Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Am J Pathol ; 185(3): 631-42, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25701883

RESUMEN

Heart failure, the leading cause of hospitalization of elderly patients, is correlated with myocardial fibrosis (ie, deposition of excess extracellular matrix proteins such as collagen). A key regulator of collagen homeostasis is lysyl oxidase (LOX), an enzyme responsible for cross-linking collagen fibers. Our objective was to ameliorate age-related myocardial fibrosis by disrupting collagen cross-linking through inhibition of LOX. The nonreversible LOX inhibitor ß-aminopropionitrile (BAPN) was administered by osmotic minipump to 38-week-old C57BL/6J male mice for 2 weeks. Sirius Red staining of myocardial cross sections revealed a reduction in fibrosis, compared with age-matched controls (5.84 ± 0.30% versus 10.17 ± 1.34%) (P < 0.05), to a level similar to that of young mice at 8 weeks (4.9 ± 1.2%). BAPN significantly reduced COL1A1 mRNA, compared with age-matched mice (3.5 ± 0.3-fold versus 15.2 ± 4.9-fold) (P < 0.05), suggesting that LOX is involved in regulation of collagen synthesis. In accord, fibrotic factor mRNA expression was reduced after BAPN. There was also a novel increase in Ly6C expression by resident macrophages. By interrupting collagen cross-linking by LOX, the BAPN treatment reduced myocardial fibrosis. A novel observation is that BAPN treatment modulated the transforming growth factor-ß pathway, collagen synthesis, and the resident macrophage population. This is especially valuable in terms of potential therapeutic targeting of collagen regulation and thereby age-related myocardial fibrosis.


Asunto(s)
Aminopropionitrilo/uso terapéutico , Colágeno/metabolismo , Cardiopatías/tratamiento farmacológico , Corazón/efectos de los fármacos , Miocardio/metabolismo , Factores de Edad , Aminopropionitrilo/farmacología , Animales , Fibrosis/metabolismo , Fibrosis/patología , Cardiopatías/metabolismo , Cardiopatías/patología , Masculino , Ratones , Miocardio/patología
2.
Am J Pathol ; 185(4): 927-42, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25794704

RESUMEN

Macrophages are increasingly recognized as a potential therapeutic target in myocardial fibrosis via interactions with fibroblasts. We have characterized macrophage depletion and inhibition of nonclassical macrophage migration, in addition to direct interactions between nonclassical macrophages and fibroblasts in angiotensin II (AngII)-mediated, hypertensive myocardial fibrosis. Macrophage depletion was achieved by daily i.v. clodronate liposomes (-1 day to +3 days) during AngII infusion. Cx3cr1(-/-) mice were used to inhibit nonclassical macrophage migration. Macrophage phenotype (F4/80, CD11b, Ly6C) was characterized by immunofluorescence and flow cytometry. Collagen was assessed by Sirius Red/Fast Green. Quantitative real-time RT-PCR was performed for transcript levels. AngII/wild-type (WT) mice displayed significant infiltrate and fibrosis compared with saline/WT, which was virtually ablated by clodronate liposomes independent of hypertension. In vitro data supported M2 macrophages promoting fibroblast differentiation and collagen production. AngII/Cx3cr1(-/-) mice, however, significantly increased macrophage infiltrate and fibrosis relative to AngII/WT. AngII/Cx3cr1(-/-) mice also showed an M1 phenotypic shift relative to WT mice in, which the predominant phenotype was Ly6C(low), CD206(+) (M2). Myocardial IL-1ß was significantly up-regulated, whereas transforming growth factor ß down-regulated with this M1 shift. We demonstrated that infiltrating macrophages are critical to AngII-mediated myocardial fibrosis by preventing the development of fibrosis after liposomal depletion of circulating monocytes. Our findings also suggest that some macrophages, namely M2, may confer a protective myocardial environment that may prevent excessive tissue injury.


Asunto(s)
Macrófagos/metabolismo , Miocardio/patología , Actinas/metabolismo , Administración Intravenosa , Angiotensina II/administración & dosificación , Angiotensina II/farmacología , Animales , Antígenos Ly/metabolismo , Receptor 1 de Quimiocinas CX3C , Ácido Clodrónico/administración & dosificación , Ácido Clodrónico/farmacología , Colágeno/biosíntesis , Electrocardiografía , Fibrosis , Mediadores de Inflamación/metabolismo , Liposomas/administración & dosificación , Liposomas/farmacología , Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Miocardio/metabolismo , Células 3T3 NIH , Receptores de Quimiocina/deficiencia , Receptores de Quimiocina/metabolismo
3.
Am J Pathol ; 183(2): 459-69, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23731726

RESUMEN

Fibroblast progenitor cells (fibrocytes) are important to the development of myocardial fibrosis and are suggested to migrate to the heart via CXCL12 and chemokine ligand (CCL) 2. We hypothesized that if these chemokines are recruiting fibrocytes, disrupting their signaling will reduce early (3-day) fibrocyte infiltration and, consequently, fibrosis in the myocardium. C57/Bl6 and CCR2(-/-) mice were infused with saline or angiotensin (Ang) II, with or without CXC receptor 4 blockade (AMD3100). Hearts were assessed for chemokine up-regulation, immunofluorescence, and histological features. AngII caused early myocardial up-regulation of CXCL12 and CCL2, which corresponded to significant myocardial infiltration and fibrosis compared with controls. Animals receiving AMD3100 and/or with the genotype CCR2(-/-) failed to demonstrate reductions in infiltrate or fibrosis after 3 days of AngII, and AngII + AMD3100 animals showed exacerbated fibrocyte infiltration and fibrosis compared with AngII alone. CCR2(-/-) mice demonstrated significant reductions in myocardial fibrosis relative to wild type, but this was after 28 days of AngII infusion and was the result of reduced infiltrating cell proliferation. An alternative CCR2 ligand, CCL12, was found to be increasing infiltrating cell proliferation in the heart after AngII infusion, which we confirmed in vitro. In conclusion, early fibrocyte recruitment cannot be inhibited through modulating CXCL12 or CCL2, as previously thought. Ablating CCR2 signaling did confer myocardial fibrosis reductions, but these benefits were not observed until much later and were likely the result of modulated proliferation through ablating the CCL12-CCR2 interaction.


Asunto(s)
Angiotensina II/farmacología , Quimiocina CCL2/fisiología , Quimiocina CXCL12/fisiología , Fibroblastos/fisiología , Miocardio/metabolismo , Vasoconstrictores/farmacología , Animales , Bencilaminas , Movimiento Celular/fisiología , Proliferación Celular/efectos de los fármacos , Ciclamas , Fibrosis/patología , Compuestos Heterocíclicos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Miocardio/patología , Receptores CXCR4/antagonistas & inhibidores , Células Madre/fisiología , Regulación hacia Arriba
4.
Am J Pathol ; 182(3): 714-26, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23287510

RESUMEN

Exposure of rodents to angiotensin II (AngII) is a common model of fibrosis. We have previously shown that cellular infiltration of bone marrow-derived progenitor cells (fibrocytes) occurs before deposition of extracellular matrix and is associated with the production of connective tissue growth factor (CTGF). In the present study, we characterized the role of CTGF in promoting fibrocyte accumulation and regulation after AngII exposure. In animals exposed to AngII using osmotic minipumps (2.0 µg/kg per min), myocardial CTGF mRNA peaked at 6 hours (21-fold; P < 0.01), whereas transforming growth factor-ß (TGF-ß) peaked at 3 days (fivefold; P < 0.05) compared with saline control. Early CTGF expression occurred before fibrocyte migration (1 day) into the myocardium or ECM deposition (3 days). CTGF protein expression was evident by day 3 of AngII exposure and seemed to be localized to resident cells. Isolated cardiomyocytes and microvascular endothelial cells responded to AngII with increased CTGF production (2.1-fold and 2.8-fold, respectively; P < 0.05), which was abolished with the addition of anti-TGF-ß neutralizing antibody. The effect of CTGF on isolated fibrocytes suggested a role in fibrocyte proliferation (twofold; P < 0.05) and collagen production (2.3-fold; P < 0.05). In summary, we provide strong evidence that AngII exposure first resulted in Smad2-dependent production of CTGF by resident cells (6 hours), well before the accumulation of fibrocytes or TGF-ß mRNA up-regulation. In addition, CTGF contributes to fibrocyte proliferation in the myocardium and enhances fibrocyte differentiation into a myofibroblast phenotype responsible for ECM deposition.


Asunto(s)
Angiotensina II/farmacología , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Miocardio/metabolismo , Miocardio/patología , Animales , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Separación Celular , Factor de Crecimiento del Tejido Conjuntivo/farmacología , Citocinas/metabolismo , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Fibrosis , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Factor de Crecimiento Transformador beta/metabolismo
5.
Int J Exp Pathol ; 93(2): 115-24, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22225615

RESUMEN

Using an established model of myocardial hypertrophy and fibrosis after angiotensin II (AngII) infusion, our aim was to characterize the early cellular element involved in the development of myocardial fibrosis in detail. Male Lewis rats were infused with saline or AngII (0.7 mg/kg per day) for up to seven days. Collagen deposition and cellular infiltration were identified by histology stains. Infiltrating cells were grown in vitro and examined by flow cytometry and immunostaining. Chemokine expression was measured using qRT-PCR. AngII infusion resulted in multifocal myocardial cellular infiltration (peak at three days) that preceded collagen deposition. Monocyte chemotactic protein (MCP)-1 transcripts peaked after one day of AngII exposure. Using a triple-labelling technique, the infiltrating cells were found to express markers of leucocyte (ED1(+)), mesenchymal [α-smooth muscle actin (SMA)(+)] and haematopeotic progenitor cells (CD133(+)) suggesting a fibroblast progenitor phenotype. In vitro, ED1(+)/SMA(+)/CD133(+) cells were isolated and grown from AngII-exposed animals. Comparatively few cells were cultured from untreated control hearts, and they were found to be ED1(-)/SMA(+)/CD133(-). We provide evidence that myocardial ECM deposition is preceded by infiltration into the myocardium by cells that express a combination of haematopoietic (ED1, CD133) and mesenchymal (SMA) cell markers, which is a characteristic of the phenotype of fibroblast precursor cells, termed fibrocytes. This suggests that fibrocytes rather than (as is often presumed) leucocytes may have effector functions in the initiation of myocardial fibrosis.


Asunto(s)
Angiotensina II/toxicidad , Fibroblastos/patología , Células Madre Mesenquimatosas/patología , Miocardio/patología , Antígeno AC133 , Actinas/metabolismo , Animales , Antígenos CD/metabolismo , Biomarcadores/metabolismo , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Quimiocina CCL2/metabolismo , Quimiocinas/metabolismo , Colágeno/metabolismo , Modelos Animales de Enfermedad , Ectodisplasinas/metabolismo , Matriz Extracelular/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibrosis , Glicoproteínas/metabolismo , Corazón/efectos de los fármacos , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Miocardio/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Péptidos/metabolismo , Ratas , Ratas Endogámicas Lew
6.
EClinicalMedicine ; 37: 100975, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34222846

RESUMEN

BACKGROUND: The SARS-CoV-2 (Severe Acute Respiratory Syndrome coronavirus 2) has led to more than 165 million COVID-19 cases and >3.4 million deaths worldwide. Epidemiological analysis has revealed that the risk of developing severe COVID-19 increases with age. Despite a disproportionate number of older individuals and long-term care facilities being affected by SARS-CoV-2 and COVID-19, very little is understood about the immune responses and development of humoral immunity in the extremely old person after SARS-CoV-2 infection. Here we conducted a serological study to investigate the development of humoral immunity in centenarians following a SARS-CoV-2 outbreak in a long-term care facility. METHODS: Extreme aged individuals and centenarians who were residents in a long-term care facility and infected with or exposed to SARS-CoV-2 were investigated between April and June 2020 for the development of antibodies to SARS-CoV-2. Blood samples were collected from positive and bystander individuals 30 and 60 days after original diagnosis of SARS-CoV-2 infection. Plasma was used to quantify IgG, IgA, and IgM isotypes and subsequent subclasses of antibodies specific for SARS-CoV-2 spike protein. The function of anti-spike was then assessed by virus neutralization assays against the native SARS-CoV-2 virus. FINDINGS: Fifteen long-term care residents were investigated for SARS-CoV-2 infection. All individuals had a Clinical Frailty scale score ≥5 and were of extreme older age or were centenarians. Six women with a median age of 98.8 years tested positive for SARS-CoV-2. Anti-spike IgG antibody titers were the highest titers observed in our cohort with all IgG positive individuals having virus neutralization ability. Additionally, 5 out of the 6 positive participants had a robust IgA anti-SARS-CoV-2 response. In all 5, antibodies were detected after 60 days from initial diagnosis.

7.
J Renin Angiotensin Aldosterone Syst ; 19(1): 1470320318759358, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29575960

RESUMEN

INTRODUCTION: Transforming growth factor-ß (TGF-ß) and connective tissue growth factor (CTGF) are often described as the initial pro-fibrotic mediators upregulated early in fibrosis models dependent on angiotensin II (Ang-II). In the present study, we explore the mechanistic link between TGF-ß and CTGF expression by using a novel TGF-ß trap. MATERIALS AND METHODS: NIH/3T3 fibroblasts were subjected to TGF-ß with or without TGF-ß trap or 1D11 antibody, CTGF or CTGF plus TGF-ß for six or 24 hours, and then used for quantitative real-time polymerase chain reaction (qRT-PCR) or immunocytochemistry. Male C57BL/6 mice were infused with Ang-II and randomly assigned TGF-ß trap for six or 24 hours. Hearts were harvested for histological analyses, qRT-PCR and western blotting. RESULTS: Exogenous TGF-ß-induced fibroblasts resulted in significant upregulation of CTGF, TGF-ß and type I collagen transcript levels in vitro. Additionally, TGF-ß promoted the differentiation of fibroblasts into α-SMA+ myofibroblasts. CTGF expression was reduced by the addition of TGF-ß trap or neutralizing antibody, confirming that its expression is dependent on TGF-ß signaling. In contrast, exogenous CTGF did not appear to have an effect on fibroblast production of pro-fibrotic transcripts or fibroblast differentiation. Ang-II infusion in vivo led to a significant increase in TGF-ß and CTGF mRNA expression at six and 24 hours with corresponding changes in Smad2 phosphorylation (pSmad2), indicative of increased TGF-ß signaling. Ang-II animals that received the TGF-ß trap demonstrated reduced CTGF mRNA levels and pSmad2 at six hours, suggesting that early CTGF expression is dependent on TGF-ß signaling. CONCLUSIONS: We demonstrated that CTGF expression is dependent on TGF-ß signaling both in vitro and in vivo in a model of myocardial fibrosis. This also suggests that early myocardial CTGF mRNA expression (six hours) after Ang-II exposure is likely dependent on latent TGF-ß activation via the canonical Smad-dependent pathway in resident cardiac cells.


Asunto(s)
Angiotensina II/farmacología , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Hipertensión/metabolismo , Miocardio/metabolismo , Miocardio/patología , Transducción de Señal , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Factor de Crecimiento del Tejido Conjuntivo/genética , Fibrosis , Masculino , Ratones , Ratones Endogámicos C57BL , Miofibroblastos/efectos de los fármacos , Miofibroblastos/metabolismo , Miofibroblastos/patología , Células 3T3 NIH , Fosforilación/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta/genética , Regulación hacia Arriba/efectos de los fármacos
8.
ESC Heart Fail ; 5(1): 107-114, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28737273

RESUMEN

AIMS: Previous studies have examined risk factors for the development of heart failure (HF) subsequent to acute coronary syndrome (ACS). Our study seeks to clarify the clinical variables that best characterize patients who remain free from HF after coronary artery bypass grafting (CABG) surgery for ACS to determine novel biological factors favouring freedom from HF in prospective translational studies. METHODS AND RESULTS: Nova Scotia residents (1995-2012) undergoing CABG within 3 weeks of ACS were included. The primary outcome was freedom from readmission to hospital due to HF. Descriptive statistics were generated, and a Cox proportional hazards model assessed outcome with adjustment for clinical characteristics. Of 11 936 Nova Scotians who underwent isolated CABG, 3264 (27%) had a recent ACS and were included. Deaths occurred in 210 (6%) of subjects prior to discharge. A total of 3054 patients were included in the long-term analysis. During follow-up, HF necessitating readmission occurred in 688 (21%) subjects with a hazard ratio of 12% at 2 years. The adjusted Cox model demonstrated significantly better freedom from HF for younger, male subjects without metabolic syndrome and no history of chronic obstructive pulmonary disease, renal insufficiency, atrial fibrillation, or HF. CONCLUSIONS: Our findings have outlined important clinical variables that predict freedom from HF. Furthermore, we have shown that 12% of patients undergoing CABG after ACS develop HF (2 years). Our findings support our next phase in which we plan to prospectively collect blood and tissue specimens from ACS patients undergoing CABG in order to determine novel biological mechanism(s) that favour resolution of post-ACS inflammation.


Asunto(s)
Síndrome Coronario Agudo/fisiopatología , Puente de Arteria Coronaria , Frecuencia Cardíaca/fisiología , Recuperación de la Función , Sistema de Registros , Medición de Riesgo , Síndrome Coronario Agudo/mortalidad , Síndrome Coronario Agudo/cirugía , Anciano , Anciano de 80 o más Años , Femenino , Estudios de Seguimiento , Insuficiencia Cardíaca , Mortalidad Hospitalaria/tendencias , Humanos , Masculino , Persona de Mediana Edad , Nueva Escocia/epidemiología , Estudios Prospectivos , Factores de Riesgo , Factores de Tiempo
9.
Cardiovasc Pathol ; 25(5): 390-8, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27327107

RESUMEN

BACKGROUND: Macrophages (MΦ) are functionally diverse and dynamic. Until recently, cardiac MΦ were assumed to be monocyte derived; however, resident cardiac MΦ (rCMΦ), present at baseline, were identified in myocardia and have been implicated in cardiac healing. Previously, we demonstrated that CCR2(-/-) mice are protected from myocardial fibrosis - an observation initially attributed to changes in infiltrating monocytes. Here, we reexplored this observation in the context of our new understanding of rCMΦ. METHODS: Male CCR2(-/-) and C57BL/6 hearts were digested and purified to a single cell suspension, incubated with fluorophore-linked antibodies (CCR2, CX3CR1, CD11b, Ly6C, TNF-α, and IL-10), and assessed by flow cytometry. Differentiated MΦ were cocultured with fibroblasts in order to characterize how MΦ phenotype influences fibroblast activation. Fibroblasts were characterized for their expression of smooth muscle cell actin (SMA). RESULTS: A significant decrease in Ly6C expression was observed in the CCR2(-/-) cardiac MΦ population relative to WT, which corresponded with significantly lower TNF-α expression and significantly higher IL-10 expression. Using in vitro coculture system, classical MΦ promoted fibroblast activation relative to nonclassical MΦ. CONCLUSION: CCR2(-/-) rCMΦ favor a more antiinflammatory phenotype relative to WT controls. Moreover, a shift toward the antiinflammatory promotes proliferation, but not activation in vitro. Together, these observations suggest that antiinflammatory cardiac MΦ populations may inhibit myocardial fibrosis in a pathological setting by preventing the activation of fibroblasts. NEWS AND NOTEWORTHY: Here, we provide novel evidence for baseline differences in rCMΦ phenotypes (i.e. classical vs. nonclassical) and how these differences could modulate cardiac healing. Importantly, we observed differences in how classical vs. nonclassical MΦ influenced fibroblast activation, which could, in turn, affect fibrosis.


Asunto(s)
Fibroblastos/patología , Cardiopatías/metabolismo , Macrófagos/patología , Miocardio/patología , Receptores CCR2/metabolismo , Animales , Proliferación Celular , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Fibroblastos/metabolismo , Fibrosis/patología , Citometría de Flujo , Cardiopatías/patología , Inmunohistoquímica , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Receptores CCR2/deficiencia
10.
Artículo en Inglés | MEDLINE | ID: mdl-24721281

RESUMEN

BACKGROUND: Myocardial fibrosis is a pathological process that is characterized by disrupted regulation of extracellular matrix proteins resulting in permanent scarring of the heart tissue and eventual diastolic heart failure. Pro-fibrotic molecules including transforming growth factor-ß and connective tissue growth factor are expressed early in the AngiotensinII (AngII)-induced and other models of myocardial fibrosis. As such, antibody-based therapies against these and other targets are currently under development. RESULTS: In the present study, C57Bl/6 mice were subcutaneously implanted with a mini-osmotic pump containing either AngII (2.0 µg/kg/min) or saline control for 3 days in combination with mIgG (1 mg/kg/d) injected through the tail vein. Fibrosis was assessed after picosirius red staining of myocardial cross-sections and was significantly increased after AngII exposure compared to saline control (11.37 ± 1.41%, 4.94 ± 1.15%; P <0.05). Non-specific mIgG treatment (1 mg/kg/d) significantly increased the amount of fibrosis (26.34 ± 3.03%; P <0.01). However, when AngII exposed animals were treated with a Fab fragment of the mIgG or mIgM, this exacerbation of fibrosis was no longer observed (14.49 ± 2.23%; not significantly different from AngII alone). CONCLUSIONS: These data suggest that myocardial fibrosis was increased by the addition of exogenous non-specific antibodies in an Fc-mediated manner. These findings could have substantial impact on the future experimental design of antibody-based therapeutics.

11.
Hypertens Res ; 35(4): 449-56, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22258030

RESUMEN

Activation of the renin-angiotensin system (RAS) is thought to promote myocardial fibrosis. However, it is unclear whether this physiological fibrotic response results from chronic hemodynamic stress or from direct cellular signaling. Male C57B/6 mice were randomly assigned to receive angiotensin II (AngII) (2.0 µg kg(-1) min(-1)), AngII+hydralazine (6.9 µg kg(-1) min(-1)) or saline (control) via osmotic pumps for 7 days. Blood pressure was measured via noninvasive plethysmography. Hearts were harvested and processed for analysis. Cellular infiltration and collagen deposition were analyzed using histological staining. Molecular mediators were assessed using quantitative RT-PCR. As previously described, animals that received AngII developed hypertension and multifocal cellular infiltration by SMA(+)/CD133(+) fibroblast progenitors followed by collagen deposition. The coadministration of hydralazine with AngII completely inhibited the hypertensive effects of AngII (P0.01) and resulted in minimal cellular infiltration and minimal collagen deposition. These findings were in the context of persistent RAS activation, which was evidenced by elevation in serum aldosterone levels in animals that received AngII or AngII+hydralazine compared with animals that received saline. At the molecular level, infusion of AngII resulted in the significant upregulation of profibrotic factors (connective tissue growth factor-7.8±0.7 fold), proinflammatory mediators (TNFα-4.6±0.8 fold; IL-1ß-6.4±2.6 fold) and chemokines (CCL2-3.8±1.0 fold; CXCL12-3.2±0.4 fold), which were inhibited when hydralazine was also infused. We provide evidence that myocardial infiltration by fibroblast progenitor cells secondary to AngII and the resultant fibrosis can be prevented by the addition of hydralazine. Furthermore, the beneficial effects of hydralazine were observed while maintaining RAS activation, suggesting that the mechanism of fibrosis is blood pressure dependent.


Asunto(s)
Angiotensina II/farmacología , Presión Sanguínea/fisiología , Movimiento Celular/fisiología , Fibroblastos/patología , Hipertensión/patología , Miocardio/patología , Aldosterona/sangre , Animales , Presión Sanguínea/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Colágeno/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibrosis , Corazón/efectos de los fármacos , Hipertensión/inducido químicamente , Hipertensión/metabolismo , Masculino , Ratones , Miocardio/metabolismo , Sistema Renina-Angiotensina/efectos de los fármacos , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Células Madre/patología
12.
PLoS One ; 7(9): e45663, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23029168

RESUMEN

AIMS: Myocardial fibrosis contributes to the development of heart failure. Activated Protein C (aPC) is a circulating anticoagulant with anti-inflammatory and cytoprotective properties. Using a model of myocardial fibrosis second to Angiotensin II (AngII) infusion, we investigated the novel therapeutic function aPC in the development of fibrosis. METHODS AND RESULTS: C57Bl/6 and Tie2-EPCR mice were infused with AngII (2.0 µg/kg/min), AngII and aPC (0.4 µg/kg/min) or saline for 3d. Hearts were harvested and processed for analysis or used for cellular isolation. Basic histology and collagen deposition were assessed using histologic stains. Transcript levels of molecular mediators were analyzed by quantitative RT-PCR. Mice infused with AngII exhibited multifocal areas of myocardial cellular infiltration associated with significant collagen deposition compared to saline control animals (p<0.01). AngII-aPC infusion inhibited this cellular infiltration and the corresponding collagen deposition. AngII-aPC infusion also inhibited significant expression of the pro-fibrotic cytokines TGF-ß1, CTGF and PDGF found in AngII only infused animals (p<0.05). aPC signals through its receptor, EPCR. Using Tie2-EPCR animals, where endothelial cells over-express EPCR and exhibit enhanced aPC-EPCR signaling, no significant reduction in cellular infiltration or fibrosis was evident with AngII infusion suggesting aPC-mediate protection is endothelial cell independent. Isolated infiltrating cells expressed significant EPCR transcripts suggesting a direct effect on infiltrating cells. CONCLUSIONS: This data indicates that aPC treatment abrogates the fibrogenic response to AngII. aPC does not appear to confer protection by stimulating the endothelium but by acting directly on the infiltrating cells, potentially inhibiting migration or activation.


Asunto(s)
Angiotensina II/administración & dosificación , Modelos Animales de Enfermedad , Fibrosis/prevención & control , Cardiopatías/prevención & control , Proteína C/farmacología , Animales , Secuencia de Bases , Citocinas/metabolismo , Cartilla de ADN , Fibrosis/metabolismo , Técnica del Anticuerpo Fluorescente , Cardiopatías/metabolismo , Etiquetado Corte-Fin in Situ , Masculino , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
J Renin Angiotensin Aldosterone Syst ; 12(3): 295-303, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21393356

RESUMEN

HYPOTHESIS: Monocytes produce pro-inflammatory cytokines in response to Angiotensin II (AngII). INTRODUCTION: AngII has been suggested by many to be pro-inflammatory and likely to contribute to the migration of leukocytes in patients with cardiovascular conditions. MATERIALS AND METHODS: Monocytes were purified from peripheral blood mononuclear cells (PBMCs) by negative selection using antibodies conjugated to magnetic beads. Detection of CD14(+) and AT(1)R expression was achieved by double-labeling flow cytometry. Highly purified monocytes were then stimulated with AngII (6 and 24 h) to assess IL-6 and TNF-α transcript levels by qRT-PCR and protein secretion by ELISA. RESULTS: Monocytes comprised 9.7 ± 2.0% of the PBMCs. Monocyte isolation by negative selection yielded a purity of up to 99.8%. We demonstrated AT(1)R expression on 9.5 ± 0.3% of highly purifed CD14(+)/CD16(-) monocytes. Stimulation of highly purified monocytes with AngII resulted in increased transcript levels of IL-6 at 6 h but not at 24 h, and increased secretion of IL-6 in a dose-dependent manner compared with controls (p <0.01). Conversely, there was no increase in TNF-α mRNA transcripts or protein secretion. CONCLUSIONS: We provide evidence that a CD14(+)/CD16(-) subset of highly purified human monocytes express AT(1)R and respond to AngII exposure in vitro by producing IL-6 but not TNF-α.


Asunto(s)
Angiotensina II/farmacología , Interleucina-6/biosíntesis , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Factor de Necrosis Tumoral alfa/biosíntesis , Adulto , Separación Celular , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Interleucina-1/genética , Interleucina-1/metabolismo , Interleucina-6/genética , Receptores de Lipopolisacáridos/metabolismo , Masculino , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Receptores de IgG/metabolismo , Factor de Necrosis Tumoral alfa/genética
14.
J Heart Lung Transplant ; 26(12): 1336-9, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18096488

RESUMEN

Implantable cardioverter-defibrillators (ICDs) have been shown to reduce sudden cardiac death in select patients with impaired left ventricular function. However, consensus guidelines on ICD use have not historically addressed patients waiting for heart transplantation, and further evidence is needed to broaden and strengthen current recommendations. The objective of the present study was to review all patients listed for heart transplantation at a single institution and evaluate the impact of ICD implantation while waiting. All consecutive patients listed for heart transplantation at the Queen Elizabeth II Health Sciences Center, Halifax, Nova Scotia, from 1995 to July 2006, were included in the study (n = 124). We observed 12 deaths while waiting among patients listed for transplantation (10%), with all deaths occurring in the non-ICD patients. In patients who did have an ICD prior to transplantation, 17% received appropriate defibrillation therapy while awaiting transplantation, and 3 of 12 patients in the non-ICD population who died while waiting died suddenly, suggesting that ICDs could be used as a "bridge to transplantation" in patients with refractory heart failure who are to be listed for heart transplantation.


Asunto(s)
Desfibriladores Implantables , Insuficiencia Cardíaca/terapia , Trasplante de Corazón , Adulto , Anciano , Muerte Súbita Cardíaca/prevención & control , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Nueva Escocia , Análisis de Supervivencia , Resultado del Tratamiento , Listas de Espera
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA