Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Arterioscler Thromb Vasc Biol ; 42(5): 632-643, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35344382

RESUMEN

BACKGROUND: Obesity increases the risk for human abdominal aortic aneurysms (AAAs) and enhances Ang II (angiotensin II)-induced AAA formation in C57BL/6J mice. Obesity is also associated with increases in perivascular fat that expresses proinflammatory markers including SAA (serum amyloid A). We previously reported that deficiency of SAA significantly reduces Ang II-induced inflammation and AAA in hyperlipidemic apoE-deficient mice. In this study. we investigated whether adipose tissue-derived SAA plays a role in Ang II-induced AAA in obese C57BL/6J mice. METHODS: The development of AAA was compared between male C57BL/6J mice (wild type), C57BL/6J mice lacking SAA1.1, SAA2.1, and SAA3 (TKO); and TKO mice harboring a doxycycline-inducible, adipocyte-specific SAA1.1 transgene (TKO-Tgfat; SAA expressed only in fat). All mice were fed an obesogenic diet and doxycycline to induce SAA transgene expression and infused with Ang II to induce AAA. RESULTS: In response to Ang II infusion, SAA expression was significantly increased in perivascular fat of obese C57BL/6J mice. Maximal luminal diameters of the abdominal aorta were determined by ultrasound before and after Ang II infusion, which indicated a significant increase in aortic luminal diameters in wild type and TKO-TGfat mice but not in TKO mice. Adipocyte-specific SAA expression was associated with MMP (matrix metalloproteinase) activity and macrophage infiltration in abdominal aortas of Ang II-infused obese mice. CONCLUSIONS: We demonstrate for the first time that SAA deficiency protects obese C57BL/6J mice from Ang II-induced AAA. SAA expression only in adipocytes is sufficient to cause AAA in obese mice infused with Ang II.


Asunto(s)
Angiotensina II , Aneurisma de la Aorta Abdominal , Adipocitos/metabolismo , Angiotensina II/farmacología , Animales , Aneurisma de la Aorta Abdominal/inducido químicamente , Aneurisma de la Aorta Abdominal/genética , Apolipoproteínas E/genética , Modelos Animales de Enfermedad , Doxiciclina/efectos adversos , Masculino , Metaloproteinasas de la Matriz , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Obesos , Obesidad/complicaciones , Proteína Amiloide A Sérica/genética , Proteína Amiloide A Sérica/metabolismo
2.
J Pharmacol Exp Ther ; 377(3): 326-335, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33707301

RESUMEN

Combined neprilysin (NEP) inhibition (sacubitril) and angiotensin type 1 receptor (AT1R) antagonism (valsartan) is used in the treatment of congestive heart failure and is gaining interest for other angiotensin II (AngII)-related cardiovascular diseases. In addition to heart failure, AngII promotes hypertension, atherosclerosis, and abdominal aortic aneurysms (AAAs). Similarly, NEP substrates or products have broad effects on the cardiovascular system. In this study, we examined NEP inhibition (with sacubitril) and AT1R antagonism (with valsartan) alone or in combination on AngII-induced hypertension, atherosclerosis, or AAAs in male low-density lipoprotein receptor-deficient mice. Preliminary studies assessed drug delivery via osmotic minipumps for simultaneous release of sacubitril and/or valsartan with AngII over 28 days. Mice were infused with AngII (1000 ng/kg per minute) in the absence (vehicle) or presence of sacubitril (1, 6, or 9 mg/kg per day), valsartan (0.3, 0.5, 1, 6, or 20 mg/kg per day), or the combination thereof (1 and 0.3, or 9 or 0.5 mg/kg per day of sacubitril and valsartan, respectively). Plasma AngII and renin concentrations increased 4-fold at higher valsartan doses, indicative of removal of AngII negative feedback on renin. Sacubitril doubled plasma AngII concentrations at lower doses (1 mg/kg per day). Valsartan dose-dependently decreased systolic blood pressure, aortic atherosclerosis, and AAAs of AngII-infused mice, whereas sacubitril had no effect on atherosclerosis or AAAs but reduced blood pressure of AngII-infused mice. Combination therapy with sacubitril and valsartan did not provide additive benefits. These results suggest limited effects of combination therapy with NEP inhibition and AT1R antagonism against AngII-induced hypertension, atherosclerosis, or AAAs. SIGNIFICANCE STATEMENT: The combination of valsartan (angiotensin type 1 receptor antagonist) and sacubitril (neprilysin inhibitor) did not provide benefit above valsartan alone on AngII-induced hypertension, atherosclerosis, or abdominal aortic aneurysms in low-density lipoprotein receptor-deficient male mice. These results do not support this drug combination in therapy of these AngII-induced cardiovascular diseases.


Asunto(s)
Antihipertensivos , Aminobutiratos , Angiotensina II , Aterosclerosis , Compuestos de Bifenilo , Neprilisina , Animales , Ratones
3.
Arterioscler Thromb Vasc Biol ; 41(1): 269-283, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33054396

RESUMEN

OBJECTIVE: Turner syndrome women (monosomy X) have high risk of aortopathies consistent with a role for sex chromosomes in disease development. We demonstrated that sex chromosomes influence regional development of Ang II (angiotensin II)-induced aortopathies in mice. In this study, we determined if the number of X chromosomes regulates regional development of Ang II-induced aortopathies. Approach and Results: We used females with varying numbers of X chromosomes (XX female mice [XXF] or XO female mice [XOF]) on an C57BL/6J (ascending aortopathies) or low-density lipoprotein receptor deficient (Ldlr-/-) background (descending and abdominal aortopathies) compared with XY males (XYM). To induce aortopathies, mice were infused with Ang II. XOF (C57BL/6J) exhibited larger percent increases in ascending aortic lumen diameters than Ang II-infused XXF or XYM. Ang II-infused XOF (Ldlr-/-) exhibited similar incidences of thoracic (XOF, 50%; XYM, 71%) and abdominal aortopathies (XOF, 83%; XYM, 71%) as XYM, which were greater than XXF (XXF, 0%). Abdominal aortic lumen diameters and maximal external diameters were similar between XOF and XYM but greater than XXF, and these effects persisted with extended Ang II infusions. Larger aortic lumen diameters, abdominal aortopathy incidence (XXF, 20%; XOF, 75%), and maximal aneurysm diameters (XXF, 1.02±0.17; XOF, 1.96±0.32 mm; P=0.027) persisted in ovariectomized Ang II-infused XOF mice. Data from RNA-seq demonstrated that X chromosome genes that escape X-inactivation (histone lysine demethylases Kdm5c and Kdm6a) exhibited lower mRNA abundance in aortas of XOF than XXF (P=0.033 and 0.024, respectively). Conversely, DNA methylation was higher in aortas of XOF than XXF (P=0.038). CONCLUSIONS: The absence of a second X chromosome promotes diffuse Ang II-induced aortopathies in females.


Asunto(s)
Angiotensina II , Aorta Abdominal/patología , Aorta Torácica/patología , Aneurisma de la Aorta Abdominal/inducido químicamente , Aneurisma de la Aorta Torácica/inducido químicamente , Síndrome de Turner/complicaciones , Animales , Aorta Abdominal/metabolismo , Aorta Torácica/metabolismo , Aneurisma de la Aorta Abdominal/genética , Aneurisma de la Aorta Abdominal/metabolismo , Aneurisma de la Aorta Abdominal/patología , Aneurisma de la Aorta Torácica/genética , Aneurisma de la Aorta Torácica/metabolismo , Aneurisma de la Aorta Torácica/patología , Metilación de ADN , Modelos Animales de Enfermedad , Femenino , Histona Demetilasas/genética , Histona Demetilasas/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Ovariectomía , Receptores de LDL/deficiencia , Receptores de LDL/genética , Índice de Severidad de la Enfermedad , Síndrome de Turner/genética
4.
Biol Sex Differ ; 10(1): 45, 2019 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-31484552

RESUMEN

BACKGROUND: Obesity increases the risk for hypertension in both sexes, but the prevalence of hypertension is lower in females than in males until menopause, despite a higher prevalence of obesity in females. We previously demonstrated that angiotensin-converting enzyme 2 (ACE2), which cleaves the vasoconstrictor, angiotensin II (AngII), to generate the vasodilator, angiotensin-(1-7) (Ang-(1-7)), contributes to sex differences in obesity-hypertension. ACE2 expression in adipose tissue was influenced by obesity in a sex-specific manner, with elevated ACE2 expression in obese female mice. Moreover, estrogen stimulated adipose ACE2 expression and reduced obesity-hypertension in females. In this study, we hypothesized that deficiency of adipocyte ACE2 contributes to obesity-hypertension of females. METHODS: We generated a mouse model of adipocyte ACE2 deficiency. Male and female mice with adipocyte ACE2 deficiency or littermate controls were fed a low (LF) or a high fat (HF) diet for 16 weeks and blood pressure was quantified by radiotelemetry. HF-fed mice of each sex and genotype were challenged by an acute AngII injection, and blood pressure response was quantified. To translate these findings to humans, we performed a proof-of-principle study in obese transwomen in which systemic angiotensin peptides and blood pressure were quantified prior to and after 12 weeks of gender-affirming 17ß-estradiol hormone therapy. RESULTS: Adipocyte ACE2 deficiency had no effect on the development of obesity in either sex. HF feeding increased systolic blood pressures (SBP) of wild-type male and female mice compared to LF-fed controls. Adipocyte ACE2 deficiency augmented obesity-induced elevations in SBP in females, but not in males. Obese female, but not obese male mice with adipocyte ACE2 deficiency, had an augmented SBP response to acute AngII challenge. In humans, plasma 17ß-estradiol concentrations increased in obese transwomen administered 17ß-estradiol and correlated positively with plasma Ang-(1-7)/AngII balance, and negatively to SBP after 12 weeks of 17ß-estradiol administration. CONCLUSIONS: Adipocyte ACE2 protects female mice from obesity-hypertension, and reduces the blood pressure response to systemic AngII. In obese transwomen undergoing gender-affirming hormone therapy, 17ß-estradiol administration may regulate blood pressure via the Ang-(1-7)/AngII balance.


Asunto(s)
Adipocitos/metabolismo , Presión Sanguínea/efectos de los fármacos , Obesidad/metabolismo , Peptidil-Dipeptidasa A/metabolismo , Angiotensina I/farmacología , Enzima Convertidora de Angiotensina 2 , Animales , Dieta Alta en Grasa , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Ratones , Fragmentos de Péptidos/farmacología , Peptidil-Dipeptidasa A/genética , Caracteres Sexuales
5.
Environ Health Perspect ; 127(7): 77004, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31306034

RESUMEN

BACKGROUND: Lipophilic polychlorinated biphenyls (PCBs) accumulate with obesity, but during weight loss, liberated PCBs act as ligands of the aryl hydrocarbon receptor (AhR) to negatively influence health. Previous studies demonstrated that PCB-77 administration to obese male mice impaired glucose tolerance during weight loss. Recent studies indicate higher toxic equivalencies of dioxin-like PCBs in exposed females than males. OBJECTIVES: We compared effects of PCB-77 on weight gain or loss and glucose homeostasis in male vs. female mice. We defined effects of AhR deficiency during weight gain or loss in male and female mice exposed to PCB-77. METHODS: Study design was vehicle (VEH) or PCB-77 administration while fed a high-fat (HF) diet for 12 wk, followed by weight loss for 4 wk. The following groups were examined: male and female C57BL/6 mice administered VEH or PCB-77, female [Formula: see text] and [Formula: see text] mice administered VEH or PCB-77, and male [Formula: see text] and [Formula: see text] mice administered PCB-77. Glucose tolerance was quantified during weight gain (week 11) and loss (week 15); liver and adipose AhR and IRS2 (insulin receptor substrate 2) mRNA abundance, and PCB-77 concentrations were quantified at week 16. RESULTS: PCB-77 attenuated development of obesity in females but not males. During weight loss, PCB-77 impaired glucose tolerance of males. AhR-deficient females (VEH) were resistant to diet-induced obesity. Compared with VEH-treated mice, HF-fed [Formula: see text] females treated with PCB-77 has less weight gain, and [Formula: see text] females had greater weight gain. During weight loss, [Formula: see text] females but not [Formula: see text] males treated with PCB-77 exhibited impaired glucose tolerance. In [Formula: see text] females administered PCB-77, IRS2 mRNA abundance was lower in adipose tissue compared with VEH-treated mice. CONCLUSION: Male and female mice responded differently to PCB-77 and AhR deficiency in body weight (BW) regulation and glucose homeostasis. AhR deficiency reversed PCB-77-induced glucose impairment of obese males losing weight but augmented glucose intolerance of females. These results demonstrate sex differences in PCB-77-induced regulation of glucose homeostasis of mice. https://doi.org/10.1289/EHP4133.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/deficiencia , Glucosa/metabolismo , Bifenilos Policlorados/efectos adversos , Receptores de Hidrocarburo de Aril/deficiencia , Pérdida de Peso/fisiología , Animales , Peso Corporal/efectos de los fármacos , Femenino , Homeostasis , Masculino , Ratones , Obesidad/inducido químicamente
6.
Nat Commun ; 10(1): 2631, 2019 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-31201301

RESUMEN

Men and women differ in circulating lipids and coronary artery disease (CAD). While sex hormones such as estrogens decrease CAD risk, hormone replacement therapy increases risk. Biological sex is determined by sex hormones and chromosomes, but effects of sex chromosomes on circulating lipids and atherosclerosis are unknown. Here, we use mouse models to separate effects of sex chromosomes and hormones on atherosclerosis, circulating lipids and intestinal fat metabolism. We assess atherosclerosis in multiple models and experimental paradigms that distinguish effects of sex chromosomes, and male or female gonads. Pro-atherogenic lipids and atherosclerosis are greater in XX than XY mice, indicating a primary effect of sex chromosomes. Small intestine expression of enzymes involved in lipid absorption and chylomicron assembly are greater in XX male and female mice with higher intestinal lipids. Together, our results show that an XX sex chromosome complement promotes the bioavailability of dietary fat to accelerate atherosclerosis.


Asunto(s)
Trastornos del Desarrollo Sexual 46, XX/metabolismo , Aterosclerosis/genética , Metabolismo de los Lípidos/genética , Lípidos/sangre , Cromosoma X/fisiología , Trastornos del Desarrollo Sexual 46, XX/sangre , Animales , Aterosclerosis/sangre , Aterosclerosis/metabolismo , Dieta Aterogénica/efectos adversos , Modelos Animales de Enfermedad , Femenino , Hormonas Esteroides Gonadales/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Intestino Delgado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ovario/metabolismo , Factores Sexuales , Proteína de la Región Y Determinante del Sexo/genética , Testículo/metabolismo
7.
Food Chem Toxicol ; 130: 219-230, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31078726

RESUMEN

Pseudomonas aeruginosa, a leading cause of sepsis, produces pyocyanin, a blue-pigmented virulence factor. Sepsis is associated with cachexia, but mechanisms are unknown and conventional nutrition approaches are not effective treatments. Pyocyanin has affinity for the aryl hydrocarbon receptor (AhR), which is expressed on adipocytes and regulates adipocyte differentiation. The purpose of this study was to define in vitro and in vivo effects of pyocyanin on adipocyte differentiation and body weight regulation as relates to septic cachexia. In 3T3-L1 preadipocytes, pyocyanin activated AhR and its downstream marker CYP1a1, and reduced differentiation. Administration of pyocyanin to male C57BL/6J mice acutely reduced body temperature with altered locomotion, but caused sustained weight loss. Chronic pyocyanin administration to male and female C57BL/6J mice resulted in sustained reductions in body weight and fat mass, with adipose-specific AhR activation. Pyocyanin-treated male mice had decreased energy expenditure and physical activity, and increased adipose explant lipolysis. In females, pyocyanin caused robust reductions in body weight, adipose-specific AhR activation, and increased expression of inflammatory cytokines in differentiated adipocytes. These results demonstrate that pyocyanin reduces adipocyte differentiation and decreases body weight and fat mass in male and female mice, suggesting that pyocyanin may play a role in septic cachexia.


Asunto(s)
Adipocitos/efectos de los fármacos , Adiposidad/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Pseudomonas aeruginosa , Piocianina/farmacología , Células 3T3-L1 , Animales , Caquexia , Diferenciación Celular/efectos de los fármacos , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Proyectos Piloto , ARN Mensajero , Sepsis
8.
J Vasc Surg ; 70(5): 1658-1668.e1, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-30850299

RESUMEN

OBJECTIVE: Previous studies demonstrated that deficiency of angiotensin-converting enzyme 2 (ACE2) augmented angiotensin II (AngII)-induced atherosclerosis and abdominal aortic aneurysm (AAA) formation in hypercholesterolemic mice. Effects of ACE2 deficiency could arise from increased concentrations of its substrate, AngII, or decreased concentrations of its product, angiotensin-(1-7) [Ang-(1-7)]. Infusion of Ang-(1-7), a Mas receptor (MasR) ligand, to hypercholesterolemic male mice reduced AngII-induced atherosclerosis, suggesting a protective role of the Ang-(1-7)/MasR axis. However, it is unclear whether endogenous Ang-(1-7) acts at MasR to influence AngII-induced vascular diseases. The purpose of this study was to define the role of MasR deficiency in AngII-induced atherosclerosis and AAA formation and severity in hypercholesterolemic male mice. METHODS: MasR+/+ and MasR-/- male mice on a low-density lipoprotein receptor-deficient (Ldlr-/-) or apolipoprotein E-deficient (Apoe-/-) background were infused with AngII at either 600 or 1000 ng/kg/min by osmotic minipump for 28 days. Atherosclerosis was quantified at study end point as percentage lesion surface area of the aortic arch in Ldlr-/- mice. Abdominal aortic internal diameters were quantified by ultrasound, and maximal external AAA diameters were quantified at study end point. Blood pressure was quantified by radiotelemetry and a tail cuff-based technique. Serum cholesterol concentrations and vascular tissue characterization were examined at study end point. RESULTS: MasR deficiency did not influence body weight, systolic blood pressure at baseline and during AngII infusion, or serum cholesterol concentrations in either Apoe-/- or Ldlr-/- mice. MasR deficiency increased AngII-induced atherosclerosis in aortic arches of Ldlr-/- mice (P < .05), associated with increased oxidative stress and apoptosis in aortic root sections (P < .05). MasR deficiency also augmented internal and external AAA diameters and increased aortic ruptures of both Ldlr-/- and Apoe-/- mice (P < .05). These effects were associated with increased elastin breaks and T-lymphocyte and macrophage accumulation into abdominal aortas of AngII-infused MasR-deficient mice (P < .05). CONCLUSIONS: These results demonstrate that MasR deficiency augmented AngII-induced atherosclerosis and AAA rupture through mechanisms involving increased oxidative stress, inflammation, and apoptosis, suggesting that MasR activation may provide therapeutic efficacy against vascular diseases.


Asunto(s)
Angiotensina II/metabolismo , Aneurisma de la Aorta Abdominal/patología , Rotura de la Aorta/patología , Aterosclerosis/complicaciones , Proteínas Proto-Oncogénicas/deficiencia , Receptores Acoplados a Proteínas G/deficiencia , Angiotensina I/metabolismo , Angiotensina II/administración & dosificación , Animales , Aorta/patología , Aneurisma de la Aorta Abdominal/sangre , Aneurisma de la Aorta Abdominal/etiología , Rotura de la Aorta/sangre , Rotura de la Aorta/etiología , Apoptosis/genética , Aterosclerosis/sangre , Colesterol , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Ratones Noqueados para ApoE , Estrés Oxidativo/genética , Fragmentos de Péptidos/metabolismo , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas/genética , Receptores Acoplados a Proteínas G/genética , Receptores de LDL/genética , Receptores de LDL/metabolismo
9.
JCI Insight ; 3(2)2018 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-29367460

RESUMEN

Mesenchymal stem cells (MSCs) can give rise to both adipocytes and osteoblasts, but the molecular mechanisms underlying MSC fate determination remain poorly understood. IκB kinase ß (IKKß), a central coordinator of inflammation and immune responses through activation of NF-κB, has been implicated as a critical molecular link between obesity and metabolic disorders. Here, we show that IKKß can reciprocally regulate adipocyte and osteoblast differentiation of murine and human MSCs through an NF-κB-independent mechanism. IKKß is a ß-catenin kinase that phosphorylates the conserved degron motif of ß-catenin to prime it for ß-TrCP-mediated ubiquitination and degradation, thereby increasing adipogenesis and inhibiting osteogenesis in MSCs. Animal studies demonstrated that deficiency of IKKß in BM mesenchymal stromal cells increased bone mass and decreased BM adipocyte formation in adult mice. In humans, IKKß expression in adipose tissue was also positively associated with increased adiposity and elevated ß-catenin phosphorylation. These findings suggest IKKß as a key molecular switch that regulates MSC fate, and they provide potentially novel mechanistic insights into the understanding of the cross-regulation between the evolutionarily conserved IKKß and Wnt/ß-catenin signaling pathways. The IKKß-Wnt axis we uncovered may also have important implications for development, homeostasis, and disease pathogenesis.


Asunto(s)
Diferenciación Celular/fisiología , Quinasa I-kappa B/metabolismo , Células Madre Mesenquimatosas/metabolismo , Obesidad/patología , beta Catenina/metabolismo , Grasa Abdominal/patología , Adipocitos/fisiología , Adipogénesis/fisiología , Adulto , Animales , Biopsia , Células Cultivadas , Femenino , Humanos , Quinasa I-kappa B/análisis , Quinasa I-kappa B/genética , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Modelos Animales , Obesidad/sangre , Osteoblastos/fisiología , Osteogénesis/fisiología , Fosforilación/fisiología , Cultivo Primario de Células , Proteolisis , Ubiquitinación/fisiología , Vía de Señalización Wnt/fisiología , Proteínas con Repetición de beta-Transducina/metabolismo
10.
Arterioscler Thromb Vasc Biol ; 38(1): 143-153, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29097367

RESUMEN

OBJECTIVE: Aortic pathologies exhibit sexual dimorphism, with aneurysms in both the thoracic and abdominal aorta (ie, abdominal aortic aneurysm [AAA]) exhibiting higher male prevalence. Women have lower prevalence of aneurysms, but when they occur, aneurysms progress rapidly. To define mechanisms for these sex differences, we determined the role of sex chromosome complement and testosterone on the location and progression of angiotensin II (AngII)-induced aortic pathologies. APPROACH AND RESULTS: We used transgenic male mice expressing Sry (sex-determining region Y) on an autosome to create Ldlr (low-density lipoprotein receptor)-deficient male mice with an XY or XX sex chromosome complement. Transcriptional profiling was performed on abdominal aortas from XY or XX males, demonstrating 1746 genes influenced by sex chromosomes or sex hormones. Males (XY or XX) were either sham-operated or orchiectomized before AngII infusions. Diffuse aortic aneurysm pathology developed in XY AngII-infused males, whereas XX males developed focal AAAs. Castration reduced all AngII-induced aortic pathologies in XY and XX males. Thoracic aortas from AngII-infused XY males exhibited adventitial thickening that was not present in XX males. We infused male XY and XX mice with either saline or AngII and quantified mRNA abundance of key genes in both thoracic and abdominal aortas. Regional differences in mRNA abundance existed before AngII infusions, which were differentially influenced by AngII between genotypes. Prolonged AngII infusions resulted in aortic wall thickening of AAAs from XY males, whereas XX males had dilated focal AAAs. CONCLUSIONS: An XY sex chromosome complement mediates diffuse aortic pathology, whereas an XX sex chromosome complement contributes to focal AngII-induced AAAs.


Asunto(s)
Angiotensina II , Aorta Abdominal/patología , Aneurisma de la Aorta Abdominal/genética , Proteína de la Región Y Determinante del Sexo/metabolismo , Cromosoma X , Cromosoma Y , Animales , Aorta Abdominal/metabolismo , Aorta Abdominal/fisiopatología , Aneurisma de la Aorta Abdominal/inducido químicamente , Aneurisma de la Aorta Abdominal/metabolismo , Aneurisma de la Aorta Abdominal/patología , Dilatación Patológica , Modelos Animales de Enfermedad , Femenino , Predisposición Genética a la Enfermedad , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Orquiectomía , Fenotipo , Receptores de LDL/deficiencia , Receptores de LDL/genética , Caracteres Sexuales , Factores Sexuales , Proteína de la Región Y Determinante del Sexo/genética , Testosterona/metabolismo , Remodelación Vascular , Rigidez Vascular
11.
Methods Mol Biol ; 1614: 1-19, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28500591

RESUMEN

The renin-angiotensin-aldosterone system (RAAS) is a complex system of enzymes, receptors, and peptides that help to control blood pressure and fluid homeostasis. Techniques in studying the RAAS can be difficult due to such factors as peptide/enzyme stability and receptor localization. This paper gives a brief account of the different components of the RAAS and current methods in measuring each component. There is also a discussion of different methods in measuring stem and immune cells by flow cytometry, hypertension, atherosclerosis, oxidative stress, energy balance, and other RAAS-activated phenotypes. While studies on the RAAS have been performed for over 100 years, new techniques have allowed scientists to come up with new insights into this system. These techniques are detailed in this Methods in Molecular Biology Series and give students new to studying the RAAS the proper controls and technical details needed to perform each procedure.


Asunto(s)
Cromatografía Liquida/métodos , Ensayo de Inmunoadsorción Enzimática/métodos , Citometría de Flujo/métodos , Espectrometría de Masas/métodos , Sistema Renina-Angiotensina/fisiología , Renina/sangre , Animales , Presión Sanguínea , Humanos , Hipertensión , Estrés Oxidativo , Peptidil-Dipeptidasa A/metabolismo
12.
Methods Mol Biol ; 1614: 69-73, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28500596

RESUMEN

The renin angiotensin system (RAS) is well known for its role in regulating blood pressure (BP). An activated RAS contributes to elevated blood pressure and is evident in both human and animal models of hypertension. Drugs that target the classic vasoconstrictive arm of the RAS (angiotensin II/AT1 receptor signaling) are potent anti-hypertensive agents in clinical setting. However, the newly discovered angiotensin-converting enzyme 2 (ACE2)/angiotensin-(1-7)/Mas receptor axis added new vitality to the hypertension field. Advances in genetic manipulation and the relative low cost made the mouse model as one of the most popular animal models to study hypertension. Since a reliable and accurate method for BP assessment is the key for such experiments, here we provide a protocol for BP measurement in mice using a noninvasive BP system. The CODA noninvasive BP system (a tail-cuff Method, Kent Scientific Corporation) enables blood pressure (BP) measurements in mice. This method uses a specialized volume pressure recording (VPR) sensor, and measures blood volume changes that are placed over the animal's tail. Mice do need to be restrained in specific holders and artificially heated to maintain normal BP.


Asunto(s)
Determinación de la Presión Sanguínea/veterinaria , Presión Sanguínea/fisiología , Ratones , Sistema Renina-Angiotensina/fisiología , Cola (estructura animal)/irrigación sanguínea , Angiotensina I/metabolismo , Animales , Determinación de la Presión Sanguínea/instrumentación , Determinación de la Presión Sanguínea/métodos , Hipertensión/diagnóstico , Hipertensión/metabolismo , Fragmentos de Péptidos/metabolismo , Programas Informáticos
13.
Methods Mol Biol ; 1614: 61-67, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28500595

RESUMEN

The use of fluorogenic substrates to measure enzymatic activity is widely used to understand function within different experimental models. ACE2 is important in understanding the balance between AngII and Ang-(1-7) and how this balance could then in turn influence hypertension or other disease outcomes. Here, we describe a method to measure ACE2 activity in abdominal aorta of hyperlipidemic mice under both saline and AngII infusion.


Asunto(s)
Aorta Abdominal/enzimología , Técnica del Anticuerpo Fluorescente/métodos , Colorantes Fluorescentes/metabolismo , Peptidil-Dipeptidasa A/análisis , Angiotensina II/química , Angiotensina II/metabolismo , Enzima Convertidora de Angiotensina 2 , Animales , Aorta Abdominal/efectos de los fármacos , Modelos Animales de Enfermedad , Hiperlipidemias/enzimología , Hiperlipidemias/patología , Ratones , Ratones Noqueados , Peptidil-Dipeptidasa A/metabolismo , Receptores de LDL/fisiología , Cloruro de Sodio/administración & dosificación , Vasoconstrictores/metabolismo
14.
Methods Mol Biol ; 1614: 75-85, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28500597

RESUMEN

The TA11PA-C10 implantable transmitter (Data Sciences International, DSI) is designed to measure blood pressure (BP) and activity in freely moving laboratory mice. The fluid filled catheter is placed in the free flowing blood of the systemic artery (inserted into the left carotid artery and extended into the aorta), and the transmitter body is placed in a benign location for long-term biocompatibility. The transmitter can be used to monitor BP in mice (as small as 17 g) under normal physiological and unrestricted conditions 24 h a day while remaining free from stress associated with human interaction. Thus, telemetry is considered the gold standard for BP monitoring in small animals such as mice. However, this methodology does require a good understanding of the system as well as appropriate training to perform the delicate transmitter implantation surgery.


Asunto(s)
Presión Sanguínea/fisiología , Ablación por Catéter/veterinaria , Ratones , Programas Informáticos , Telemetría/veterinaria , Animales , Arterias Carótidas/cirugía , Ablación por Catéter/instrumentación , Ablación por Catéter/métodos , Telemetría/métodos
15.
Diabetes ; 66(2): 505-518, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27856608

RESUMEN

The angiotensin (ANG)-(1-7)/Mas receptor (MasR) pathway activates vascular repair-relevant functions of bone marrow progenitor cells. We tested the effects of ANG-(1-7) on mobilization and vasoreparative functions of progenitor cells that are impaired in diabetes. The study was performed in streptozotocin-induced diabetic (db/db) mice. Diabetes resulted in a decreased number of Lineage-Sca-1+c-Kit+ (LSK) cells in the circulation, which was normalized by ANG-(1-7). Diabetes-induced depletion of LSK cells in the bone marrow was reversed by ANG-(1-7). ρ-Kinase (ROCK) activity was increased specifically in bone marrow LSK cells by ANG-(1-7) in diabetes, and the beneficial effects of ANG-(1-7) were prevented by fasudil. ANG-(1-7) increased Slit3 levels in the bone marrow supernatants, which activated ROCK in LSK cells and sensitized them for stromal-derived factor-1α (SDF)-induced migration. Diabetes prevented the mobilization of LSK cells in response to ischemia and impaired the recovery of blood flow, both of which were reversed by ANG-(1-7) in both models of diabetes. Genetic ablation of MasR prevented ischemia-induced mobilization of LSK cells and impaired blood flow recovery, which was associated with decreased proliferation and migration of LSK cells in response to SDF or vascular endothelial growth factor. These results suggest that MasR is a promising target for the treatment of diabetic bone marrow mobilopathy and vascular disease.


Asunto(s)
Angiotensina I/farmacología , Vasos Sanguíneos/efectos de los fármacos , Células de la Médula Ósea/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Diabetes Mellitus Experimental/fisiopatología , Neovascularización Fisiológica/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Células Madre/efectos de los fármacos , Vasodilatadores/farmacología , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/farmacología , Animales , Vasos Sanguíneos/fisiopatología , Médula Ósea/efectos de los fármacos , Médula Ósea/fisiopatología , Linaje de la Célula , Quimiocina CXCL12/efectos de los fármacos , Quimiocina CXCL12/metabolismo , Isquemia , Masculino , Proteínas de la Membrana/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Ratones , Inhibidores de Proteínas Quinasas/farmacología , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas/genética , Receptores Acoplados a Proteínas G/genética , Recuperación de la Función/efectos de los fármacos , Regeneración , Factor A de Crecimiento Endotelial Vascular/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo , Quinasas Asociadas a rho/efectos de los fármacos , Quinasas Asociadas a rho/metabolismo
16.
Circulation ; 135(4): 379-391, 2017 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-27815372

RESUMEN

BACKGROUND: Abdominal aortic aneurysms (AAAs) are a deadly pathology with strong sexual dimorphism. Similar to humans, female mice exhibit far lower incidences of angiotensin II-induced AAAs than males. In addition to sex hormones, the X and Y sex chromosomes, and their unique complements of genes, may contribute to sexually dimorphic AAA pathology. Here, we defined the effect of female (XX) versus male (XY) sex chromosome complement on angiotensin II-induced AAA formation and rupture in phenotypically female mice. METHODS: Female low-density lipoprotein receptor (Ldlr) deficient mice with an XX or XY sex chromosome complement were infused with angiotensin II for 28 days to induce AAAs. Abdominal aortic lumen diameters were quantified by ultrasound, whereas AAA diameters were quantified at study end point. DNA microarrays were performed on abdominal aortas. To mimic males, female mice were administered a single dose of testosterone as neonates or as adults before angiotensin II infusions. RESULTS: Female Ldlr-/- deficient mice with an XX and XY sex chromosome complement had similar sex organ weights and low serum testosterone concentrations. Abdominal aortas from female XY mice selectively expressed Y chromosome genes, whereas genes known to escape X inactivation were higher in XX females. The majority of aortic gene differences in XY versus XX females fell within inflammatory pathways. AAA incidences doubled and aneurysms ruptured in XY females. AAAs from XY females exhibited inflammation, and plasma interleukin-1ß concentrations were increased in XY females. Moreover, aortas from XY females had augmented matrix metalloproteinase activity and increased oxidative stress. Last, testosterone exposure applied chronically, or as a single bolus at postnatal day 1, markedly worsened AAA outcomes in XY in comparison with XX adult females. CONCLUSIONS: An XY sex chromosome complement in phenotypic females profoundly influenced aortic gene expression profiles and promoted AAA severity. When XY females were exposed to testosterone, aneurysm rupture rates were striking. Mechanisms for augmented AAA severity in XY females include increased inflammation, augmented matrix metalloproteineases, and oxidative stress. Our results demonstrate that genes on the sex chromosomes regulate aortic vascular biology and contribute to sexual dimorphism of AAAs. Sex chromosome genes may serve as novel targets for sex-specific AAA therapeutics.


Asunto(s)
Angiotensina II/efectos adversos , Aneurisma de la Aorta Abdominal/inducido químicamente , Vasoconstrictores/efectos adversos , Angiotensina II/farmacología , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Cromosomas Sexuales , Vasoconstrictores/farmacología
18.
Biol Sex Differ ; 6: 12, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26131353

RESUMEN

BACKGROUND: Abdominal aortic aneurysms (AAAs) occur predominately in males. However, AAAs in females have rapid growth rates and rupture at smaller sizes. Mechanisms contributing to AAA progression in females are undefined. We defined effects of ovariectomy, with and without 17-ß estradiol (E2), on progression of established angiotensin II (AngII)-induced AAAs in female mice. METHODS: We used neonatal testosterone exposures at 1 day of age to promote susceptibility to AngII-induced AAAs in adult female Ldlr (-/-) mice. Females were infused with AngII for 28 days to induce AAAs, and then stratified into groups that were sham, ovariectomized (Ovx, vehicle), or Ovx with E2 administration for 2 months of continued AngII infusions. Aortic lumen diameters were quantified by ultrasound and analyzed by linear mixed model, and maximal AAA diameters were analyzed by one-way ANOVA. Atherosclerosis was quantified en face in the aortic arch. AAA tissue sections were analyzed for cellular composition. We quantified effects of E2 on abdominal aortic smooth muscle cell (SMC) growth, α-actin and transforming growth factor-beta (TGF-ß) production, and wound healing. RESULTS: Serum E2 concentrations were increased significantly by E2. Aortic lumen diameters increased over time in sham-operated and Ovx (vehicle) females, but not in Ovx females administered E2. At day 70, E2 administration decreased significantly aortic lumen diameters compared to Ovx vehicle and sham-operated females. Compared to Ovx females (vehicle), maximal AAA diameters were reduced significantly by E2. AAA tissue sections from Ovx females administered E2 exhibited significant increases in α-actin and decreases in neutrophils compared to Ovx females administered vehicle. In abdominal aortic SMCs, E2 resulted in a concentration-dependent increase in α-actin, elevated TGF-ß, and more rapid wound healing. E2 administration to Ovx females also significantly reduced atherosclerotic lesions compared to sham-operated females. This effect was accompanied by significant reductions in serum cholesterol concentrations. CONCLUSIONS: E2 administration to Ovx females abolished progressive growth and decreased severity of AngII-induced AAAs. These effects were accompanied by increased SMC α-actin, elevated TGF-ß, and reduced neutrophils. Similarly, E2 administration reduced AngII-induced atherosclerosis. These results suggest that loss of E2 in post-menopausal females may contribute to progressive growth of AAAs.

19.
Am J Physiol Endocrinol Metab ; 308(12): E1066-75, 2015 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-26078188

RESUMEN

We recently demonstrated that female mice are resistant to the development of obesity-induced hypertension through a sex hormone-dependent mechanism that involved adipose angiotensin-converting enzyme 2 (ACE2). In this study, we hypothesized that provision of 17ß-estradiol (E2) to ovariectomized (OVX) high-fat (HF)-fed female hypertensive mice would reverse obesity-hypertension through an ACE2-dependent mechanism. Pilot studies defined dose-dependent effects of E2 in OVX female mice on serum E2 concentrations and uterine weights. An E2 dose of 36 µg/ml restored normal serum E2 concentrations and uterine weights. Therefore, HF-fed OVX female Ace2(+/+) and Ace2(-/-) mice were administered vehicle or E2 (36 µg/ml) for 16 wk. E2 administration significantly decreased body weights of HF-fed OVX female Ace2(+/+) and Ace2(-/-) mice of either genotype. At 15 wk, E2 administration decreased systolic blood pressure (SBP) of OVX HF-fed Ace2(+/+) but not Ace2(-/-) females during the light but not the dark cycle. E2-mediated reductions in SBP in Ace2(+/+) females were associated with significant elevations in adipose ACE2 mRNA abundance and activity and reduced plasma ANG II concentrations. In contrast to females, E2 administration had no effect on any parameter quantified in HF-fed male hypertensive mice. In 3T3-L1 adipocytes, E2 promoted ACE2 mRNA abundance through effects at estrogen receptor-α (ERα) and resulted in ERα-mediated binding at the ACE2 promoter. These results demonstrate that E2 administration to OVX females reduces obesity-induced elevations in SBP (light cycle) through an ACE2-dependent mechanism. Beneficial effects of E2 to decrease blood pressure in OVX obese females may result from stimulation of adipose ACE2.


Asunto(s)
Estradiol/administración & dosificación , Hipertensión/tratamiento farmacológico , Hipertensión/etiología , Obesidad/complicaciones , Peptidil-Dipeptidasa A/fisiología , Adipocitos/metabolismo , Adiposidad/efectos de los fármacos , Adiposidad/genética , Enzima Convertidora de Angiotensina 2 , Animales , Presión Sanguínea/efectos de los fármacos , Femenino , Hipertensión/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Ratones Transgénicos , Ovariectomía , Peptidil-Dipeptidasa A/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
20.
J Pharmacol Sci ; 128(2): 78-82, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25986486

RESUMEN

The role of myosin light chain kinase (MLCK) in inducing podosomes was examined by confocal and electron microscopy. Removal of myosin from the actin core of podosomes using blebbistatin, a myosin inhibitor, resulted in the formation of smaller podosomes. Downregulation of MLCK by the transfection of MLCK small interfering RNA (siRNA) led to the failure of podosome formation. However, ML-7, an inhibitor of the kinase activity of MLCK, failed to inhibit podosome formation. Based on our previous report (Thatcher et al. J.Pharm.Sci. 116 116-127, 2011), we outlined the important role of the actin-binding activity of MLCK in producing smaller podosomes.


Asunto(s)
Quinasa de Cadena Ligera de Miosina/fisiología , Forbol 12,13-Dibutirato/farmacología , Podosomas/efectos de los fármacos , Podosomas/ultraestructura , Actinas/metabolismo , Animales , Azepinas/farmacología , Células Cultivadas , Regulación hacia Abajo , Microscopía Inmunoelectrónica , Quinasa de Cadena Ligera de Miosina/antagonistas & inhibidores , Quinasa de Cadena Ligera de Miosina/genética , Quinasa de Cadena Ligera de Miosina/metabolismo , Naftalenos/farmacología , Podosomas/genética , Unión Proteica , ARN Interferente Pequeño , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...