Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Artículo en Inglés | MEDLINE | ID: mdl-39207487

RESUMEN

PURPOSE: This study aimed to evaluate a novel technique for cell tracking by visualising the activity of the human sodium/iodide symporter (hNIS) after transplantation of hNIS-expressing multilayered cell sheets in a rat model of chronic myocardial infarction. METHODS: Triple-layered cell sheets were generated from mouse embryonic fibroblasts (MEFs) derived from mice overexpressing hNIS (hNIS-Tg). Myocardial infarction was induced by permanent ligation of the left anterior descending coronary artery in F344 athymic rats, and a triple-layered MEFs sheets were transplanted to the infarcted area two weeks after surgery. To validate the temporal tracking and kinetic analysis of the transplanted MEFs sheets, sequential cardiac single-photon emission computed tomography (SPECT) examinations with a 99mTcO4- injection were performed. The cell sheets generated using MEFs of wild-type mice (WT) served as controls. RESULTS: A significantly higher amount of 99mTcO4- was taken into the hNIS-Tg MEFs than into WT MEFs (146.1 ± 30.9-fold). The obvious accumulation of 99mTcO4- was observed in agreement with the region where hNIS-Tg MEFs were transplanted, and these radioactivities peaked 40-60 min after 99mTcO4- administration. The volume of distribution of the hNIS-Tg MEF sheets declined gradually after transplantation, implying cellular malfunction and a loss in the number of transplanted cells. CONCLUSION: The reporter gene imaging with hNIS enables the serial tracking and quantitative kinetic analysis of cell sheets transplanted to infarcted hearts.

2.
Biology (Basel) ; 11(9)2022 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-36138830

RESUMEN

Natriuretic peptides, including atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP), and C-type natriuretic peptide (CNP), have cardioprotective effects and regulate blood pressure in mammals. ANP and BNP are hormones secreted from the heart into the bloodstream in response to increased preload and afterload. Both hormones act through natriuretic peptide receptor 1 (NPR1). In contrast, CNP acts through natriuretic peptide receptor 2 (NPR2) and was found to be produced by the vascular endothelium, chondrocytes, and cardiac fibroblasts. Based on its relatively low plasma concentration compared with ANP and BNP, CNP is thought to function as both an autocrine and a paracrine factor in the vasculature, bone, and heart. The cytoplasmic domains of both NPR1 and NPR2 display a guanylate cyclase activity that catalyzes the formation of cyclic GMP. NPR3 lacks this guanylate cyclase activity and is reportedly coupled to Gi-dependent signaling. Recently, we reported that the continuous infusion of the peptide osteocrin, an endogenous ligand of NPR3 secreted by bone and muscle cells, lowered blood pressure in wild-type mice, suggesting that endogenous natriuretic peptides play major roles in the regulation of blood pressure. Neprilysin is a neutral endopeptidase that degrades several vasoactive peptides, including natriuretic peptides. The increased worldwide clinical use of the angiotensin receptor-neprilysin inhibitor for the treatment of chronic heart failure has brought renewed attention to the physiological effects of natriuretic peptides. In this review, we provide an overview of the discovery of ANP and its translational research. We also highlight our recent findings on the blood pressure regulatory effects of ANP, focusing on its molecular mechanisms.

3.
Hypertension ; 79(7): 1409-1422, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35534926

RESUMEN

BACKGROUND: ANP (atrial natriuretic peptide), acting through NPR1 (natriuretic peptide receptor 1), provokes hypotension. Such hypotension is thought to be due to ANP inducing vasodilation via NPR1 in the vasculature; however, the underlying mechanism remains unclear. Here, we investigated the mechanisms of acute and chronic blood pressure regulation by ANP. METHODS AND RESULTS: Immunohistochemical analysis of rat tissues revealed that NPR1 was abundantly expressed in endothelial cells and smooth muscle cells of small arteries and arterioles. Intravenous infusion of ANP significantly lowered systolic blood pressure in wild-type mice. ANP also significantly lowered systolic blood pressure in smooth muscle cell-specific Npr1-knockout mice but not in endothelial cell-specific Npr1-knockout mice. Moreover, ANP significantly lowered systolic blood pressure in Nos3-knockout mice. In human umbilical vein endothelial cells, treatment with ANP did not influence nitric oxide production or intracellular Ca2+ concentration, but it did hyperpolarize the cells. ANP-induced hyperpolarization of human umbilical vein endothelial cells was inhibited by several potassium channel blockers and was also abolished under knockdown of RGS2 (regulator of G-protein signaling 2), an GTPase activating protein in G-protein α-subunit. ANP increased Rgs2 mRNA expression in human umbilical vein endothelial cells but failed to lower systolic blood pressure in Rgs2-knockout mice. Endothelial cell-specific Npr1-overexpressing mice exhibited lower blood pressure than did wild-type mice independent of RGS2, and showed dilation of arterial vessels on synchrotron radiation microangiography. CONCLUSIONS: Together, these results indicate that vascular endothelial NPR1 plays a crucial role in ANP-mediated blood pressure regulation, presumably by a mechanism that is RGS2-dependent in the acute phase and RGS2-independent in the chronic phase.


Asunto(s)
Factor Natriurético Atrial , Presión Sanguínea , Receptores del Factor Natriurético Atrial , Animales , Factor Natriurético Atrial/farmacología , Presión Sanguínea/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Proteínas de Unión al GTP/metabolismo , Ratones , Ratones Noqueados , Ratas , Receptores del Factor Natriurético Atrial/metabolismo
4.
Circulation ; 141(7): 571-588, 2020 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-31665900

RESUMEN

BACKGROUND: The maternal circulatory system and hormone balance both change dynamically during pregnancy, delivery, and the postpartum period. Although atrial natriuretic peptides and brain natriuretic peptides produced in the heart control circulatory homeostasis through their common receptor, NPR1, the physiologic and pathophysiologic roles of endogenous atrial natriuretic peptide/brain natriuretic peptide in the perinatal period are not fully understood. METHODS: To clarify the physiologic and pathophysiologic roles of the endogenous atrial natriuretic peptide/brain natriuretic peptide-NPR1 system during the perinatal period, the phenotype of female wild-type and conventional or tissue-specific Npr1-knockout mice during the perinatal period was examined, especially focusing on maternal heart weight, blood pressure, and cardiac function. RESULTS: In wild-type mice, lactation but not pregnancy induced reversible cardiac hypertrophy accompanied by increases in fetal cardiac gene mRNAs and ERK1/2 (extracellular signaling-regulated kinase) phosphorylation. Npr1-knockout mice exhibited significantly higher plasma aldosterone level than did wild-type mice, severe cardiac hypertrophy accompanied by fibrosis, and left ventricular dysfunction in the lactation period. Npr1-knockout mice showed a high mortality rate over consecutive pregnancy-lactation cycles. In the hearts of Npr1-knockout mice during or after the lactation period, an increase in interleukin-6 mRNA expression, phosphorylation of signal transducer and activator of transcription 3, and activation of the calcineurin-nuclear factor of the activated T cells pathway were observed. Pharmacologic inhibition of the mineralocorticoid receptor or neuron-specific deletion of the mineralocorticoid receptor gene significantly ameliorated cardiac hypertrophy in lactating Npr1-knockout mice. Anti-interleukin-6 receptor antibody administration tended to reduce cardiac hypertrophy in lactating Npr1-knockout mice. CONCLUSIONS: These results suggest that the characteristics of lactation-induced cardiac hypertrophy in wild-type mice are different from exercise-induced cardiac hypertrophy, and that the endogenous atrial natriuretic peptide/brain natriuretic peptide-NPR1 system plays an important role in protecting the maternal heart from interleukin-6-induced inflammation and remodeling in the lactation period, a condition mimicking peripartum cardiomyopathy.


Asunto(s)
Factor Natriurético Atrial/deficiencia , Cardiomegalia/metabolismo , Lactancia , Sistema de Señalización de MAP Quinasas , Periodo Periparto , Receptores del Factor Natriurético Atrial/deficiencia , Animales , Cardiomegalia/genética , Cardiomegalia/patología , Femenino , Ratones , Ratones Noqueados
5.
Lipids ; 54(8): 465-470, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31273804

RESUMEN

Lysophosphatidylcholine (lysoPtdCho) is produced mainly by the phospholipase A2-dependent hydrolysis of phosphatidylcholine (PtdCho) and can induce inflammatory activation and osteogenic gene expression in vascular smooth muscle cells. However, the mechanisms mediating these processes have not been fully elucidated. In this study, we investigated whether inhibition of protein kinase A (PKA) signaling suppressed lysoPtdCho-induced calcification of human aortic smooth muscle cells (HASMC). Calcium levels and alkaline phosphatase activity were significantly increased in HASMC treated with lysoPtdCho, but not PtdCho, compared with those in phosphate-buffered saline-treated HASMC. However, the addition of a PKA inhibitor (H-89) or PKA siRNA blocked lysoPtdCho-induced HASMC calcification. These results showed that lysoPtdCho could activate PKA-mediated HASMC calcification and that PKA may be a therapeutic target for lysoPtdCho-mediated vascular smooth muscle cell calcification.


Asunto(s)
Aorta/efectos de los fármacos , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Isoquinolinas/farmacología , Lisofosfatidilcolinas/antagonistas & inhibidores , Células Musculares/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Sulfonamidas/farmacología , Aorta/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Relación Dosis-Respuesta a Droga , Humanos , Isoquinolinas/química , Lisofosfatidilcolinas/farmacología , Células Musculares/metabolismo , Inhibidores de Proteínas Quinasas/química , Relación Estructura-Actividad , Sulfonamidas/química
6.
Ultrasound Med Biol ; 45(6): 1455-1465, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30857759

RESUMEN

Arginine-glycine-aspartate (RGD)-carrying microbubbles (MBs) have been utilized as a specific contrast agent for glycoprotein IIb/IIIa (αIIbß3 integrin)-expressing activated platelets in ultrasound molecular imaging. Recently, we found that surface modification with lactadherin provides the RGD motif on the surface of phosphatidylserine-containing clinically available MBs, Sonazoid. Here, we examined the potential of lactadherin-bearing Sonazoid MBs to be targeted MBs for glycoprotein IIb/IIIa using the custom-designed in vitro settings with recombinant αIIbß3 integrin, activated platelets or erythrocyte-rich human clots. By modification of the surface with lactadherin, a large number of Sonazoid MBs were attached to the αIIbß3 integrin-coated and platelet-immobilized plate. Additionally, the video intensity of clots after incubation with lactadherin-bearing Sonazoid MBs was significantly higher than that with unmodified Sonazoid MBs, implying the number of attached Sonazoid MBs was increased by the modification with lactadherin. Our results suggest that the lactadherin-bearing Sonazoid MBs have the potential to be thrombus-targeted MBs.


Asunto(s)
Antígenos de Superficie/farmacología , Medios de Contraste/farmacocinética , Compuestos Férricos/farmacocinética , Aumento de la Imagen/métodos , Hierro/farmacocinética , Microburbujas , Proteínas de la Leche/farmacología , Óxidos/farmacocinética , Ultrasonografía/métodos , Femenino , Humanos , Masculino , Imagen Molecular/métodos , Valores de Referencia
8.
Peptides ; 111: 42-46, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29791869

RESUMEN

Ghrelin, a growth hormone-releasing peptide that was first discovered in the stomach of rats in 1999, is an endogenous ligand of growth hormone secretagogue receptor. Ghrelin exerts its potent growth hormone-releasing and orexigenic activities by binding to specific receptors in the brain. Subsequent studies showed that ghrelin participates in the regulation of diverse processes, including energy balance, body weight maintenance, and glucose and fat metabolism, and demonstrated that ghrelin is beneficial for treatment of cardiac diseases. In animal models of chronic heart failure, administration of ghrelin improves cardiac function and remodeling, and these findings were recapitulated in human patients with heart failure. Also in animal models, ghrelin administration effectively diminishes pulmonary hypertension induced by monocrotaline or chronic hypoxia. In addition, repeated administration of ghrelin to cachectic chronic obstructive pulmonary disease patients has positive effects on body composition, including amelioration of muscle wasting, improvement of functional capacity, and sympathetic activity. Moreover, administration of ghrelin early after myocardial infarction decreases the frequency of fatal arrhythmia and improved the survival rate. In ghrelin-deficient mice, both exogenous and endogenous ghrelin protects against fatal arrhythmia and promotes remodeling after myocardial infarction. Although the mechanisms underlying the effects of ghrelin on the cardiovascular system have not been fully elucidated, some evidence suggests that its beneficial effects are mediated through both direct actions on cardiovascular cells and regulation of autonomic nervous system activity. Therefore, ghrelin is a promising novel therapeutic agent for cardiac disease.


Asunto(s)
Ghrelina/farmacología , Corazón/efectos de los fármacos , Animales , Sistema Nervioso Autónomo/efectos de los fármacos , Sistema Nervioso Autónomo/metabolismo , Sistema Cardiovascular/efectos de los fármacos , Sistema Cardiovascular/metabolismo , Ghrelina/uso terapéutico , Corazón/fisiología , Corazón/fisiopatología , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/metabolismo , Humanos , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/metabolismo , Receptores de Ghrelina/metabolismo
9.
Life Sci ; 209: 466-471, 2018 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-30144450

RESUMEN

AIMS: Smooth muscle cells (SMCs) play a role in medial vascular calcification, which can be stimulated by high levels of serum phosphate and inflammatory mediators. The aim of this study was to investigate whether mitogen-activated protein kinases (MAPKs) (p38 MAPK, ERK1/2, and JNK) and protein kinase A (PKA) can participate in inorganic phosphate (Pi)- and inflammation response-stimulated SMC calcification. MAIN METHODS: We examined the change of Pi- and/or inflammation-stimulated human aortic smooth muscle cell (HASMC) calcification in the presence and absence of inhibitors or small interfering RNAs. KEY FINDINGS: Ca levels were increased in HASMCs incubated with 1.5-3.9 mM Pi, but not with 0.9 mM Pi or compared with non-Pi-treated HASMCs. Furthermore, the addition of interferon-γ (IFN-γ) increased pro-inflammatory cytokines [interleukin (IL)-1α, IL-6, and tumor necrosis factor-α (TNF-α)] in media containing Raw 264.7 cells. Ca levels were significantly increased in HASMCs cultured in IFN-γ-treated medium, compared with non-IFN-γ-treated medium in the presence of Pi (0.9-2.4 mM). The inhibition of p38 MAPK and PKA decreased HASMC calcification stimulated by Pi and IFN-γ-treated medium, though PKA inhibition produced a more significant reduction in calcification than p38 MAPK inhibition. SIGNIFICANCE: These results indicate that PKA inhibition can efficiently reduce Pi- and inflammation-stimulated SMC calcification.


Asunto(s)
Aorta/efectos de los fármacos , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Inflamación/fisiopatología , Interferón gamma/farmacología , Músculo Liso Vascular/efectos de los fármacos , Fosfatos/farmacología , Calcificación Vascular/tratamiento farmacológico , Aorta/inmunología , Aorta/metabolismo , Aorta/patología , Células Cultivadas , Humanos , Mediadores de Inflamación/metabolismo , Péptidos y Proteínas de Señalización Intracelular/farmacología , Músculo Liso Vascular/inmunología , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Factor de Necrosis Tumoral alfa/metabolismo , Calcificación Vascular/inmunología , Calcificación Vascular/metabolismo , Calcificación Vascular/patología
10.
Ultrasound Med Biol ; 44(5): 1063-1073, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29501282

RESUMEN

The usefulness of ultrasound molecular imaging with αvß3 integrin-targeted microbubbles for detecting tumor angiogenesis has been demonstrated. Recently, we developed αvß3 integrin-targeted microbubbles by modifying clinically available microbubbles (Sonazoid, Daiichi-Sankyo Pharmaceuticals, Tokyo, Japan) with a secreted glycoprotein (lactadherin). The aims of our present study were to simplify the preparation of lactadherin-bearing Sonazoid and to examine the diagnostic utility of lactadherin-bearing Sonazoid for αvß3 integrin-expressing tumor vessels by using SK-OV-3-tumor-bearing mice. By incubating 1.2 × 107 Sonazoid microbubbles with 1.0 µg lactadherin, the complicated washing and centrifugation steps during the microbubble preparation could be omitted with no significant reduction in labeling ratio of lactadherin-bearing Sonazoid. In addition, the number of Sonazoid microbubbles accumulated in the SK-OV-3 tumor was significantly increased by modifying Sonazoid with lactadherin. Our data suggest that the lactadherin-bearing Sonazoid is an easily prepared and potentially clinically translatable targeted microbubble for αvß3 integrin-expressing vessels.


Asunto(s)
Medios de Contraste , Aumento de la Imagen/métodos , Integrina alfaVbeta3 , Microburbujas , Neoplasias Ováricas/diagnóstico por imagen , Ultrasonografía/métodos , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Compuestos Férricos , Hierro , Ratones , Ratones Endogámicos BALB C , Óxidos , Reproducibilidad de los Resultados
11.
Circ Res ; 122(5): 742-751, 2018 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-29326144

RESUMEN

RATIONALE: An increase of severe ischemic heart diseases results in an increase of the patients with congestive heart failure (CHF). Therefore, new therapies are expected in addition to recanalization of coronary arteries. Previous clinical trials using natriuretic peptides (NPs) prove the improvement of CHF by NPs. OBJECTIVE: We aimed at investigating whether OSTN (osteocrin) peptide potentially functioning as an NPR (NP clearance receptor) 3-blocking peptide can be used as a new therapeutic peptide for treating CHF after myocardial infarction (MI) using animal models. METHODS AND RESULTS: We examined the effect of OSTN on circulation using 2 mouse models; the continuous intravenous infusion of OSTN after MI and the OSTN-transgenic (Tg) mice with MI. In vitro studies revealed that OSTN competitively bound to NPR3 with atrial NP. In both OSTN-continuous intravenous infusion model and OSTN-Tg model, acute inflammation within the first week after MI was reduced. Moreover, both models showed the improvement of prognosis at 28 days after MI by OSTN. Consistent with the in vitro study binding of OSTN to NPR3, the OSTN-Tg exhibited an increased plasma atrial NP and C-type NP, which might result in the improvement of CHF after MI as indicated by the reduced weight of hearts and lungs and by the reduced fibrosis. CONCLUSIONS: OSTN might suppress the worsening of CHF after MI by inhibiting clearance of NP family peptides.


Asunto(s)
Insuficiencia Cardíaca/tratamiento farmacológico , Proteínas Musculares/uso terapéutico , Infarto del Miocardio/tratamiento farmacológico , Factores de Transcripción/uso terapéutico , Animales , Factor Natriurético Atrial/metabolismo , Células HEK293 , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Musculares/metabolismo , Infarto del Miocardio/complicaciones , Infarto del Miocardio/metabolismo , Unión Proteica , Receptores del Factor Natriurético Atrial/metabolismo , Factores de Transcripción/metabolismo
12.
Sci Rep ; 6: 39377, 2016 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-27996031

RESUMEN

Severe intrauterine ischemia is detrimental to the developing brain. The impact of mild intrauterine hypoperfusion on neurological development, however, is still unclear. We induced mild intrauterine hypoperfusion in rats on embryonic day 17 via arterial stenosis with metal microcoils wrapped around the uterine and ovarian arteries. All pups were born with significantly decreased birth weights. Decreased gray and white matter areas were observed without obvious tissue damage. Pups presented delayed newborn reflexes, muscle weakness, and altered spontaneous activity. The levels of proteins indicative of inflammation and stress in the vasculature, i.e., RANTES, vWF, VEGF, and adiponectin, were upregulated in the placenta. The levels of mRNA for proteins associated with axon and astrocyte development were downregulated in fetal brains. The present study demonstrates that even mild intrauterine hypoperfusion can alter neurological development, which mimics the clinical signs and symptoms of children with neurodevelopmental disorders born prematurely or with intrauterine growth restriction.


Asunto(s)
Enfermedades del Prematuro/patología , Trastornos del Neurodesarrollo/patología , Animales , Animales Recién Nacidos , Femenino , Retardo del Crecimiento Fetal/patología , Inflamación/patología , Parto/fisiología , Placenta/patología , Embarazo , Ratas , Ratas Sprague-Dawley , Reproducción/fisiología , Útero/patología , Sustancia Blanca/patología
13.
Endocrinology ; 157(1): 358-67, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26517044

RESUMEN

Atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP) bind to the receptor guanylyl cyclase (GC)-A, leading to diuresis, natriuresis, and blood vessel dilation. In addition, ANP and BNP have various angiogenic properties in ischemic tissue. When breeding mice devoid of GC-A, we noted significant skewing of the Mendelian ratio in the offspring, suggesting embryonic lethality due to knockout of GC-A. Consequently, we here investigated the roles of endogenous ANP and BNP in embryonic neovascularization and organ morphogenesis. Embryos resulting from GC-A(-/-) × GC-A(+/-) crosses developed hydrops fetalis (HF) beginning at embryonic day (E)14.5. All embryos with HF had the genotype GC-A(-/-). At E17.5, 33.3% (12 of 36) of GC-A(-/-) embryos had HF, and all GC-A(-/-) embryos with HF were dead. Beginning at E16.0, HF-GC-A(-/-) embryos demonstrated poorly developed superficial vascular vessels and sc hemorrhage, the fetal side of the placenta appeared ischemic, and vitelline vessels on the yolk sac were poorly developed. Furthermore, HF-GC-A(-/-) embryos also showed abnormal constriction of umbilical cord vascular vessels, few cardiac trabeculae and a thin compact zone, hepatic hemorrhage, and poor bone development. Electron microscopy of E16.5 HF-GC-A(-/-) embryos revealed severe vacuolar degeneration in endothelial cells, and the expected 3-layer structure of the smooth muscle wall of the umbilical artery was indistinct. These data demonstrate the importance of the endogenous ANP/BNP-GC-A system not only in the neovascularization of ischemic tissues but also in embryonic vascular development and organ morphogenesis.


Asunto(s)
Factor Natriurético Atrial/metabolismo , Embrión de Mamíferos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Péptido Natriurético Encefálico/metabolismo , Neovascularización Fisiológica , Organogénesis , Receptores del Factor Natriurético Atrial/metabolismo , Animales , Factor Natriurético Atrial/genética , Células Cultivadas , Cruzamientos Genéticos , Embrión de Mamíferos/citología , Embrión de Mamíferos/patología , Embrión de Mamíferos/ultraestructura , Femenino , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/ultraestructura , Humanos , Hidropesía Fetal/genética , Hidropesía Fetal/patología , Hidropesía Fetal/veterinaria , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones Noqueados , Microscopía Electrónica de Transmisión , Péptido Natriurético Encefálico/genética , Embarazo , Receptores del Factor Natriurético Atrial/agonistas , Receptores del Factor Natriurético Atrial/deficiencia , Receptores del Factor Natriurético Atrial/genética , Transducción de Señal
14.
Hypertension ; 65(6): 1238-44, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25870195

RESUMEN

Cardiac hypertrophy, which is commonly caused by hypertension, is a major risk factor for heart failure and sudden death. Endogenous ghrelin has been shown to exert a beneficial effect on cardiac dysfunction and postinfarction remodeling via modulation of the autonomic nervous system. However, ghrelin's ability to attenuate cardiac hypertrophy and its potential mechanism of action are unknown. In this study, cardiac hypertrophy was induced by transverse aortic constriction in ghrelin knockout mice and their wild-type littermates. After 12 weeks, the ghrelin knockout mice showed significantly increased cardiac hypertrophy compared with wild-type mice, as evidenced by their significantly greater heart weight/tibial length ratios (9.2±1.9 versus 7.9±0.8 mg/mm), left ventricular anterior wall thickness (1.3±0.2 versus 1.0±0.2 mm), and posterior wall thickness (1.1±0.3 versus 0.9±0.1 mm). Furthermore, compared with wild-type mice, ghrelin knockout mice showed suppression of the cholinergic anti-inflammatory pathway, as indicated by reduced parasympathetic nerve activity and higher plasma interleukin-1ß and interleukin-6 levels. The administration of either nicotine or ghrelin activated the cholinergic anti-inflammatory pathway and attenuated cardiac hypertrophy in ghrelin knockout mice. In conclusion, our results show that endogenous ghrelin plays a crucial role in the progression of pressure overload-induced cardiac hypertrophy via a mechanism that involves the activation of the cholinergic anti-inflammatory pathway.


Asunto(s)
Cardiomegalia/metabolismo , Colinérgicos , Ghrelina/farmacología , Transducción de Señal/efectos de los fármacos , Presión Ventricular/fisiología , Análisis de Varianza , Animales , Derivados de Atropina/farmacología , Cardiomegalia/fisiopatología , Modelos Animales de Enfermedad , Ghrelina/metabolismo , Ratones , Ratones Noqueados , Nicotina/farmacología , Distribución Aleatoria , Valores de Referencia , Transducción de Señal/fisiología
15.
Proc Natl Acad Sci U S A ; 112(13): 4086-91, 2015 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-25775533

RESUMEN

Most patients suffering from cancer die of metastatic disease. Surgical removal of solid tumors is performed as an initial attempt to cure patients; however, surgery is often accompanied with trauma, which can promote early recurrence by provoking detachment of tumor cells into the blood stream or inducing systemic inflammation or both. We have previously reported that administration of atrial natriuretic peptide (ANP) during the perioperative period reduces inflammatory response and has a prophylactic effect on postoperative cardiopulmonary complications in lung cancer surgery. Here we demonstrate that cancer recurrence after curative surgery was significantly lower in ANP-treated patients than in control patients (surgery alone). ANP is known to bind specifically to NPR1 [also called guanylyl cyclase-A (GC-A) receptor]. In mouse models, we found that metastasis of GC-A-nonexpressing tumor cells (i.e., B16 mouse melanoma cells) to the lung was increased in vascular endothelium-specific GC-A knockout mice and decreased in vascular endothelium-specific GC-A transgenic mice compared with control mice. We examined the effect of ANP on tumor metastasis in mice treated with lipopolysaccharide, which mimics systemic inflammation induced by surgical stress. ANP inhibited the adhesion of cancer cells to pulmonary arterial and micro-vascular endothelial cells by suppressing the E-selectin expression that is promoted by inflammation. These results suggest that ANP prevents cancer metastasis by inhibiting the adhesion of tumor cells to inflamed endothelial cells.


Asunto(s)
Factor Natriurético Atrial/farmacología , Células Endoteliales/citología , Neoplasias/metabolismo , Animales , Adhesión Celular , Línea Celular Tumoral , Supervivencia sin Enfermedad , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Inflamación , Estimación de Kaplan-Meier , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/terapia , Melanoma Experimental , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Metástasis de la Neoplasia , Recurrencia Local de Neoplasia , Neoplasias/patología , Estudios Retrospectivos
16.
Clin Exp Nephrol ; 19(2): 197-207, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24845230

RESUMEN

BACKGROUND AND AIM: The infusion of chronic angiotensin II (Ang II) has been shown to promote renal interstitial fibrosis. To evaluate the pathophysiological significance of the natriuretic peptide-GC-A system, we infused Ang II (1.0 mg/kg/day) in GC-A-deficient mice (GC-A-KO). METHODS: We used 5 groups (Wild-Saline n = 12, Wild-Ang II n = 14, GC-A-KO-Saline n = 11, GC-A-KO-Ang II n = 13, and GC-A-KO-Ang II-Hydralazine n = 10). Saline or Ang II was infused subcutaneously using an osmotic minipump for 3 weeks. Hydralazine was administered orally (0.05 g/L in drinking water). RESULTS: Systolic blood pressure was significantly higher in the GC-A-KO-Saline group (130 ± 12 mmHg) than in the Wild-Saline group (105 ± 30 mmHg), and was similar to that in the Wild-Ang II (141 ± 17 mmHg) and GC-A-KO-Ang II-Hydralazine (140 ± 20 mmHg) groups. Systolic blood pressure was significantly higher in the GC-A-KO-Ang II group (159 ± 21 mmHg) than in the 4 other groups. Renal tubular atrophy and interstitial fibrosis were significantly more severe in the GC-A-KO-Ang II group (atrophy 13.4 %, fibrosis 12.0 %) than in the Wild-Saline (0, 2.0 %), Wild-Ang II (2.9, 4.4 %), and GC-A-KO-Saline (0, 2.6 %) groups. Hydralazine could not inhibit this aggravation (GC-A-KO-Ang II-Hydralazine 13.5, 11.3 %). The expression of monocyte chemotactic protein-1 in tubular cells, and F4/80 and alpha-smooth muscle actin in the interstitium was clearly detected in the Ang II-infused wild and GC-A-KO groups and was associated with renal tubular atrophy and interstitial fibrosis. The expression of E-cadherin in tubular cells was absent in the Ang II-infused wild and GC-A-KO groups and was associated with renal tubular atrophy. CONCLUSIONS: The natriuretic peptide-GC-A system may play an inhibitory role in Ang II-induced renal tubular atrophy, interstitial fibrosis, and phenotypic transformation in renal tubular cells and fibroblasts.


Asunto(s)
Angiotensina II/farmacología , Antihipertensivos/farmacología , Hidralazina/farmacología , Túbulos Renales/patología , Receptores del Factor Natriurético Atrial/deficiencia , Vasoconstrictores/farmacología , Actinas/análisis , Animales , Antígenos de Diferenciación/análisis , Antígenos de Diferenciación/genética , Atrofia , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/genética , Cadherinas/análisis , Quimiocina CCL2/análisis , Fibrosis , Expresión Génica/efectos de los fármacos , Péptidos y Proteínas de Señalización Intracelular/genética , Túbulos Renales/química , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Osteopontina/genética , ARN Mensajero/metabolismo , Receptores del Factor Natriurético Atrial/genética , Cloruro de Sodio/farmacología
17.
Peptides ; 56: 156-62, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24747279

RESUMEN

Both hexarelin and its natural analog ghrelin exert comparable cardioprotective activities. A single dose of ghrelin administered at the very acute phase after experimental myocardial infarction positively affects cardiac function in chronic heart failure. Therefore, this study aimed to determine whether a single dose of oral hexarelin has the same effect in the chronic disease phase. Myocardial infarction or sham operation was generated by left coronary artery ligation in male C57BL/6J mice, which subsequently received one dose of hexarelin or vehicle treatment by oral gavage 30 min after operation. Although the mortality within 14 days after myocardial infarction did not differ between the groups, hexarelin treatment protected cardiac function in the chronic phase as evidenced by higher ejection fraction and fractional shortening, as well as lower lung weight/body weight and lung weight/tibial length ratios, compared with vehicle treatment. Hexarelin treatment concurrently lowered plasma epinephrine and dopamine levels, and shifted the balance of autonomic nervous activity toward parasympathetic nervous activity as evidenced by a smaller low/high-frequency power ratio and larger normalized high-frequency power on heart rate variability analysis. The results first demonstrate that one dose of oral hexarelin treatment potentially protects chronic cardiac function after acute myocardial infarction, and implicate that activating growth hormone secretagogue receptor 1a might be beneficial for cardioprotection, although other mechanism may also be involved.


Asunto(s)
Infarto del Miocardio/tratamiento farmacológico , Oligopéptidos/uso terapéutico , Administración Oral , Animales , Dopamina/sangre , Epinefrina/sangre , Frecuencia Cardíaca/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Norepinefrina/sangre , Oligopéptidos/administración & dosificación
18.
PLoS One ; 9(4): e95771, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24752134

RESUMEN

Cardiac left ventricle hypertrophy (LVH) constitutes a major risk factor for heart failure. Although LVH is most commonly caused by chronic elevation in arterial blood pressure, reduction of blood pressure to normal levels does not always result in regression of LVH, suggesting that additional factors contribute to the development of this pathology. We tested whether genetic preconditions associated with the imbalance in sodium homeostasis could trigger the development of LVH without concomitant increases in blood pressure. The results showed that the presence of a hypertensive variant of α-adducin gene in Milan rats (before they become hypertensive) resulted in elevated expression of genes associated with LVH, and of salt-inducible kinase 2 (SIK2) in the left ventricle (LV). Moreover, the mRNA expression levels of SIK2, α-adducin, and several markers of cardiac hypertrophy were positively correlated in tissue biopsies obtained from human hearts. In addition, we found in cardiac myocytes that α-adducin regulates the expression of SIK2, which in turn mediates the effects of adducin on hypertrophy markers gene activation. Furthermore, evidence that SIK2 is critical for the development of LVH in response to chronic high salt diet (HS) was obtained in mice with ablation of the sik2 gene. Increases in the expression of genes associated with LVH, as well as increases in LV wall thickness upon HS, occurred only in sik2+/+ but not in sik2-/- mice. Thus LVH triggered by HS or the presence of a genetic variant of α-adducin requires SIK2 and is independent of elevated blood pressure. Inhibitors of SIK2 may constitute part of a novel therapeutic regimen aimed at prevention/regression of LVH.


Asunto(s)
Cardiomegalia/prevención & control , Hipertrofia Ventricular Izquierda/prevención & control , Proteínas Serina-Treonina Quinasas/metabolismo , Cloruro de Sodio Dietético/farmacología , Animales , Presión Sanguínea/efectos de los fármacos , Cardiomegalia/enzimología , Humanos , Hipertrofia Ventricular Izquierda/enzimología , Inmunohistoquímica , Técnicas In Vitro , Masculino , Ratones , Ratones Noqueados , Proteínas Serina-Treonina Quinasas/genética , Ratas
19.
PLoS One ; 9(2): e88319, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24551087

RESUMEN

Although mesenchymal stem cells (MSCs) can be obtained from the fetal membrane (FM), little information is available regarding biological differences in MSCs derived from different layers of the FM or their therapeutic potential. Isolated MSCs from both amnion and chorion layers of FM showed similar morphological appearance, multipotency, and cell-surface antigen expression. Conditioned media obtained from amnion- and chorion-derived MSCs inhibited cell death caused by serum starvation or hypoxia in endothelial cells and cardiomyocytes. Amnion and chorion MSCs secreted significant amounts of angiogenic factors including HGF, IGF-1, VEGF, and bFGF, although differences in the cellular expression profile of these soluble factors were observed. Transplantation of human amnion or chorion MSCs significantly increased blood flow and capillary density in a murine hindlimb ischemia model. In addition, compared to human chorion MSCs, human amnion MSCs markedly reduced T-lymphocyte proliferation with the enhanced secretion of PGE2, and improved the pathological situation of a mouse model of acute graft-versus-host disease. Our results highlight that human amnion- and chorion-derived MSCs, which showed differences in their soluble factor secretion and angiogenic/immuno-suppressive function, could be ideal cell sources for regenerative medicine.


Asunto(s)
Amnios/citología , Corion/citología , Citoprotección , Terapia de Inmunosupresión , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Neovascularización Fisiológica , Animales , Femenino , Miembro Posterior/irrigación sanguínea , Miembro Posterior/patología , Células Endoteliales de la Vena Umbilical Humana/citología , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Isquemia/terapia , Masculino , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Desnudos , Miocitos Cardíacos/citología
20.
Cell Transplant ; 23(7): 889-99, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-23562186

RESUMEN

Mesenchymal stem cells (MSCs) are an attractive therapeutic cell source for treating renal diseases. MSC administration has been shown to improve renal function, although the underlying mechanisms are not completely understood. We recently showed that allogenic fetal membrane-derived MSCs (FM-MSCs), which are available noninvasively in large amounts, had a renoprotective effect in an experimental glomerulonephritis model. Here we investigated whether allogenic FM-MSC administration could protect kidneys from ischemia/reperfusion (I/R) injury. Lewis rats were subjected to right nephrectomy and left renal I/R injury by clamping the left renal artery as an acute kidney injury (AKI) model. After declamping, FM-MSCs (5 × 10(5) cells) obtained from major histocompatibility complex (MHC)-mismatched ACI rats were intravenously administered. I/R-injured rats exhibited increased serum creatinine and BUN, whereas FM-MSC administration significantly ameliorated renal function. Histological analysis revealed that FM-MSC administration significantly suppressed tubular apoptosis and infiltration of macrophages and T-cells. Administration of FM-MSCs mainly homed into the lung, but increased serum IL-10 levels. Of interest is that renoprotective effects of FM-MSCs were abolished by using anti-IL-10 neutralization antibody, suggesting that IL-10 would be one of the candidate factors to protect rat kidney from I/R injury in this model. We concluded that allogenic FM-MSC transplantation is a potent therapeutic strategy for the treatment of AKI.


Asunto(s)
Lesión Renal Aguda/prevención & control , Membranas Extraembrionarias/citología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Lesión Renal Aguda/etiología , Lesión Renal Aguda/patología , Animales , Anticuerpos Neutralizantes/inmunología , Nitrógeno de la Urea Sanguínea , Creatinina/sangre , Femenino , Interleucina-10/sangre , Interleucina-10/inmunología , Interleucina-10/metabolismo , Riñón/fisiopatología , Macrófagos/inmunología , Macrófagos/fisiología , Masculino , Ratas , Ratas Endogámicas ACI , Ratas Endogámicas Lew , Daño por Reperfusión/complicaciones , Daño por Reperfusión/patología , Linfocitos T/inmunología , Linfocitos T/fisiología , Trasplante Homólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA