Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Physiol Regul Integr Comp Physiol ; 324(4): R589-R600, 2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-36878484

RESUMEN

Androgen-deprivation therapy (ADT) is the primary systemic therapy for treating advanced or metastatic prostate cancer (PCa), which has improved survival outcomes in patients with PCa. However, ADT may develop metabolic and cardiovascular adverse events that impact the quality of life and lifespan in PCa survivors. The present study was designed to establish a murine model of ADT with a gonadotropin-releasing hormone (GnRH) agonist leuprolide and to investigate its effects on metabolism and cardiac function. We also examined the potential cardioprotective role of sildenafil (inhibitor of phosphodiesterase 5) under chronic ADT. Middle-aged male C57BL/6J mice received a 12-wk subcutaneous infusion via osmotic minipumps containing either saline or 18 mg/4 wk leuprolide with or without 1.3 mg/4 wk sildenafil cotreatment. Compared with saline controls, leuprolide treatment significantly reduced prostate weight and serum testosterone levels, confirming chemical castration in these mice. The ADT-induced chemical castration was not affected by sildenafil. Leuprolide significantly increased the weight of abdominal fat after 12-wk treatment without a change in total body weight, and sildenafil did not block the proadipogenic effect of leuprolide. No signs of left ventricular systolic and diastolic dysfunction were observed throughout the leuprolide treatment period. Interestingly, leuprolide treatment significantly elevated serum levels of cardiac troponin I (cTn-I), a biomarker of cardiac injury, and sildenafil did not abolish this effect. We conclude that long-term ADT with leuprolide increases abdominal adiposity and cardiac injury biomarker without cardiac contractile dysfunction. Sildenafil did not prevent ADT-associated adverse changes.


Asunto(s)
Cardiopatías , Neoplasias de la Próstata , Humanos , Masculino , Animales , Ratones , Leuprolida/efectos adversos , Citrato de Sildenafil/farmacología , Neoplasias de la Próstata/inducido químicamente , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/terapia , Antagonistas de Andrógenos/efectos adversos , Andrógenos , Adiposidad , Calidad de Vida , Ratones Endogámicos C57BL , Cardiopatías/inducido químicamente , Hormona Liberadora de Gonadotropina
2.
Int J Mol Sci ; 24(10)2023 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-37240345

RESUMEN

Robust activation of mTOR (mammalian target of rapamycin) signaling in diabetes exacerbates myocardial injury following lethal ischemia due to accelerated cardiomyocyte death with cardiac remodeling and inflammatory responses. We examined the effect of rapamycin (RAPA, mTOR inhibitor) on cardiac remodeling and inflammation following myocardial ischemia/reperfusion (I/R) injury in diabetic rabbits. Diabetic rabbits (DM) were subjected to 45 min of ischemia and 10 days of reperfusion by inflating/deflating a previously implanted hydraulic balloon occluder. RAPA (0.25 mg/kg, i.v.) or DMSO (vehicle) was infused 5 min before the onset of reperfusion. Post-I/R left ventricular (LV) function was assessed by echocardiography and fibrosis was evaluated by picrosirius red staining. Treatment with RAPA preserved LV ejection fraction and reduced fibrosis. Immunoblot and real-time PCR revealed that RAPA treatment inhibited several fibrosis markers (TGF-ß, Galectin-3, MYH, p-SMAD). Furthermore, immunofluorescence staining revealed the attenuation of post-I/R NLRP3-inflammasome formation with RAPA treatment as shown by reduced aggregation of apoptosis speck-like protein with a caspase recruitment domain and active-form of caspase-1 in cardiomyocytes. In conclusion, our study suggests that acute reperfusion therapy with RAPA may be a viable strategy to preserve cardiac function with the alleviation of adverse post-infarct myocardial remodeling and inflammation in diabetic patients.


Asunto(s)
Diabetes Mellitus , Daño por Reperfusión Miocárdica , Animales , Conejos , Sirolimus/farmacología , Sirolimus/uso terapéutico , Remodelación Ventricular , Miocitos Cardíacos/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Diabetes Mellitus/patología , Inflamación/tratamiento farmacológico , Inflamación/patología , Isquemia/patología , Fibrosis , Mamíferos/metabolismo
3.
Int Wound J ; 13(4): 572-84, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26290474

RESUMEN

Vitamin C (VitC) or ascorbic acid (AscA), a cofactor for collagen synthesis and a primary antioxidant, is rapidly consumed post-wounding. Parenteral VitC administration suppresses pro-inflammatory responses while promoting anti-inflammatory and pro-resolution effects in human/murine sepsis. We hypothesised that VitC could promote wound healing by altering the inflammatory, proliferative and remodelling phases of wound healing. Mice unable to synthesise VitC (Gulo(-/-) ) were used in this study. VitC was provided in the water (sufficient), withheld from another group (deficient) and supplemented by daily intra-peritoneal infusion (200 mg/kg, deficient + AscA) in a third group. Full thickness excisional wounds (6 mm) were created and tissue collected on days 7 and 14 for histology, quantitative polymerase chain reaction (qPCR) and Western blotting. Human neonatal dermal fibroblasts (HnDFs) were used to assess effects of In conclusion, VitC favorably on proliferation. Histological analysis showed improved wound matrix deposition and organisation in sufficient and deficient +AscA mice. Wounds from VitC sufficient and deficient + AscA mice had reduced expression of pro-inflammatory mediators and higher expression of wound healing mediators. Supplementation of HnDF with AscA induced the expression of self-renewal genes and promoted fibroblast proliferation. VitC favourably impacts the spatiotemporal expression of transcripts associated with early resolution of inflammation and tissue remodelling.


Asunto(s)
Cicatrización de Heridas , Animales , Antioxidantes , Ácido Ascórbico , Fibroblastos , Humanos , Inflamación , Ratones
4.
Eur Respir J ; 45(2): 449-62, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25261325

RESUMEN

Prostacyclin and its analogues improve cardiac output and functional capacity in patients with pulmonary arterial hypertension (PAH); however, the underlying mechanism is not fully understood. We hypothesised that prostanoids have load-independent beneficial effects on the right ventricle (RV). Angio-obliterative PAH and RV failure were induced in rats with a single injection of SU5416 followed by 4 weeks of exposure to hypoxia. Upon confirmation of RV dysfunction and PAH, rats were randomised to 0.1 µg·kg(-1) nebulised iloprost or drug-free vehicle, three times daily for 2 weeks. RV function and treadmill running time were evaluated pre- and post-iloprost/vehicle treatment. Pulmonary artery banded rats were treated 8 weeks after surgery to allow for significant RV hypertrophy. Inhaled iloprost significantly improved tricuspid annulus plane systolic excursion and increased exercise capacity, while mean pulmonary artery pressure and the percentage of occluded pulmonary vessels remained unchanged. Rats treated with iloprost had a striking reduction in RV collagen deposition, procollagen mRNA levels and connective tissue growth factor expression in both SU5416/hypoxia and pulmonary artery banded rats. In vitro, cardiac fibroblasts treated with iloprost showed a reduction in transforming growth factor (TGF)-ß1-induced connective tissue growth factor expression, in a protein kinase A-dependent manner. Iloprost decreased TGF-ß1-induced procollagen mRNA expression as well as cardiac fibroblast activation and migration. Iloprost significantly induced metalloproteinase-9 gene expression and activity and increased the expression of autophagy genes associated with collagen degradation. Inhaled iloprost improves RV function and reverses established RV fibrosis partially by preventing collagen synthesis and by increasing collagen turnover.


Asunto(s)
Hipertrofia Ventricular Derecha/tratamiento farmacológico , Iloprost/uso terapéutico , Animales , Colágeno/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Ecocardiografía , Fibroblastos/citología , Fibrosis , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/patología , Hemodinámica , Hipertensión Pulmonar/inducido químicamente , Hipoxia/fisiopatología , Indoles , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Microscopía de Contraste de Fase , Condicionamiento Físico Animal , Procolágeno/metabolismo , Pirroles , ARN Mensajero/metabolismo , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Factor de Crecimiento Transformador beta1/metabolismo , Vasodilatadores/uso terapéutico , Función Ventricular Derecha
5.
Am J Respir Cell Mol Biol ; 51(3): 413-25, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24684441

RESUMEN

NF-κB and IL-6, a NF-κB downstream mediator, play a central role in the inflammatory response of tissues. We aimed to determine the role of the classical NF-κB pathway in severe pulmonary arterial hypertension (PAH) induced by SU5416 and chronic hypoxia (SuHx) in rats. Tissue samples from patients with idiopathic PAH (iPAH) and control subjects were investigated. SuHx rats were treated from Days 1 to 3, 1 to 21, and 29 to 42 with the NF-κB inhibitor pyrrolidine dithiocarbamate (PDTC) and/or from Days 1 to 21 with anti-IL-6 antibody. Nuclear staining for NF-κB, an indicator of the activation of the classical NF-κB pathway, was detected in pulmonary arterial lesions of patients with iPAH and SuHx rats. NF-κB inhibition with PDTC prevented and reduced pulmonary arterial obliteration without reducing muscularization. However, the elevated lung levels of IL-6 were not reduced in PDTC-treated SuHx animals. PDTC treatment prevented or reduced apoptosis of pulmonary artery wall cells and pulmonary arterial obliteration. IL-6 inhibition had only a partial effect on apoptosis and obliteration. Pulmonary arterial media wall thickness was not affected by any of these treatments. Preventive and therapeutic PDTC treatment promoted immune regulation by increasing the number of perivascular CD4(+) T cells, in particular regulatory T cells (early treatment), and by reducing the number of perivascular CD8(+) T lymphocytes and CD45RA(+) B lymphocytes. Therapeutic PDTC treatment further preserved right ventricular function in SuHx animals. Inhibition of NF-κB may represent a therapeutic option for pulmonary arterial obliteration via reduced vessel wall cell apoptosis and improved regulation of the immune system.


Asunto(s)
Hipertensión Pulmonar/metabolismo , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Animales , Apoptosis , Linfocitos T CD4-Positivos/metabolismo , Hipertensión Pulmonar Primaria Familiar , Humanos , Hipertensión Pulmonar/patología , Hipoxia/metabolismo , Indoles/química , Inflamación , Interleucina-6/metabolismo , Antígenos Comunes de Leucocito/metabolismo , Pulmón/patología , Arteria Pulmonar/patología , Pirroles/química , Pirrolidinas/química , Ratas , Transducción de Señal , Tiocarbamatos/química , Factores de Tiempo
6.
Physiol Genomics ; 45(12): 449-61, 2013 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-23632417

RESUMEN

Right ventricular failure (RVF) is the most frequent cause of death in patients with pulmonary arterial hypertension (PAH); however, specific therapies targeted to treat RVF have not been developed. Chronic treatment with carvedilol has been shown to reduce established maladaptive right ventricle (RV) hypertrophy and to improve RV function in experimental PAH. However, the mechanisms by which carvedilol improves RVF are unknown. We have previously demonstrated by microarray analysis that RVF is characterized by a distinct gene expression profile when compared with functional, compensatory hypertrophy. We next sought to identify the effects of carvedilol on gene expression on a genome-wide basis. PAH and RVF were induced in male Sprague-Dawley rats by the combination of VEGF-receptor blockade and chronic hypoxia. After RVF was established, rats were treated with carvedilol or vehicle for 4 wk. RNA was isolated from RV tissue and hybridized for microarray analysis. An initial prediction analysis of carvedilol-treated RVs showed that the gene expression profile resembled the RVF prediction set. However, a more extensive analysis revealed a small group of genes differentially expressed after carvedilol treatment. Further analysis categorized these genes in pathways involved in cardiac hypertrophy, mitochondrial dysfunction, and protein ubiquitination. Genes encoding proteins in the cardiac hypertrophy and protein ubiquitination pathways were downregulated in the RV by carvedilol, while genes encoding proteins in the mitochondrial dysfunction pathway were upregulated by carvedilol. These gene expression changes may explain some of the mechanisms that underlie the functional improvement of the RV after carvedilol treatment.


Asunto(s)
Carbazoles/administración & dosificación , Carbazoles/uso terapéutico , Perfilación de la Expresión Génica , Hipertensión Pulmonar/complicaciones , Propanolaminas/administración & dosificación , Propanolaminas/uso terapéutico , Transcripción Genética , Disfunción Ventricular Derecha/tratamiento farmacológico , Disfunción Ventricular Derecha/genética , Animales , Carbazoles/farmacología , Cardiomegalia/complicaciones , Cardiomegalia/genética , Carvedilol , Análisis por Conglomerados , Regulación de la Expresión Génica/efectos de los fármacos , Hipertensión Pulmonar/genética , Masculino , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética , Propanolaminas/farmacología , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transcripción Genética/efectos de los fármacos , Ubiquitinación/efectos de los fármacos , Ubiquitinación/genética , Disfunción Ventricular Derecha/etiología
7.
Eur Respir J ; 41(1): 104-14, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22835607

RESUMEN

Epidemiological evidence links pulmonary arterial hypertension (PAH) with thyroid disease, but a mechanistic explanation for this association is lacking. Because a central hallmark of vascular remodelling in pulmonary hypertension is lumen obliteration by endothelial cell growth and because thyroid hormones are known to be angiogenic, we hypothesised that thyroid hormones play a role in the control of endothelial cell proliferation in experimental PAH in rats. Hypothyroidism was induced by subtotal thyroidectomy and treatment with propylthiouracil (PTU) in rats with experimental PAH after combined exposure to vascular endothelial growth factor receptor inhibition and hypoxia (the Sugen-chronic hypoxia (SuHx) model). Subtotal thyroidectomy prevented and PTU treatment reversed the development of severe experimental PAH. Thyroxin repletion restored the PAH phenotype in thyroidectomised SuHx rats. The prevention of PAH by thyroidectomy was associated with a reduced rate of cell turnover, reduced extracellular signal-regulated protein kinases 1 and 2 phosphorylation, and reduced expression of α(v)ß(3) integrin, fibroblast growth factor (FGF)-2 and FGF receptor. Thyroidectomy mitigated hypoxia-induced pulmonary hypertension, but this effect was not associated with a decreased pulmonary vascular resistance. These data suggest that thyroid hormone permits endothelial cell proliferation in PAH. A causal link between thyroid diseases and the onset or progression of vascular remodelling in PAH patients remains to be determined.


Asunto(s)
Hipertensión Pulmonar/patología , Hormonas Tiroideas/fisiología , Animales , Hipertensión Pulmonar Primaria Familiar , Hipertensión Pulmonar/complicaciones , Hipertensión Pulmonar/etiología , Masculino , Neovascularización Patológica/etiología , Ratas , Ratas Sprague-Dawley
8.
Cells ; 12(24)2023 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-38132140

RESUMEN

The inhibition of mammalian target of rapamycin (mTOR) with rapamycin (RAPA) provides protection against myocardial ischemia/reperfusion (I/R) injury in diabetes. Since interactions between transcripts, including long non-coding RNA (lncRNA), microRNA(miRNA) and mRNA, regulate the pathophysiology of disease, we performed unbiased miRarray profiling in the heart of diabetic rabbits following I/R injury with/without RAPA treatment to identify differentially expressed (DE) miRNAs and their predicted targets of lncRNAs/mRNAs. Results showed that among the total of 806 unique miRNAs targets, 194 miRNAs were DE after I/R in diabetic rabbits. Specifically, eight miRNAs, including miR-199a-5p, miR-154-5p, miR-543-3p, miR-379-3p, miR-379-5p, miR-299-5p, miR-140-3p, and miR-497-5p, were upregulated and 10 miRNAs, including miR-1-3p, miR-1b, miR-29b-3p, miR-29c-3p, miR-30e-3p, miR-133c, miR-196c-3p, miR-322-5p, miR-499-5p, and miR-672-5p, were significantly downregulated after I/R injury. Interestingly, RAPA treatment significantly reversed these changes in miRNAs. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis indicated the participation of miRNAs in the regulation of several signaling pathways related to I/R injury, including MAPK signaling and apoptosis. Furthermore, in diabetic hearts, the expression of lncRNAs, HOTAIR, and GAS5 were induced after I/R injury, but RAPA suppressed these lncRNAs. In contrast, MALAT1 was significantly reduced following I/R injury, with the increased expression of miR-199a-5p and suppression of its target, the anti-apoptotic protein Bcl-2. RAPA recovered MALAT1 expression with its sponging effect on miR-199-5p and restoration of Bcl-2 expression. The identification of novel targets from the transcriptome analysis in RAPA-treated diabetic hearts could potentially lead to the development of new therapeutic strategies for diabetic patients with myocardial infarction.


Asunto(s)
Diabetes Mellitus , Lagomorpha , MicroARNs , Daño por Reperfusión Miocárdica , ARN Largo no Codificante , Animales , Humanos , Conejos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , ARN Mensajero , MicroARNs/genética , MicroARNs/metabolismo , Lagomorpha/genética , Lagomorpha/metabolismo , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/genética , Isquemia , Proteínas Proto-Oncogénicas c-bcl-2
9.
Am J Respir Cell Mol Biol ; 46(5): 582-91, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22162909

RESUMEN

Obliteration of the vascular lumen by endothelial cell growth is a hallmark of many forms of severe pulmonary arterial hypertension. Copper plays a significant role in the control of endothelial cell proliferation in cancer and wound-healing. We sought to determine whether angioproliferation in rats with experimental pulmonary arterial hypertension and pulmonary microvascular endothelial cell proliferation in humans depend on the proangiogenic action of copper. A copper-depleted diet prevented, and copper chelation with tetrathiomolybdate reversed, the development of severe experimental pulmonary arterial hypertension. The copper chelation-induced reopening of obliterated vessels was caused by caspase-independent apoptosis, reduced vessel wall cell proliferation, and a normalization of vessel wall structure. No evidence was found for a role of super oxide-1 inhibition or lysyl-oxidase-1 inhibition in the reversal of angioproliferation. Tetrathiomolybdate inhibited the proliferation of human pulmonary microvascular endothelial cells, isolated from explanted lungs from control subjects and patients with pulmonary arterial hypertension. These data suggest that the inhibition of endothelial cell proliferation by a copper-restricting strategy could be explored as a new therapeutic approach in pulmonary arterial hypertension. It remains to be determined, however, whether potential toxicity to the right ventricle is offset by the beneficial pulmonary vascular effects of antiangiogenic treatment in patients with pulmonary arterial hypertension.


Asunto(s)
Cobre/fisiología , Endotelio Vascular/patología , Hipertensión Pulmonar/patología , Microvasos/patología , Neovascularización Patológica/patología , Animales , Caspasas/metabolismo , División Celular , Células Cultivadas , Quelantes/química , Quelantes/uso terapéutico , Cobre/administración & dosificación , Dieta , Activación Enzimática , Humanos , Hipertensión Pulmonar/tratamiento farmacológico , Hipertensión Pulmonar/etiología , Hipoxia/complicaciones , Inmunohistoquímica , Indoles/farmacología , Masculino , Molibdeno/química , Molibdeno/uso terapéutico , Neovascularización Patológica/metabolismo , Pirroles/farmacología , Ratas , Ratas Sprague-Dawley
10.
Am J Respir Cell Mol Biol ; 47(5): 679-87, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22842496

RESUMEN

The combination of chronic hypoxia and treatment of rats with the vascular endothelial growth factor (VEGF) receptor blocker, SU5416, induces pulmonary angio-obliteration, resulting in severe pulmonary arterial hypertension (PAH). Inflammation is thought to contribute to the pathology of PAH. Allergic inflammation caused by ovalbumin (OVA) immunization causes muscularization of pulmonary arteries, but not severe PAH. Whether disturbance of the immune system and allergic inflammation in the setting of lung endothelial cell apoptosis causes PAH is unknown. We investigated the effects of OVA-allergic inflammation on the development of PAH initiated by VEGF blockade-induced lung endothelial cell apoptosis. OVA-immunized rats were treated with SU5416 to induce pulmonary vascular endothelial cell apoptosis. The combination of OVA and SU5416 treatment resulted in severe angio-obilterative PAH, accompanied by increased IL-6 expression in the lungs. c-Kit(+) and Sca-1(+) cells were found in and around the lung vascular lesions. Pan-caspase inhibiton, dexamethasone treatment, and depletion of B-lymphocytes using an anti-CD20 antibody suppressed this remodeling. OVA immunization also increased lung tissue hypoxia-induced factor-1α and VEGF expression. Our results also suggest that the increased expression of hypoxia-induced factor-1α and IL-6 induced by the allergic lung inflammation may be a component of the pathogenesis of PAH.


Asunto(s)
Modelos Animales de Enfermedad , Hipertensión Pulmonar/inducido químicamente , Inmunización , Indoles/efectos adversos , Ovalbúmina/inmunología , Pirroles/efectos adversos , Animales , Suero Antilinfocítico/farmacología , Apoptosis/efectos de los fármacos , Asparagina/análogos & derivados , Asparagina/farmacología , Asparagina/uso terapéutico , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Inhibidores de Caspasas/farmacología , Inhibidores de Caspasas/uso terapéutico , Proliferación Celular , Dexametasona/farmacología , Dexametasona/uso terapéutico , Hemodinámica , Hipertensión Pulmonar/inmunología , Hipertensión Pulmonar/prevención & control , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Indoles/farmacología , Interleucina-6/metabolismo , Pulmón/irrigación sanguínea , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/patología , Masculino , Antígeno Nuclear de Célula en Proliferación/metabolismo , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/patología , Arteria Pulmonar/fisiopatología , Pirroles/farmacología , Ratas , Ratas Sprague-Dawley , Receptores de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
11.
Am J Physiol Lung Cell Mol Physiol ; 302(4): L363-9, 2012 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21964406

RESUMEN

Severe forms of pulmonary arterial hypertension (PAH) are characterized by various degrees of remodeling of the pulmonary arterial vessels, which increases the pulmonary vascular resistance and right ventricular afterload, thus contributing to the development of right ventricle dysfunction and failure. Recent years have seen advances in the understanding of the pathobiology of PAH; however, many important questions remain unanswered. Elucidating the pathobiology of PAH continues to be critical to design new effective therapeutic strategies, and appropriate animal models of PAH are necessary to achieve the task. Although the monocrotaline rat model of PAH has contributed to a better understanding of vascular remodeling in pulmonary hypertension, we question the validity of this model as a preclinically relevant model of severe plexogenic PAH. Here we review pertinent publications that either have been forgotten or ignored, and we reexamine the monocrotaline model in the context of human forms of PAH.


Asunto(s)
Hipertensión Pulmonar/inducido químicamente , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/fisiopatología , Animales , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Enfermedad Hepática Inducida por Sustancias y Drogas/fisiopatología , Modelos Animales de Enfermedad , Humanos , Hipertensión Pulmonar/patología , Hipertensión Pulmonar/fisiopatología , Pulmón/irrigación sanguínea , Pulmón/patología , Monocrotalina , Miocarditis/inducido químicamente , Miocarditis/patología , Miocarditis/fisiopatología
12.
Am J Physiol Lung Cell Mol Physiol ; 302(10): L977-91, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22307907

RESUMEN

Many chronic pulmonary diseases are associated with pulmonary hypertension (PH) and pulmonary vascular remodeling, which is a term that continues to be used to describe a wide spectrum of vascular abnormalities. Pulmonary vascular structural changes frequently increase pulmonary vascular resistance, causing PH and right heart failure. Although rat models had been standard models of PH research, in more recent years the availability of genetically engineered mice has made this species attractive for many investigators. Here we review a large amount of data derived from experimental PH reports published since 1996. These studies using wild-type and genetically designed mice illustrate the challenges and opportunities provided by these models. Hemodynamic measurements are difficult to obtain in mice, and right heart failure has not been investigated in mice. Anatomical, cellular, and genetic differences distinguish mice and rats, and pharmacogenomics may explain the degree of PH and the particular mode of pulmonary vascular adaptation and also the response of the right ventricle.


Asunto(s)
Modelos Animales de Enfermedad , Ventrículos Cardíacos/fisiopatología , Hipertensión Pulmonar/fisiopatología , Hipertrofia Ventricular Derecha/fisiopatología , Arteria Pulmonar/fisiopatología , Animales , Presión Sanguínea , Hipertensión Pulmonar Primaria Familiar , Ventrículos Cardíacos/patología , Hipertensión Pulmonar/patología , Hipertrofia Ventricular Derecha/patología , Ratones , Ratones Transgénicos , Arteria Pulmonar/patología , Ratas , Resistencia Vascular , Función Ventricular Derecha , Remodelación Ventricular
13.
Am J Physiol Lung Cell Mol Physiol ; 303(1): L20-32, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22523283

RESUMEN

Bacterial infections of the lungs and abdomen are among the most common causes of sepsis. Abdominal peritonitis often results in acute lung injury (ALI). Recent reports demonstrate a potential benefit of parenteral vitamin C [ascorbic acid (AscA)] in the pathogenesis of sepsis. Therefore we examined the mechanisms of vitamin C supplementation in the setting of abdominal peritonitis-mediated ALI. We hypothesized that vitamin C supplementation would protect lungs by restoring alveolar epithelial barrier integrity and preventing sepsis-associated coagulopathy. Male C57BL/6 mice were intraperitoneally injected with a fecal stem solution to induce abdominal peritonitis (FIP) 30 min prior to receiving either AscA (200 mg/kg) or dehydroascorbic acid (200 mg/kg). Variables examined included survival, extent of ALI, pulmonary inflammatory markers (myeloperoxidase, chemokines), bronchoalveolar epithelial permeability, alveolar fluid clearance, epithelial ion channel, and pump expression (aquaporin 5, cystic fibrosis transmembrane conductance regulator, epithelial sodium channel, and Na(+)-K(+)-ATPase), tight junction protein expression (claudins, occludins, zona occludens), cytoskeletal rearrangements (F-actin polymerization), and coagulation parameters (thromboelastography, pro- and anticoagulants, fibrinolysis mediators) of septic blood. FIP-mediated ALI was characterized by compromised lung epithelial permeability, reduced alveolar fluid clearance, pulmonary inflammation and neutrophil sequestration, coagulation abnormalities, and increased mortality. Parenteral vitamin C infusion protected mice from the deleterious consequences of sepsis by multiple mechanisms, including attenuation of the proinflammatory response, enhancement of epithelial barrier function, increasing alveolar fluid clearance, and prevention of sepsis-associated coagulation abnormalities. Parenteral vitamin C may potentially have a role in the management of sepsis and ALI associated with sepsis.


Asunto(s)
Lesión Pulmonar Aguda/tratamiento farmacológico , Ácido Ascórbico/farmacología , Sepsis/tratamiento farmacológico , Abdomen/microbiología , Abdomen/patología , Lesión Pulmonar Aguda/metabolismo , Lesión Pulmonar Aguda/microbiología , Lesión Pulmonar Aguda/fisiopatología , Animales , Biomarcadores/sangre , Coagulación Sanguínea/efectos de los fármacos , Lavado Broncoalveolar/métodos , Línea Celular , Proteínas del Citoesqueleto/metabolismo , Humanos , Inflamación/sangre , Inflamación/metabolismo , Inflamación/fisiopatología , Canales Iónicos/metabolismo , Transporte Iónico/efectos de los fármacos , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Neutrófilos/fisiología , Peritonitis/tratamiento farmacológico , Peritonitis/metabolismo , Peritonitis/microbiología , Peritonitis/fisiopatología , Permeabilidad/efectos de los fármacos , Alveolos Pulmonares/efectos de los fármacos , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/fisiopatología , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/fisiopatología , Sepsis/sangre , Sepsis/metabolismo , Sepsis/fisiopatología , ATPasa Intercambiadora de Sodio-Potasio/metabolismo
14.
Lung ; 190(1): 69-82, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22015802

RESUMEN

BACKGROUND: We postulate that in adults there is an established lung structure maintenance program and that lung alveolar septal cells are undergoing both continuous apoptosis and proliferation. Whereas lung cell apoptosis has been recognized in human emphysema, little is known about cell proliferation. METHODS: Using a novel rat model of emphysema, induced by intratracheal instillation of cigarette smoke extract (CSE), we investigated the dynamics of emphysematous lung destruction. Emphysematous lung destruction was determined by measuring mean linear intercept and destructive index. Lung injury and repair were assessed by immunohistochemistry and Western blot analysis for active caspase-3 and proliferating cell nuclear antigen (PCNA) after 4, 8, and 12 weeks of CSE instillations. RESULTS: The emphysematous lung tissue destruction was present at 4 weeks of CSE treatment and progressed to 8 weeks. Spontaneous repair began at 12 weeks. Treatment with a peroxisome proliferator activated receptor (PPAR)α+γ agonist or granulocyte and macrophage-colony stimulating factor (GM-CSF) for 4 weeks prevented the progression of emphysematous lung destruction and decreased the number of caspase-3-positive cells. CONCLUSION: Apoptosis and cell proliferation occur in this new model of emphysema. Treatment with a PPARα+γ agonist or GM-CSF can inhibit the progression of emphysematous alveolar septal destruction by decreasing alveolar cell apoptosis.


Asunto(s)
Apoptosis , Proliferación Celular , Enfisema/metabolismo , Enfisema/patología , Análisis de Varianza , Animales , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Enfisema/inducido químicamente , Enfisema/enzimología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Metaloproteinasa 2 de la Matriz/efectos de los fármacos , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/efectos de los fármacos , Metaloproteinasa 9 de la Matriz/metabolismo , PPAR alfa/agonistas , PPAR gamma/agonistas , Antígeno Nuclear de Célula en Proliferación/metabolismo , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/patología , Ratas , Humo , Nicotiana , Factor A de Crecimiento Endotelial Vascular/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo , alfa 1-Antitripsina/farmacología
15.
Am J Respir Crit Care Med ; 183(10): 1402-10, 2011 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-21297075

RESUMEN

RATIONALE: Inhibitors of histone deacetylases (HDACs) reduce pressure-overload-induced left ventricular hypertrophy and dysfunction, but their effects on right ventricular (RV) adaptation to pressure overload are unknown. OBJECTIVES: Determine the effect of the broad-spectrum HDAC inhibitors trichostatin A (TSA) and valproic acid (VPA) on RV function and remodeling after pulmonary artery banding (PAB) in rats. METHODS: Chronic progressive RV pressure-overload was induced in rats by PAB. After establishment of adaptive RV hypertrophy 4 weeks after surgery, rats were treated for 2 weeks with vehicle, TSA, or VPA. RV function and remodeling were determined using echocardiography, invasive hemodynamic measurements, immunohistochemistry, and molecular analyses after 2 weeks of HDAC inhibition. The effects of TSA were determined on the expression of proangiogenic and prohypertrophic genes in human myocardial fibroblasts and microvascular endothelial cells. MEASUREMENTS AND MAIN RESULTS: TSA treatment did not prevent the development of RV hypertrophy and was associated with RV dysfunction, capillary rarefaction, fibrosis, and increased rates of myocardial cell death. Similar results were obtained with the structurally unrelated HDAC inhibitor VPA. With TSA treatment, a reduction was found in expression of vascular endothelial growth factor and angiopoietin-1, which proteins are involved in vascular adaptation to pressure-overload. TSA dose-dependently suppressed vascular endothelial growth factor, endothelial nitric oxide synthase, and angiopoietin-1 expression in cultured myocardial endothelial cells, which effects were mimicked by selective gene silencing of several class I and II HDACs. CONCLUSIONS: HDAC inhibition is associated with dysfunction and worsened remodeling of the pressure-overloaded RV. The detrimental effects of HDAC inhibition on the pressure-overloaded RV may come about via antiangiogenic or proapoptotic effects.


Asunto(s)
Inhibidores de Histona Desacetilasas/farmacología , Ácidos Hidroxámicos/farmacología , Arteria Pulmonar/cirugía , Ácido Valproico/farmacología , Procedimientos Quirúrgicos Vasculares , Disfunción Ventricular Derecha/fisiopatología , Angiopoyetina 1 , Animales , Western Blotting , Células Cultivadas , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Ecocardiografía Doppler , Ventrículos Cardíacos/diagnóstico por imagen , Histona Desacetilasas , Humanos , Hipertensión Pulmonar/complicaciones , Hipertensión Pulmonar/fisiopatología , Hipertrofia Ventricular Derecha/complicaciones , Hipertrofia Ventricular Derecha/fisiopatología , Ligadura , Masculino , Óxido Nítrico Sintasa/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Factor A de Crecimiento Endotelial Vascular/efectos de los fármacos , Disfunción Ventricular Derecha/complicaciones , Remodelación Ventricular/efectos de los fármacos
16.
Am J Physiol Lung Cell Mol Physiol ; 300(3): L402-13, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21224215

RESUMEN

In patients with chronic obstructive pulmonary disease (COPD), histone deacetylase (HDAC) expression and activity are reduced in the lung tissue. However, whether HDAC activity controls the maintenance of the lung alveolar septal structures has not been investigated. To explore the consequences of HDAC inhibition and address the question of whether HDAC inhibition causes lung cell apoptosis and emphysema, male Sprague-Dawley rats and human pulmonary microvascular endothelial cells (HPMVEC) were treated with trichostatin A (TSA), a specific inhibitor of HDACs. Chronic TSA treatment increased the alveolar air space area, mean linear intercept, and the number of caspase-3-positive cells in rat lungs. TSA suppressed hypoxia-inducible factor-1α (HIF-1α), VEGF, and lysyl oxidase (LOX) and increased microtubule-associated protein-1 light chain 3 (LC3), p53, and miR34a microRNA expression in both rat lungs and cultured HPMVEC. Gene silencing of HDAC2 using small interfering RNA (siRNA) in cultured HPMVEC resulted in the suppression of HIF-1α, VEGF, and LOX and an increase of p53 expression. These data indicate that HDAC inhibition causes emphysema and that HDAC-dependent mechanisms contribute to the maintenance of the adult lung structure. Our results also suggest that the increase in apoptosis, as a consequence of HDAC inhibition, is associated with decreased VEGF and HIF-1α expression.


Asunto(s)
Histona Desacetilasas/metabolismo , Enfisema Pulmonar/enzimología , Enfisema Pulmonar/patología , Animales , Células Endoteliales/efectos de los fármacos , Células Endoteliales/enzimología , Células Endoteliales/patología , Regulación de la Expresión Génica/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Ácidos Hidroxámicos/farmacología , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Masculino , MicroARNs/metabolismo , Microvasos/patología , Modelos Biológicos , Alveolos Pulmonares/irrigación sanguínea , Alveolos Pulmonares/efectos de los fármacos , Alveolos Pulmonares/patología , Estabilidad del ARN/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Proteína p53 Supresora de Tumor/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
17.
Am J Physiol Lung Cell Mol Physiol ; 300(5): L753-61, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21335523

RESUMEN

Chronic hypoxia induces pulmonary arterial remodeling, resulting in pulmonary hypertension and right ventricular hypertrophy. Hypoxia has been implicated as a physiological stimulus for p53 induction and hypoxia-inducible factor-1α (HIF-1α). However, the subcellular interactions between hypoxic exposure and expression of p53 and HIF-1α remain unclear. To examine the role of p53 and HIF-1α expression on hypoxia-induced pulmonary arterial remodeling, wild-type (WT) and p53 knockout (p53KO) mice were exposed to either normoxia or hypoxia for 8 wk. Following chronic hypoxia, both genotypes demonstrated elevated right ventricular pressures, right ventricular hypertrophy as measured by the ratio of the right ventricle to the left ventricle plus septum weights, and vascular remodeling. However, the right ventricular systolic pressures, the ratio of the right ventricle to the left ventricle plus septum weights, and the medial wall thickness of small vessels were significantly greater in the p53KO mice than in the WT mice. The p53KO mice had lower levels of p21 and miR34a expression, and higher levels of HIF-1α, VEGF, and PDGF expression than WT mice following chronic hypoxic exposure. This was associated with a higher proliferating cell nuclear antigen expression of pulmonary artery in p53KO mice. We conclude that p53 plays a critical role in the mitigation of hypoxia-induced small pulmonary arterial remodeling. By interacting with p21 and HIF-1α, p53 may suppress hypoxic pulmonary arterial remodeling and pulmonary arterial smooth muscle cell proliferation under hypoxia.


Asunto(s)
Genes p53/fisiología , Hipertensión Pulmonar/fisiopatología , Hipoxia/fisiopatología , Animales , Hipertensión Pulmonar/etiología , Hipertrofia Ventricular Derecha/fisiopatología , Hipoxia/complicaciones , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones , Ratones Noqueados , MicroARNs/biosíntesis , Quinasas p21 Activadas/biosíntesis
18.
Crit Care Med ; 39(6): 1454-60, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21358394

RESUMEN

OBJECTIVE: Sepsis-induced lung injury is a persisting clinical problem with no direct therapy. Recent work suggests that intravenously infused ascorbic acid improves the circulatory dysfunction of sepsis. We used a model of endotoxin-induced acute lung injury to determine whether parenteral ascorbic acid modulates the dysregulated proinflammatory, procoagulant state that leads to lung injury. DESIGN: C57BL/6 mice were exposed to lethal lipopolysaccharide doses (10 µg/g of body weight) to induce acute lung injury. SETTING: Laboratory investigation. SUBJECTS: Wild-type C57BL/6 mice. INTERVENTIONS: Ascorbic acid or its oxidized form (dehydroascorbic acid) was administered intraperitoneally at 200 mg/kg 30 mins after the lethal lipopolysaccharide dose. MEASUREMENTS AND MAIN RESULTS: We quantified survival, lung capillary leak, proinflammatory chemokine expression, and lung microvascular thrombosis. Lipopolysaccharide induced 100% lethality in mice within 28 hrs of exposure and in lung we observed intense neutrophil sequestration, loss of capillary barrier function, exuberant pulmonary inflammation, and extensive microthrombus formation. A time-delayed infusion protocol of both ascorbic acid and dehydroascorbic acid significantly prolonged survival. Both ascorbic acid and dehydroascorbic acid preserved lung architecture and barrier function while attenuating proinflammatory chemokine expression and microvascular thrombosis. Ascorbic acid and dehydroascorbic acid attenuated nuclear factor kappa B activation and normalized coagulation parameters. CONCLUSIONS: Ascorbic acid administered in an interventional manner following lipopolysaccharide infusion attenuates proinflammatory, procoagulant states that induce lung vascular injury in an animal model of sepsis.


Asunto(s)
Lesión Pulmonar Aguda/tratamiento farmacológico , Antioxidantes/uso terapéutico , Ácido Ascórbico/uso terapéutico , Escherichia coli , Lipopolisacáridos , Lesión Pulmonar Aguda/etiología , Lesión Pulmonar Aguda/patología , Animales , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Sepsis/complicaciones , Sepsis/patología , Sepsis/terapia
19.
Am J Respir Crit Care Med ; 182(5): 652-60, 2010 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-20508210

RESUMEN

RATIONALE: Most patients with pulmonary arterial hypertension (PAH) die from right heart failure. Beta-adrenergic receptor blockade reduces mortality by about 30% in patients with left-sided systolic heart failure, but is not used in PAH. OBJECTIVES: To assess the effect of the adrenergic receptor blocker carvedilol on the pulmonary circulation and right heart in experimental pulmonary hypertension in rats. METHODS: Angioproliferative pulmonary hypertension was induced in rats by combined exposure to the vascular endothelial growth factor-receptor antagonist SU5416 and hypoxia. Carvedilol treatment was started after establishment of pulmonary hypertension and right heart dysfunction. MEASUREMENTS AND MAIN RESULTS: Compared with vehicle-treated animals, treatment with carvedilol resulted in increased exercise endurance; improved right ventricular (RV) function (increased tricuspid annular plane systolic excursion and decreased RV dilatation); and an increased cardiac output. The morphology of the pulmonary vessels and the RV afterload were not affected by carvedilol. Carvedilol treatment was associated with enhancement of RV fetal gene reactivation, increased protein kinase G (PKG) activity, and a reduction in capillary rarefaction and fibrosis. Metoprolol had similar but less pronounced effects in the SU5416 and hypoxia model. Cardioprotective effects were noted of both carvedilol and metoprolol in the monocrotaline model. In the case of carvedilol, but not metoprolol, part of these effects resulted from a prevention of monocrotaline-induced lung remodeling. CONCLUSIONS: Adrenergic receptor blockade reverses RV remodeling and improves RV function in experimental pulmonary hypertension. Beta-adrenergic receptor blockers are not recommended in humans with PAH before their safety and efficacy are assessed in well-designed clinical trials.


Asunto(s)
Antagonistas Adrenérgicos/farmacología , Carbazoles/farmacología , Hipertensión Pulmonar/tratamiento farmacológico , Propanolaminas/farmacología , Disfunción Ventricular Derecha/tratamiento farmacológico , Remodelación Ventricular/efectos de los fármacos , Animales , Carvedilol , Modelos Animales de Enfermedad , Hipertensión Pulmonar/inducido químicamente , Indoles/administración & dosificación , Masculino , Circulación Pulmonar/efectos de los fármacos , Pirroles/administración & dosificación , Ratas , Ratas Sprague-Dawley , Factor A de Crecimiento Endotelial Vascular/administración & dosificación , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Disfunción Ventricular Derecha/inducido químicamente
20.
STAR Protoc ; 2(3): 100772, 2021 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-34485939

RESUMEN

We developed a preclinical model of myocardial ischemia/reperfusion (I/R) injury in conscious diabetic rabbits to identify an early pharmacological intervention for patients with diabetes and acute myocardial infarction (AMI). Here, we describe a reproducible protocol for induction of diabetes with subsequent manifestation of myocardial I/R injury in conscious rabbits to mimic the real-life scenario observed in clinical settings. Further, we demonstrate the efficacy of rapamycin at the onset of reperfusion to limit the adverse effect of AMI. For complete details on the use and execution of this protocol, please refer to Samidurai et al. (2020).


Asunto(s)
Cardiotónicos/farmacología , Diabetes Mellitus Tipo 1/fisiopatología , Daño por Reperfusión Miocárdica/etiología , Daño por Reperfusión Miocárdica/prevención & control , Sirolimus/farmacología , Aloxano , Animales , Apoptosis , Oclusión con Balón/métodos , Diabetes Mellitus Experimental/fisiopatología , Diabetes Mellitus Tipo 1/complicaciones , Modelos Animales de Enfermedad , Electrocardiografía , Masculino , Daño por Reperfusión Miocárdica/patología , Conejos , Troponina I/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA