Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Physiol Rep ; 12(14): e16143, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39034131

RESUMEN

Inflammation through activation of caspase-1, seems to play a role in pulmonary hypertension induced by alveolar hypoxia. Whether alveolar hypoxia induces caspase-1-mediated inflammation and influx of leukocytes in other organs than the lungs, is not known. Our aim was to explore sites of caspase-1-related inflammation in alveolar hypoxia. Wild type (WT) mice were exposed to environmental hypoxia or room-air, and organs were analyzed. Right heart catheterization was performed after 14 days of alveolar hypoxia in WT mice and mice transplanted with WT or caspase-1-/- bone marrow. Hypoxia induced leukocyte accumulation and increased caspase-1 protein in the lungs, not in other organs. WT mice transplanted with WT or caspase-1-/- bone marrow showed no difference in pulmonary leukocyte accumulation or development of pulmonary hypertension after alveolar hypoxia. Caspase-1 and IL-18 were detected in bronchial epithelium in WT mice, and hypoxia induced IL-18 secretion from bronchial epithelial cells. IL-18 stimulation generated IL-6 mRNA in monocytes. Phosphorylated STAT3 was increased in hypoxic lungs, not in other organs. Alveolar hypoxia induces caspase-1 activation and leukocyte accumulation specific to the lungs, not in other organs. Caspase-1 activation and IL-18 secretion from bronchial epithelial cells might initiate hypoxia-induced inflammation, leading to pulmonary hypertension.


Asunto(s)
Caspasa 1 , Hipoxia , Inflamasomas , Interleucina-18 , Pulmón , Ratones Endogámicos C57BL , Animales , Masculino , Inflamasomas/metabolismo , Ratones , Caspasa 1/metabolismo , Caspasa 1/genética , Pulmón/metabolismo , Pulmón/patología , Interleucina-18/metabolismo , Interleucina-18/genética , Hipoxia/metabolismo , Inflamación/metabolismo , Inflamación/patología , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/patología , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT3/genética , Ratones Noqueados , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/etiología , Hipertensión Pulmonar/patología
2.
Physiol Rep ; 10(3): e15184, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35146955

RESUMEN

Mountain climbing at high altitude implies exposure to low levels of oxygen, low temperature, wind, physical and psychological stress, and nutritional insufficiencies. We examined whether right ventricular (RV) and left ventricular (LV) myocardial masses were reversibly altered by exposure to extreme altitude. Magnetic resonance imaging and echocardiography of the heart, dual x-ray absorptiometry scan of body composition, and blood samples were obtained from ten mountain climbers before departure to Mount Everest or Dhaulagiri (baseline), 13.5 ± 1.5 days after peaking the mountain (post-hypoxia), and six weeks and six months after expeditions exceeding 8000 meters above sea level. RV mass was unaltered after extreme altitude, in contrast to a reduction in LV mass by 11.8 ± 3.4 g post-hypoxia (p = 0.001). The reduction in LV mass correlated with a reduction in skeletal muscle mass. After six weeks, LV myocardial mass was restored to baseline values. Extreme altitude induced a reduction in LV end-diastolic volume (20.8 ± 7.7 ml, p = 0.011) and reduced E', indicating diastolic dysfunction, which were restored after six weeks follow-up. Elevated circulating interleukin-18 after extreme altitude compared to follow-up levels, might have contributed to reduced muscle mass and diastolic dysfunction. In conclusion, the mass of the RV, possibly exposed to elevated afterload, was not changed after extreme altitude, whereas LV mass was reduced. The reduction in LV mass correlated with reduced skeletal muscle mass, indicating a common denominator, and elevated circulating interleukin-18 might be a mechanism for reduced muscle mass after extreme altitude.


Asunto(s)
Mal de Altura/fisiopatología , Ventrículos Cardíacos/diagnóstico por imagen , Adulto , Diástole , Femenino , Ventrículos Cardíacos/anatomía & histología , Ventrículos Cardíacos/metabolismo , Humanos , Interleucina-18/metabolismo , Masculino , Persona de Mediana Edad , Tamaño de los Órganos , Función Ventricular
3.
Int J Cancer ; 147(11): 3189-3198, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32525564

RESUMEN

The randomized "Testicular cancer and Aerobic and Strength Training trial" (TAST-trial) aimed to evaluate the effect of high-intensity interval training (HIIT) on cardiorespiratory fitness during cisplatin-based chemotherapy (CBCT) for testicular cancer (TC). Here, we report on an unexpected high number of thromboembolic (TE) events among patients randomized to the intervention arm, and on a review of the literature on TE events in TC patients undergoing CBCT. Patients aged 18 to 60 years with a diagnosis of metastatic germ cell TC, planned for 3 to 4 CBCT cycles, were randomized to a 9 to 12 weeks exercise intervention, or to a single lifestyle counseling session. The exercise intervention included two weekly HIIT sessions, each with 2 to 4 intervals of 2 to 4 minutes at 85% to 95% of peak heart rate. The study was prematurely discontinued after inclusion of 19 of the planned 94 patients, with nine patients randomized to the intervention arm and 10 to the control arm. Three patients in the intervention arm developed TE complications; two with pulmonary embolism and one with myocardial infarction. All three patients had clinical stage IIA TC. No TE complications were observed among patients in the control arm. Our observations indicate that high-intensity aerobic training during CBCT might increase the risk of TE events in TC patients, leading to premature closure of the TAST-trial.


Asunto(s)
Cisplatino/uso terapéutico , Entrenamiento de Intervalos de Alta Intensidad/efectos adversos , Neoplasias de Células Germinales y Embrionarias/tratamiento farmacológico , Neoplasias de Células Germinales y Embrionarias/rehabilitación , Neoplasias Testiculares/tratamiento farmacológico , Neoplasias Testiculares/rehabilitación , Tromboembolia/inducido químicamente , Adulto , Capacidad Cardiovascular , Consejo , Humanos , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Neoplasias de Células Germinales y Embrionarias/patología , Ensayos Clínicos Controlados Aleatorios como Asunto , Neoplasias Testiculares/patología , Adulto Joven
4.
Am J Physiol Lung Cell Mol Physiol ; 316(6): L999-L1012, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30908936

RESUMEN

Lung diseases with hypoxia are complicated by pulmonary hypertension, leading to heart failure and death. No pharmacological treatment exists. Increased proinflammatory cytokines are found in hypoxic patients, suggesting an inflammatory pathogenesis. Caspase-1, the effector of the inflammasome, mediates inflammation through activation of the proinflammatory cytokines interleukin (IL)-18 and IL-1ß. Here, we investigate inflammasome-related mechanisms that can trigger hypoxia-induced pulmonary hypertension. Our aim was to examine whether caspase-1 induces development of hypoxia-related pulmonary hypertension and is a suitable target for therapy. Wild-type (WT) and caspase-1-/- mice were exposed to 10% oxygen for 14 days. Hypoxic caspase-1-/- mice showed lower pressure and reduced muscularization in pulmonary arteries, as well as reduced right ventricular remodeling compared with WT. Smooth muscle cell (SMC) proliferation was reduced in caspase-1-deficient pulmonary arteries and in WT arteries treated with a caspase-1 inhibitor. Impaired inflammation was shown in hypoxic caspase-1-/- mice by abolished pulmonary influx of immune cells and lower levels of IL-18, IL-1ß, and IL-6, which were also reduced in the medium surrounding caspase-1 abrogated pulmonary arteries. By adding IL-18 or IL-1ß to caspase-1-deficient pulmonary arteries, SMC proliferation was retained. Furthermore, inhibition of both IL-6 and phosphorylated STAT3 reduced proliferation of SMC in vitro, indicating IL-18, IL-6, and STAT3 as downstream mediators of caspase-1-induced SMC proliferation in pulmonary arteries. Caspase-1 induces SMC proliferation in pulmonary arteries through the caspase-1/IL-18/IL-6/STAT3 pathway, leading to pulmonary hypertension in mice exposed to hypoxia. We propose that caspase-1 inhibition is a potential target for treatment of pulmonary hypertension.


Asunto(s)
Caspasa 1/genética , Hipoxia de la Célula/fisiología , Hipertensión Pulmonar/patología , Miocitos del Músculo Liso/fisiología , Función Ventricular Derecha/fisiología , Animales , Línea Celular , Proliferación Celular/genética , Humanos , Inflamasomas/metabolismo , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/citología , Músculo Liso Vascular/crecimiento & desarrollo , Arteria Pulmonar/citología , Arteria Pulmonar/patología , Factor de Transcripción STAT3/metabolismo
5.
Am J Physiol Lung Cell Mol Physiol ; 309(4): L378-87, 2015 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-26071556

RESUMEN

Pulmonary hypertension is a serious condition that can lead to premature death. The mechanisms involved are incompletely understood although a role for the immune system has been suggested. Inflammasomes are part of the innate immune system and consist of the effector caspase-1 and a receptor, where nucleotide-binding oligomerization domain-like receptor pyrin domain-containing 3 (NLRP3) is the best characterized and interacts with the adaptor protein apoptosis-associated speck-like protein containing a caspase-recruitment domain (ASC). To investigate whether ASC and NLRP3 inflammasome components are involved in hypoxia-induced pulmonary hypertension, we utilized mice deficient in ASC and NLRP3. Active caspase-1, IL-18, and IL-1ß, which are regulated by inflammasomes, were measured in lung homogenates in wild-type (WT), ASC(-/-), and NLRP3(-/-) mice, and phenotypical changes related to pulmonary hypertension and right ventricular remodeling were characterized after hypoxic exposure. Right ventricular systolic pressure (RVSP) of ASC(-/-) mice was significantly lower than in WT exposed to hypoxia (40.8 ± 1.5 mmHg vs. 55.8 ± 2.4 mmHg, P < 0.001), indicating a substantially reduced pulmonary hypertension in mice lacking ASC. Magnetic resonance imaging further supported these findings by demonstrating reduced right ventricular remodeling. RVSP of NLRP3(-/-) mice exposed to hypoxia was not significantly altered compared with WT hypoxia. Whereas hypoxia increased protein levels of caspase-1, IL-18, and IL-1ß in WT and NLRP3(-/-) mice, this response was absent in ASC(-/-) mice. Moreover, ASC(-/-) mice displayed reduced muscularization and collagen deposition around arteries. In conclusion, hypoxia-induced elevated right ventricular pressure and remodeling were attenuated in mice lacking the inflammasome adaptor protein ASC, suggesting that inflammasomes play an important role in the pathogenesis of pulmonary hypertension.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Hipertensión Pulmonar/metabolismo , Inflamasomas/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Arterias/patología , Proteínas Adaptadoras de Señalización CARD , Hipoxia de la Célula , Colágeno/metabolismo , Expresión Génica , Hipertrofia Ventricular Derecha/metabolismo , Interleucina-18/sangre , Leucocitos/inmunología , Pulmón/inmunología , Pulmón/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/patología , Remodelación Ventricular
6.
Exp Lung Res ; 38(8): 406-19, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22908875

RESUMEN

Interleukin (IL)-18 is a pro-inflammatory cytokine suggested to be involved in the development of pulmonary emphysema and inflammation. Studies involving immunology and cancer have revealed that IL-18 can have synergistic effects with IL-12. We have studied the presence of IL-18 and IL-12 receptors (IL-18R/IL-12R) in the lungs and whether IL-18 and IL-12, alone or in combination, have the ability to initiate the induction of mediators related to the development of emphysema and inflammation. The expression of the IL-18R was abundant in lungs compared to other organs (heart, liver, and spleen), and the IL-12R was also expressed in lung tissue. Mice treated with i.p. injection of recombinant murine IL-18 or IL-12 expressed significantly higher pulmonary mRNA levels of the matrix degrading enzymes metalloproteinase (MMP) 12 and cathepsin S, in addition to interferon-γ, tumor necrosis factor-α, and CXC chemokine ligand 9 (CXCL9) (all P < .05) than controls (received PBS). Treatment with IL-18 and IL-12 in combination showed an even more pronounced induction of these mediators, as well as a significant increase in MMP-9, IL-6, IL-1ß, and transforming growth factor-ß (P < .05). Furthermore, cellular apoptosis in lung tissue was induced. Immunohistochemical analysis revealed T-cell infiltration in pulmonary vessels following co-stimulation. In summary, IL-18 and IL-12 exert a synergistic effect on the lungs by inducing MMPs, cathepsins S, and pro-inflammatory cytokines, which may promote pulmonary emphysema and inflammation. The synergy between IL-18 and IL-12 involves infiltration of T-cells in the lungs, possibly induced by the T-cell chemoattractant CXCL9.


Asunto(s)
Catepsinas/biosíntesis , Interleucina-12/farmacología , Interleucina-18/farmacología , Pulmón/efectos de los fármacos , Metaloproteinasa 12 de la Matriz/biosíntesis , Animales , Apoptosis/efectos de los fármacos , Catepsinas/genética , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Quimioterapia Combinada , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Masculino , Metaloproteinasa 12 de la Matriz/genética , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/metabolismo , Receptores de Interleucina-12/metabolismo , Receptores de Interleucina-18/metabolismo , Proteínas Recombinantes/farmacología , Linfocitos T/metabolismo , Linfocitos T/patología , Regulación hacia Arriba/efectos de los fármacos
7.
Am J Physiol Lung Cell Mol Physiol ; 301(1): L50-9, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21498626

RESUMEN

The chemokine receptor CCR7 regulates lymphocyte trafficking, and CCR7 deficiency induces infiltration of T and B cells adjacent to vessels in mouse lungs. Perivascular infiltration of T and B cells has also been found in human pulmonary arterial hypertension, and downregulation of the CCR7 receptor in circulating leukocytes of such patients has been observed. To investigate whether changes in the CCR7 system contribute to the pathogenesis of pulmonary hypertension, we utilized mice deficient of the CCR7 receptor. The cardiopulmonary and inflammatory responses of CCR7 depletion were evaluated in CCR7-deficient and wild-type mice. Measurements of cytokines upregulated in the animal model were also performed in patients with pulmonary hypertension and controls and in vascular smooth muscle cells. We found that mice lacking CCR7 had increased right ventricular systolic pressure, reduced pulmonary artery acceleration time, increased right ventricular/tibial length ratio, Rho kinase-mediated pulmonary vasoconstriction, and increased muscularization of distal arteries, indicating pulmonary hypertension. These mice also showed increased perivascular infiltration of leukocytes, consisting mainly of T and B cells, and increased mRNA levels of the inflammatory cytokines interleukin-12 and CX3CL1 within pulmonary tissue. Increased serum levels of interleukin-12 and CX3CL1 were also observed in patients with pulmonary hypertension, particularly in those with pulmonary hypertension associated with connective tissue disorder. In smooth muscle cells, interleukin-12 induced secretion of the angiogenic cytokine interleukin-8. We conclude that these results suggest a role for CCR7 in the development of pulmonary arterial hypertension, at least in some subgroups, possibly via pulmonary infiltration of lymphocytes and secretion of interleukin-12 and CX3CL1.


Asunto(s)
Movimiento Celular , Leucocitos/patología , Neumonía/complicaciones , Neumonía/patología , Receptores CCR7/deficiencia , Adulto , Animales , Quimiocina CX3CL1/sangre , Hipertensión Pulmonar Primaria Familiar , Femenino , Regulación de la Expresión Génica , Hemodinámica , Humanos , Hipertensión Pulmonar/sangre , Hipertensión Pulmonar/complicaciones , Hipertensión Pulmonar/patología , Hipertensión Pulmonar/fisiopatología , Interleucina-12/sangre , Interleucina-8/metabolismo , Pulmón/patología , Pulmón/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Tamaño de los Órganos , Neumonía/sangre , Neumonía/fisiopatología , Arteria Pulmonar/metabolismo , Arteria Pulmonar/patología , Arteria Pulmonar/fisiopatología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores CCR7/metabolismo
8.
J Appl Physiol (1985) ; 106(4): 1356-64, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19196915

RESUMEN

Activin A, a member of the transforming growth factor (TGF)-beta superfamily, is involved in regulation of tissue remodeling and inflammation. Herein, we wanted to explore a role for activin A in pulmonary hypertension (PH). Circulating levels of activin A and its binding protein follistatin were measured in patients with PH (n = 47) and control subjects (n = 14). To investigate synthesis and localization of pulmonary activin A, we utilized an experimental model of hypoxia-induced PH. In mouse lungs, we also explored signaling pathways that can be activated by activin A, such as phosphorylation of Smads, which are mediators of TGF-beta signaling. Possible pathophysiological mechanisms initiated by activin A were explored by exposing pulmonary arterial smooth muscle cells in culture to this cytokine. Elevated levels of activin A and follistatin were found in patients with PH, and activin A levels were significantly related to mortality. Immunohistochemistry of lung autopsies from PH patients and lungs with experimental PH localized activin A primarily to alveolar macrophages and bronchial epithelial cells. Mice with PH exhibited increased pulmonary levels of mRNA for activin A and follistatin in the lungs, and also elevated pulmonary levels of phosphorylated Smad2. Finally, we found that activin A increased proliferation and induced gene expression of endothelin-1 and plasminogen activator inhibitor-1 in pulmonary artery smooth muscle cells, mediators that could contribute to vascular remodeling. Our findings in both clinical and experimental studies suggest a role for activin A in the development of various types of PH.


Asunto(s)
Activinas/metabolismo , Hipertensión Pulmonar/metabolismo , Adulto , Animales , Presión Sanguínea/fisiología , Western Blotting , Proliferación Celular , Citocinas/biosíntesis , Células Endoteliales/metabolismo , Endotelina-1/biosíntesis , Endotelina-1/genética , Femenino , Folistatina/sangre , Humanos , Hipertensión Pulmonar/patología , Técnicas para Inmunoenzimas , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Fosforilación , Arteria Pulmonar/citología , Arteria Pulmonar/metabolismo , Arteria Pulmonar/patología , ARN/biosíntesis , ARN/genética , Proteína Smad2/metabolismo , Regulación hacia Arriba
9.
Cardiovasc Res ; 80(1): 47-54, 2008 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-18599478

RESUMEN

AIMS: Chronic obstructive pulmonary disease with alveolar hypoxia is associated with diastolic dysfunction in the right and left ventricle (LV). LV diastolic dysfunction is not caused by increased afterload, and we recently showed that reduced phosphorylation of phospholamban at serine (Ser) 16 may explain the reduced relaxation of the myocardium. Here, we study the mechanisms leading to the hypoxia-induced reduction in phosphorylation of phospholamban at Ser16. METHODS AND RESULTS: In C57Bl/6j mice exposed to 10% oxygen, signalling molecules were measured in cardiac tissue, sarcoplasmic reticulum (SR)-enriched membrane preparations, and serum. Cardiomyocytes isolated from neonatal mice were exposed to interleukin (IL)-18 for 24 h. The beta-adrenergic pathway in the myocardium was not altered by alveolar hypoxia, as assessed by measurements of beta-adrenergic receptor levels, adenylyl cyclase activity, and subunits of cyclic AMP-dependent protein kinase. However, alveolar hypoxia led to a significantly higher amount (124%) and activity (234%) of protein phosphatase (PP) 2A in SR-enriched membrane preparations from LV compared with control. Serum levels of an array of cytokines were assayed, and a pronounced increase in IL-18 was observed. In isolated cardiomyocytes, treatment with IL-18 increased the amount and activity of PP2A, and reduced phosphorylation of phospholamban at Ser16 to 54% of control. CONCLUSION: Our results indicate that the diastolic dysfunction observed in alveolar hypoxia might be caused by increased circulating IL-18, thereby inducing an increase in PP2A and a reduction in phosphorylation of phospholamban at Ser16.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Insuficiencia Cardíaca Diastólica/metabolismo , Hipoxia/metabolismo , Interleucina-18/sangre , Proteína Fosfatasa 2/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Adenilil Ciclasas/metabolismo , Animales , Peso Corporal , Calcio/metabolismo , Colágeno/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Citocinas/metabolismo , Insuficiencia Cardíaca Diastólica/etiología , Hipoxia/etiología , Hipoxia/patología , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/metabolismo , Tamaño de los Órganos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/complicaciones , Receptores Adrenérgicos beta/metabolismo
10.
Am J Physiol Heart Circ Physiol ; 291(2): H507-16, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16582020

RESUMEN

Chronic obstructive pulmonary disease (COPD) may lead to pulmonary hypertension (PH) and reduced function of the right ventricle (RV). However, COPD patients may also develop left ventricular (LV) diastolic dysfunction. We hypothesized that alveolar hypoxia induces LV diastolic dysfunction and changes in proteins governing Ca(2+) removal from cytosol during diastole. Mice exposed to 10% oxygen for 1, 2, or 4 wk were compared with controls. Cardiac hemodynamics were assessed with Doppler echocardiography and a microtransducer catheter under general anesthesia. The pulmonary artery blood flow acceleration time was shorter and RV pressure was higher after 4 wk of hypoxia compared with controls (both P < 0.05). In the RV and LV, 4 wk of hypoxia induced a prolongation of the time constant of isovolumic pressure decay (51% RV, 43% LV) and a reduction in the maximum rate of decline in pressure compared with control (42% RV, 42% LV, all P < 0.05), indicating impaired relaxation and diastolic dysfunction. Alveolar hypoxia induced a 38%, 47%, and 27% reduction in Ser16-phosphorylated phospholamban (PLB) in the RV after 1, 2, and 4 wk of hypoxia, respectively, and at the same time points, Ser16-phosphorylated PLB in the LV was downregulated by 32%, 34%, and 25% (all P < 0.05). The amounts of PLB and sarco(endo)plasmic reticulum Ca(2+) ATPase (SERCA2a) were not changed. In conclusion, chronic alveolar hypoxia induces hypophosphorylation of PLB at Ser16, which might be a mechanism for impaired relaxation and diastolic dysfunction in both the RV and LV.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Hipoxia/fisiopatología , Alveolos Pulmonares/metabolismo , Disfunción Ventricular Izquierda/fisiopatología , Adenilil Ciclasas/metabolismo , Animales , Presión Sanguínea/fisiología , Western Blotting , Peso Corporal/fisiología , Citosol/metabolismo , Diástole/fisiología , Ecocardiografía , Hemodinámica/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Tamaño de los Órganos/fisiología , Fosforilación , Alveolos Pulmonares/citología , Ensayo de Unión Radioligante , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Disfunción Ventricular Izquierda/diagnóstico por imagen , Disfunción Ventricular Derecha/diagnóstico por imagen , Disfunción Ventricular Derecha/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...