Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Basic Res Cardiol ; 119(2): 243-260, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38329499

RESUMEN

Inflammaging, a pro-inflammatory status that characterizes aging and primarily involving macrophages, is a master driver of age-related diseases. Mineralocorticoid receptor (MR) activation in macrophages critically regulates inflammatory and fibrotic processes. However, macrophage-specific mechanisms and the role of the macrophage MR for the regulation of inflammation and fibrotic remodeling in the aging heart have not yet been elucidated. Transcriptome profiling of cardiac macrophages from male/female young (4 months-old), middle (12 months-old) and old (18 and 24 months-old) mice revealed that myeloid cell-restricted MR deficiency prevents macrophage differentiation toward a pro-inflammatory phenotype. Pathway enrichment analysis showed that several biological processes related to inflammation and cell metabolism were modulated by the MR in aged macrophages. Further, transcriptome analysis of aged cardiac fibroblasts revealed that macrophage MR deficiency reduced the activation of pathways related to inflammation and upregulation of ZBTB16, a transcription factor involved in fibrosis. Phenotypic characterization of macrophages showed a progressive replacement of the TIMD4+MHC-IIneg/low macrophage population by TIMD4+MHC-IIint/high and TIMD4-MHC-IIint/high macrophages in the aging heart. By integrating cell sorting and transwell experiments with TIMD4+/TIMD4-macrophages and fibroblasts from old MRflox/MRLysMCre hearts, we showed that the inflammatory crosstalk between TIMD4- macrophages and fibroblasts may imply the macrophage MR and the release of mitochondrial superoxide anions. Macrophage MR deficiency reduced the expansion of the TIMD4- macrophage population and the emergence of fibrotic niches in the aging heart, thereby protecting against cardiac inflammation, fibrosis, and dysfunction. This study highlights the MR as an important mediator of cardiac macrophage inflammaging and age-related fibrotic remodeling.


Asunto(s)
Miocardio , Receptores de Mineralocorticoides , Animales , Femenino , Masculino , Ratones , Fibrosis , Inflamación/metabolismo , Macrófagos/metabolismo , Miocardio/patología , Receptores de Mineralocorticoides/genética , Receptores de Mineralocorticoides/metabolismo
2.
J Am Heart Assoc ; 13(3): e033553, 2024 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-38293923

RESUMEN

BACKGROUND: Alveolar hypoxia is protective in the context of cardiovascular and ischemic heart disease; however, the underlying mechanisms are incompletely understood. The present study sought to test the hypothesis that hypoxia is cardioprotective in left ventricular pressure overload (LVPO)-induced heart failure. We furthermore aimed to test that overlapping mechanisms promote cardiac recovery in heart failure patients following left ventricular assist device-mediated mechanical unloading and circulatory support. METHODS AND RESULTS: We established a novel murine model of combined chronic alveolar hypoxia and LVPO following transverse aortic constriction (HxTAC). The HxTAC model is resistant to cardiac hypertrophy and the development of heart failure. The cardioprotective mechanisms identified in our HxTAC model include increased activation of HIF (hypoxia-inducible factor)-1α-mediated angiogenesis, attenuated induction of genes associated with pathological remodeling, and preserved metabolic gene expression as identified by RNA sequencing. Furthermore, LVPO decreased Tbx5 and increased Hsd11b1 mRNA expression under normoxic conditions, which was attenuated under hypoxic conditions and may induce additional hypoxia-mediated cardioprotective effects. Analysis of samples from patients with advanced heart failure that demonstrated left ventricular assist device-mediated myocardial recovery revealed a similar expression pattern for TBX5 and HSD11B1 as observed in HxTAC hearts. CONCLUSIONS: Hypoxia attenuates LVPO-induced heart failure. Cardioprotective pathways identified in the HxTAC model might also contribute to cardiac recovery following left ventricular assist device support. These data highlight the potential of our novel HxTAC model to identify hypoxia-mediated cardioprotective mechanisms and therapeutic targets that attenuate LVPO-induced heart failure and mediate cardiac recovery following mechanical circulatory support.


Asunto(s)
Estenosis de la Válvula Aórtica , Insuficiencia Cardíaca , Humanos , Ratones , Animales , Insuficiencia Cardíaca/etiología , Cardiomegalia/metabolismo , Miocardio/metabolismo , Hipoxia/complicaciones , Remodelación Ventricular , Modelos Animales de Enfermedad
3.
Am J Physiol Heart Circ Physiol ; 323(6): H1352-H1364, 2022 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-36399384

RESUMEN

Perturbed vitamin-A metabolism is associated with type 2 diabetes and mitochondrial dysfunction that are pathophysiologically linked to the development of diabetic cardiomyopathy (DCM). However, the mechanism, by which vitamin A might regulate mitochondrial energetics in DCM has previously not been explored. To test the hypothesis that vitamin-A deficiency accelerates the onset of cardiomyopathy in diet-induced obesity (DIO), we subjected mice with lecithin retinol acyltransferase (Lrat) germline deletion, which exhibit impaired vitamin-A stores, to vitamin A-deficient high-fat diet (HFD) feeding. Wild-type mice fed with a vitamin A-sufficient HFD served as controls. Cardiac structure, contractile function, and mitochondrial respiratory capacity were preserved despite vitamin-A deficiency following 20 wk of HFD feeding. Gene profiling by RNA sequencing revealed that vitamin A is required for the expression of genes involved in cardiac fatty acid oxidation, glycolysis, tricarboxylic acid cycle, and mitochondrial oxidative phosphorylation in DIO as expression of these genes was relatively preserved under vitamin A-sufficient HFD conditions. Together, these data identify a transcriptional program, by which vitamin A preserves cardiac energetic gene expression in DIO that might attenuate subsequent onset of mitochondrial and contractile dysfunction.NEW & NOTEWORTHY The relationship between vitamin-A status and the pathogenesis of diabetic cardiomyopathy has not been studied in detail. We assessed cardiac mitochondrial respiratory capacity, contractile function, and gene expression by RNA sequencing in a murine model of combined vitamin-A deficiency and diet-induced obesity. Our study identifies a role for vitamin A in preserving cardiac energetic gene expression that might attenuate subsequent development of mitochondrial and contractile dysfunction in diet-induced obesity.


Asunto(s)
Diabetes Mellitus Tipo 2 , Cardiomiopatías Diabéticas , Ratones , Animales , Vitamina A , Modelos Animales de Enfermedad , Dieta , Obesidad/genética , Expresión Génica , Vitaminas
4.
Mol Cell Biol ; 42(10): e0016322, 2022 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-36125265

RESUMEN

Insulin and insulin-like growth factor 1 (IGF1) signaling is transduced by insulin receptor substrate 1 (IRS1) and IRS2. To elucidate physiological and redundant roles of insulin and IGF1 signaling in adult hearts, we generated mice with inducible cardiomyocyte-specific deletion of insulin and IGF1 receptors or IRS1 and IRS2. Both models developed dilated cardiomyopathy, and most mice died by 8 weeks post-gene deletion. Heart failure was characterized by cardiomyocyte loss and disarray, increased proapoptotic signaling, and increased autophagy. Suppression of autophagy by activating mTOR signaling did not prevent heart failure. Transcriptional profiling revealed reduced serum response factor (SRF) transcriptional activity and decreased mRNA levels of genes encoding sarcomere and gap junction proteins as early as 3 days post-gene deletion, in concert with ultrastructural evidence of sarcomere disruption and intercalated discs within 1 week after gene deletion. These data confirm conserved roles for constitutive insulin and IGF1 signaling in suppressing autophagic and apoptotic signaling in the adult heart. The present study also identifies an unexpected role for insulin and IGF1 signaling in regulating an SRF-mediated transcriptional program, which maintains expression of genes encoding proteins that support sarcomere integrity in the adult heart, reduction of which results in rapid development of heart failure.


Asunto(s)
Insuficiencia Cardíaca , Factor I del Crecimiento Similar a la Insulina , Ratones , Animales , Proteínas Sustrato del Receptor de Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/genética , Insulina/metabolismo , Factor de Respuesta Sérica/metabolismo , Sarcómeros/metabolismo , Miocitos Cardíacos/metabolismo , Insuficiencia Cardíaca/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , ARN Mensajero/metabolismo , Conexinas/metabolismo
5.
Elife ; 102021 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-34289931

RESUMEN

Immature neutrophils and HLA-DRneg/low monocytes expand in cancer, autoimmune diseases and viral infections, but their appearance and immunoregulatory effects on T-cells after acute myocardial infarction (AMI) remain underexplored. We found an expansion of circulating immature CD16+CD66b+CD10neg neutrophils and CD14+HLA-DRneg/low monocytes in AMI patients, correlating with cardiac damage, function and levels of immune-inflammation markers. Immature CD10neg neutrophils expressed high amounts of MMP-9 and S100A9, and displayed resistance to apoptosis. Moreover, we found that increased frequency of CD10neg neutrophils and elevated circulating IFN-γ levels were linked, mainly in patients with expanded CD4+CD28null T-cells. Notably, the expansion of circulating CD4+CD28null T-cells was associated with cytomegalovirus (CMV) seropositivity. Using bioinformatic tools, we identified a tight relationship among the peripheral expansion of immature CD10neg neutrophils, CMV IgG titers, and circulating levels of IFN-γ and IL-12 in patients with AMI. At a mechanistic level, CD10neg neutrophils enhanced IFN-γ production by CD4+ T-cells through a contact-independent mechanism involving IL-12. In vitro experiments also highlighted that HLA-DRneg/low monocytes do not suppress T-cell proliferation but secrete high levels of pro-inflammatory cytokines after differentiation to macrophages and IFN-γ stimulation. Lastly, using a mouse model of AMI, we showed that immature neutrophils (CD11bposLy6GposCD101neg cells) are recruited to the injured myocardium and migrate to mediastinal lymph nodes shortly after reperfusion. In conclusion, immunoregulatory functions of CD10neg neutrophils play a dynamic role in mechanisms linking myeloid cell compartment dysregulation, Th1-type immune responses and inflammation after AMI.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Antígenos HLA-DR/inmunología , Monocitos/inmunología , Infarto del Miocardio/inmunología , Neprilisina/inmunología , Neutrófilos/inmunología , Anciano , Animales , Biomarcadores , Diferenciación Celular , Proliferación Celular , Citocinas , Femenino , Humanos , Inflamación , Activación de Linfocitos , Masculino , Ratones , Persona de Mediana Edad , Infarto del Miocardio/patología , Linfocitos T/inmunología
6.
PLoS One ; 16(3): e0248196, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33667270

RESUMEN

INTRODUCTION: Regulating excessive activation of fibroblasts may be a promising target to optimize extracellular matrix deposition and myocardial stiffness. Fibroblast activation protein alpha (FAP) is upregulated in activated fibroblasts after myocardial infarction (MI), and alters fibroblast migration in vitro. We hypothesized that FAP depletion may have a protective effect on left ventricular (LV) remodeling after MI. MATERIALS AND METHODS: We used the model of chronic MI in homozygous FAP deficient mice (FAP-KO, n = 51) and wild type mice (WT, n = 55) to analyze wound healing by monocyte and myofibroblast infiltration. Heart function and remodeling was studied by echocardiography, morphometric analyses including capillary density and myocyte size, collagen content and in vivo cell-proliferation. In non-operated healthy mice up to 6 months of age, morphometric analyses and collagen content was assessed (WT n = 10, FAP-KO n = 19). RESULTS: Healthy FAP-deficient mice did not show changes in LV structure or differences in collagen content or cardiac morphology. Infarct size, survival and cardiac function were not different between FAP-KO and wildtype mice. FAP-KO animals showed less LV-dilation and a thicker scar, accompanied by a trend towards lower collagen content. Wound healing, assessed by infiltration with inflammatory cells and myofibroblasts were not different between groups. CONCLUSION: We show that genetic ablation of FAP does not impair cardiac wound healing, and attenuates LV dilation after MI in mice. FAP seems dispensable for normal cardiac function and homeostasis.


Asunto(s)
Endopeptidasas/deficiencia , Ventrículos Cardíacos/metabolismo , Proteínas de la Membrana/deficiencia , Monocitos/metabolismo , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Miofibroblastos/metabolismo , Animales , Dilatación Patológica , Endopeptidasas/metabolismo , Femenino , Ventrículos Cardíacos/patología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Monocitos/patología , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Miocardio/patología , Miofibroblastos/patología
7.
Hypertension ; 73(1): 102-111, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30543467

RESUMEN

Myocardial infarction (MI) is a major cause of death worldwide. Here, we identify the macrophage MR (mineralocorticoid receptor) as a crucial pathogenic player in cardiac wound repair after MI. Seven days after left coronary artery ligation, mice with myeloid cell-restricted MR deficiency compared with WT (wild type) controls displayed improved cardiac function and remodeling associated with enhanced infarct neovascularization and scar maturation. Gene expression profiling of heart-resident and infarct macrophages revealed that MR deletion drives macrophage differentiation in the ischemic microenvironment toward a phenotype outside the M1/M2 paradigm, with regulation of multiple interrelated factors controlling wound healing and tissue repair. Mechanistic and functional data suggest that inactivation of the macrophage MR promotes myocardial infarct healing through enhanced efferocytosis of neutrophils, the suppression of free radical formation, and the modulation of fibroblast activation state. Crucially, targeted delivery of MR antagonists to macrophages, with a single administration of RU28318 or eplerenone-containing liposomes at the onset of MI, improved the healing response and protected against cardiac remodeling and functional deterioration, offering an effective and unique therapeutic strategy for cardiac repair.


Asunto(s)
Eplerenona/farmacología , Corazón/fisiopatología , Infarto del Miocardio , Miocardio , Receptores de Mineralocorticoides , Cicatrización de Heridas , Animales , Diferenciación Celular/efectos de los fármacos , Microambiente Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica/métodos , Liposomas , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Antagonistas de Receptores de Mineralocorticoides/farmacología , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocardio/metabolismo , Miocardio/patología , Receptores de Mineralocorticoides/deficiencia , Receptores de Mineralocorticoides/metabolismo , Remodelación Ventricular/efectos de los fármacos , Remodelación Ventricular/fisiología , Cicatrización de Heridas/efectos de los fármacos , Cicatrización de Heridas/fisiología
8.
FASEB J ; 31(11): 5122-5132, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28768721

RESUMEN

Cell- and tissue-specific actions of glucocorticoids are mediated by the glucocorticoid receptor. Here, we demonstrate that the glucocorticoid receptor (GR) in macrophages is essential for cardiac healing after myocardial infarction. Compared with GRflox (wild-type controls), GRLysMCre mice that lacked GR in myeloid cells showed increased acute mortality as a result of cardiac rupture. Seven days after left coronary artery ligation, GRLysMCre mice exhibited worse cardiac function and adverse remodeling associated with impaired scar formation and angiogenic response to ischemic injury. Inactivation of GR altered the functional differentiation/maturation of monocyte-derived macrophages in the infarcted myocardium. Mechanistically, CD45+/CD11b+/Ly6G-/F4/80+ macrophages isolated from GRLysMCre infarcts showed deregulation of factors that control inflammation, neovascularization, collagen degradation, and scar tissue formation. Moreover, we demonstrate that cardiac fibroblasts sorted from the ischemic myocardium of GRLysMCre mice compared with cells isolated from injured GRflox hearts displayed higher matrix metalloproteinase 2 expression, and we provide evidence that the macrophage GR regulates myofibroblast differentiation in the infarct microenvironment during the early phase of wound healing. In summary, GR signaling in macrophages, playing a crucial role in tissue-repairing mechanisms, could be a potential therapeutic target during wound healing after ischemic myocardial injury.-Galuppo, P., Vettorazzi, S., Hövelmann, J., Scholz, C.-J., Tuckermann, J. P., Bauersachs, J., Fraccarollo, D. The glucocorticoid receptor in monocyte-derived macrophages is critical for cardiac infarct repair and remodeling.


Asunto(s)
Macrófagos/metabolismo , Monocitos/metabolismo , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Receptores de Glucocorticoides/metabolismo , Transducción de Señal , Animales , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/metabolismo , Macrófagos/patología , Ratones , Ratones Transgénicos , Monocitos/patología , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Miocardio/patología , Miofibroblastos/metabolismo , Miofibroblastos/patología , Receptores de Glucocorticoides/genética
9.
PLoS One ; 12(8): e0183670, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28829829

RESUMEN

BACKGROUND: Transcatheter aortic valve replacement (TAVR) is the method of choice for patients with severe aortic valve stenosis, who are ineligible or at high risk for surgery. Though TAVR leads to a significant reduction in mortality, a notable amount of patients are re-hospitalized early after TAVR. Parameters or biomarkers predicting outcome are therefore needed to identify patients who benefit most. Specific monocyte subsets have been associated with cardiovascular diseases and were shown to possess prognostic value. METHODS: Peripheral blood was drawn before and after transfemoral TAVR with the self-expanding CoreValve, Boston Lotus or the balloon-expanding Edwards Sapien prosthesis. Classical (CD14++CD16-), intermediate (CD14++CD16+) and non-classical (CD14+CD16++) monocyte subsets were determined by flow cytometry. Transthoracic echocardiography was performed before, early after as well as 3 months after the TAVR procedure. RESULTS: No significant differences in the absolute monocyte counts were found after TAVR. A significant decline in the intermediate monocyte population was though observed early after TAVR (pre 4.01±0.38%, post 2.803±0.34%, p≤0.05). Creatinine levels stayed stable after TAVR procedure and intermediate monocytes were associated with worse renal function. Monocyte decline was not related to changes in CRP-, noradrenaline, cortisol or aldosterone-levels. The amount of intermediate monocytes correlated with worse cardiac function and predicted the possibility to reach an improvement in NYHA functional class at 3 months after TAVR. CONCLUSIONS: A significant decline of intermediate monocytes occurs shortly after TAVR. High levels of intermediate monocytes were associated with worse cardiac function and predicted poor functional capacity, hinting at a possible prognostic value.


Asunto(s)
Receptores de Lipopolisacáridos/inmunología , Monocitos/inmunología , Receptores de IgG/inmunología , Sístole , Reemplazo de la Válvula Aórtica Transcatéter , Función Ventricular Izquierda , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Masculino
10.
Methods Mol Biol ; 1627: 123-137, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28836199

RESUMEN

Cardiac (myo)fibroblasts play a key role in the regulation of wound healing and pathogenic remodeling after myocardial infarction. Impaired scar formation and alterations of the extracellular matrix network precipitate cardiac dysfunction leading to increased morbidity and mortality. Therapeutic approaches addressing (myo)fibroblast phenotype appear to be useful in preventing progressive structural, electrical, and functional impairment and heart failure.Permanent ligation of the left anterior descending coronary artery has proven to be a valuable experimental model to investigate the arrays of pathways/mechanisms involved in cardiac repair and extracellular matrix remodeling in ischemic heart failure. Here we describe the surgical procedure to occlude the left coronary artery in mice. Moreover, we present an accurate method to isolate (myo)fibroblasts from ischemic myocardium, with maintenance of the functional phenotype, using the specific marker for mouse cardiac fibroblasts mEF-SK4. The protocol can be completed within a few hours, and the isolated fibroblasts/myofibroblasts are suitable for downstream molecular biology applications, like gene expression profiling and cell culture.


Asunto(s)
Cardiomiopatías/metabolismo , Cardiomiopatías/patología , Modelos Animales de Enfermedad , Miofibroblastos/metabolismo , Fenotipo , Animales , Biomarcadores , Cardiomiopatías/etiología , Separación Celular , Células Cultivadas , Fibroblastos/metabolismo , Fibrosis , Citometría de Flujo , Isquemia/complicaciones , Ratones , Miofibroblastos/patología
11.
Int J Cardiol ; 232: 155-159, 2017 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-28082091

RESUMEN

BACKGROUND: Fibroblast activation protein alpha (FAP) is a membrane-bound serine protease expressed by activated fibroblasts after myocardial infarction (MI). Reduced circulating FAP levels were associated with increased mortality in patients with acute coronary syndrome. We hypothesized that FAP concentrations are altered after acute ST-elevation MI (STEMI), and related to myocardial damage. METHODS: We measured circulating FAP concentrations in blood plasma of 60 patients on admission, day 1, day 3 and day 5 after STEMI, and in 25 apparently healthy blood donors as controls. RESULTS: Plasma FAP concentrations were lower in STEMI patients on admission (71ng/mL) than in blood donors (101ng/mL, P<0.0001). FAP concentrations declined in STEMI patients from admission to day 3 (66ng/mL, P<0.05) and day 5 (57ng/mL, P<0.05). FAP concentrations on day 5 were inversely correlated with maximum CK and maximum CRP levels. In a multiple linear regression analysis, maximum CRP was independently associated with low FAP concentrations on day 5 after STEMI. When stratified according to the absolute amount of FAP change from admission to day 5 (ΔFAP), patients with high ΔFAP (-22ng/mL) had worse left ventricular function, higher levels of hs-cTnT, CK on admission, maximum CK and CRP than patients with low ΔFAP (-3ng/mL). CONCLUSIONS: Our study first demonstrates alterations of circulating FAP concentrations acutely after STEMI. A greater decline of circulating FAP concentrations in the first 5days after STEMI is associated with increased myocardial damage and inflammation. Measurement of circulating FAP might help to better understand the relation of myocardial injury and inflammatory response in the individual patient.


Asunto(s)
Gelatinasas/sangre , Proteínas de la Membrana/sangre , Infarto del Miocardio con Elevación del ST/sangre , Serina Endopeptidasas/sangre , Adulto , Anciano , Anciano de 80 o más Años , Antígenos de Superficie , Biomarcadores/sangre , Endopeptidasas , Ensayo de Inmunoadsorción Enzimática , Femenino , Estudios de Seguimiento , Alemania/epidemiología , Humanos , Masculino , Persona de Mediana Edad , Pronóstico , Estudios Retrospectivos , Infarto del Miocardio con Elevación del ST/mortalidad , Tasa de Supervivencia/tendencias
12.
Hypertension ; 66(5): 978-87, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26351025

RESUMEN

Reduced nitric oxide bioavailability contributes to progression of cardiac dysfunction and remodeling in ischemic heart failure. Clinical use of organic nitrates as nitric oxide donors is limited by development of nitrate tolerance and reactive oxygen species formation. We investigated the effects of long-term therapy with pentaerythritol tetranitrate (PETN), an organic nitrate devoid of tolerance, in rats with congestive heart failure after extensive myocardial infarction. Seven days after coronary artery ligation, rats were randomly allocated to treatment with PETN (80 mg/kg BID) or placebo for 9 weeks. Long-term PETN therapy prevented the progressive left ventricular dilatation and improved left ventricular contractile function and relaxation in rats with congestive heart failure. Mitochondrial superoxide anion production was markedly increased in the failing left ventricular myocardium and nearly normalized by PETN treatment. Gene set enrichment analysis revealed that PETN beneficially modulated the dysregulation of mitochondrial genes involved in energy metabolism, paralleled by prevention of uncoupling protein-3, thioredoxin-2, and superoxide dismutase-2 downregulation. Moreover, PETN provided a remarkable protective effect against reactive fibrosis in chronically failing hearts. Mechanistically, induction of heme oxygenase-1 by PETN prevented mitochondrial superoxide generation, NOX4 upregulation, and ensuing formation of extracellular matrix proteins in fibroblasts from failing hearts. In summary, PETN targeting reactive oxygen species generation prevented the changes of mitochondrial antioxidant enzymes and progressive fibrotic remodeling, leading to amelioration of cardiac functional performance. Therefore, PETN might be a promising therapeutic option in the treatment of ischemic heart diseases involving oxidative stress and impairment in nitric oxide bioactivity.


Asunto(s)
Insuficiencia Cardíaca/tratamiento farmacológico , Ventrículos Cardíacos/fisiopatología , Miocardio/metabolismo , Tetranitrato de Pentaeritritol/farmacología , Tetranitrato de Pentaeritritol/uso terapéutico , Especies Reactivas de Oxígeno/metabolismo , Remodelación Ventricular/efectos de los fármacos , Animales , Disponibilidad Biológica , Modelos Animales de Enfermedad , Insuficiencia Cardíaca/fisiopatología , Ventrículos Cardíacos/efectos de los fármacos , Masculino , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Ratas , Superóxido Dismutasa/metabolismo , Superóxidos/metabolismo , Resultado del Tratamiento , Vasodilatadores/farmacología , Vasodilatadores/uso terapéutico , Remodelación Ventricular/fisiología
13.
J Mol Cell Cardiol ; 87: 194-203, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26319660

RESUMEN

INTRODUCTION: Fibroblast activation protein α (FAP) is a membrane-bound serine protease expressed by activated fibroblasts during wound healing in the skin. Expression of FAP after myocardial infarction (MI) and potential effects on cardiac wound healing are largely unknown. METHODS: MI was induced in rats and FAP expression was analyzed at 3, 7 and 28 days post-MI by microarray, Western blot and immunohistochemistry. In human hearts after MI, a FAP(+) fibroblast population was identified, and characterized by immunohistochemistry for prolyl-4-hydroxylase ß, α-smooth muscle actin, Thy-1 and vimentin. Signaling pathways leading to FAP expression were studied in human cardiac fibroblasts by Western blot and ELISA using TGFß1, TGF-beta type I-receptor (TGFbR1)-inhibitor SB431542 or the MAPK-inhibitor U0126 as well as siRNA targeting SMAD2 and SMAD3. Finally, fibroblasts were assayed for FAP-dependent migration (modified Boyden-chamber), proliferation (BrdU-assay) and gelatinolytic activity by gelatin zymography. RESULTS: In rats, FAP expression was increased after MI especially in the peri-infarct area peaking at 7 days post-MI. Co-localization analysis identified the majority of FAP(+) cells as activated proto-myofibroblasts and myofibroblasts. Concordantly, FAP(+) fibroblasts were abundant in ischemic tissue of human hearts after MI, but not in healthy control hearts. In vitro, FAP was induced by TGFß1 via the canonical SMAD2/SMAD3 pathway. Depletion of FAP in fibroblasts reduced migratory capacity, while proliferation was not affected. Gelatin zymography revealed gelatinase activity by fibroblast-derived FAP. CONCLUSION: In this study, we show for the first time the expression of FAP in activated fibroblasts after MI and its activation by TGFß1. Effects of FAP on fibroblast migration and gelatinolytic activity indicate a potential role in cardiac wound healing and remodeling.


Asunto(s)
Proteínas de la Matriz Extracelular/metabolismo , Gelatinasas/biosíntesis , Inflamación/genética , Proteínas de la Membrana/biosíntesis , Infarto del Miocardio/genética , Serina Endopeptidasas/biosíntesis , Factor de Crecimiento Transformador beta/metabolismo , Animales , Endopeptidasas , Proteínas de la Matriz Extracelular/genética , Gelatinasas/genética , Regulación de la Expresión Génica , Humanos , Inflamación/patología , Proteínas de la Membrana/genética , Infarto del Miocardio/patología , Miocardio/metabolismo , Miocardio/patología , Miofibroblastos/metabolismo , Miofibroblastos/patología , Ratas , Serina Endopeptidasas/genética , Transducción de Señal , Factor de Crecimiento Transformador beta/genética , Cicatrización de Heridas/genética
14.
ESC Heart Fail ; 2(3): 150-158, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28834677

RESUMEN

AIMS: The selective mineralocorticoid receptor (MR) antagonist eplerenone given early in patients with acute myocardial infarction (MI) improves clinical outcome, whereas little is known about the effectiveness of early spironolactone therapy. We aimed to compare the ability of eplerenone and spironolactone to promote cardiac repair after experimental MI. METHODS AND RESULTS: Starting immediately after coronary artery ligation, C57BL/6J mice were treated with placebo, eplerenone, or spironolactone. At 7 days, treatment with eplerenone or spironolactone reduced thinning and expansion of healing infarct and improved early left ventricular chamber enlargement. Remarkably, eplerenone therapy resulted in significantly greater improvement than spironolactone of left ventricular contractile function and relaxation, associated with a more considerable leftward and downward shift of the pressure volume curve. Seven-day survival rate was significantly increased only in eplerenone treated mice. Moreover, eplerenone was superior to spironolactone in ameliorating neovessel formation in the injured myocardium. Optimized flow cytometry analysis of the monocyte differentiation marker Ly6C revealed predominant accumulation of Ly6Chigh monocytes/macrophages at the site of ischemic injury during the early inflammatory phase in placebo-treated mice. In contrast, MR antagonism, especially by eplerenone, led to a skewing of the monocyte/macrophage population toward a higher frequency of healing promoting Ly6Clow cells. CONCLUSION: The MR antagonist eplerenone versus spironolactone showed superior efficacy during the acute MI phase with more beneficial effects on survival, early cardiac dilation, and functional decline. Modulation of monocyte maturation and enhanced infarct neovessel formation appears to play a pivotal role.

15.
Basic Res Cardiol ; 109(4): 421, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24907870

RESUMEN

Impaired nitric oxide (NO)-soluble guanylate cyclase (sGC)-cGMP signaling is involved in the pathogenesis of ischemic heart diseases, yet the impact of long-term sGC activation on progressive cardiac remodeling and heart failure after myocardial infarction (MI) has not been explored. Moreover, it is unknown whether stimulating the NO/heme-independent sGC provides additional benefits to ACE inhibition in chronic ischemic heart failure. Starting 10 days after MI, rats were treated with placebo, the sGC activator ataciguat (10 mg/kg/twice daily), ramipril (1 mg/kg/day), or a combination of both for 9 weeks. Long-term ataciguat therapy reduced left ventricular (LV) diastolic filling pressure and pulmonary edema, improved the rightward shift of the pressure-volume curve, LV contractile function and diastolic stiffness, without lowering blood pressure. NO/heme-independent sGC activation provided protection over ACE inhibition against mitochondrial superoxide production and progressive fibrotic remodeling, ultimately leading to a further improvement of cardiac performance, hypertrophic growth and heart failure. We found that ataciguat stimulating sGC activity was potentiated in (myo)fibroblasts during hypoxia-induced oxidative stress and that NO/heme-independent sGC activation modulated fibroblast-cardiomyocyte crosstalk in the context of heart failure and hypoxia. In addition, ataciguat inhibited human cardiac fibroblast differentiation and extracellular matrix protein production in response to TGF-ß1. Overall, long-term sGC activation targeting extracellular matrix homeostasis conferred cardioprotection against progressive cardiac dysfunction, pathological remodeling and heart failure after myocardial infarction. NO/heme-independent sGC activation may prove to be a useful therapeutic target in patients with chronic heart failure and ongoing fibrotic remodeling.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Cardiotónicos/farmacología , Activadores de Enzimas/farmacología , Guanilato Ciclasa/metabolismo , Insuficiencia Cardíaca/tratamiento farmacológico , Infarto del Miocardio/complicaciones , Miocitos Cardíacos/efectos de los fármacos , Ramipril/farmacología , Receptores Citoplasmáticos y Nucleares/metabolismo , Sulfonamidas/farmacología , Remodelación Ventricular/efectos de los fármacos , ortoaminobenzoatos/farmacología , Animales , Comunicación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Quimioterapia Combinada , Activación Enzimática , Matriz Extracelular/metabolismo , Fibrosis , Insuficiencia Cardíaca/enzimología , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Hemodinámica/efectos de los fármacos , Humanos , Masculino , Ratones , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Cardíacas/enzimología , Contracción Miocárdica/efectos de los fármacos , Infarto del Miocardio/enzimología , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/patología , Miofibroblastos/efectos de los fármacos , Miofibroblastos/enzimología , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Estrés Oxidativo/efectos de los fármacos , Ratas Wistar , Guanilil Ciclasa Soluble , Superóxidos/metabolismo , Factores de Tiempo , Función Ventricular Izquierda/efectos de los fármacos
16.
Int J Cardiol ; 168(4): 3926-31, 2013 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-23932048

RESUMEN

BACKGROUND: Fibroblast activation protein α (FAP) is a membrane glycoprotein with dipeptidyl-peptidase and collagenase activity and is expressed in cancer, arthritis, and atherosclerotic plaques. We hypothesized that FAP can be measured quantitatively in the circulation and provide prognostic information in acute coronary syndrome (ACS). METHODS: We assessed the performance of a commercially available FAP ELISA and the pre-analytic characteristics of the marker. We determined FAP concentrations in EDTA plasma samples from 101 apparently healthy blood donors and 407 patients with ACS. Patients were followed for 12 months regarding all-cause mortality and non-fatal myocardial infarction (MI). RESULTS: FAP was stable at room temperature (for 1 day) and 4°C (3 days) and resistant to 3 freeze/thaw cycles. Recovery of recombinant human FAP ranged from 78 to 103% and serial dilutions of spiked samples resulted in measurements within 91 to 120% of expected values. Patients with ACS had lower plasma FAP concentrations compared with blood donors [median (25th-75th percentiles): 84 (69-101) ng/mL vs. 108 (87-124) ng/mL, P < 0.001]. Patients presenting with FAP concentrations in the first quartile had a 3.0-fold higher risk of death (95% confidence interval 1.4-6.2) compared with patients in the second to fourth quartiles (P = 0.004). FAP concentration was not related to the risk of MI. CONCLUSIONS: Our study is the first to associate FAP with prognosis in ACS. The favorable pre-analytic characteristics of FAP will facilitate future studies of the marker in other disease settings associated with altered FAP expression.


Asunto(s)
Síndrome Coronario Agudo/sangre , Síndrome Coronario Agudo/diagnóstico , Circulación Sanguínea/fisiología , Gelatinasas/sangre , Proteínas de la Membrana/sangre , Serina Endopeptidasas/sangre , Síndrome Coronario Agudo/mortalidad , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores/sangre , Endopeptidasas , Ensayo de Inmunoadsorción Enzimática/métodos , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven
17.
Age (Dordr) ; 35(3): 747-62, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22538858

RESUMEN

MicroRNAs (miRs) are small non- coding RNA molecules controlling a plethora of biological processes such as development, cellular survival and senescence. We here determined miRs differentially regulated during cardiac postnatal development and aging. Cardiac function, morphology and miR expression profiles were determined in neonatal, 4 weeks, 6 months and 19 months old normotensive male healthy C57/Bl6N mice. MiR-22 was most prominently upregulated during cardiac aging. Cardiac expression of its bioinformatically predicted target mimecan (osteoglycin, OGN) was gradually decreased with advanced age. Luciferase reporter assays validated mimecan as a bona fide miR-22 target. Both, miR-22 and its target mimecan were co- expressed in cardiac fibroblasts and smooth muscle cells. Functionally, miR-22 overexpression induced cellular senescence and promoted migratory activity of cardiac fibroblasts. Small interference RNA-mediated silencing of mimecan in cardiac fibroblasts mimicked the miR-22-mediated effects. Rescue experiments revealed that the effects of miR-22 on cardiac fibroblasts were only partially mediated by mimecan. In conclusion, miR-22 upregulation in the aging heart contributed at least partly to accelerated cardiac fibroblast senescence and increased migratory activity. Our results suggest an involvement of miR-22 in age-associated cardiac changes, such as cardiac fibrosis.


Asunto(s)
Envejecimiento/genética , Fibroblastos/patología , Regulación del Desarrollo de la Expresión Génica , MicroARNs/genética , ARN/genética , Regulación hacia Arriba/genética , Envejecimiento/metabolismo , Envejecimiento/patología , Animales , Animales Recién Nacidos , Fibroblastos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/biosíntesis , Reacción en Cadena de la Polimerasa
18.
Eur J Clin Invest ; 42(10): 1112-20, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22536780

RESUMEN

The classical view of aldosterone actions via the mineralocorticoid receptor (MR) limited to control of fluid balance and blood pressure homoeostasis has been progressively overcome by clinical and experimental evidence emphasizing the pleiotropic role of MR activation in the pathogenesis of cardiovascular disease. Clinical studies have shown the benefit of MR blockade in patients with left ventricular dysfunction and heart failure after myocardial infarction (MI), hypertension or diabetic nephropathy. Deleterious effects of MR activation include cardiac structural and electrical remodelling, cardiovascular fibrosis, inflammation and oxidative stress. Complexity of pathophysiological role of MR derives from the presence of circulating glucocorticoids at higher concentrations than aldosterone and the equal affinity of the MR for aldosterone, cortisol and corticosterone. Recent experimental studies using different animal models and genetic tools have deeply explored the cell-specific functional role of MR in cardiovascular pathology. This review addresses emerging preclinical studies as well as ongoing clinical trials regarding MR activation in MI and failure.


Asunto(s)
Insuficiencia Cardíaca/etiología , Infarto del Miocardio/etiología , Receptores de Mineralocorticoides/metabolismo , Aldosterona/fisiología , Técnicas Genéticas , Humanos , Hiperpotasemia/inducido químicamente , Antagonistas de Receptores de Mineralocorticoides/farmacología , Antagonistas de Receptores de Mineralocorticoides/uso terapéutico , Ensayos Clínicos Controlados Aleatorios como Asunto , Receptores de Mineralocorticoides/genética , Disfunción Ventricular Izquierda/etiología
19.
Cardiovasc Res ; 94(2): 293-303, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22387461

RESUMEN

Adverse cardiac remodelling is a major cause of morbidity and mortality following acute myocardial infarction (MI). Mechanical and neurohumoral factors involved in structural and molecular post-infarction remodelling were important targets in research and treatment for years. More recently, therapeutic strategies that address myocardial regeneration and pathophysiological mechanisms of infarct wound healing appear to be useful novel tools to prevent progressive ventricular dilation, functional deterioration, life-threatening arrhythmia, and heart failure. This review provides an overview of future and emerging therapies for cardiac wound healing and remodelling after MI.


Asunto(s)
Terapia Genética/métodos , MicroARNs/uso terapéutico , Infarto del Miocardio/complicaciones , Isquemia Miocárdica/terapia , Trasplante de Células Madre/métodos , Remodelación Ventricular/efectos de los fármacos , Humanos , Infarto del Miocardio/tratamiento farmacológico , Isquemia Miocárdica/tratamiento farmacológico , Remodelación Ventricular/fisiología
20.
Circulation ; 124(6): 720-30, 2011 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-21788589

RESUMEN

BACKGROUND: Myocardial infarction leads to cardiac remodeling and development of heart failure. Insufficient myocardial capillary density after myocardial infarction has been identified as a critical event in this process, although the underlying mechanisms of cardiac angiogenesis are mechanistically not well understood. METHODS AND RESULTS: Here, we show that the small noncoding RNA microRNA-24 (miR-24) is enriched in cardiac endothelial cells and considerably upregulated after cardiac ischemia. MiR-24 induces endothelial cell apoptosis, abolishes endothelial capillary network formation on Matrigel, and inhibits cell sprouting from endothelial spheroids. These effects are mediated through targeting of the endothelium-enriched transcription factor GATA2 and the p21-activated kinase PAK4, which were identified by bioinformatic predictions and validated by luciferase gene reporter assays. Respective downstream signaling cascades involving phosphorylated BAD (Bcl-XL/Bcl-2-associated death promoter) and Sirtuin1 were identified by transcriptome, protein arrays, and chromatin immunoprecipitation analyses. Overexpression of miR-24 or silencing of its targets significantly impaired angiogenesis in zebrafish embryos. Blocking of endothelial miR-24 limited myocardial infarct size of mice via prevention of endothelial apoptosis and enhancement of vascularity, which led to preserved cardiac function and survival. CONCLUSIONS: Our findings indicate that miR-24 acts as a critical regulator of endothelial cell apoptosis and angiogenesis and is suitable for therapeutic intervention in the setting of ischemic heart disease.


Asunto(s)
Células Endoteliales/metabolismo , MicroARNs/fisiología , Infarto del Miocardio/fisiopatología , Animales , Apoptosis/efectos de los fármacos , Arteriolas/patología , Capilares/patología , Hipoxia de la Célula , Células Cultivadas/efectos de los fármacos , Células Cultivadas/metabolismo , Colágeno , Combinación de Medicamentos , Evaluación Preclínica de Medicamentos , Células Endoteliales/patología , Factor de Transcripción GATA2/biosíntesis , Factor de Transcripción GATA2/genética , Perfilación de la Expresión Génica , Insuficiencia Cardíaca/etiología , Hemo-Oxigenasa 1/biosíntesis , Hemo-Oxigenasa 1/genética , Laminina , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Infarto del Miocardio/complicaciones , Infarto del Miocardio/genética , Neovascularización Fisiológica/efectos de los fármacos , Neovascularización Fisiológica/genética , Oligorribonucleótidos/farmacología , Proteoglicanos , Interferencia de ARN , ARN Interferente Pequeño/farmacología , ARN Interferente Pequeño/uso terapéutico , Esferoides Celulares , Remodelación Ventricular , Pez Cebra/embriología , Proteínas de Pez Cebra/biosíntesis , Proteínas de Pez Cebra/genética , Quinasas p21 Activadas/biosíntesis , Quinasas p21 Activadas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...