Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Exp Cell Res ; 335(2): 238-47, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-26033363

RESUMEN

BACKGROUND AND PURPOSE: Angiotensin II is a vaso-constrictive peptide that regulates blood pressure homeostasis. Even though the inflammatory effects of AngII in renal pathophysiology have been studied, there still exists a paucity of data with regard to the mechanism of action of AngII-mediated kidney injury. The objective of this study was to elucidate the mechanistic role of HMGB1-TLR4 signaling in AngII-induced inflammation in the kidney. EXPERIMENTAL APPROACH: Rat tubular epithelial cells (NRK52E) were treated with AngII over a preset time-course. In another set of experiments, HMGB1 was neutralized and TLR4 was knocked down using small interfering RNA targeting TLR4. Cell extracts were subjected to RT-PCR, immunoblotting, flow cytometry, and ELISA. KEY RESULTS: AngII-induced inflammation in NRK52E cells increased gene and protein expression of TLR4, HMGB1 and key proinflammatory cytokines (TNFα and IL1ß). Pretreatment with Losartan (an AT1 receptor blocker) attenuated the AngII-induced expression of TLR4 and inflammatory cytokines. TLR4 silencing was used to elucidate the specific role played by TLR4 in AngII-induced inflammation. TLR4siRNA treatment in these cells significantly decreased the AngII-induced inflammatory effect. Consistent observations were made when the Ang II treated cells were pretreated with anti-HMGB1. Downstream activation of NFκB and rate of generation of ROS was also decreased on gene silencing of TLR4 and exposure to anti-HMGB1. CONCLUSIONS AND IMPLICATIONS: These results indicate a key role for HMGB1-TLR4 signaling in AngII-mediated inflammation in the renal epithelial cells. Our data also reveal that AngII-induced effects could be alleviated by HMGB1-TLR4 inhibition, suggesting this pathway as a potential therapeutic target for hypertensive renal dysfunctions.


Asunto(s)
Células Epiteliales/metabolismo , Proteína HMGB1/metabolismo , Hipertensión/metabolismo , Fallo Renal Crónico/metabolismo , Receptor Toll-Like 4/metabolismo , Angiotensina II , Animales , Antihipertensivos/farmacología , Línea Celular , Células Epiteliales/inmunología , Hipertensión/inducido químicamente , Hipertensión/complicaciones , Fallo Renal Crónico/etiología , Fallo Renal Crónico/inmunología , Túbulos Renales/patología , Losartán/farmacología , FN-kappa B/metabolismo , Ratas , Transducción de Señal
2.
Am J Physiol Heart Circ Physiol ; 309(7): H1115-22, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26254332

RESUMEN

Exercise training (ExT) is recommended to treat hypertension along with pharmaceutical antihypertensive therapies. Effects of ExT in hypothalamic content of high mobility box 1 (HMGB1) and microglial activation remain unknown. We examined whether ExT would decrease autonomic and cardiovascular abnormalities in spontaneously hypertensive rats (SHR), and whether these effects were associated with decreased HMGB1 content, microglial activation, and inflammation in the hypothalamic paraventricular nucleus (PVN). Normotensive Wistar-Kyoto (WKY) rats and SHR underwent moderate-intensity ExT for 2 wk. After ExT, cardiovascular (heart rate and arterial pressure) and autonomic parameters (arterial pressure and heart rate variability, peripheral sympathetic activity, cardiac vagal activity, and baroreflex function) were measured in conscious and freely-moving rats through chronic arterial and venous catheterization. Cerebrospinal fluid, plasma, and brain were collected for molecular and immunohistochemistry analyses of the PVN. In addition to reduced heart rate variability, decreased vagal cardiac activity and increased mean arterial pressure, heart rate, arterial pressure variability, cardiac, and vasomotor sympathetic activity, SHR had higher HMGB1 protein expression, IκB-α phosphorylation, TNF-α and IL-6 protein expression, and microglia activation in the PVN. These changes were accompanied by higher plasma and cerebrospinal fluid levels of HMGB1. The ExT + SHR group had decreased expression of HMGB1, CXCR4, SDF-1, and phosphorylation of p42/44 and IκB-α. ExT reduced microglial activation and proinflammatory cytokines content in the PVN, and improved autonomic control as well. Data suggest that training-induced downregulation of activated HMGB1/CXCR4/microglia/proinflammatory cytokines axis in the PVN of SHR is a prompt neural adaptation to counterbalance the deleterious effects of inflammation on autonomic control.


Asunto(s)
Citocinas/metabolismo , Proteína HMGB1/metabolismo , Microglía/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Condicionamiento Físico Animal , Sistema Nervioso Simpático/fisiopatología , Animales , Presión Arterial , Sistema Nervioso Autónomo/fisiología , Sistema Nervioso Autónomo/fisiopatología , Barorreflejo/fisiología , Citocinas/inmunología , Frecuencia Cardíaca/fisiología , Proteínas I-kappa B/metabolismo , Inflamación , Interleucina-6/inmunología , Interleucina-6/metabolismo , Microglía/fisiología , Inhibidor NF-kappaB alfa , Núcleo Hipotalámico Paraventricular/inmunología , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Receptores CXCR4/inmunología , Receptores CXCR4/metabolismo , Transducción de Señal , Sistema Nervioso Simpático/fisiología , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Nervio Vago/fisiología , Nervio Vago/fisiopatología
3.
Front Cardiovasc Med ; 10: 1074700, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37034342

RESUMEN

Toll-like receptor 4 (TLR4) is an integral factor in the initiation of the innate immune response and plays an important role in cardiovascular diseases such as hypertension and myocardial infarction. Previous studies from our lab demonstrated that central TLR4 blockade reduced cardiac TLR4 expression, attenuated hypertension, and improved cardiac function. However, the contribution of cardiac specific TLR4 to the development of hypertension and cardiac remodeling is unknown. Therefore, we hypothesized that cardiomyocyte specific knockdown of TLR4 would have beneficial effects on hypertension, cardiac hypertrophy, and remodeling. To test this hypothesis, cardiomyocyte-specific TLR4 knockdown (cTLR4KO) mice were generated by crossing floxed TLR4 mice with Myh6-Cre mice, and subjected to angiotensin II (Ang II, 1 µg/kg/min or vehicle for 14 days) hypertension model. Blood pressure measurements using radio telemetry revealed no differences in baseline mean arterial pressure between control littermates and cTLR4KO mice (103 ± 2 vs. 105 ± 3 mmHg, p > 0.05). Ang II-induced hypertension (132 ± 2 vs. 151 ± 3 mmHg, p < 0.01) was attenuated and cardiac hypertrophy (heart/body weight; 4.7 vs. 5.8 mg/g, p < 0.01) was prevented in cTLR4KO mice when compared with control mice. In addition, the level of myocardial fibrosis was significantly reduced, and the cardiac function was improved in cTLR4KO mice infused with Ang II. Furthermore, cardiac inflammation, as evidenced by elevated gene expression of TNF, IL-6, and MCP-1 in the left ventricle, was attenuated in cTLR4KO mice infused with Ang II. Together, this data revealed a protective role for cardiomyocyte-specific deletion of TLR4 against Ang II-induced hypertension and cardiac dysfunction through inhibition of proinflammatory cytokines.

4.
Function (Oxf) ; 3(3): zqac017, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35493997

RESUMEN

Human hypertension caused by in-frame deletion of CULLIN3 exon-9 (Cul3∆9) is driven by renal and vascular mechanisms. We bred conditionally activatable Cul3∆9 transgenic mice with tamoxifen-inducible Tie2-CREERT2 mice to test the importance of endothelial Cul3. The resultant mice (E-Cul3∆9) trended towards elevated nighttime blood pressure (BP) correlated with increased nighttime activity, but displayed no difference in daytime BP or activity. Male and female E-Cul3∆9 mice together exhibited a decline in endothelial-dependent relaxation in carotid artery. Male but not female E-Cul3∆9 mice displayed severe endothelial dysfunction in cerebral basilar artery. There was no impairment in mesenteric artery and no difference in smooth muscle function, suggesting the effects of Cul3∆9 are arterial bed-specific and sex-dependent. Expression of Cul3∆9 in primary mouse aortic endothelial cells decreased endogenous Cul3 protein, phosphorylated (S1177) endothelial nitric oxide synthase (eNOS) and nitric oxide (NO) production. Protein phosphatase (PP) 2A, a known Cul3 substrate, dephosphorylates eNOS. Cul3∆9-induced impairment of eNOS activity was rescued by a selective PP2A inhibitor okadaic acid, but not by a PP1 inhibitor tautomycetin. Because NO deficiency contributes to salt-induced hypertension, we tested the salt-sensitivity of E-Cul3∆9 mice. While both male and female E-Cul3∆9 mice developed salt-induced hypertension and renal injury, the pressor effect of salt was greater in female mutants. The increased salt-sensitivity in female E-Cul3∆9 mice was associated with decreased renovascular relaxation and impaired natriuresis in response to a sodium load. Thus, CUL3 mutations in the endothelium may contribute to human hypertension in part through decreased endothelial NO bioavailability, renovascular dysfunction, and increased salt-sensitivity of BP.


Asunto(s)
Hipertensión , Vasodilatación , Animales , Humanos , Masculino , Ratones , Células Endoteliales/metabolismo , Endotelio/metabolismo , Hipertensión/inducido químicamente , Ratones Transgénicos , Mutación , Óxido Nítrico/efectos adversos , Cloruro de Sodio/efectos adversos , Cloruro de Sodio Dietético/efectos adversos , Femenino
5.
J Am Coll Cardiol ; 79(1): 35-48, 2022 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-34991787

RESUMEN

BACKGROUND: Reperfusion therapy for acute myocardial infarction (MI) is lifesaving. However, the benefit of reperfusion therapy can be paradoxically diminished by reperfusion injury, which can increase MI size. OBJECTIVES: Hemorrhage is known to occur in reperfused MIs, but whether hemorrhage plays a role in reperfusion-mediated MI expansion is not known. METHODS: We studied cardiac troponin kinetics (cTn) of ST-segment elevation MI patients (n = 70) classified by cardiovascular magnetic resonance to be hemorrhagic (70%) or nonhemorrhagic following primary percutaneous coronary intervention. To isolate the effects of hemorrhage from ischemic burden, we performed controlled canine studies (n = 25), and serially followed both cTn and MI size with time-lapse imaging. RESULTS: CTn was not different before reperfusion; however, an increase in cTn following primary percutaneous coronary intervention peaked earlier (12 hours vs 24 hours; P < 0.05) and was significantly higher in patients with hemorrhage (P < 0.01). In hemorrhagic animals, reperfusion led to rapid expansion of myocardial necrosis culminating in epicardial involvement, which was not present in nonhemorrhagic cases (P < 0.001). MI size and salvage were not different at 1 hour postreperfusion in animals with and without hemorrhage (P = 0.65). However, within 72 hours of reperfusion, a 4-fold greater loss in salvageable myocardium was evident in hemorrhagic MIs (P < 0.001). This paralleled observations in patients with larger MIs occurring in hemorrhagic cases (P < 0.01). CONCLUSIONS: Myocardial hemorrhage is a determinant of MI size. It drives MI expansion after reperfusion and compromises myocardial salvage. This introduces a clinical role of hemorrhage in acute care management, risk assessment, and future therapeutics.


Asunto(s)
Hemorragia/diagnóstico por imagen , Daño por Reperfusión Miocárdica/diagnóstico por imagen , Infarto del Miocardio con Elevación del ST/diagnóstico por imagen , Animales , Modelos Animales de Enfermedad , Perros , Humanos , Imagen por Resonancia Cinemagnética , Miocardio/patología , Necrosis , Intervención Coronaria Percutánea , Tomografía de Emisión de Positrones , Estudios Prospectivos , Infarto del Miocardio con Elevación del ST/terapia , Terapia Recuperativa , Tiempo de Tratamiento , Troponina/sangre
6.
Nat Commun ; 13(1): 6394, 2022 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-36302906

RESUMEN

Sudden blockage of arteries supplying the heart muscle contributes to millions of heart attacks (myocardial infarction, MI) around the world. Although re-opening these arteries (reperfusion) saves MI patients from immediate death, approximately 50% of these patients go on to develop chronic heart failure (CHF) and die within a 5-year period; however, why some patients accelerate towards CHF while others do not remains unclear. Here we show, using large animal models of reperfused MI, that intramyocardial hemorrhage - the most damaging form of reperfusion injury (evident in nearly 40% of reperfused ST-elevation MI patients) - drives delayed infarct healing and is centrally responsible for continuous fatty degeneration of the infarcted myocardium contributing to adverse remodeling of the heart. Specifically, we show that the fatty degeneration of the hemorrhagic MI zone stems from iron-induced macrophage activation, lipid peroxidation, foam cell formation, ceroid production, foam cell apoptosis and iron recycling. We also demonstrate that timely reduction of iron within the hemorrhagic MI zone reduces fatty infiltration and directs the heart towards favorable remodeling. Collectively, our findings elucidate why some, but not all, MIs are destined to CHF and help define a potential therapeutic strategy to mitigate post-MI CHF independent of MI size.


Asunto(s)
Insuficiencia Cardíaca , Infarto del Miocardio , Animales , Miocardio , Infarto del Miocardio/complicaciones , Infarto del Miocardio/terapia , Hemorragia , Corazón , Insuficiencia Cardíaca/etiología , Hierro , Remodelación Ventricular , Modelos Animales de Enfermedad
7.
Cardiovasc Res ; 117(1): 308-319, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-32428209

RESUMEN

AIMS: Salt-sensitive (SS) hypertension is accompanied by impaired vasodilation in the systemic and renal circulation. However, the causal relationship between vascular dysfunction and salt-induced hypertension remains controversial. We sought to determine whether primary vascular dysfunction, characterized by a failure to vasodilate during salt loading, plays a causal role in the pathogenesis of SS hypertension. METHODS AND RESULTS: Mice selectively expressing a peroxisome proliferator-activated receptor γ dominant-negative mutation in vascular smooth muscle (S-P467L) exhibited progressive SS hypertension during a 4 week high salt diet (HSD). This was associated with severely impaired vasodilation in systemic and renal vessels. Salt-induced impairment of vasodilation occurred as early as 3 days after HSD, which preceded the onset of SS hypertension. Notably, the overt salt-induced hypertension in S-P467L mice was not driven by higher cardiac output, implying elevations in peripheral vascular resistance. In keeping with this, HSD-fed S-P467L mice exhibited decreased smooth muscle responsiveness to nitric oxide (NO) in systemic vessels. HSD-fed S-P467L mice also exhibited elevated albuminuria and a blunted increase in urinary NO metabolites which was associated with blunted renal blood flow and increased sodium retention mediated by a lack of HSD-induced suppression of NKCC2. Blocking NKCC2 function prevented the salt-induced increase in blood pressure in S-P467L mice. CONCLUSION: We conclude that failure to vasodilate in response to salt loading causes SS hypertension by restricting renal perfusion and reducing renal NO through a mechanism involving NKCC2 in a mouse model of vascular peroxisome proliferator-activated receptor γ impairment.


Asunto(s)
Presión Sanguínea , Hipertensión/fisiopatología , Riñón/irrigación sanguínea , Músculo Liso Vascular/fisiopatología , Circulación Renal , Vasodilatación , Animales , Arterias Carótidas/metabolismo , Arterias Carótidas/fisiopatología , Modelos Animales de Enfermedad , Hipertensión/etiología , Hipertensión/genética , Hipertensión/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Músculo Liso Vascular/metabolismo , Mutación , Óxido Nítrico/metabolismo , PPAR gamma/genética , PPAR gamma/metabolismo , Arteria Renal/metabolismo , Arteria Renal/fisiopatología , Cloruro de Sodio Dietético , Miembro 1 de la Familia de Transportadores de Soluto 12/metabolismo
8.
PLoS One ; 15(12): e0243207, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33264359

RESUMEN

BACKGROUND: Intramyocardial hemorrhage following reperfusion is strongly associated with major adverse cardiovascular events in myocardial infarction (MI) patients; yet the mechanisms contributing to these outcomes are not well understood. Large animal models have been used to investigate intramyocardial hemorrhage, but they are exorbitantly expensive and difficult to use for mechanistic studies. In contrast, rat models are widely used to investigate mechanistic aspects of cardiovascular physiology, but a rat model that consistently recapitulates the characteristics of an hemorrhagic MI does not exist. To bridge this gap, we investigated the physiological conditions of MI that would create intramyocardial hemorrhage in rats so that a reliable model of hemorrhagic MI would become available for basic research. METHODS & RESULTS: Sprague-Dawley rats underwent either a 90-minute (90-min) ischemia and then reperfusion (I/R) (n = 22) or 30-minute (30-min) I/R (n = 18) of the left anterior descending coronary artery. Sham rats (n = 12) were used as controls. 90-min I/R consistently yielded hemorrhagic MI, while 30-min I/R consistently yielded non-hemorrhagic MI. Twenty-four hours post-reperfusion, ex-vivo late-gadolinium-enhancement (LGE) and T2* cardiac MRI performed on excised hearts from 90-min I/R rats revealed colocalization of iron deposits within the scarred tissue; however, in 30-min I/R rats scar was evident on LGE but no evidence of iron was found on T2* CMR. Histological studies verified tissue damage (H&E) detected on LGE and the presence of iron (Perl's stain) observed on T2*-CMR. At week 4 post-reperfusion, gene and protein expression of proinflammatory markers (TNF-α, IL-1ß and MMP-9) were increased in the 90-min I/R group when compared to 30-min I/R groups. Further, transmission electron microscopy performed on 90-min I/R myocardium that were positive for iron on T2* CMR and Perl's stain showed accumulation of granular iron particles within the phagosomes. CONCLUSION: Ischemic time prior to reperfusion is a critical factor in determining whether a MI is hemorrhagic or non-hemorrhagic in rats. Specifically, a period of 90-min of ischemia prior to reperfusion can produce rat models of hemorrhagic MI, while 30-minutes of ischemia prior to reperfusion can ensure that the MIs are non-hemorrhagic. Hemorrhagic MIs in rats result in marked increase in iron deposition, proinflammatory burden and adverse left-ventricular remodeling compared to rats with non-hemorrhagic MIs.


Asunto(s)
Biomarcadores/metabolismo , Hemorragia/diagnóstico por imagen , Infarto del Miocardio/diagnóstico por imagen , Daño por Reperfusión Miocárdica/complicaciones , Animales , Modelos Animales de Enfermedad , Gadolinio/administración & dosificación , Hemorragia/etiología , Hemorragia/genética , Hemorragia/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Imagen por Resonancia Cinemagnética , Masculino , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Infarto del Miocardio/etiología , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Daño por Reperfusión Miocárdica/diagnóstico por imagen , Ratas , Ratas Sprague-Dawley , Factores de Tiempo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba
9.
Hypertension ; 76(2): 468-477, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32507043

RESUMEN

Several cardiac and renal diseases are attributed to a dysregulation of the renin-angiotensin system. Renin, the rate-limiting enzyme of the renin-angiotensin system, has 2 isoforms. The classical renin isoform (renin-a) encoding preprorenin is mainly confined to the juxtaglomerular cells and released into the circulation upon stimulation. Alternatively, renin-b is predicted to remain intracellular and is expressed in the brain, heart, and adrenal gland. In the brain, ablation of renin-b (Ren-bNull mice) results in increased brain renin-angiotensin system activity. However, the consequences of renin-b ablation in tissues outside the brain remain unknown. Therefore, we hypothesized that renin-b protects from hypertensive cardiac and renal end-organ damage in mice. Ren-bNull mice exhibited normal blood pressure at baseline. Thus, we induced hypertension by using a slow pressor dose of Ang II (angiotensin II). Ang II increased blood pressure in both wild type and Ren-bNull to the same degree. Although the blood pressure between Ren-bNull and wild-type mice was elevated equally, 4-week infusion of Ang II resulted in exacerbated cardiac remodeling in Ren-bNull mice compared with wild type. Ren-bNull mice also exhibited a modest increase in renal glomerular matrix deposition, elevated plasma aldosterone, and a modestly enhanced dipsogenic response to Ang II. Interestingly, ablation of renin-b strongly suppressed plasma renin, but renal cortical renin mRNA was preserved. Altogether, these data indicate that renin-b might play a protective role in the heart, and thus renin-b could be a potential target to treat hypertensive heart disease.


Asunto(s)
Presión Sanguínea/fisiología , Predisposición Genética a la Enfermedad , Riñón/metabolismo , Isoformas de Proteínas/genética , Sistema Renina-Angiotensina/fisiología , Renina/genética , Angiotensina II , Animales , Hipertensión/inducido químicamente , Hipertensión/genética , Hipertensión/metabolismo , Masculino , Ratones , Ratones Noqueados , Isoformas de Proteínas/metabolismo , Renina/sangre , Renina/metabolismo
10.
JCI Insight ; 52019 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-31184598

RESUMEN

Patients with mutations in Cullin-3 (CUL3) exhibit severe early onset hypertension but the contribution of the smooth muscle remains unclear. Conditional genetic ablation of CUL3 in vascular smooth muscle (S-CUL3KO) causes progressive impairment in responsiveness to nitric oxide (NO), rapid development of severe hypertension, and increased arterial stiffness. Loss of CUL3 in primary aortic smooth muscle cells or aorta resulted in decreased expression of the NO receptor, soluble guanylate cyclase (sGC), causing a marked reduction in cGMP production and impaired vasodilation to cGMP analogues. Vasodilation responses to a selective large conductance Ca2+-activated K+-channel activator were normal suggesting that downstream signals which promote smooth muscle-dependent relaxation remained intact. We conclude that smooth muscle specific CUL3 ablation impairs both cGMP production and cGMP responses and that loss of CUL3 function selectively in smooth muscle is sufficient to cause severe hypertension by interfering with the NO-sGC-cGMP pathway. Our study provides compelling evidence for the sufficiency of vascular smooth muscle CUL3 as a major regulator of BP. CUL3 mutations cause severe vascular dysfunction, arterial stiffness and hypertension due to defects in vascular smooth muscle.


Asunto(s)
Proteínas Cullin/genética , Proteínas Cullin/metabolismo , Predisposición Genética a la Enfermedad/genética , Hipertensión/genética , Hipertensión/metabolismo , Músculo Liso/metabolismo , Animales , Aorta/metabolismo , Aorta/patología , GMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Hipertensión/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/metabolismo , Mutación , Miocitos del Músculo Liso/metabolismo , Óxido Nítrico , Guanilil Ciclasa Soluble/metabolismo , Transcriptoma , Rigidez Vascular , Vasodilatación
11.
J Clin Invest ; 129(6): 2318-2332, 2019 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-30896450

RESUMEN

Mice selectively expressing PPARγ dominant negative mutation in vascular smooth muscle exhibit RhoBTB1-deficiency and hypertension. Our rationale was to employ genetic complementation to uncover the mechanism of action of RhoBTB1 in vascular smooth muscle. Inducible smooth muscle-specific restoration of RhoBTB1 fully corrected the hypertension and arterial stiffness by improving vasodilator function. Notably, the cardiovascular protection occurred despite preservation of increased agonist-mediated contraction and RhoA/Rho kinase activity, suggesting RhoBTB1 selectively controls vasodilation. RhoBTB1 augmented the cGMP response to nitric oxide by restraining the activity of phosphodiesterase 5 (PDE5) by acting as a substrate adaptor delivering PDE5 to the Cullin-3 E3 Ring ubiquitin ligase complex for ubiquitination inhibiting PDE5. Angiotensin-II infusion also caused RhoBTB1-deficiency and hypertension which was prevented by smooth muscle specific RhoBTB1 restoration. We conclude that RhoBTB1 protected from hypertension, vascular smooth muscle dysfunction, and arterial stiffness in at least two models of hypertension.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/metabolismo , Hipertensión/prevención & control , Músculo Liso Vascular/metabolismo , Rigidez Vascular , Vasodilatación , Proteínas de Unión al GTP rho/metabolismo , Angiotensina II/efectos adversos , Angiotensina II/farmacología , Animales , Proteínas Cullin/genética , Proteínas Cullin/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/genética , Modelos Animales de Enfermedad , Células HEK293 , Humanos , Hipertensión/inducido químicamente , Hipertensión/genética , Hipertensión/metabolismo , Ratones , Ratones Transgénicos , Músculo Liso Vascular/patología , Óxido Nítrico/genética , Óxido Nítrico/metabolismo , Proteínas de Unión al GTP rho/genética , Quinasas Asociadas a rho/genética , Quinasas Asociadas a rho/metabolismo
12.
Hypertension ; 74(1): 173-183, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31104564

RESUMEN

Preeclampsia is a hypertensive disorder of pregnancy associated with vascular dysfunction and cardiovascular risk to offspring. We hypothesize that endothelial PPARγ (peroxisome proliferator-activated receptor-γ) provides cardiovascular protection in offspring from pregnancies complicated by hypertension. C57BL/6J dams were bred with E-V290M sires, which express a dominant-negative allele of PPARγ selectively in the endothelium. Arginine vasopressin was infused throughout gestation. Vasopressin elevated maternal blood pressure at gestational day 14 to 15 and urinary protein at day 17 consistent. Systolic blood pressure and vasodilation responses to acetylcholine were similar in vasopressin-exposed offspring compared to offspring from control pregnancies. We treated offspring with a subpressor dose of angiotensin II to test if hypertension during pregnancy predisposes offspring to hypertension. Male and female angiotensin II-treated E-V290M offspring from vasopressin-exposed but not control pregnancy exhibited significant impairment in acetylcholine-induced relaxation in carotid artery. Endothelial dysfunction in angiotensin II-treated E-V290M vasopressin-exposed offspring was attenuated by tempol, an effect which was more prominent in male offspring. Nrf2 (nuclear factor-E2-related factor) protein levels were significantly elevated in aorta from male E-V290M offspring, but not female offspring compared to controls. Blockade of ROCK (Rho-kinase) signaling and incubation with a ROCK2-specific inhibitor improved endothelial function in both male and female E-V290M offspring from vasopressin-exposed pregnancy. Our data suggest that interference with endothelial PPARγ in offspring from vasopressin-exposed pregnancies increases the risk for endothelial dysfunction on exposure to a cardiovascular stressor in adulthood. This implies that endothelial PPARγ provides protection to cardiovascular stressors in offspring of a pregnancy complicated by hypertension and perhaps in preeclampsia.


Asunto(s)
Angiotensina II/farmacología , Endotelio Vascular/metabolismo , Hipertensión Inducida en el Embarazo/genética , FN-kappa B/metabolismo , PPAR gamma/genética , Preñez , Acetilcolina/farmacología , Animales , Animales Recién Nacidos , Femenino , Regulación del Desarrollo de la Expresión Génica , Humanos , Hipertensión Inducida en el Embarazo/fisiopatología , Ratones Endogámicos C57BL , Estrés Oxidativo/fisiología , Embarazo , Sustancias Protectoras/farmacología , Transducción de Señal/genética
13.
Hypertension ; 72(5): 1227-1235, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30354810

RESUMEN

Low-salt diet is beneficial in salt-sensitive hypertension but may provoke cardiovascular risk in patients with heart failure, diabetes mellitus, or other cardiovascular abnormalities because of endogenous renin-angiotensin system activation. PPAR (peroxisome proliferator-activated receptor)-γ is a transcription factor which promotes an antioxidant pathway in the endothelium. We studied transgenic mice expressing a dominant-negative mutation in PPAR-γ selectively in the endothelium (E-V290M) to test the hypothesis that endothelial PPAR-γ plays a protective role in response to low salt-mediated renin-angiotensin system activation. Plasma renin and Ang II (angiotensin II) were significantly and equally increased in all mice fed low salt for 6 weeks. Vasorelaxation to acetylcholine was not affected in basilar artery from E-V290M at baseline but was significantly and selectively impaired in E-V290M after low salt. Unlike basilar artery, low salt was not sufficient to induce vascular dysfunction in carotid artery or aorta. Endothelial dysfunction in the basilar artery from E-V290M mice fed low salt was attenuated by scavengers of superoxide, inhibitors of NADPH oxidase, or blockade of the Ang II AT1 (angiotensin type-1) receptor. Simultaneous AT1 and AT2 receptor blockade revealed that the restoration of endothelial function after AT1 receptor blockade was not a consequence of AT2 receptor activation. We conclude that interference with PPAR-γ in the endothelium produces endothelial dysfunction in the cerebral circulation in response to low salt-mediated activation of the endogenous renin-angiotensin system, mediated at least in part, through AT1 receptor activation and perturbed redox homeostasis. Moreover, our data suggest that the cerebral circulation may be particularly sensitive to inhibition of PPAR-γ activity and renin-angiotensin system activation.


Asunto(s)
Endotelio Vascular/metabolismo , Coactivadores de Receptor Nuclear/metabolismo , Sistema Renina-Angiotensina/fisiología , Acetilcolina/farmacología , Angiotensina II/sangre , Animales , Arteria Basilar/efectos de los fármacos , Arteria Basilar/metabolismo , Endotelio Vascular/efectos de los fármacos , Masculino , Ratones , Ratones Transgénicos , Coactivadores de Receptor Nuclear/genética , Renina/sangre , Sistema Renina-Angiotensina/efectos de los fármacos , Vasodilatación/efectos de los fármacos
14.
JCI Insight ; 3(19)2018 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-30282823

RESUMEN

Copeptin, a marker of arginine vasopressin (AVP) secretion, is elevated throughout human pregnancies complicated by preeclampsia (PE), and AVP infusion throughout gestation is sufficient to induce the major phenotypes of PE in mice. Thus, we hypothesized a role for AVP in the pathogenesis of PE. AVP infusion into pregnant C57BL/6J mice resulted in hypertension, renal glomerular endotheliosis, intrauterine growth restriction, decreased placental growth factor (PGF), altered placental morphology, placental oxidative stress, and placental gene expression consistent with human PE. Interestingly, these changes occurred despite a lack of placental hypoxia or elevations in placental fms-like tyrosine kinase-1 (FLT1). Coinfusion of AVP receptor antagonists and time-restricted infusion of AVP uncovered a mid-gestational role for the AVPR1A receptor in the observed renal pathologies, versus mid- and late-gestational roles for the AVPR2 receptor in the blood pressure and fetal phenotypes. These findings demonstrate that AVP is sufficient to initiate phenotypes of PE in the absence of placental hypoxia, and indicate that AVP may mechanistically (independently, and possibly synergistically with hypoxia) contribute to the development of clinical signs of PE in specific subtypes of human PE. Additionally, they identify divergent and gestational time-specific signaling mechanisms that mediate the development of PE phenotypes in response to AVP.


Asunto(s)
Antagonistas de los Receptores de Hormonas Antidiuréticas/administración & dosificación , Neurofisinas/metabolismo , Preeclampsia/etiología , Precursores de Proteínas/metabolismo , Vasopresinas/metabolismo , Animales , Presión Sanguínea/efectos de los fármacos , Determinación de la Presión Sanguínea , Hipoxia de la Célula/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Neurofisinas/administración & dosificación , Placenta/efectos de los fármacos , Placenta/patología , Pletismografía , Preeclampsia/diagnóstico , Preeclampsia/patología , Embarazo , Precursores de Proteínas/administración & dosificación , Receptores de Vasopresinas/metabolismo , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/metabolismo , Vasopresinas/administración & dosificación
15.
Food Funct ; 8(11): 4118-4128, 2017 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-29019365

RESUMEN

BACKGROUND: Blueberries (BB) have been shown to improve insulin sensitivity and endothelial function in obese and pre-diabetic humans, and decrease oxidative stress and inflammation, and ameliorate cardio-renal damage in rodents. This indicates that blueberries have a systemic effect and are not limited to a particular organ system. In order for blueberries to exert beneficial effects on the whole body, the mechanism would logically have to operate through modulation of cellular humoral factors. OBJECTIVE: This study investigated the role of blueberries in modulating immune cell levels and attenuating circulatory and monocyte inflammation and oxidative stress in metabolic syndrome (MetS) subjects. DESIGN: A double-blind, randomized and placebo-controlled study was conducted in adults with MetS, in which they received a blueberry (22.5 g freeze-dried) or placebo smoothie twice daily for six weeks. Free radical production in the whole blood and monocytes, dendritic cell (DC) levels, expression of cytokines in monocytes and serum inflammatory markers were assessed pre- and post-intervention. RESULTS: Baseline free radical levels in MetS subjects' samples were not different between groups. Treatment with blueberries markedly decreased superoxide and total reactive oxygen species (ROS) in whole blood and monocytes compared to the placebo (p ≤ 0.05). The baseline DC numbers in MetS subjects' samples in both groups were not different, however treatment with blueberries significantly increased myeloid DC (p ≤ 0.05) and had no effect on plasmacytoid cells. Blueberry treatment decreased monocyte gene expression of TNFα, IL-6, TLR4 and reduced serum GMCSF in MetS subjects when compared to the placebo treatment (p ≤ 0.05). CONCLUSIONS: The findings of the current study demonstrate that blueberries exert immunomodulatory effects and attenuate oxidative stress and inflammation in adults with MetS.


Asunto(s)
Arándanos Azules (Planta)/química , Suplementos Dietéticos/análisis , Síndrome Metabólico/tratamiento farmacológico , Monocitos/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Extractos Vegetales/administración & dosificación , Adulto , Método Doble Ciego , Femenino , Humanos , Interleucina-6/inmunología , Masculino , Síndrome Metabólico/genética , Síndrome Metabólico/inmunología , Síndrome Metabólico/metabolismo , Persona de Mediana Edad , Monocitos/inmunología , Especies Reactivas de Oxígeno/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
17.
PLoS One ; 11(9): e0160923, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27603014

RESUMEN

Post-traumatic stress disorder (PTSD) is a trauma and stressor-related disorder that results in a prolonged stress response. It is associated with increased oxidative stress and inflammation in the prefrontal cortex (PFC) and hippocampus (HC). The only approved therapy for PTSD is selective serotonin re-uptake inhibitors (SSRIs), but their efficacy is marginal. Recently, we demonstrated that over-production of norepinephrine (NE) as the possible reason for the lack of efficacy of SSRIs. Hence, there is a need for novel therapeutic approaches for the treatment of PTSD. In this study, we investigated the anti-inflammatory role of blueberries in modulating inflammatory markers and neurotransmitter levels in PTSD. Rats were fed either a blueberry enriched (2%) or a control diet. Rats were exposed to cats for one hour on days 1 and 11 of a 31-day schedule to simulate traumatic conditions. The rats were also subjected to psychosocial stress via daily cage cohort changes. At the end of the study, the rats were euthanized and the PFC and HC were isolated. Monoamines were measured by high-performance liquid chromatography. Reactive oxygen species (ROS), gene and protein expression levels of inflammatory cytokines were also measured. In our PTSD model, NE levels were increased and 5-HT levels were decreased when compared to control. In contrast, a blueberry enriched diet increased 5-HT without affecting NE levels. The rate limiting enzymes tyrosine hydroxylase and tryptophan hydroxylase were also studied and they confirmed our findings. The enhanced levels free radicals, gene and protein expression of inflammatory cytokines seen in the PTSD group were normalized with a blueberry enriched diet. Decreased anxiety in this group was shown by improved performance on the elevated plus-maze. These findings indicate blueberries can attenuate oxidative stress and inflammation and restore neurotransmitter imbalances in a rat model of PTSD.


Asunto(s)
Arándanos Azules (Planta) , Hipocampo/efectos de los fármacos , Inflamación/dietoterapia , Corteza Prefrontal/efectos de los fármacos , Trastornos por Estrés Postraumático/dietoterapia , Animales , Cromatografía Líquida de Alta Presión , Modelos Animales de Enfermedad , Humanos , Inflamación/genética , Inflamación/fisiopatología , Neurotransmisores , Norepinefrina/metabolismo , Estrés Oxidativo/efectos de los fármacos , Corteza Prefrontal/fisiopatología , Ratas , Especies Reactivas de Oxígeno/metabolismo , Serotonina/metabolismo , Inhibidores Selectivos de la Recaptación de Serotonina/administración & dosificación , Trastornos por Estrés Postraumático/fisiopatología , Triptófano Hidroxilasa
18.
PLoS One ; 10(3): e0122850, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25811788

RESUMEN

BACKGROUND & PURPOSE: Toll-like receptor 4 (TLR4) signaling induces tissue pro-inflammatory cytokine release and endoplasmic reticulum (ER) stress. We examined the role of TLR4 in autonomic dysfunction and the contribution of ER stress. EXPERIMENTAL APPROACH: Our study included animals divided in 6 experimental groups: rats treated with saline (i.v., 0.9%), LPS (i.v., 10mg/kg), VIPER (i.v., 0.1 mg/kg), or 4-PBA (i.p., 10 mg/kg). Two other groups were pretreated either with VIPER (TLR4 viral inhibitory peptide) LPS + VIPER (i.v., 0.1 mg/kg) or 4-Phenyl butyric acid (4-PBA) LPS + PBA (i.p., 10 mg/kg). Arterial pressure (AP) and heart rate (HR) were measured in conscious Sprague-Dawley rats. AP, HR variability, as well as baroreflex sensitivity (BrS), was determined after LPS or saline treatment for 2 hours. Immunofluorescence staining for NeuN, Ib1a, TLR4 and GRP78 in the hypothalamic paraventricular nucleus (PVN) was performed. TNF-α, TLR4 and GRP78 protein expression in the PVN were evaluated by western blot. Plasma norepinephrine levels were determined by ELISA. KEY RESULTS: Acute LPS treatment increased HR and plasma norepinephrine concentration. It also decreased HR variability and high frequency (HF) components of HR variability, as well BrS. Acute LPS treatment increased TLR4 and TNF-α protein expression in the PVN. These hemodynamic and molecular effects were partially abrogated with TLR4 blocker or ER stress inhibitor pretreatment. In addition, immunofluorescence study showed that TLR4 is co-localized with GRP78in the neurons. Further inhibition of TLR4 or ER stress was able to attenuate the LPS-induced microglia activation. CONCLUSIONS & IMPLICATIONS: TLR4 signaling promotes autonomic dysfunction, inflammation and microglia activation, through neuronal ER stress, in the PVN.


Asunto(s)
Estrés del Retículo Endoplásmico , Inflamación/metabolismo , Microglía/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico/efectos de los fármacos , Expresión Génica , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Lipopolisacáridos/farmacología , Neuronas/metabolismo , Norepinefrina/biosíntesis , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Ratas , Transducción de Señal , Factor de Necrosis Tumoral alfa/farmacología
19.
PLoS One ; 9(11): e111976, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25372283

RESUMEN

BACKGROUND: Metabolic syndrome (MetS) is characterized by a cluster of health factors that indicate a higher risk for cardio-renal diseases. Recent evidence indicates that antioxidants from berries are alternative to attenuate oxidative stress and inflammation. We tested the hypothesis that inflammation-induced renal damage is triggered by the activation of TLR4, and subsequent modulation of redox-sensitive molecules and mitogen-activated protein kinase (MAPK) pathway. METHODS: Five-week old lean and obese Zucker rats (LZR and OZR) were fed a blueberry-enriched diet or an isocaloric control diet for 15 weeks. A glucose tolerance test and acute renal clearance experiments were performed. Gene and protein expression levels for TLR4, cytokines and phosphorylation of ERK and p38MAPK were measured. Kidney redox status and urinary albumin levels were quantified. Renal pathology was evaluated histologically. RESULTS: Control OZR exhibited lower glucose tolerance; exacerbated renal function parameters; increased oxidative stress. Gene and protein expression levels of TLR4 were higher and this was accompanied by increased renal pathology with extensive albuminuria and deterioration in antioxidant levels in OZR. In addition, OZR had increased phosphorylation of ERK and p38MAPK. Blueberry-fed OZR exhibited significant improvements in all these parameters compared to OZR. CONCLUSION: TLR4-MAPK signaling pathway is a key to the renal structural injury and dysfunction in MetS and blueberry (BB) protect against this damage by inhibiting TLR4. SIGNIFICANCE: This is the first study to put forth a potential mechanism of TLR4-induced kidney damage in a model of MetS and to elucidate a downstream mechanism by which blueberry exert their reno-protective effects.


Asunto(s)
Arándanos Azules (Planta)/química , Dieta , Riñón/metabolismo , Síndrome Metabólico/metabolismo , Estrés Oxidativo , Animales , Peso Corporal , Modelos Animales de Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Expresión Génica , Glucosa/metabolismo , Hemodinámica , Riñón/patología , Riñón/fisiopatología , Pruebas de Función Renal , Glomérulos Renales/metabolismo , Glomérulos Renales/patología , Masculino , Síndrome Metabólico/genética , Síndrome Metabólico/fisiopatología , Ratones Noqueados , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Oxidación-Reducción , Fosforilación , Ratas , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos
20.
Free Radic Biol Med ; 71: 16-25, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24657730

RESUMEN

Inflammation has been implicated in the pathophysiology of kidney disorders. Previous studies have documented the contributions of various inflammatory cascades in the development of kidney and other organ dysfunctions. The Toll-like receptor 4 (TLR4) inflammatory pathway is a major contributor of inflammation in the kidney. Interestingly, lipopolysaccharide (LPS), a specific ligand for TLR4, has been shown to induce acute kidney injury (AKI) in animal models. We have previously studied the beneficial effects of nonpharmacological agents, particularly blueberries (BB), in attenuating inflammation and oxidative stress. We hypothesize that BB protect against the LPS-induced AKI by inhibiting TLR4 activation and kidney injury markers. Twelve-week-old male Sprague-Dawley rats received a BB solution or saline intragastric gavage for 2 days. One group of BB and saline-gavaged animals was injected with LPS (10 mg/kg bw). Another group of rats was injected with VIPER (0.1 mg/kg iv), a TLR4-specific inhibitory peptide, 2 h before LPS administration. Compared to LPS-administered rats, the BB-pretreated animals exhibited improved glomerular filtration rate, elevated renal blood flow, and a reduced renal vascular resistance. In addition, a reduction in the rate of production of free radicals, namely total reactive oxygen species (ROS) and superoxide, was observed in the BB-supplemented LPS group. Gene and protein expressions for TLR4, proinflammatory cytokine, and acute kidney injury markers were also attenuated in animals that were pretreated with BB as measured by real time RT-PCR and Western blotting, respectively. These results in the BB-pretreated group were consistent with those in the VIPER-treated rats, and indicate that BB protects against AKI by inhibiting TLR4 and its subsequent effect on inflammatory and oxidative stress pathways.


Asunto(s)
Lesión Renal Aguda/tratamiento farmacológico , Antiinflamatorios no Esteroideos/farmacología , Arándanos Azules (Planta)/química , Extractos Vegetales/farmacología , Receptor Toll-Like 4/antagonistas & inhibidores , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/genética , Lesión Renal Aguda/metabolismo , Animales , Expresión Génica , Tasa de Filtración Glomerular/efectos de los fármacos , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Inflamación/genética , Inflamación/metabolismo , Lipopolisacáridos , Masculino , Péptidos/farmacología , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , Circulación Renal/efectos de los fármacos , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología , Resistencia Vascular/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA