independent effects of clopidogrel, we pretreated P2Y12 null mice. Interestingly, the number of circulating neutrophils was reduced in treated septic P2Y12 null mice, suggesting neutrophils as a target for clopidogrel pleiotropic effects. No difference was observed in P2Y1 null mice during sepsis, indicating that the P2Y12 receptor is responsible for the effects. CONCLUSIONS: P2Y12 null mice are refractory to sepsis-induced lung injury, suggesting a key role for activated platelets and the P2Y12 receptor during sepsis.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Plaquetas/metabolismo , Pulmón/metabolismo , Activación Plaquetaria , Neumonía/metabolismo , Receptores Purinérgicos P2Y12/metabolismo , Sepsis/metabolismo , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/patología , Lesión Pulmonar Aguda/prevención & control , Animales , Plaquetas/efectos de los fármacos , Clopidogrel , Citocinas/sangre , Predisposición Genética a la Enfermedad , Leucocitos/metabolismo , Pulmón/efectos de los fármacos , Pulmón/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Infiltración Neutrófila/efectos de los fármacos , Selectina-P/sangre , Fenotipo , Activación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/farmacología , Neumonía/genética , Neumonía/patología , Neumonía/prevención & control , Antagonistas del Receptor Purinérgico P2Y/farmacología , Receptores Purinérgicos P2Y12/deficiencia , Receptores Purinérgicos P2Y12/genética , Sepsis/tratamiento farmacológico , Sepsis/genética , Sepsis/microbiología , Transducción de Señal , Ticlopidina/análogos & derivados , Ticlopidina/farmacología
9.
J Cell Commun Signal ; 9(1): 37-54, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25617052

RESUMEN

Key clinical features of cumulative trauma disorders include pain, muscle weakness, and tissue fibrosis, although the etiology is still under investigation. Here, we characterized the temporal pattern of altered sensorimotor behaviors and inflammatory and fibrogenic processes occurring in forearm muscles and serum of young adult, female rats performing an operant, high repetition high force (HRHF) reaching and grasping task for 6, 12, or 18 weeks. Palmar mechanical sensitivity, cold temperature avoidance and spontaneous behavioral changes increased, while grip strength declined, in 18-week HRHF rats, compared to controls. Flexor digitorum muscles had increased MCP-1 levels after training and increased TNFalpha in 6-week HRHF rats. Serum had increased IL-1beta, IL-10 and IP-10 after training. Yet both muscle and serum inflammation resolved by week 18. In contrast, IFNγ increased at week 18 in both muscle and serum. Given the anti-fibrotic role of IFNγ, and to identify a mechanism for the continued grip strength losses and behavioral sensitivities, we evaluated the fibrogenic proteins CCN2, collagen type I and TGFB1, as well as the nociceptive/fibrogenic peptide substance P. Each increased in and around flexor digitorum muscles and extracellular matrix in the mid-forearm, and in nerves of the forepaw at 18 weeks. CCN2 was also increased in serum at week 18. At a time when inflammation had subsided, increases in fibrogenic proteins correlated with sensorimotor declines. Thus, muscle and nerve fibrosis may be critical components of chronic work-related musculoskeletal disorders. CCN2 and substance P may serve as potential targets for therapeutic intervention, and CCN2 as a serum biomarker of fibrosis progression.

10.
J Leukoc Biol ; 95(2): 313-23, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24142066

RESUMEN

Thienopyridines are a class of antiplatelet drugs that are metabolized in the liver to several metabolites, of which only one active metabolite can irreversibly antagonize the platelet P2Y12 receptor. Possible effects of these drugs and the role of activated platelets in inflammatory responses have also been investigated in a variety of animal models, demonstrating that thienopyridines could alter inflammation. However, it is not clear whether it is caused only by the P2Y12 antagonism or whether off-target effects of other metabolites also intervene. To address this question, we investigated P2Y12 KO mice during a LPS-induced model of systemic inflammation, and we treated these KO mice with a thienopyridine drug (clopidogrel). Contrary to the reported effects of clopidogrel, numbers of circulating WBCs and plasma levels of cytokines were increased in LPS-exposed KO mice compared with WT in this inflammation model. Moreover, both spleen and bone marrow show an increase in cell content, suggesting a role for P2Y12 in regulation of bone marrow and spleen cellular composition. Finally, the injury was more severe in the lungs of KO mice compared with WT. Interestingly, clopidogrel treatments also exerted protective effects in KO mice, suggesting off-target effects for this drug. In conclusion, the P2Y12 receptor plays an important role during LPS-induced inflammation, and this signaling pathway may be involved in regulating cell content in spleen and bone marrow during LPS systemic inflammation. Furthermore, clopidogrel may have effects that are independent of P2Y12 receptor blockade.


Asunto(s)
Inflamación/patología , Receptores Purinérgicos P2Y12/deficiencia , Animales , Peso Corporal/efectos de los fármacos , Médula Ósea/patología , Citocinas/sangre , Inflamación/sangre , Inflamación/inmunología , Recuento de Leucocitos , Leucocitos/patología , Lipopolisacáridos , Lesión Pulmonar/inmunología , Lesión Pulmonar/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Tamaño de los Órganos , Receptores Purinérgicos P2Y12/metabolismo , Esplenomegalia/sangre , Esplenomegalia/inmunología , Esplenomegalia/patología , Análisis de Supervivencia
11.
Retina ; 34(3): 504-8, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23945636

RESUMEN

PURPOSE: To evaluate the high-speed anterior segment optical coherence tomography (AS-OCT) grading of the AS inflammation in patients with ocular inflammation. METHODS: A retrospective consecutive case series study. Patients with clinically visible AS inflammation in at least one eye underwent AS-OCT (Visante; Zeiss Meditec) with three to eight line scans per eye, performed by a trained masked examiner. The images were reviewed for hyperreflective spots, noise, and artifact, and these were correlated to clinical examination. RESULTS: Seventy-eight eyes of 41 patients were imaged. Forty-seven eyes had anterior chamber cells on clinical examination, and 68 had hyperreflective spots visible on AS-OCT. There was a significant correlation (Spearman r = 0.7274) between clinical examination and Visante OCT images. Several patterns of inflammation and artifacts were apparent. CONCLUSION: The AS-OCT is a promising technique for grading anterior chamber cells. There was a significant correlation between clinical examination and Visante grading.


Asunto(s)
Cámara Anterior , Tomografía de Coherencia Óptica/métodos , Uveítis Anterior/diagnóstico , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Niño , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Adulto Joven
12.
PLoS One ; 8(7): e69093, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23861957

RESUMEN

Administration of the thienopyridine P2Y12 receptor antagonist, clopidogrel, increased the erosive arthritis induced by peptidoglycan polysaccharide (PG-PS) in rats or by injection of the arthritogenic K/BxN serum in mice. To determine if the detrimental effects are caused exclusively by clopidogrel, we evaluated prasugrel, a third-generation thienopyridine pro-drug, that contrary to clopidogrel is mostly metabolized into its active metabolite in the intestine. Prasugrel effects were examined on the PG-PS-induced arthritis rat model. Erosive arthritis was induced in Lewis rats followed by treatment with prasugrel for 21 days. Prasugrel treated arthritic animals showed a significant increase in the inflammatory response, compared with untreated arthritic rats, in terms of augmented macroscopic joint diameter associated with significant signs of inflammation, histomorphometric measurements of the hind joints and elevated platelet number. Moreover, fibrosis at the pannus, assessed by immunofluorescence of connective tissue growth factor, was increased in arthritic rats treated with prasugrel. In addition to the arthritic manifestations, hepatomegaly, liver granulomas and giant cell formation were observed after PG-PS induction and even more after prasugrel exposure. Cytokine plasma levels of IL-1 beta, IL-6, MIP1 alpha, MCP1, IL-17 and RANTES were increased in arthritis-induced animals. IL-10 plasma levels were significantly decreased in animals treated with prasugrel. Overall, prasugrel enhances inflammation in joints and liver of this animal model. Since prasugrel metabolites inhibit neutrophil function ex-vivo and the effects of both clopidogrel and prasugrel metabolites on platelets are identical, we conclude that the thienopyridines metabolites might exert non-platelet effects on other immune cells to aggravate inflammation.


Asunto(s)
Artritis Experimental/patología , Granuloma/patología , Articulaciones/patología , Hígado/patología , Piperazinas/efectos adversos , Inhibidores de Agregación Plaquetaria/efectos adversos , Profármacos/efectos adversos , Tiofenos/efectos adversos , Animales , Artritis Experimental/inducido químicamente , Artritis Experimental/inmunología , Clopidogrel , Citocinas/biosíntesis , Citocinas/inmunología , Femenino , Granuloma/inducido químicamente , Granuloma/inmunología , Inflamación/inducido químicamente , Inflamación/inmunología , Inflamación/patología , Articulaciones/inmunología , Hígado/inmunología , Peptidoglicano , Clorhidrato de Prasugrel , Ratas , Ratas Endogámicas Lew , Ticlopidina/efectos adversos , Ticlopidina/análogos & derivados
13.
J Pharmacol Exp Ther ; 344(1): 231-43, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23097214

RESUMEN

Clopidogrel and prasugrel belong to a thienopyridine class of oral antiplatelet drugs that, after having been metabolized in the liver, can inhibit platelet function by irreversibly antagonizing the P2Y(12) receptor. Furthermore, thienopyridines influence numerous inflammatory conditions, but their effects on neutrophils have not been evaluated, despite the important role of these cells in inflammation. Therefore, we investigated the effect of prasugrel metabolites on neutrophils to further clarify the role of thienopyridines in inflammation. Interestingly, a prasugrel metabolite mixture, produced in vitro using rat liver microsomes, significantly inhibited N-formyl-methionyl-leucyl-phenylalanine (fMLP)- and platelet-activating factor (PAF)-induced neutrophil activation. More specifically, prasugrel metabolites inhibited neutrophil transmigration, CD16 surface expression, and neutrophil-platelet aggregation. Moreover, prasugrel metabolite pretreatment also significantly decreased fMLP- or PAF-induced extracellular-signal-regulated kinase phosphorylation as well as calcium mobilization. To determine the target of prasugrel in neutrophils, the role of both P2Y(12) and P2Y(13) receptors was studied using specific reversible antagonists, AR-C69931MX and MRS2211, respectively. Neither antagonist had any direct effect on the agonist-induced neutrophil functional responses. Our findings indicate that prasugrel metabolites may directly target neutrophils and inhibit their activation, suggesting a possible explanation for their anti-inflammatory effects previously observed. However, these metabolites do not act through either the P2Y(12) or P2Y(13) receptor in neutrophils.


Asunto(s)
Neutrófilos/efectos de los fármacos , Piperazinas/farmacología , Antagonistas del Receptor Purinérgico P2Y/farmacología , Tiofenos/farmacología , Animales , Western Blotting , Antígeno CD11b/biosíntesis , Calcio/metabolismo , Separación Celular , Supervivencia Celular/efectos de los fármacos , Quimiotaxis de Leucocito/efectos de los fármacos , Humanos , Inflamación/inducido químicamente , Inflamación/patología , Lipopolisacáridos , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Microsomas Hepáticos , Peroxidasa/metabolismo , Piperazinas/metabolismo , Agregación Plaquetaria/efectos de los fármacos , Clorhidrato de Prasugrel , Antagonistas del Receptor Purinérgico P2/farmacología , Antagonistas del Receptor Purinérgico P2Y/metabolismo , Receptores Purinérgicos P2Y12/efectos de los fármacos , Tiofenos/metabolismo
14.
PLoS One ; 7(5): e38359, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22675458

RESUMEN

BACKGROUND: This study elucidates exposure-response relationships between performance of repetitive tasks, grip strength declines, and fibrogenic-related protein changes in muscles, and their link to inflammation. Specifically, we examined forearm flexor digitorum muscles for changes in connective tissue growth factor (CTGF; a matrix protein associated with fibrosis), collagen type I (Col1; a matrix component), and transforming growth factor beta 1 (TGFB1; an upstream modulator of CTGF and collagen), in rats performing one of two repetitive tasks, with or without anti-inflammatory drugs. METHODOLOGY/RESULTS: To examine the roles of force versus repetition, rats performed either a high repetition negligible force food retrieval task (HRNF), or a high repetition high force handle-pulling task (HRHF), for up to 9 weeks, with results compared to trained only (TR-NF or TR-HF) and normal control rats. Grip strength declined with both tasks, with the greatest declines in 9-week HRHF rats. Quantitative PCR (qPCR) analyses of HRNF muscles showed increased expression of Col1 in weeks 3-9, and CTGF in weeks 6 and 9. Immunohistochemistry confirmed PCR results, and also showed greater increases of CTGF and collagen matrix in 9-week HRHF rats than 9-week HRNF rats. ELISA, and immunohistochemistry revealed greater increases of TGFB1 in TR-HF and 6-week HRHF, compared to 6-week HRNF rats. To examine the role of inflammation, results from 6-week HRHF rats were compared to rats receiving ibuprofen or anti-TNF-α treatment in HRHF weeks 4-6. Both treatments attenuated HRHF-induced increases in CTGF and fibrosis by 6 weeks of task performance. Ibuprofen attenuated TGFB1 increases and grip strength declines, matching our prior results with anti-TNFα. CONCLUSIONS/SIGNIFICANCE: Performance of highly repetitive tasks was associated with force-dependent declines in grip strength and increased fibrogenic-related proteins in flexor digitorum muscles. These changes were attenuated, at least short-term, by anti-inflammatory treatments.


Asunto(s)
Fuerza de la Mano , Músculo Esquelético/metabolismo , Animales , Antiinflamatorios no Esteroideos/farmacología , Colágeno Tipo I/metabolismo , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Femenino , Fibrosis , Miembro Anterior/efectos de los fármacos , Miembro Anterior/metabolismo , Miembro Anterior/fisiopatología , Ibuprofeno/farmacología , Inflamación/metabolismo , Ratas , Ratas Sprague-Dawley , Factor de Crecimiento Transformador beta1/metabolismo
15.
Arterioscler Thromb Vasc Biol ; 32(5): 1211-9, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22362759

RESUMEN

OBJECTIVE: Members of the protein kinase C (PKC) family are shown to positively and negatively regulate platelet activation. Although positive regulatory roles are extensively studied, negative regulatory roles of PKCs are poorly understood. We investigated the mechanism and specific isoforms involved in PKC-mediated negative regulation of ADP-induced functional responses. METHODS AND RESULTS: A pan-PKC inhibitor, GF109203X, potentiated ADP-induced cPLA(2) phosphorylation and thromboxane generation as well as ERK activation and intracellular calcium (Ca(2+)(i)) mobilization, 2 signaling molecules, upstream of cPLA(2) activation. Thus, PKCs inhibit cPLA(2) activation by inhibiting ERK and Ca(2+)(i) mobilization. Because the inhibitor of classic PKC isoforms, GO-6976, did not affect ADP-mediated thromboxane generation, we investigated the role of novel class of PKC isoforms. ADP-induced thromboxane generation, calcium mobilization, and ERK phosphorylation were potentiated in PKCε null murine platelets compared with platelets from wild-type littermates. Interestingly, when thromboxane release is blocked, ADP-induced aggregation in PKCε knockout and wild-type was similar, suggesting that PKCε does not affect ADP-induced aggregation directly. PKCε knockout mice exhibited shorter times to occlusion in an FeCl(3)-induced arterial injury model and shorter bleeding times in tail-bleeding experiments. CONCLUSIONS: We conclude that PKCε negatively regulates ADP-induced thromboxane generation in platelets and offers protection against thrombosis.


Asunto(s)
Adenosina Difosfato/farmacología , Plaquetas/metabolismo , Calcio/metabolismo , Agregación Plaquetaria/fisiología , Proteína Quinasa C-epsilon/metabolismo , Tromboxanos/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos , Líquido Intracelular/metabolismo , Ratones , Agregación Plaquetaria/efectos de los fármacos , Recuento de Plaquetas , Transducción de Señal , Trombosis/metabolismo , Trombosis/patología
16.
Thromb Res ; 129(6): 801-6, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21889790

RESUMEN

Activated factor X (FXa) and thrombin can up-regulate gene expression of connective tissue growth factor (CTGF/CCN2) on fibroblasts. Since tissue factor (TF) is expressed on these cells, we hypothesized that they may assemble the prothrombinase complex leading to CTGF/CCN2 upregulation. In addition, the effect of thrombospondin-1 (TSP1) on this reaction was evaluated. Human foreskin fibroblasts were incubated with purified factor VII (FVII), factor X (FX), factor V (FV), prothrombin and calcium in the presence and absence of TSP1. Generation of FXa and of thrombin were assessed using chromogenic substrates. SMAD pathway phosphorylation was detected via Western-blot analysis. Pre-incubation of fibroblasts with FVII led to its auto-activation by cell-surface expressed TF, which in turn in the presence of FX, FVa, prothrombin and calcium led to FXa (9.7±0.8nM) and thrombin (7.9±0.04 U/mL×10-3) generation. Addition of TSP1 significantly enhanced thrombin (23.3±0.7 U/mL×10-3) but not FXa (8.5±0.6nM) generation. FXa and thrombin generation leads to upregulation of CTGF/CCN2. TSP1 alone upregulated CTGF/CCN2, an effect mediated via activation of transforming growth factor beta (TGFß) as shown by phosphorylation of the SMAD pathway, an event blunted by using a TGFß receptor I inhibitor (TGFßRI). FXa- and thrombin-induced upregulation of CTGF/CCN2 was not blocked by TGFßRI. In summary, assembly of the prothrombinase complex occurs on fibroblast's surface leading to serine proteases generation, an event enhanced by TSP1 and associated with CTGF/CCN2 upregulation. These mechanisms may play an important role in human diseases associated with fibrosis.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Factor V/metabolismo , Factor Xa/metabolismo , Fibroblastos/metabolismo , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Línea Celular , Factor de Crecimiento del Tejido Conjuntivo/biosíntesis , Factor de Crecimiento del Tejido Conjuntivo/genética , Factor V/genética , Factor VII/metabolismo , Factor Xa/genética , Fibroblastos/enzimología , Prepucio/citología , Expresión Génica , Humanos , Masculino , Trombina/biosíntesis , Tromboplastina/biosíntesis , Tromboplastina/genética , Tromboplastina/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
17.
J Health Care Poor Underserved ; 22(4): 1144-50, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22080699

RESUMEN

This Report from the Field documents a series of interventions developed by Temple University Health System and School of Medicine through participation in the RWJF initiative entitled Hablamos Juntos. The report delineates outcomes to date demonstrating that these interventions have met the challenge of improving patient provider communication for Latinos.


Asunto(s)
Barreras de Comunicación , Comunicación , Multilingüismo , Relaciones Profesional-Paciente , Habilitación Profesional , Curriculum , Educación de Pregrado en Medicina/métodos , Evaluación Educacional , Hispánicos o Latinos , Hospitales Universitarios , Humanos , Philadelphia , Proyectos Piloto , Desarrollo de Programa
18.
PLoS One ; 6(10): e26035, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22028806

RESUMEN

The P2Y12 receptor plays a crucial role in the regulation of platelet activation by several agonists, which is irreversibly antagonized by the active metabolite of clopidogrel, a widely used anti-thrombotic drug. In this study, we investigated whether reduction of platelet reactivity leads to reduced inflammatory responses using a rat model of erosive arthritis. We evaluated the effect of clopidogrel on inflammation in Lewis rats in a peptidoglycan polysaccharide (PG-PS)-induced arthritis model with four groups of rats: 1) untreated, 2) clopidogrel-treated, 3) PG-PS-induced, and 4) PG-PS-induced and clopidogrel-treated. There were significant differences between the PG-PS+clopidogrel group when compared to the PG-PS group including: increased joint diameter and clinical manifestations of inflammation, elevated plasma levels of pro-inflammatory cytokines (IL-1 beta, interferon (IFN) gamma, and IL-6), an elevated neutrophil blood count and an increased circulating platelet count. Plasma levels of IL-10 were significantly lower in the PG-PS+clopidogrel group compared to the PG-PS group. Plasma levels of platelet factor 4 (PF4) were elevated in both the PG-PS and the PG-PS+clopidogrel groups, however PF4 levels showed no difference upon clopidogrel treatment, suggesting that the pro- inflammatory effect of clopidogrel may be due to its action on cells other than platelets. Histology indicated an increase in leukocyte infiltration at the inflammatory area of the joint, increased pannus formation, blood vessel proliferation, subsynovial fibrosis and cartilage erosion upon treatment with clopidogrel in PG-PS-induced arthritis animals. In summary, animals treated with clopidogrel showed a pro-inflammatory effect in the PG-PS-induced arthritis animal model, which might not be mediated by platelets. Elucidation of the mechanism of clopidogrel-induced cell responses is important to understand the role of the P2Y12 receptor in inflammation.


Asunto(s)
Artritis Experimental/inducido químicamente , Peptidoglicano/efectos adversos , Antagonistas del Receptor Purinérgico P2Y/farmacología , Receptores Purinérgicos P2Y12/metabolismo , Ticlopidina/análogos & derivados , Animales , Artritis Experimental/sangre , Artritis Experimental/complicaciones , Artritis Experimental/patología , Clopidogrel , Citocinas/sangre , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Femenino , Inflamación/sangre , Inflamación/inducido químicamente , Inflamación/complicaciones , Inflamación/patología , Leucocitosis/complicaciones , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Agregación Plaquetaria/efectos de los fármacos , Ratas , Trombocitosis/complicaciones , Ticlopidina/farmacología
19.
J Cell Physiol ; 226(11): 2943-52, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21302290

RESUMEN

Current osteoinductive protein therapy utilizes bolus administration of large doses of bone morphogenetic proteins (BMPs), which is costly, and may not replicate normal bone healing. The limited in vivo biologic activity of BMPs requires the investigation of growth factors that may enhance this activity. In this study, we utilized the C3H10T1/2 murine mesenchymal stem cell line to test the hypotheses that osteoactivin (OA) has comparable osteoinductive effects to bone morphogenetic protein-2 (BMP-2), and that sustained administration of either growth factor would result in increased osteoblastic differentiation as compared to bolus administration. Sustained release biodegradable hydrogels were designed, and C3H10T1/2 cells were grown on hydrogels loaded with BMP-2 or OA. Controls were grown on unloaded hydrogels, and positive controls were exposed to bolus growth factor administration. Cells were harvested at several time points to assess osteoblastic differentiation. Alkaline phosphatase (ALP) staining and activity, and gene expression of ALP and osteocalcin were assessed. Treatment with OA or BMP-2 resulted in comparable effects on osteoblastic marker expression. However, cells grown on hydrogels demonstrated osteoblastic differentiation that was not as robust as cells treated with bolus administration. This study shows that OA has comparable effects to BMP-2 on osteoblastic differentiation using both bolus administration and continuous release, and that bolus administration of OA has a more profound effect than administration using hydrogels for sustained release. This study will lead to a better understanding of appropriate delivery methods of osteogenic growth factors like OA for repair of fractures and segmental bone defects.


Asunto(s)
Proteína Morfogenética Ósea 2/administración & dosificación , Diferenciación Celular/efectos de los fármacos , Proteínas del Ojo/administración & dosificación , Glicoproteínas de Membrana/administración & dosificación , Células Madre Mesenquimatosas/efectos de los fármacos , Fosfatasa Alcalina/biosíntesis , Fosfatasa Alcalina/genética , Animales , Diferenciación Celular/genética , Línea Celular , Preparaciones de Acción Retardada , Expresión Génica/efectos de los fármacos , Hidrogel de Polietilenoglicol-Dimetacrilato/administración & dosificación , Células Madre Mesenquimatosas/citología , Ratones , Osteocalcina/biosíntesis , Osteocalcina/genética
20.
Curr Vasc Pharmacol ; 8(3): 338-43, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-19485899

RESUMEN

Biologic therapy for rheumatoid arthritis (RA) targets specific molecules that mediate and sustain the clinical manifestations of this complex illness. Compared with the general population, patients with RA die prematurely, in part due to associated cardiovascular disease. Even though the mechanisms by which premature atherosclerosis develops in RA is unknown, chronic inflammation may play a major role. This review connects current knowledge of the pathophysiology of RA with data available in the literature related to thrombospondin-1 (TSP1), transforming growth factor beta (TGFbeta and connective tissue growth factor (CTGF) and their relationship with cardiovascular disease in RA. The TSP1/TGFbeta/CTGF axis may contribute in the pro-inflammatory and pro-atherogenic state in patients affected with RA. In fact, increased TSP1 plasma levels are found in patients of RA. TGFbeta is activated by TSP1 through a non-enzymatic mechanism and is constitutively overexpressed by synovial fibroblasts from RA patients. Activation of TGFbeta pathway in synovial fibroblasts and other cells including neutrophils leads to downstream upregulation of CTGF. Overexpression of CTGF is associated with angiogenesis, fibrosis, atherosclerotic blood vessels and erosive arthritis lesions. Recent RA therapies emphasize the need for aggressive control of the activity of the disease to prevent premature atherosclerosis in RA patients. The complexity and heterogeneity of RA as judged by response to a wide spectrum of treatments mandates the elucidation of unknown pro-inflammatory pathways playing a major role in this disease. The TSP1/TGFbeta/CTFG axis represents one of these pro-inflammatory pathways that may result in the development of promising therapeutic strategies to prevent chronic inflammation and thus premature atherosclerosis in RA.


Asunto(s)
Antirreumáticos/metabolismo , Artritis Reumatoide/metabolismo , Factor de Crecimiento del Tejido Conjuntivo/fisiología , Sistemas de Liberación de Medicamentos/tendencias , Trombospondina 1/fisiología , Factor de Crecimiento Transformador beta/fisiología , Animales , Antirreumáticos/administración & dosificación , Artritis Reumatoide/complicaciones , Artritis Reumatoide/tratamiento farmacológico , Aterosclerosis/complicaciones , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/metabolismo , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Humanos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Trombospondina 1/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...