Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Hypertens Res ; 46(5): 1210-1220, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36788301

RESUMEN

Angiotensin converting enzyme 2 (ACE2) functions as an enzyme that produces angiotensin 1-7 (A1-7) from angiotensin II (AII) in the renin-angiotensin system (RAS). We evaluated aging phenotypes, especially skeletal muscle aging, in ACE2 systemically deficient (ACE2 KO) mice and found that ACE2 has an antiaging function. The characteristic aging phenotype observed in ACE2 KO mice was not reproduced in mice deficient in the A1-7 receptor Mas or in Tsukuba hypertensive mice, a model of chronic AII overproduction, suggesting that ACE2 has a RAS-independent antiaging function. In this review, the results we have obtained and related studies on the aging regulatory mechanism mediated by RAS components will be presented and summarized. We evaluated the aging phenotype of ACE2 systemically deficient (ACE2 KO) mice, particularly skeletal muscle aging, and found that ACE2 has an antiaging function. The characteristic aging phenotype observed in ACE2 KO mice was not reproduced in Mas KO mice, angiotensin 1-7 receptor-deficient mice or in Tsukuba hypertensive mice, a model of chronic angiotensin II overproduction, suggesting that the antiaging functions of ACE2 are independent of the renin-angiotensin system (RAS).


Asunto(s)
Angiotensina II , Sistema Renina-Angiotensina , Ratones , Animales , Angiotensina II/farmacología , Angiotensina II/metabolismo , Enzima Convertidora de Angiotensina 2 , Peptidil-Dipeptidasa A/genética , Peptidil-Dipeptidasa A/metabolismo , Envejecimiento , Angiotensina I/metabolismo
2.
FASEB J ; 36(10): e22555, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36125010

RESUMEN

Although α-synuclein (SNCA) is a well-known pathological molecule involved in synucleinopathy in neurons, its physiological roles remain largely unknown. We reported that serum SNCA levels have a close inverse correlation with blood pressure and age, which indicates the involvement of SNCA in age-related endothelial dysfunction. Therefore, this study aimed to elucidate the molecular functions of SNCA in the endothelium. We confirmed that SNCA was expressed in and secreted from endothelial cells (ECs). Exogenous treatment with recombinant SNCA (rSNCA) activated the Akt-eNOS axis and increased nitric oxide production in ECs. Treatment with rSNCA also suppressed TNF-α- and palmitic acid-induced NF-κB activation, leading to the suppression of VCAM-1 upregulation and restoration of eNOS downregulation in ECs. As for endogenous SNCA expression, replicative senescence resulted in the attenuation of SNCA expression in cultured ECs, similar to the effects of physiological aging on mice aortas. The siRNA-mediated silencing of SNCA consistently resulted in senescent phenotypes, such as eNOS downregulation, increased ß-gal activity, decreased Sirt1 expression, and increased p53 expression, in ECs. Ex vivo assessment of endothelial functions using aortic rings revealed impaired endothelium-dependent acetylcholine-induced relaxation in SNCA knockout (KO) mice. Furthermore, SNCA KO mice, especially those on a high-fat diet, displayed elevated blood pressure compared with wild-type mice; this could be eNOS dysfunction-dependent because of the lower difference caused by L-NAME administration. These results indicate that exogenous and endogenous SNCA in ECs might physiologically maintain vascular integrity, and age-related endothelial dysfunction might be partially ascribed to loss-of-function of SNCA in ECs.


Asunto(s)
Enfermedades Vasculares , alfa-Sinucleína/metabolismo , Acetilcolina/metabolismo , Animales , Células Endoteliales/metabolismo , Ratones , Ratones Noqueados , FN-kappa B/metabolismo , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico/metabolismo , Ácido Palmítico/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño/metabolismo , Sirtuina 1/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Molécula 1 de Adhesión Celular Vascular/metabolismo , Enfermedades Vasculares/metabolismo
3.
Front Nutr ; 9: 868845, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35463998

RESUMEN

The severity of coronavirus disease 2019 (COVID-19) is characterized by systemic damage to organs, including skeletal muscle, due to excessive secretion of inflammatory cytokines. Clinical studies have suggested that the kynurenine pathway of tryptophan metabolism is selectively enhanced in patients with severe COVID-19. In addition to acting as a receptor for severe acute respiratory syndrome coronavirus 2, the causative virus of COVID-19, angiotensin converting enzyme 2 (ACE2) contributes to tryptophan absorption and inhibition of the renin-angiotensin system. In this article, we review previous studies to assess the potential for a link between tryptophan metabolism, ACE2, and skeletal muscle damage in patients with COVID-19.

4.
J Am Heart Assoc ; 10(13): e021030, 2021 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-34212761

RESUMEN

Background The activation of AT2 (angiotensin II type 2 receptor ) and Mas receptor by angiotensin II and angiotensin-(1-7), respectively, is the primary process that counteracts activation of the canonical renin-angiotensin system (RAS). Although inhibition of canonical RAS could delay the progression of physiological aging, we recently reported that deletion of Mas had no impact on the aging process in mice. Here, we used male mice with a deletion of only AT2 or a double deletion of AT2 and Mas to clarify whether these receptors contribute to the aging process in a complementary manner, primarily by focusing on aging-related muscle weakness. Methods and Results Serial changes in grip strength of these mice up to 24 months of age showed that AT2/Mas knockout mice, but not AT2 knockout mice, had significantly weaker grip strength than wild-type mice from the age of 18 months. AT2/Mas knockout mice exhibited larger sizes, but smaller numbers and increased frequency of central nucleation (a marker of aged muscle) of single skeletal muscle fibers than AT2 knockout mice. Canonical RAS-associated genes, inflammation-associated genes, and senescence-associated genes were highly expressed in skeletal muscles of AT2/Mas knockout mice. Muscle angiotensin II content increased in AT2/Mas knockout mice. Conclusions Double deletion of AT2 and Mas in mice exaggerated aging-associated muscle weakness, accompanied by signatures of activated RAS, inflammation, and aging in skeletal muscles. Because aging-associated phenotypes were absent in single deletions of the receptors, AT2 and Mas could complement each other in preventing local activation of RAS during aging.


Asunto(s)
Fuerza Muscular , Debilidad Muscular/fisiopatología , Músculo Esquelético/fisiopatología , Proteínas Proto-Oncogénicas/deficiencia , Receptor de Angiotensina Tipo 2/deficiencia , Receptores Acoplados a Proteínas G/deficiencia , Factores de Edad , Animales , Fibrosis , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Fuerza de la Mano , Mediadores de Inflamación/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Fuerza Muscular/genética , Debilidad Muscular/genética , Debilidad Muscular/metabolismo , Debilidad Muscular/patología , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Fenotipo , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas/genética , Receptor de Angiotensina Tipo 2/genética , Receptores Acoplados a Proteínas G/genética , Sistema Renina-Angiotensina/genética
5.
Sci Rep ; 11(1): 5759, 2021 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-33707701

RESUMEN

The receptor for advanced glycation end-products (RAGE) and the G protein-coupled angiotensin II (AngII) type I receptor (AT1) play a central role in cardiovascular diseases. It was recently reported that RAGE modifies AngII-mediated AT1 activation via the membrane oligomeric complex of the two receptors. In this study, we investigated the presence of the different directional crosstalk in this phenomenon, that is, the RAGE/AT1 complex plays a role in the signal transduction pathway of RAGE ligands. We generated Chinese hamster ovary (CHO) cells stably expressing RAGE and AT1, mutated AT1, or AT2 receptor. The activation of two types of G protein α-subunit, Gq and Gi, was estimated through the accumulation of inositol monophosphate and the inhibition of forskolin-induced cAMP production, respectively. Rat kidney epithelial cells were used to assess RAGE ligand-induced cellular responses. We determined that RAGE ligands activated Gi, but not Gq, only in cells expressing RAGE and wildtype AT1. The activation was inhibited by an AT1 blocker (ARB) as well as a RAGE inhibitor. ARBs inhibited RAGE ligand-induced ERK phosphorylation, NF-κB activation, and epithelial-mesenchymal transition of rat renal epithelial cells. Our findings suggest that the activation of AT1 plays a central role in RAGE-mediated cellular responses and elucidate the role of a novel molecular mechanism in the development of cardiovascular diseases.


Asunto(s)
Membrana Celular/metabolismo , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Animales , Células CHO , Cricetulus , Transición Epitelial-Mesenquimal , Proteínas de Unión al GTP/metabolismo , Productos Finales de Glicación Avanzada/metabolismo , Humanos , Ligandos , Unión Proteica , Ratas , Albúmina Sérica Bovina/metabolismo , Transducción de Señal , Transgenes
6.
iScience ; 24(2): 102076, 2021 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-33659870

RESUMEN

Arrestin-dependent activation of a G-protein-coupled receptor (GPCR) triggers endocytotic internalization of the receptor complex. We analyzed the interaction between the pattern recognition receptor (PRR) lectin-like oxidized low-density lipoprotein (oxLDL) receptor (LOX-1) and the GPCR angiotensin II type 1 receptor (AT1) to report a hitherto unidentified mechanism whereby internalization of the GPCR mediates cellular endocytosis of the PRR ligand. Using genetically modified Chinese hamster ovary cells, we found that oxLDL activates Gαi but not the Gαq pathway of AT1 in the presence of LOX-1. Endocytosis of the oxLDL-LOX-1 complex through the AT1-ß-arrestin pathway was demonstrated by real-time imaging of the membrane dynamics of LOX-1 and visualization of endocytosis of oxLDL. Finally, this endocytotic pathway involving GPCR kinases (GRKs), ß-arrestin, and clathrin is relevant in accumulating oxLDL in human vascular endothelial cells. Together, our findings indicate that oxLDL activates selective G proteins and ß-arrestin-dependent internalization of AT1, whereby the oxLDL-LOX-1 complex undergoes endocytosis.

7.
FASEB J ; 35(3): e21419, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33566370

RESUMEN

In the early phase of the Coronavirus disease 2019 (COVID-19) pandemic, it was postulated that the renin-angiotensin-system inhibitors (RASi) increase the infection risk. This was primarily based on numerous reports, which stated that the RASi could increase the organ Angiotensin-converting enzyme 2 (ACE2), the receptor of Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), in rodents. RASi can theoretically antagonize the potential influence of angiotensin II (Ang II) on ACE2. However, while Ang II decreases the ACE2 levels in cultured cells, there is little evidence that supports this phenomenon in living animals. In this study, we tested whether Ang II or Ang II combined with its antagonist would alter the ACE2 and other molecules associated with the infection of SARS-CoV-2. Male C57BL6/J mice were administered vehicle, Ang II (400 ng/kg/min), or Ang II with losartan (10 mg/kg/min) for 2 weeks. ACE2 knockout mice were used as a negative control for the ACE2 assay. We found that both Ang II, which elevated blood pressure by 30 mm Hg, and Ang II with losartan, had no effect on the expression or protein activity of ACE2 in the lung, left ventricle, kidney, and ileum. Likewise, these interventions had no effect on the expression of Transmembrane Protease Serine 2 (TMPRSS2) and Furin, proteases that facilitate the virus-cell fusion, and the expression or activity of Tumor Necrosis Factor α-Convertase (TACE) that cleaves cell-surface ACE2. Collectively, physiological concentrations of Ang II do not modulate the molecules associated with SARS-CoV-2 infection. These results support the recent observational studies suggesting that the use of RASi is not a risk factor for COVID-19.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Angiotensina II/farmacología , Enzima Convertidora de Angiotensina 2/metabolismo , COVID-19/metabolismo , Losartán/farmacología , SARS-CoV-2 , Proteína ADAM17/genética , Proteína ADAM17/metabolismo , Angiotensina II/administración & dosificación , Bloqueadores del Receptor Tipo 1 de Angiotensina II/administración & dosificación , Enzima Convertidora de Angiotensina 2/genética , Animales , Furina/genética , Furina/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Losartán/administración & dosificación , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Vasoconstrictores/farmacología
8.
Clin Sci (Lond) ; 134(22): 3047-3062, 2020 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-33231620

RESUMEN

Angiotensin converting enzyme-2 (ACE2) is a multifunctional transmembrane protein recently recognised as the entry receptor of the virus causing COVID-19. In the renin-angiotensin system (RAS), ACE2 cleaves angiotensin II (Ang II) into angiotensin 1-7 (Ang 1-7), which is considered to exert cellular responses to counteract the activation of the RAS primarily through a receptor, Mas, in multiple organs including skeletal muscle. Previous studies have provided abundant evidence suggesting that Ang 1-7 modulates multiple signalling pathways leading to protection from pathological muscle remodelling and muscle insulin resistance. In contrast, there is relatively little evidence to support the protective role of ACE2 in skeletal muscle. The potential contribution of endogenous ACE2 to the regulation of Ang 1-7-mediated protection of these muscle pathologies is discussed in this review. Recent studies have suggested that ACE2 protects against ageing-associated muscle wasting (sarcopenia) through its function to modulate molecules outside of the RAS. Thus, the potential association of sarcopenia with ACE2 and the associated molecules outside of RAS is also presented herein. Further, we introduce the transcriptional regulation of muscle ACE2 by drugs or exercise, and briefly discuss the potential role of ACE2 in the development of COVID-19.


Asunto(s)
Angiotensina I/metabolismo , COVID-19/metabolismo , Músculo Esquelético/enzimología , Fragmentos de Péptidos/metabolismo , Angiotensina II/metabolismo , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/metabolismo , Animales , COVID-19/enzimología , COVID-19/genética , Humanos , SARS-CoV-2/fisiología
9.
Nat Commun ; 11(1): 4150, 2020 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-32811819

RESUMEN

The systemic decline in autophagic activity with age impairs homeostasis in several tissues, leading to age-related diseases. A mechanistic understanding of adipocyte dysfunction with age could help to prevent age-related metabolic disorders, but the role of autophagy in aged adipocytes remains unclear. Here we show that, in contrast to other tissues, aged adipocytes upregulate autophagy due to a decline in the levels of Rubicon, a negative regulator of autophagy. Rubicon knockout in adipocytes causes fat atrophy and hepatic lipid accumulation due to reductions in the expression of adipogenic genes, which can be recovered by activation of PPARγ. SRC-1 and TIF2, coactivators of PPARγ, are degraded by autophagy in a manner that depends on their binding to GABARAP family proteins, and are significantly downregulated in Rubicon-ablated or aged adipocytes. Hence, we propose that age-dependent decline in adipose Rubicon exacerbates metabolic disorders by promoting excess autophagic degradation of SRC-1 and TIF2.


Asunto(s)
Adipocitos/metabolismo , Envejecimiento/fisiología , Autofagia/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Enfermedades Metabólicas/metabolismo , Adipocitos/patología , Adipogénesis/genética , Tejido Adiposo/citología , Tejido Adiposo/metabolismo , Adiposidad/genética , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Autofagia/fisiología , Hígado Graso/genética , Hígado Graso/metabolismo , Técnicas de Inactivación de Genes , Glucosa/genética , Glucosa/metabolismo , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Metabolismo de los Lípidos/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/metabolismo , Coactivador 1 de Receptor Nuclear/metabolismo , Coactivador 2 del Receptor Nuclear/metabolismo , PPAR gamma/metabolismo
10.
FASEB J ; 34(2): 2792-2811, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31912559

RESUMEN

While adipose tissue is required to maintain glucose metabolism, excessive calorie intake induces obesity via mechanisms including accelerated proliferation and differentiation of preadipocytes, leading to insulin resistance. Here, we investigated the role of myoferlin (MYOF), a ferlin family protein, in regulating glucose metabolism by mainly focusing on its unknown role in adipose tissue. Whereas young MYOF knockout (KO) mice on a normal diet showed aggravated glucose tolerance and insulin sensitivity, those on a high-fat diet (HFD) showed preserved glucose tolerance with an attenuated gain of body weight, reduced visceral fat deposits, and less severe fatty liver. The Adipose MYOF expression was reduced by aging but was restored by an HFD along with the retained expression of NFAT transcription factors. Loss-of-function of MYOF in preadipocytes suppressed proliferation and differentiation into mature adipocytes along with the decreased expression of genes involved in adipogenesis. The MYOF expression in preadipocytes was reduced with differentiation. Attenuated obesity in MYOF KO mice on an HFD was also accompanied with increased oxygen consumption by an unidentified mechanism and with reduced adipose inflammation due to less inflammatory macrophages. These insights suggest that the multifunctional roles of MYOF involve the regulation of preadipocyte function and affect glucose metabolism bidirectionally depending on consumed calories.


Asunto(s)
Adipocitos/metabolismo , Adipogénesis/fisiología , Adiposidad/fisiología , Glucosa/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Musculares/metabolismo , Tejido Adiposo Pardo/metabolismo , Animales , Diferenciación Celular , Inflamación/metabolismo , Resistencia a la Insulina/fisiología , Macrófagos/metabolismo , Masculino , Ratones Endogámicos C57BL
11.
Hypertens Res ; 43(4): 296-304, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31853045

RESUMEN

Inhibition of the renin-angiotensin system (RAS) has been shown to alleviate muscle atrophy both under pathological conditions and during physiological aging. We recently reported that the deletion of angiotensin converting enzyme 2 (ACE2), which converts Angiotensin II to Angiotensin-(1-7) in mice, leads to the early manifestation of aging-associated muscle weakness along with the increased expression of p16INK4a, a senescence-associated gene, and increased central nuclei in the tibialis anterior (TA) muscle in middle age. As ACE2 is multifunctional and functions beyond its role in the RAS, we investigated whether activation of the RAS primarily contributes to muscle weakness in ACE2 knockout (KO) mice by comparing these mice to Tsukuba hypertensive (TH) mice that overproduce human angiotensin II. The grip strength of young (6 months) and middle-aged (15 months) TH mice was consistently lower than that of wild-type mice at the same ages. Middle-aged TH mice were continuously lean with extremely reduced adiposity. Central nuclei in the gastrocnemius (GM) muscle were increased in ACE2KO mice, while no apparent morphological change was observed in the GM muscles of TH mice. Increased expression of p16INK4a along with alterations in the expression of several sarcopenia-associated genes were observed in the GM muscles of ACE2KO mice but not TH mice. These findings suggest that chronic overactivation of the RAS does not primarily contribute to the early aging phenotypes of skeletal muscle in ACE2KO mice.


Asunto(s)
Enzima Convertidora de Angiotensina 2/genética , Fuerza de la Mano/fisiología , Debilidad Muscular/genética , Sistema Renina-Angiotensina/fisiología , Adiposidad/fisiología , Envejecimiento/fisiología , Enzima Convertidora de Angiotensina 2/metabolismo , Animales , Hipertensión/genética , Hipertensión/metabolismo , Hipertensión/fisiopatología , Ratones , Ratones Noqueados , Debilidad Muscular/metabolismo , Debilidad Muscular/fisiopatología , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatología
12.
Clin Sci (Lond) ; 133(18): 2005-2018, 2019 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-31519791

RESUMEN

The angiotensin-converting enzyme 2 (ACE2)-angiotensin 1-7 (A1-7)-A1-7 receptor (Mas) axis plays a protective role in the renin-angiotensin system (RAS). We recently found that ACE2 knockout (ACE2KO) mice exhibit earlier aging-associated muscle weakness, and that A1-7 alleviates muscle weakness in aging mice. In the present study, we investigated the role of the A1-7-Mas pathway in the effect of ACE2 on physiological aging. Male wild-type, ACE2KO, and Mas knockout (MasKO) mice were subjected to periodical grip strength measurement, followed by administration of A1-7 or vehicle for 4 weeks at 24 months of age. ACE2KO mice exhibited decreased grip strength after 6 months of age, while grip strength of MasKO mice was similar to that of wild-type mice. A1-7 improved grip strength in ACE2KO and wild-type mice, but not in MasKO mice. Muscle fibre size was smaller in ACE2KO mice than that in wild-type and MasKO mice, and increased with A1-7 in ACE2KO and WT mice, but not in MasKO mice. Centrally nucleated fibres (CNFs) and expression of the senescence-associated gene p16INK4a in skeletal muscles were enhanced only in ACE2KO mice and were not altered by A1-7. ACE2KO mice, but not MasKO mice, exhibited thinning of peripheral fat along with increased adipose expression of p16INK4a A1-7 significantly increased bone volume in wild-type and ACE2KO mice, but not in MasKO mice. Our findings suggest that the impact of ACE2 on physiological aging does not depend on the endogenous production of A1-7 by ACE2, while overactivation of the A1-7-Mas pathway could alleviate sarcopenia and osteoporosis in aged mice.


Asunto(s)
Envejecimiento/patología , Angiotensina I/uso terapéutico , Resorción Ósea/tratamiento farmacológico , Debilidad Muscular/tratamiento farmacológico , Fragmentos de Péptidos/uso terapéutico , Peptidil-Dipeptidasa A/deficiencia , Tejido Adiposo/patología , Angiotensina I/farmacología , Enzima Convertidora de Angiotensina 2 , Animales , Peso Corporal/efectos de los fármacos , Resorción Ósea/complicaciones , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Miembro Anterior/fisiopatología , Eliminación de Gen , Fuerza de la Mano , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Debilidad Muscular/complicaciones , Debilidad Muscular/diagnóstico por imagen , Músculos/diagnóstico por imagen , Músculos/efectos de los fármacos , Músculos/patología , Tamaño de los Órganos/efectos de los fármacos , Factor de Transcripción PAX3/metabolismo , Fragmentos de Péptidos/farmacología , Peptidil-Dipeptidasa A/metabolismo , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas/metabolismo , Receptores Acoplados a Proteínas G/deficiencia , Receptores Acoplados a Proteínas G/metabolismo , Sistema Renina-Angiotensina/efectos de los fármacos , Factores de Tiempo
13.
J Cardiovasc Pharmacol ; 73(5): 282-289, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30829732

RESUMEN

BACKGROUND: Sacubitril/valsartan was shown to attenuate the development of cardiac hypertrophy with enhanced blood pressure reduction compared with valsartan alone in animal models. We investigated whether a low-dose sacubitril/valsartan has blood pressure-independent effects on cardiac hypertrophy and pulmonary edema using a rat model of hypertension and obesity. METHODS AND RESULTS: In plan 1, male SHR/NDmcr-cp rats fed normal or phase-increased high salt were treated with vehicle, 6-mg/kg sacubitril/valsartan or 3-mg/kg valsartan, for 6 months. In plan 2, after high-salt loading for 6 months, drugs were administered for 4 months. Antihypertensive effects of the 2 drugs were similar during all study periods. In plan 1 with normal salt, there were no differences between treatments in the left ventricle weight/body weight (BW), or lung weight/BW as an index of cardiac hypertrophy or pulmonary edema, respectively. These indexes were smaller in high-salt-fed rats with sacubitril/valsartan than vehicle. In plan 2, both indexes did not differ between vehicle and sacubitril/valsartan. Ventricle weight/BW was lower in valsartan than sacubitril/valsartan. In plan 2, gene markers of cardiac dysfunction were upregulated by sacubitril/valsartan compared with the other groups. CONCLUSIONS: Low-dose sacubitril/valsartan may have different effects depending on the stage of cardiac hypertrophy in rats.


Asunto(s)
Aminobutiratos/administración & dosificación , Bloqueadores del Receptor Tipo 1 de Angiotensina II/administración & dosificación , Cardiomegalia/prevención & control , Inhibidores de Proteasas/administración & dosificación , Cloruro de Sodio Dietético , Tetrazoles/administración & dosificación , Animales , Biomarcadores/sangre , Biomarcadores/orina , Compuestos de Bifenilo , Presión Sanguínea/efectos de los fármacos , Cardiomegalia/metabolismo , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Modelos Animales de Enfermedad , Combinación de Medicamentos , Regulación de la Expresión Génica , Riñón/fisiopatología , Masculino , Neprilisina/antagonistas & inhibidores , Edema Pulmonar/metabolismo , Edema Pulmonar/patología , Edema Pulmonar/fisiopatología , Edema Pulmonar/prevención & control , Ratas Endogámicas SHR , Valsartán
14.
Biochem Biophys Res Commun ; 508(4): 1168-1174, 2019 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-30554661

RESUMEN

Cardiovascular disease is one of the leading causes of death in the elderly, and novel therapeutic targets against atherogenesis are urgent. The initiation of atherosclerotic changes of monocyte adhesion on the vascular endothelium and subsequent foam cell formation are noteworthy pathophysiologies when searching for strategies to prevent the progression of age-related atherosclerosis. We report the significance of the deubiquitinating enzyme cylindromatosis (CYLD) in vascular remodeling by interference with inflammatory responses regulated by NF-κB signaling. The purpose of this study was to elucidate the pathological functions of CYLD in the early phase of atherogenesis associated with aging. Treatment with inflammatory cytokines induced endogenous CYLD in aortic endothelial cells (HAECs) and THP-1 cells. siRNA-mediated CYLD silencing led to enhanced monocyte adhesion along with increased adhesion molecules in HAECs treated with TNFα. In siRNA-mediated CYLD silenced RAW 264.7 macrophages treated with oxidized LDL (oxLDL), augmented lipid accumulation was observed, along with increased expression of the class A macrophage scavenger receptor (SR-A), lectin-like oxidized LDL receptor-1 (LOX-1), CD36, fatty acid binding protein 4 (FABP4), the cholesterol ester synthase acyl-CoA cholesterol acyltransferase (ACAT1), MCP-1, and IL-1ß and decreased expression of scavenger receptor class B type I (SR-BI). Intriguingly, CYLD gene expression was significantly reduced in bone marrow-derived macrophages of aged mice compared that of young mice, as well as in senescent HAECs compared with young cells. These findings suggest that age-related attenuation of CYLD expression in endothelial cells (ECs) and macrophages triggers the initiation of age-related atherogenesis by exacerbating monocyte adhesion on the endothelium and foam cell formation. CYLD in the vasculature may be a novel therapeutic target, especially in the early preventive intervention against the initiation of age-related atherogenesis.


Asunto(s)
Envejecimiento/patología , Aterosclerosis/fisiopatología , Cisteína Endopeptidasas/metabolismo , Enzima Desubiquitinante CYLD/metabolismo , Endotelio Vascular/metabolismo , Endotelio Vascular/fisiopatología , Macrófagos/metabolismo , Envejecimiento/genética , Animales , Aterosclerosis/genética , Aterosclerosis/patología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Adhesión Celular/efectos de los fármacos , Citocinas/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Endotelio Vascular/efectos de los fármacos , Células Espumosas/efectos de los fármacos , Células Espumosas/metabolismo , Silenciador del Gen/efectos de los fármacos , Humanos , Mediadores de Inflamación/metabolismo , Lipoproteínas LDL/farmacología , Macrófagos/efectos de los fármacos , Masculino , Ratones , Células RAW 264.7 , ARN Interferente Pequeño/metabolismo , Células THP-1 , Regulación hacia Arriba/efectos de los fármacos
15.
J Cachexia Sarcopenia Muscle ; 9(5): 975-986, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30207087

RESUMEN

BACKGROUND: A pharmacologic strategy for age-related muscle weakness is desired to improve mortality and disability in the elderly. Angiotensin-converting enzyme 2 (ACE2) cleaves angiotensin II into angiotensin 1-7, a peptide known to protect against acute and chronic skeletal muscle injury in rodents. Since physiological aging induces muscle weakness via mechanisms distinct from other muscle disorders, the role of ACE2-angiotensin 1-7 in age-related muscle weakness remains undetermined. Here, we investigated whether deletion of ACE2 alters the development of muscle weakness by aging and whether angiotensin 1-7 reverses muscle weakness in older mice. METHODS: After periodic measurement of grip strength and running distance in male ACE2KO and wild-type mice until 24 months of age, we infused angiotensin 1-7 or vehicle for 4 weeks, and measured grip strength, and excised tissues. Tissues were also excised from younger (3-month-old) and middle-aged (15-month-old) mice. Microarray analysis of RNA was performed using tibialis anterior (TA) muscles from middle-aged mice, and some genes were further tested using RT-PCR. RESULTS: Grip strength of ACE2KO mice was reduced at 6 months and was persistently lower than that of wild-type mice (p < 0.01 at 6, 12, 18, and 24-month-old). Running distance of ACE2KO mice was shorter than that of wild-type mice only at 24 months of age [371 ± 26 vs. 479 ± 24 (m), p < 0.01]. Angiotensin 1-7 improved grip strength in both types of older mice, with larger effects observed in ACE2KO mice (% increase, 3.8 ± 1.5 and 13.3 ± 3.1 in wild type and ACE2KO mice, respectively). Older, but not middle-aged ACE2KO mice had higher oxygen consumption assessed by a metabolic cage than age-matched wild-type mice. Angiotensin 1-7 infusion modestly increased oxygen consumption in older mice. There was no difference in a wheel-running activity or glucose tolerance between ACE2KO and wild-type mice and between mice with vehicle and angiotensin 1-7 infusion. Analysis of TA muscles revealed that p16INK4a, a senescence-associated gene, and central nuclei of myofibers increased in middle-aged, but not younger ACE2KO mice. p16INK4a and central nuclei increased in TA muscles of older wild-type mice, but the differences between ACE2KO and wild-type mice remained significant (p < 0.01). Angiotensin 1-7 did not alter the expression of p16INK4a or central nuclei in TA muscles of both types of mice. Muscle ACE2 expression of wild-type mice was the lowest at middle age (2.6 times lower than younger age, p < 0.05). CONCLUSIONS: Deletion of ACE2 induced the early manifestation of muscle weakness with signatures of muscle senescence. Angiotensin 1-7 improved muscle function in older mice, supporting future application of the peptide or its analogues in the treatment of muscle weakness in the elderly population.


Asunto(s)
Angiotensina I/metabolismo , Debilidad Muscular/etiología , Debilidad Muscular/metabolismo , Músculo Esquelético/metabolismo , Fragmentos de Péptidos/metabolismo , Peptidil-Dipeptidasa A/deficiencia , Factores de Edad , Enzima Convertidora de Angiotensina 2 , Animales , Biomarcadores , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Prueba de Tolerancia a la Glucosa , Ratones , Ratones Noqueados , Debilidad Muscular/fisiopatología , Músculo Esquelético/fisiopatología , Consumo de Oxígeno , Condicionamiento Físico Animal , Transcriptoma
16.
Sci Rep ; 8(1): 5145, 2018 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-29572511

RESUMEN

Chronic obstructive pulmonary disease (COPD) has been recently characterized as a disease of accelerated lung aging, but the mechanism remains unclear. Tetraspanins have emerged as key players in malignancy and inflammatory diseases. Here, we found that CD9/CD81 double knockout (DKO) mice with a COPD-like phenotype progressively developed a syndrome resembling human aging, including cataracts, hair loss, and atrophy of various organs, including thymus, muscle, and testis, resulting in shorter survival than wild-type (WT) mice. Consistent with this, DNA microarray analysis of DKO mouse lungs revealed differential expression of genes involved in cell death, inflammation, and the sirtuin-1 (SIRT1) pathway. Accordingly, expression of SIRT1 was reduced in DKO mouse lungs. Importantly, siRNA knockdown of CD9 and CD81 in lung epithelial cells additively decreased SIRT1 and Foxo3a expression, but reciprocally upregulated the expression of p21 and p53, leading to reduced cell proliferation and elevated apoptosis. Furthermore, deletion of these tetraspanins increased the expression of pro-inflammatory genes and IL-8. Hence, CD9 and CD81 might coordinately prevent senescence and inflammation, partly by maintaining SIRT1 expression. Altogether, CD9/CD81 DKO mice represent a novel model for both COPD and accelerated senescence.


Asunto(s)
Envejecimiento Prematuro , Pulmón , Enfermedad Pulmonar Obstructiva Crónica , Tetraspanina 28/deficiencia , Tetraspanina 29/deficiencia , Envejecimiento Prematuro/genética , Envejecimiento Prematuro/metabolismo , Envejecimiento Prematuro/patología , Animales , Modelos Animales de Enfermedad , Proteína Forkhead Box O3/biosíntesis , Proteína Forkhead Box O3/genética , Regulación de la Expresión Génica , Humanos , Pulmón/metabolismo , Pulmón/patología , Ratones , Ratones Noqueados , Enfermedad Pulmonar Obstructiva Crónica/genética , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/patología , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología , Sirtuina 1/biosíntesis , Sirtuina 1/genética , Síndrome , Proteína p53 Supresora de Tumor/biosíntesis , Proteína p53 Supresora de Tumor/genética
17.
Sci Rep ; 7: 42323, 2017 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-28176863

RESUMEN

The conventional forelimb grip strength test is a widely used method to assess skeletal muscle function in rodents; in this study, we modified this method to improve its variability and consistency. The modified test had lower variability among trials and days than the conventional test in young C57BL6 mice, especially by improving the variabilities in male. The modified test was more sensitive than the conventional test to detect a difference in motor function between female and male mice, or between young and old male mice. When the modified test was performed on male mice during the aging process, reduction of grip strength manifested between 18 and 24 months of age at the group level and at the individual level. The modified test was similar to the conventional test in detecting skeletal muscle dysfunction in young male dystrophic mice. Thus, the modified forelimb grip strength test, with its improved validity and reliability may be an ideal substitute for the conventional method.


Asunto(s)
Envejecimiento/fisiología , Miembro Anterior/fisiopatología , Fuerza de la Mano/fisiología , Músculo Esquelético/fisiopatología , Animales , Peso Corporal , Masculino , Ratones Endogámicos C57BL , Actividad Motora/fisiología , Distrofia Muscular Animal/fisiopatología , Reproducibilidad de los Resultados
18.
FASEB J ; 29(8): 3342-56, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25877213

RESUMEN

The angiotensin II type 1 receptor (AT1) is a 7-transmembrane domain GPCR that when activated by its ligand angiotensin II, generates signaling events promoting vascular dysfunction and the development of cardiovascular disease. Here, we show that the single-transmembrane oxidized LDL (oxLDL) receptor (LOX-1) resides in proximity to AT1 on cell-surface membranes and that binding of oxLDL to LOX-1 can allosterically activate AT1-dependent signaling events. oxLDL-induced signaling events in human vascular endothelial cells were abolished by knockdown of AT1 and inhibited by AT1 blockade (ARB). oxLDL increased cytosolic G protein by 350% in Chinese hamster ovary (CHO) cells with genetically induced expression of AT1 and LOX-1, whereas little increase was observed in CHO cells expressing only LOX-1. Immunoprecipitation and in situ proximity ligation assay (PLA) assays in CHO cells revealed the presence of cell-surface complexes involving LOX-1 and AT1. Chimeric analysis showed that oxLDL-induced AT1 signaling events are mediated via interactions between the intracellular domain of LOX-1 and AT1 that activate AT1. oxLDL-induced impairment of endothelium-dependent vascular relaxation of vascular ring from mouse thoracic aorta was abolished by ARB or genetic deletion of AT1. These findings reveal a novel pathway for AT1 activation and suggest a new mechanism whereby oxLDL may be promoting risk for cardiovascular disease.


Asunto(s)
Lectinas/metabolismo , Lipoproteínas LDL/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Receptores de LDL Oxidadas/metabolismo , Animales , Células CHO , Células COS , Línea Celular , Línea Celular Tumoral , Chlorocebus aethiops , Cricetulus , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Humanos , Transducción de Señal/fisiología
19.
Diabetes ; 62(1): 223-33, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22933108

RESUMEN

ACE type 2 (ACE2) functions as a negative regulator of the renin-angiotensin system by cleaving angiotensin II (AII) into angiotensin 1-7 (A1-7). This study assessed the role of endogenous ACE2 in maintaining insulin sensitivity. Twelve-week-old male ACE2 knockout (ACE2KO) mice had normal insulin sensitivities when fed a standard diet. AII infusion or a high-fat, high-sucrose (HFHS) diet impaired glucose tolerance and insulin sensitivity more severely in ACE2KO mice than in their wild-type (WT) littermates. The strain difference in glucose tolerance was not eliminated by an AII receptor type 1 (AT1) blocker but was eradicated by A1-7 or an AT1 blocker combined with the A1-7 inhibitor (A779). The expression of GLUT4 and a transcriptional factor, myocyte enhancer factor (MEF) 2A, was dramatically reduced in the skeletal muscles of the standard diet-fed ACE2KO mice. The expression of GLUT4 and MEF2A was increased by A1-7 in ACE2KO mice and decreased by A779 in WT mice. A1-7 enhanced upregulation of MEF2A and GLUT4 during differentiation of myoblast cells. In conclusion, ACE2 protects against high-calorie diet-induced insulin resistance in mice. This mechanism may involve the transcriptional regulation of GLUT4 via an A1-7-dependent pathway.


Asunto(s)
Transportador de Glucosa de Tipo 4/genética , Resistencia a la Insulina , Peptidil-Dipeptidasa A/fisiología , Angiotensina I/farmacología , Angiotensina II/farmacología , Enzima Convertidora de Angiotensina 2 , Animales , Dieta Alta en Grasa , Carbohidratos de la Dieta/administración & dosificación , Ingestión de Energía , Glucosa/metabolismo , Intolerancia a la Glucosa , Transportador de Glucosa de Tipo 4/fisiología , Homeostasis , Factores de Transcripción MEF2 , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/metabolismo , Mioblastos/metabolismo , Factores Reguladores Miogénicos/genética , Fragmentos de Péptidos/farmacología
20.
Hypertens Res ; 36(5): 433-9, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23254392

RESUMEN

The development of atherosclerosis is associated with disturbances in mitochondrial function that impair effective adenosine triphosphate (ATP) production, increase generation of superoxide and induce subsequent apoptosis in vascular smooth muscle cells (VSMCs). As peroxisome proliferator-activated receptor gamma (PPARγ) has a potentially important role in the regulation of mitochondrial metabolism, we studied effects of the partial PPARγ agonist and angiotensin receptor blocker telmisartan, on mitochondria-related cellular responses in VSMC. In human VSMC, telmisartan increased ATP levels and activation of mitochondrial complex II, succinate dehydrogenase, reduced the release of H2O2 and attenuated H2O2-induced increases in caspase 3/7 activity, a marker of cellular apoptosis. Eprosartan, an angiotensin II receptor blocker that lacks the ability to activate PPARγ, had no effect on these mitochondria-related cellular responses in VSMC. Studies in PPARγ-deficient VSMC revealed that the effects of telmisartan on mitochondrial function were largely independent of PPARγ although the presence of PPARγ modulated effects of telmisartan on H2O2 levels. These findings demonstrate that telmisartan can have significant effects on mitochondrial metabolism in VSMC that are potentially relevant to the pathogenesis of cardiovascular disease and that involve more than just angiotensin receptor blockade and activation of PPARγ.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Bencimidazoles/farmacología , Benzoatos/farmacología , Mitocondrias/efectos de los fármacos , Mitocondrias/fisiología , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Adenosina Trifosfato/metabolismo , Animales , Apoptosis/efectos de los fármacos , Células Cultivadas , Variaciones en el Número de Copia de ADN/efectos de los fármacos , Humanos , Peróxido de Hidrógeno/metabolismo , Ratones , Ratones Noqueados , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , PPAR gamma/deficiencia , PPAR gamma/metabolismo , Telmisartán
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA