Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Front Physiol ; 10: 1350, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31736778

RESUMEN

Acclimatization to hypoxia or high altitude involves physiological adaptation processes, to influence oxygen (O2) transport and utilization. Several natural products, including aromatic aldehydes and isothiocyanates stabilize the R-state of hemoglobin (Hb), increasing Hb-O2 affinity and Hb-O2 saturation. These products are a counter intuitive therapeutic strategy to increase O2 delivery during hypoxia. 5-Hydroxymethylfurfural (5-HMF) is well known Amadori compound formed during the Maillard reaction (the non-enzymatic browning and caramelization of carbohydrate-containing foods after thermal treatment), with well documented effects in Hb-O2 affinity. This study explores the therapeutic potential of 5-HMF on left ventricular (LV) cardiac function (LVCF) during hypoxia. Anesthetized Golden Syrian hamsters received 5-HMF i.v., at 100 mg/kg and were subjected to stepwise increased hypoxia (15, 10, and 5%) every 30 min. LVCF was assessed using a closed chest method with a miniaturized conductance catheter via continuous LV pressure-volume (PV) measurements. Heart hypoxic areas were studied using pimonidazole staining. 5-HMF improved cardiac indices, including stroke volume (SV), cardiac output (CO), ejection fraction (EF), and stroke work (SW) compared to the vehicle group. At 5% O2, SV, CO, EF, and SW were increased by 53, 42, 33, and 51% with 5-HMF relative to vehicle. Heart chronotropic activity was not statistically changed, suggesting that differences in LV-CF during hypoxia by 5-HMF were driven by volume dependent effects. Analysis of coronary blood flow and cardiac muscle metabolism suggest no direct pharmacological effects from 5-HMF, therefore these results can be attributed to 5-HMF-dependent increase in Hb-O2 affinity. These studies establish that naturally occurring aromatic aldehydes, such as 5-HMF, produce modification of hemoglobin oxygen affinity with promising therapeutic potential to increase O2 delivery during hypoxic hypoxia.

2.
Mol Pharm ; 16(7): 2929-2934, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31038318

RESUMEN

Anthracycline chemotherapy (e.g., doxorubicin or DOX) is associated with a cumulative dose-dependent cardiac dysfunction that may lead to congestive heart failure, which limits both its use and usefulness in the clinic. The cardiotoxicity may manifest acutely and/or months or years after treatment with doxorubicin has ended. Experimental and human data have demonstrated that angiotensin-converting enzyme/angiotensin-receptor antagonists mediate a cardioprotective effect against anthracycline toxicity. In this study, with the angiotensin receptor blocker, candesartan, as a positive control, we evaluated whether pretreatment with the hypoxic nitric oxide generating anticancer agent, RRx-001, could reduce acute DOX-induced cardiotoxicity. A total of 24 BALB/c mice were randomized for prophylactic treatment with vehicle, RRx-001, candesartan, or no-intervention control. Within each of the three intervention arms, mice received treatment with DOX. Murine pressure-volume analysis was performed with microconductance catheters to characterize the degree of cardiovascular dysfunction within each group. The following hemodynamic parameters were monitored: left ventricular systolic pressure (LVSP), heart rate, and maximal rate of increase of left ventricular pressure (±d P/d tmax). Five days after doxorubicin injection, untreated (with RRx-001) mice displayed significantly impaired systolic (LVSP, -27%; d P/d tmax, -25%; left ventricular developed pressure (LVDP), +33%; P < 0.05) and global (stroke volume (SV), -52%; ejection fraction (EF), -20%; stroke work (SW), -62.5%; heart rate (HR), -18%; cardiac output (CO), -57%; mean blood arterial pressure (MAP), -30%; systemic vascular resistance (SVR), +20%; P < 0.05) LV functions when compared with the untreated (with RRx-001) group. In contrast, RRx-001-treated mice showed improved variables of systolic (LVSP, +27%; d P/d tmax, +25%; LVDP, -33%; P < 0.05) and global (SV, +52%; EF, +20%; SW, +62.5%; HR, +18%; CO, +57%; MAP, +30%; SVR, -20%; P < 0.05) LV functions compared with untreated doxorubicin mice. Similar to the positive control, candesartan, the cardiotoxic effects of DOX in mice were partially attenuated by the prophylactic administration of RRx-001. These results suggest that RRx-001 as a multifunctional anticancer agent, which sensitizes cancer cells to the cytotoxic effects of chemotherapy and radiation, may also have beneficial cardioprotective effects.


Asunto(s)
Antibióticos Antineoplásicos/uso terapéutico , Azetidinas/uso terapéutico , Cardiomiopatías/inducido químicamente , Cardiotónicos/uso terapéutico , Doxorrubicina/toxicidad , Doxorrubicina/uso terapéutico , Neoplasias/tratamiento farmacológico , Nitrocompuestos/uso terapéutico , Enfermedad Aguda , Animales , Antibióticos Antineoplásicos/administración & dosificación , Azetidinas/administración & dosificación , Bencimidazoles/uso terapéutico , Compuestos de Bifenilo , Presión Sanguínea/efectos de los fármacos , Cardiotónicos/administración & dosificación , Cardiotoxicidad , Doxorrubicina/administración & dosificación , Frecuencia Cardíaca/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos BALB C , Nitrocompuestos/administración & dosificación , Distribución Aleatoria , Volumen Sistólico/efectos de los fármacos , Tetrazoles/uso terapéutico
3.
Am J Physiol Heart Circ Physiol ; 313(2): H381-H391, 2017 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-28526710

RESUMEN

Adaptation to hypoxia requires compensatory mechanisms that affect O2 transport and utilization. Decreased hemoglobin (Hb) O2 affinity is considered part of the physiological adaptive process to chronic hypoxia. However, this study explores the hypothesis that increased Hb O2 affinity can complement acute physiological responses to hypoxia by increasing O2 uptake and delivery compared with normal Hb O2 affinity during acute severe hypoxia. To test this hypothesis, Hb O2 affinity in mice was increased by oral administration of 2-hydroxy-6-{[(2S)-1-(pyridine-3-carbonyl)piperidin-2yl] methoxy}benzaldehyde (GBT1118; 70 or 140 mg/kg). Systemic and microcirculatory hemodynamics and oxygenation parameters were studied during hypoxia in awake-instrumented mice. GBT1118 increased Hb O2 affinity and decreased the Po2 at which 50% of Hb is saturated with O2 (P50) from 43 ± 1.1 to 18.3 ± 0.9 mmHg (70 mg/kg) and 7.7 ± 0.2 mmHg (140 mg/kg). In a dose-dependent fashion, GBT1118 increased arterial O2 saturation by 16% (70 mg/kg) and 40% (140 mg/kg) relative to the control group during 5% O2 hypoxia. In addition, a GBT1118-induced increase in Hb O2 affinity reduced hypoxia-induced hypotension compared with the control group. Moreover, microvascular blood flow was higher during hypoxia in GBT1118-treated groups than the control group. The increased O2 saturation and improved blood flow in GBT1118-treated groups preserved higher interstitial tissue Po2 than in the control group during 5% O2 hypoxia. In conclusion, increased Hb O2 affinity enhanced physiological tolerance to hypoxia, as evidenced by improved hemodynamics and tissue oxygenation. Therefore, pharmacologically induced increases in Hb O2 affinity become a potential therapeutic approach to improve tissue oxygenation in pulmonary diseases characterized by severe hypoxemia.NEW & NOTEWORTHY This study establishes that pharmacological modification of hemoglobin O2 affinity can be a promising and novel therapeutic strategy for the treatment of hypoxic hypoxia and paves the way for the clinical development of molecules that prevent hypoxemia.


Asunto(s)
Benzaldehídos/farmacología , Hipoxia/tratamiento farmacológico , Niacinamida/análogos & derivados , Oxígeno/sangre , Oxihemoglobinas/metabolismo , Piel/irrigación sanguínea , Adaptación Fisiológica , Administración Oral , Animales , Benzaldehídos/administración & dosificación , Benzaldehídos/farmacocinética , Biomarcadores/sangre , Velocidad del Flujo Sanguíneo , Presión Sanguínea , Modelos Animales de Enfermedad , Frecuencia Cardíaca , Hipoxia/sangre , Hipoxia/fisiopatología , Masculino , Ratones Endogámicos C57BL , Microcirculación/efectos de los fármacos , Niacinamida/administración & dosificación , Niacinamida/farmacocinética , Niacinamida/farmacología , Flujo Sanguíneo Regional , Índice de Severidad de la Enfermedad
4.
Artif Cells Nanomed Biotechnol ; 45(4): 686-693, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27881020

RESUMEN

Hemorrhage impairs myocardial contractile function and decreases oxygen delivery. This study investigates how polymerized bovine hemoglobin (PolyHb) solutions affect cardiac function after resuscitation from hemorrhagic shock (HS). Hamsters were hemorrhaged and resuscitated with PolyHb at 8.5 g/dL and 11.5 g/dL. Left ventricle (LV) function was assessed during shock and resuscitation using a miniaturize conductance catheter. PolyHb resuscitation had no beneficial effects in cardiac function; it increased cardiac afterload and systemic vascular resistance (SVR) of 46 and 116% for 8.5 and 11.5 g/dL, respectively. Study findings indicate that preclinical evaluation of cardiac function is essential to develop safe and efficacious alternatives to blood transfusion.


Asunto(s)
Corazón/efectos de los fármacos , Corazón/fisiopatología , Hemoglobinas/farmacología , Oxígeno/metabolismo , Polímeros/farmacología , Resucitación , Choque Hemorrágico/fisiopatología , Choque Hemorrágico/terapia , Animales , Bovinos , Cricetinae , Hemodinámica/efectos de los fármacos , Hemoglobinas/uso terapéutico , Masculino , Polímeros/uso terapéutico , Choque Hemorrágico/sangre , Choque Hemorrágico/metabolismo
5.
Proc Natl Acad Sci U S A ; 112(33): 10425-30, 2015 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-26240367

RESUMEN

To better understand human adaptation to stress, and in particular to hypoxia, we took advantage of one of nature's experiments at high altitude (HA) and studied Ethiopians, a population that is well-adapted to HA hypoxic stress. Using whole-genome sequencing, we discovered that EDNRB (Endothelin receptor type B) is a candidate gene involved in HA adaptation. To test whether EDNRB plays a critical role in hypoxia tolerance and adaptation, we generated EdnrB knockout mice and found that when EdnrB (-/+) heterozygote mice are treated with lower levels of oxygen (O2), they tolerate various levels of hypoxia (even extreme hypoxia, e.g., 5% O2) very well. For example, they maintain ejection fraction, cardiac contractility, and cardiac output in severe hypoxia. Furthermore, O2 delivery to vital organs was significantly higher and blood lactate was lower in EdnrB (-/+) compared with wild type in hypoxia. Tissue hypoxia in brain, heart, and kidney was lower in EdnrB (-/+) mice as well. These data demonstrate that a lower level of EDNRB significantly improves cardiac performance and tissue perfusion under various levels of hypoxia. Transcriptomic profiling of left ventricles revealed three specific genes [natriuretic peptide type A (Nppa), sarcolipin (Sln), and myosin light polypeptide 4 (Myl4)] that were oppositely expressed (q < 0.05) between EdnrB (-/+) and wild type. Functions related to these gene networks were consistent with a better cardiac contractility and performance. We conclude that EDNRB plays a key role in hypoxia tolerance and that a lower level of EDNRB contributes, at least in part, to HA adaptation in humans.


Asunto(s)
Corazón/fisiología , Hipoxia/patología , Receptor de Endotelina B/fisiología , Aclimatación/genética , Altitud , Animales , Factor Natriurético Atrial/fisiología , Gasto Cardíaco/fisiología , Etiopía , Femenino , Heterocigoto , Humanos , Ácido Láctico/química , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Musculares/fisiología , Contracción Miocárdica , Cadenas Ligeras de Miosina/fisiología , Oxígeno/química , Proteolípidos/fisiología , Sitios de Carácter Cuantitativo , Receptor de Endotelina B/genética , Análisis de Secuencia de ADN , Distribución Tisular
6.
Nanotechnology ; 25(26): 265102, 2014 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-24920392

RESUMEN

Hypoxia is the major hindrance to successful radiation therapy of tumors. Attempts to increase the oxygen (O2) tension (PO2) of tissue by delivering more O2 have been clinically disappointing, largely due to the way O2 is transported and released by the hemoglobin (Hb) within the red blood cells (RBCs). Systemic manipulation of O2 transport increases vascular resistance due to metabolic autoregulation of blood flow to prevent over oxygenation. This study investigates a new technology to increase O2 delivery to a target tissue by decreasing the Hb-O2 affinity of the blood circulating within the targeted tissue. As the Hb-O2 affinity decreases, the tissue PO2 to satisfy tissue O2 metabolic needs increases without increasing O2 delivery or extraction. Paramagnetic nanoparticles (PMNPs), synthetized using gadolinium oxide, were coated with the cell permeable Hb allosteric effector L35 (3,5-trichlorophenylureido-phenoxy-methylpropionic acid). L35 decreases Hb affinity for O2 and favors the release of O2. The L35-coated PMNPs (L35-PMNPs) were intravenously infused (10 mg kg(-1)) to hamsters instrumented with the dorsal window chamber model. A magnetic field of 3 mT was applied to localize the effects of the L35-PMNPs to the window chamber. Systemic O2 transport characteristics and microvascular tissue oxygenation were measured after administration of L35-PMNPs with and without magnetic field. The tissue PO2 in untreated control animals was 25.2 mmHg. L35-PMNPs without magnetic field decreased tissue PO2 to 23.4 mmHg, increased blood pressure, and reduced blood flow, largely due to systemic modification of Hb-O2 affinity. L35-PMNPs with magnetic field increased tissue PO2 to 27.9 mmHg, without systemic or microhemodynamic changes. These results indicate that localized modification of Hb-O2 affinity can increase PO2 of target tissue without affecting systemic O2 delivery or triggering O2 autoregulation mechanisms. This technology can be used to treat local hypoxia and to increase O2 in tumors, enhancing the efficacy of radiation therapies.


Asunto(s)
Permeabilidad Capilar , Sistemas de Liberación de Medicamentos/instrumentación , Eritrocitos/metabolismo , Microvasos/metabolismo , Nanopartículas/administración & dosificación , Nanopartículas/química , Oxígeno/metabolismo , Compuestos de Fenilurea/administración & dosificación , Animales , Análisis Químico de la Sangre , Eritrocitos/química , Hematócrito , Hemodinámica/efectos de los fármacos , Hemoglobinas/metabolismo , Campos Magnéticos , Masculino , Mesocricetus , Nanopartículas/ultraestructura , Compuestos de Fenilurea/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA