Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 102
Filter
Add more filters

Complementary Medicines
Publication year range
1.
J Am Heart Assoc ; 10(9): e018716, 2021 05 04.
Article in English | MEDLINE | ID: mdl-33870711

ABSTRACT

Background Increased potassium intake lowers blood pressure in patients with hypertension, but increased potassium intake also elevates plasma concentrations of the blood pressure-raising hormone aldosterone. Besides its well-described renal effects, aldosterone is also believed to have vascular effects, acting through mineralocorticoid receptors present in endothelial and vascular smooth muscle cells, although mineralocorticoid receptors-independent actions are also thought to be involved. Methods and Results To gain further insight into the effect of increased potassium intake and potassium-stimulated hyperaldosteronism on the human cardiovascular system, we conducted a randomized placebo-controlled double-blind crossover study in 25 healthy normotensive men, where 4 weeks treatment with a potassium supplement (90 mmol/day) was compared with 4 weeks on placebo. At the end of each treatment period, we measured potassium and aldosterone in plasma and performed an angiotensin II (AngII) infusion experiment, during which we assessed the aldosterone response in plasma. Hemodynamics were also monitored during the AngII infusion using ECG, impedance cardiography, finger plethysmography (blood pressure-monitoring), and Doppler ultrasound. The study showed that higher potassium intake increased plasma potassium (mean±SD, 4.3±0.2 versus 4.0±0.2 mmol/L; P=0.0002) and aldosterone (median [interquartile range], 440 [336-521] versus 237 [173-386] pmol/L; P<0.0001), and based on a linear mixed model for repeated measurements, increased potassium intake potentiated AngII-stimulated aldosterone secretion (P=0.0020). In contrast, the hemodynamic responses (blood pressure, total peripheral resistance, cardiac output, and renal artery blood flow) to AngII were similar after potassium and placebo. Conclusions Increased potassium intake potentiates AngII-stimulated aldosterone secretion without affecting systemic cardiovascular hemodynamics in healthy normotensive men. Registration EudraCT Number: 2013-004460-66; URL: https://www.ClinicalTrials.gov; Unique identifier: NCT02380157.


Subject(s)
Angiotensin II/administration & dosage , Blood Pressure/physiology , Hypertension/therapy , Potassium, Dietary/pharmacokinetics , Potassium/blood , Adult , Aldosterone/blood , Biomarkers/blood , Cross-Over Studies , Double-Blind Method , Female , Follow-Up Studies , Healthy Volunteers , Humans , Hypertension/physiopathology , Infusions, Intravenous , Male , Middle Aged , Treatment Outcome , Vasoconstrictor Agents/administration & dosage , Young Adult
2.
Life Sci ; 252: 117624, 2020 Jul 01.
Article in English | MEDLINE | ID: mdl-32259602

ABSTRACT

AIMS: Cardiac hypertrophy is one of most important risk factors for cardiovascular mortality. Activation of Wnt/ß-catenin signaling pathway is acknowledged to be an important mechanism for pathogenesis of cardiac hypertrophy. Polyphyllin I (PPI), a component in the traditional Chinese medicinal herb, has shown anticancer effect partially via interruption of Wnt/ß-catenin signaling pathway. Our aim was to test whether PPI attenuates cardiac hypertrophy. MATERIALS AND METHODS: Adult male C57BL/6J mice were subjected to either pressure overload generated by transverse aortic constriction (TAC) or sham surgery (control group). Angiotensin-II (Ang-II) was used to induce cardiomyocyte hypertrophy in vitro. PPI was intraperitoneally administrated daily for 4 weeks after TAC surgery and then cardiac function was determined by echocardiography and histological analysis was performed. KEY FINDINGS: PPI significantly ameliorated cardiac dysfunction of mice subjected to TAC. Meanwhile, PPI attenuated TAC induced cardiac hypertrophy indicated by blunted increase in heart mass, cross section area of cardiomyocyte, cardiac fibrosis and expression of hypertrophic biomarkers ANP, BNP and ß-MHC. In addition, PPI also ameliorated Ang-II induced cardiomyocyte hypertrophy in vitro. Importantly, PPI decreased protein expression of active ß-catenin/total ß-catenin, phosphorylation of GSK3ß and Wnt target genes c-myc, c-jun, c-fos and cyclin D1 and its anti-hypertrophic effect was blunted by supplementation of Wnt 3a. SIGNIFICANCE: Our results suggest that PPI attenuates cardiac dysfunction and attenuate development of pressure over-load induced cardiac hypertrophic via suppressing Wnt/ß-catenin signaling pathway. PPI might be a candidate drug for treatment of cardiac hypertrophy.


Subject(s)
Cardiomegaly/prevention & control , Diosgenin/analogs & derivatives , Myocytes, Cardiac/drug effects , Wnt Signaling Pathway/drug effects , Angiotensin II/administration & dosage , Animals , Diosgenin/pharmacology , Disease Models, Animal , Echocardiography , Male , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/pathology , Rats
3.
Am J Physiol Regul Integr Comp Physiol ; 318(2): R338-R350, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31850818

ABSTRACT

Exposure to different stressors in utero is linked to adult diseases such as obesity and hypertension. In this study, the impact of prenatal infection (PNI) on adult body weight and cardiovascular function was evaluated using a naturally occurring rodent pathogen, Mycoplasma pulmonis (MP). Pregnant Sprague-Dawley rats were infected with MP on gestationalday 14 and gave birth naturally. Adult PNI offspring weighed more than controls, but resting mean arterial pressure (MAP) was unchanged. Subcutaneous injection of angiotensin II (10 µg/kg) elicited a rise in MAP that was greater in both male and female PNI offspring compared with controls (P < 0.03). The accompanying reflex bradycardia was similar to the controls, suggesting that PNI induced baroreflex dysfunction. Subcutaneous nicotine administration, a potent cardiorespiratory stimulus, also elicited a transient rise in MAP that was generally greater in the PNI group, but the change in MAP from baseline was only significant in the PNI females compared with controls (P < 0.03). Elevated body weight and cardiovascular reactivity in the PNI offspring was associated with an increase in the ratio of hypothalamic corticotrophin-releasing hormone receptors type 1 to type 2 gene expression in both sexes compared with controls. These findings support previous studies demonstrating that PNI induces alterations in cardiovascular function and body weight. Yet, unlike previous studies utilizing other models of PNI (e.g., endotoxin), MP PNI did not induce resting hypertension. Thus, our study provides a foundation for future studies evaluating the cardiovascular risks of offspring exposed to microbial challenges in utero.


Subject(s)
Angiotensin II/administration & dosage , Arterial Pressure/drug effects , Baroreflex/drug effects , Cardiovascular Diseases/etiology , Mycoplasma Infections/complications , Mycoplasma pulmonis/pathogenicity , Prenatal Exposure Delayed Effects , Age Factors , Animals , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/physiopathology , Disease Models, Animal , Female , Gestational Age , Hypothalamus/metabolism , Hypothalamus/physiopathology , Injections, Subcutaneous , Male , Mycoplasma Infections/microbiology , Pregnancy , Rats, Sprague-Dawley , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Corticotropin-Releasing Hormone/metabolism , Weight Gain
4.
J Ethnopharmacol ; 220: 75-86, 2018 Jun 28.
Article in English | MEDLINE | ID: mdl-29501845

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: A recent ethnomedical survey on medicinal plants grown in Mexico revealed that Sechium edule (Jacq.) Sw. (Cucurbitaceae) is one of the most valued plant species to treat cardiovascular diseases, including hypertension. Fruits, young leaves, buds, stems, and tuberous roots of the plant are edible. Considering that endothelial dysfunction induced by Angiotensin II plays an important role in the pathogenesis of hypertension and is accompanied by a prooxidative condition, which in turn induces an inflammatory state, vascular remodeling, and tissue damage, and that S. edule has been reported to possess antioxidant, anti-inflammatory and antihypertensive activity, its capability to control endothelial dysfunction was also assessed. AIM OF THE STUDY: To assess in vivo the anti-endothelial dysfunction activity of the acetone fraction (rSe-ACE) of the hydroalcoholic extract from S. edule roots. MATERIALS AND METHODS: Endothelial dysfunction was induced in female C57BL/6 J mice by a daily intraperitoneal injection of angiotensin II for 10 weeks. Either rSe-ACE or losartan (as a control) were co-administered with angiotensin II for the same period. Blood pressure was measured at weeks 0, 5, and 10. Kidney extracts were prepared to determine IL1ß, IL4, IL6, IL10, IL17, IFNγ, TNFα, and TGFß levels by ELISA, along with the prooxidative status as assessed by the activity of antioxidant enzymes. The expression of ICAM-1 was evaluated by immunohistochemistry in kidney histological sections. Kidney and hepatic damage, as well as vascular tissue remodeling, were studied. RESULTS: The rSe-ACE fraction administered at a dose of 10 mg/kg was able to control hypertension, as well as the prooxidative and proinflammatory status in kidney as efficiently as losartan, returning mice to normotensive levels. Additionally, the fraction was more efficient than losartan to prevent liver and kidney damage. Phytochemical characterization identified cinnamic acid as a major compound, and linoleic, palmitic, and myristic acids as the most abundant non-polar components in the mixture, previously reported to aid in the control of hypertension, inflammation, and oxidative stress, three important components of endothelial dysfunction. IN CONCLUSION: this study demonstrated that rSe-ACE has anti-endothelial dysfunction activity in an experimental model and highlights the role of cinnamic acid and fatty acids in the observed effects.


Subject(s)
Cucurbitaceae/chemistry , Endothelium, Vascular/drug effects , Plant Extracts/pharmacology , Vascular Diseases/prevention & control , Acetone/chemistry , Angiotensin II/administration & dosage , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Antioxidants/metabolism , Cinnamates/isolation & purification , Cinnamates/pharmacology , Disease Models, Animal , Endothelium, Vascular/pathology , Fatty Acids/isolation & purification , Fatty Acids/pharmacology , Female , Losartan/pharmacology , Mexico , Mice , Mice, Inbred C57BL , Plant Roots , Vascular Diseases/pathology
5.
J Recept Signal Transduct Res ; 37(5): 493-499, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28812967

ABSTRACT

Cardiomyopathy involves changes in myocardial ultrastructure and cardiac hypertrophy. Angiotensin II (AngII) has previously been shown to stimulate the expression of IGF-2 and IGF-2R in H9c2 cardiomyoblasts and increase of blood pressure, and cardiac hypertrophy. Estrogen receptors (ERs) exert protective effects, such as anti-hypertrophy in cadiomyocytes. Tanshinone IIA (TSN), a main active ingredient from a Chinese medical herb, Salvia miltiorrhiza Bunge (Danshen), was shown to protect cardiomyocytes hypertrophy by different stress signals. We aimed to investigate whether TSN protected H9c2 cardiomyocytes from AngII-induced activation of IGF-2R pathway and hypertrophy by mediating through ERs. AngII resulted in H9c2 cardiomyoblast hypertrophy and increased inflammatory molecular markers. These were down-regulated by TSN via estrogen receptors. AngII resulted in elevation in MAPKs, IGF-2R and hypertrophic protein markers. These, again, were reduced by addition of the phytoestrogen with activation of ERs. Finally, AngII induced phosphorylation of heat shock factor-1 (HSF1) and decreased sirtuin-1 (SIRT1). In addition, AngII also caused an increase in distribution of IGF-2R molecules on cell membrane. In contrast, TSN reduced HSF1 phosphorylation and cell surface IGF-2R while elevating SIRT1 via ERs. TSN was capable of attenuating AngII-induced IGF-2R pathway and hypertrophy through ERs in H9c2 cardiomyoblast cells.


Subject(s)
Abietanes/administration & dosage , Cardiomegaly/drug therapy , Insulin-Like Growth Factor II/genetics , Receptor, IGF Type 2/genetics , Angiotensin II/administration & dosage , Angiotensin II/metabolism , Animals , Cardiomegaly/genetics , Cardiomegaly/pathology , Cell Line , Drugs, Chinese Herbal/administration & dosage , Gene Expression/drug effects , Heat Shock Transcription Factors/genetics , Humans , Insulin-Like Growth Factor II/metabolism , Mitogen-Activated Protein Kinase Kinases/genetics , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Rats , Receptor, IGF Type 2/metabolism , Receptors, Estrogen/genetics , Signal Transduction/drug effects , Sirtuin 1/genetics
6.
Neuroscience ; 329: 112-21, 2016 08 04.
Article in English | MEDLINE | ID: mdl-27163380

ABSTRACT

Activation of angiotensinergic pathways by central aldosterone (Aldo)-mineralocorticoid receptor (MR) pathway plays a critical role in angiotensin II (Ang II)-induced hypertension. The subfornical organ (SFO) contains both MR and angiotensin II type 1 receptors (AT1R) and can relay the signals of circulating Ang II to downstream nuclei such as the paraventricular nucleus (PVN), supraoptic nucleus (SON) and rostral ventrolateral medulla (RVLM). In Wistar rats, subcutaneous (sc) infusion of Ang II at 500ng/min/kg for 1 or 2weeks increased reactive oxygen species (ROS) as measured by dihydroethidium (DHE) staining in a nucleus - specific pattern. Intra-SFO infusion of AAV-MR- or AT1aR-siRNA prevented the Ang II-induced increase in AT1R mRNA expression in the SFO and decreased MR mRNA. Both MR- and AT1aR-siRNA prevented increases in ROS in the PVN and RVLM. MR- but not AT1aR-siRNA in the SFO prevented the Ang II-induced ROS in the SON. Both MR- and AT1aR-siRNA in the SFO prevented most of the Ang II-induced hypertension as assessed by telemetry. These results indicate that Aldo-MR signaling in the SFO is needed for the activation of Ang II-AT1R-ROS signaling from the SFO to the PVN and RVLM. Activation of Aldo-MR signaling from the SFO to the SON may enhance AT1R dependent activation of pre-sympathetic neurons in the PVN.


Subject(s)
Angiotensin II/metabolism , Hypothalamus/metabolism , Receptor, Angiotensin, Type 1/metabolism , Receptors, Mineralocorticoid/metabolism , Subfornical Organ/metabolism , Aldosterone/metabolism , Angiotensin II/administration & dosage , Animals , Blood Pressure/physiology , Dependovirus , Gene Knockdown Techniques , Genetic Vectors , Heart Rate/physiology , Hypertension/metabolism , Male , Medulla Oblongata/metabolism , RNA, Messenger/metabolism , RNA, Small Interfering , Random Allocation , Rats, Wistar , Reactive Oxygen Species/metabolism , Receptor, Angiotensin, Type 1/genetics , Receptors, Mineralocorticoid/genetics
7.
J Med Food ; 18(8): 872-81, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25799023

ABSTRACT

The beneficial effects of edible mushrooms for improving chronic intractable diseases have been documented. However, the antiatherogenic activity of the new medicinal mushroom Grifola gargal is unknown. Therefore, we evaluated whether Grifola gargal can prevent or delay the progression of atherosclerosis. Atherosclerosis was induced in ApoE lipoprotein-deficient mice by subcutaneous infusion of angiotensin II. Grifola gargal extract (GGE) was prepared and intraperitoneally injected. The weight of heart and vessels, dilatation/atheroma formation of thoracic and abdominal aorta, the percentage of peripheral granulocytes, and the blood concentration of MCP-1/CCL2 were significantly reduced in mice treated with GGE compared to untreated mice. By contrast, the percentage of regulatory T cells and the plasma concentration of SDF-1/CXCL12 were significantly increased in mice treated with the mushroom extract compared to untreated mice. In vitro, GGE significantly increased the secretion of SDF-1/CXCL12, VEGF, and TGF-ß1 from fibroblasts compared to control. This study demonstrated for the first time that Grifola gargal therapy can enhance regulatory T cells and ameliorate atherosclerosis in mice.


Subject(s)
Agaricales/chemistry , Atherosclerosis/diet therapy , Biological Products/pharmacology , Blood Vessels/drug effects , Grifola/chemistry , Heart/drug effects , Angiotensin II/administration & dosage , Angiotensin II/toxicity , Animals , Apolipoproteins E/deficiency , Atherosclerosis/chemically induced , Atherosclerosis/metabolism , Atherosclerosis/pathology , Biological Products/administration & dosage , Biological Products/chemistry , Blood Vessels/pathology , Chemokine CCL2/metabolism , Chemokine CXCL12/metabolism , Disease Models, Animal , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Granulocytes/drug effects , Heart/physiopathology , Injections, Intraperitoneal , Mice , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta1/metabolism , Treatment Outcome , Vascular Endothelial Growth Factor A/metabolism
8.
Ann Vasc Surg ; 29(2): 328-40, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25449986

ABSTRACT

BACKGROUND: The blotchy mouse caused by mutations of ATP7A develops low blood copper and aortic aneurysm and rupture. Although the aortic pathologies are believed primarily due to congenital copper deficiencies in connective tissue, perinatal copper supplementation does not produce significant therapeutic effects, hinting additional mechanisms in the symptom development, such as an independent effect of the ATP7A mutations during adulthood. METHODS: We investigated if bone marrow from blotchy mice contributes to these symptoms. For these experiments, bone marrow from blotchy mice (blotchy marrow group) and healthy littermate controls (control marrow group) was used to reconstitute recipient mice (irradiated male low-density lipoprotein receptor -/- mice), which were then infused with angiotensin II (1,000 ng/kg/min) for 4 weeks. RESULTS: By using Mann-Whitney U test, our results showed that there was no significant difference in the copper concentrations in plasma and hematopoietic cells between these 2 groups. And plasma level of triglycerides was significantly reduced in blotchy marrow group compared with that in control marrow group (P < 0.05), whereas there were no significant differences in cholesterol and phospholipids between these 2 groups. Furthermore, a bead-based multiplex immunoassay showed that macrophage inflammatory protein (MIP)-1ß, monocyte chemotactic protein (MCP)-1, MCP-3, MCP-5, tissue inhibitor of metalloproteinases (TIMP)-1, and vascular endothelial growth factor (VEGF)-A production was significantly reduced in the plasma of blotchy marrow group compared with that in control marrow group (P < 0.05). More important, although angiotensin II infusion increased maximal external aortic diameters in thoracic and abdominal segments, there was no significant difference in the aortic diameters between these 2 groups. Furthermore, aortic ruptures, including transmural breaks of the elastic laminae in the abdominal segment and lethal rupture in the thoracic segment, were observed in blotchy marrow group but not in control marrow group; however, there was no significant difference in the incidence of aortic ruptures between these 2 groups (P = 0.10; Fisher's exact test). CONCLUSIONS: Overall, our study indicated that the effect of bone marrow from blotchy mice during adulthood is dispensable in the regulation of blood copper, plasma cholesterol and phospholipids levels, and aortic pathologies, but contributes to a reduction of MIP-1ß, MCP-1, MCP-3, MCP-5, TIMP-1, and VEGF-A production and triglycerides concentration in plasma. Our study also hints that bone marrow transplantation cannot serve as an independent treatment option.


Subject(s)
Aortic Aneurysm/physiopathology , Bone Marrow/metabolism , Copper/metabolism , Adenosine Triphosphatases/genetics , Angiotensin II/administration & dosage , Animals , Aortic Aneurysm/blood , Aortic Aneurysm/metabolism , Aortic Rupture/blood , Aortic Rupture/metabolism , Aortic Rupture/physiopathology , Biomarkers/blood , Bone Marrow/physiopathology , Bone Marrow Transplantation , Cardiovascular Agents/administration & dosage , Cation Transport Proteins/genetics , Copper/blood , Copper-Transporting ATPases , Cytokines/blood , Disease Models, Animal , Enzymes/blood , Female , Lipids/blood , Male , Mice , Mice, Inbred Strains , Receptors, LDL/genetics
9.
PLoS One ; 9(2): e89307, 2014.
Article in English | MEDLINE | ID: mdl-24558493

ABSTRACT

BACKGROUND: Recent observational clinical and ex-vivo studies suggest that inflammation and in particular leukocyte activation predisposes to atrial fibrillation (AF). However, whether local binding and extravasation of leukocytes into atrial myocardium is an essential prerequisite for the initiation and propagation of AF remains elusive. Here we investigated the role of atrial CD11b/CD18 mediated infiltration of polymorphonuclear neutrophils (PMN) for the susceptibility to AF. METHODS AND RESULTS: C57bl/6J wildtype (WT) and CD11b/CD18 knock-out (CD11b(-/-)) mice were treated for 14 days with subcutaneous infusion of angiotensin II (Ang II), a known stimulus for PMN activation. Atria of Ang II-treated WT mice were characterized by increased PMN infiltration assessed in immunohistochemically stained sections. In contrast, atrial sections of CD11b(-/-) mice lacked a significant increase in PMN infiltration upon Ang II infusion. PMN infiltration was accompanied by profoundly enhanced atrial fibrosis in Ang II treated WT as compared to CD11b(-/-) mice. Upon in-vivo electrophysiological investigation, Ang II treatment significantly elevated the susceptibility for AF in WT mice if compared to vehicle treated animals given an increased number and increased duration of AF episodes. In contrast, animals deficient of CD11b/CD18 were entirely protected from AF induction. Likewise, epicardial activation mapping revealed decreased electrical conduction velocity in atria of Ang II treated WT mice, which was preserved in CD11b(-/-) mice. In addition, atrial PMN infiltration was enhanced in atrial appendage sections of patients with persistent AF as compared to patients without AF. CONCLUSIONS: The current data critically link CD11b-integrin mediated atrial PMN infiltration to the formation of fibrosis, which promotes the initiation and propagation of AF. These findings not only reveal a mechanistic role of leukocytes in AF but also point towards a potential novel avenue of treatment in AF.


Subject(s)
Angiotensin II/pharmacology , Atrial Fibrillation/etiology , Atrial Fibrillation/metabolism , CD11b Antigen/metabolism , CD18 Antigens/metabolism , Neutrophils/metabolism , Analysis of Variance , Angiotensin II/administration & dosage , Animals , CD11b Antigen/genetics , CD18 Antigens/genetics , Electrophysiologic Techniques, Cardiac/methods , Fluorescent Antibody Technique , Heart Atria/metabolism , Immunoblotting , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/drug effects
11.
Am J Physiol Cell Physiol ; 306(8): C736-44, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24401846

ABSTRACT

The existence of a local renin-angiotensin system (RAS) in neurons was first postulated 40 years ago. Further studies indicated intraneuronal generation of ANG II. However, the function and signaling mechanisms of intraneuronal ANG II remained elusive. Since ANG II type 1 receptor (AT1R) is the major type of receptor mediating the effects of ANG II, we used intracellular microinjection and concurrent Ca(2+) and voltage imaging to examine the functionality of intracellular AT1R in neurons. We show that intracellular administration of ANG II produces a dose-dependent elevation of cytosolic Ca(2+) concentration ([Ca(2+)]i) in hypothalamic neurons that is sensitive to AT1R antagonism. Endolysosomal, but not Golgi apparatus, disruption prevents the effect of microinjected ANG II on [Ca(2+)]i. Additionally, the ANG II-induced Ca(2+) response is dependent on microautophagy and sensitive to inhibition of PLC or antagonism of inositol 1,4,5-trisphosphate receptors. Furthermore, intracellular application of ANG II produces AT1R-mediated depolarization of hypothalamic neurons, which is dependent on [Ca(2+)]i increase and on cation influx via transient receptor potential canonical channels. In summary, we provide evidence that intracellular ANG II activates endolysosomal AT1Rs in hypothalamic neurons. Our results point to the functionality of a novel intraneuronal angiotensinergic pathway, extending the current understanding of intracrine ANG II signaling.


Subject(s)
Angiotensin II/metabolism , Neurons/physiology , Signal Transduction/physiology , Angiotensin II/administration & dosage , Angiotensin II/pharmacology , Angiotensin II Type 1 Receptor Blockers/administration & dosage , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Animals, Newborn , Calcium/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , Female , Gene Expression Regulation , Humans , Hypothalamus/cytology , Inositol 1,4,5-Trisphosphate/metabolism , Inositol 1,4,5-Trisphosphate Receptors/genetics , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Male , Microinjections , Neurons/drug effects , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1/drug effects , Receptor, Angiotensin, Type 1/metabolism
12.
Hypertension ; 63(2): 273-80, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24191286

ABSTRACT

Observational studies in primary hyperaldosteronism suggest a positive relationship between aldosterone and parathyroid hormone (PTH); however, interventions to better characterize the physiological relationship between the renin-angiotensin-aldosterone system (RAAS) and PTH are needed. We evaluated the effect of individual RAAS components on PTH using 4 interventions in humans without primary hyperaldosteronism. PTH was measured before and after study (1) low-dose angiotensin II (Ang II) infusion (1 ng/kg per minute) and captopril administration (25 mg×1); study (2) high-dose Ang II infusion (3 ng/kg per minute); study (3) blinded crossover randomization to aldosterone infusion (0.7 µg/kg per hour) and vehicle; and study (4) blinded randomization to spironolactone (50 mg/daily) or placebo for 6 weeks. Infusion of Ang II at 1 ng/kg per minute acutely increased aldosterone (+148%) and PTH (+10.3%), whereas Ang II at 3 ng/kg per minute induced larger incremental changes in aldosterone (+241%) and PTH (+36%; P<0.01). Captopril acutely decreased aldosterone (-12%) and PTH (-9.7%; P<0.01). In contrast, aldosterone infusion robustly raised serum aldosterone (+892%) without modifying PTH. However, spironolactone therapy during 6 weeks modestly lowered PTH when compared with placebo (P<0.05). In vitro studies revealed the presence of Ang II type I and mineralocorticoid receptor mRNA and protein expression in normal and adenomatous human parathyroid tissues. We observed novel pleiotropic relationships between RAAS components and the regulation of PTH in individuals without primary hyperaldosteronism: the acute modulation of PTH by the RAAS seems to be mediated by Ang II, whereas the long-term influence of the RAAS on PTH may involve aldosterone. Future studies to evaluate the impact of RAAS inhibitors in treating PTH-mediated disorders are warranted.


Subject(s)
Angiotensin II/administration & dosage , Captopril/administration & dosage , Hypertension/drug therapy , Parathyroid Hormone/blood , Renin-Angiotensin System/drug effects , Spironolactone/administration & dosage , Adult , Aldosterone/administration & dosage , Aldosterone/metabolism , Antihypertensive Agents/administration & dosage , Diuretics/administration & dosage , Dose-Response Relationship, Drug , Female , Humans , Hyperaldosteronism/drug therapy , Hyperaldosteronism/metabolism , Hyperaldosteronism/physiopathology , Hypertension/metabolism , Hypertension/physiopathology , Male , Middle Aged , Parathyroid Glands/drug effects , Parathyroid Glands/physiology , Renin-Angiotensin System/physiology , Vasoconstrictor Agents/administration & dosage , Vitamin D/blood
13.
Regul Pept ; 185: 37-43, 2013 Aug 10.
Article in English | MEDLINE | ID: mdl-23816464

ABSTRACT

The aim of the present study was to determine if insulin is able to modulate the pressor response to intracerebroventricularly administered angiotensin II in insulin resistant fructose overloaded rats. Male Sprague-Dawley rats were divided into two groups: 1) Control group (C) with tap water to drink for 6 weeks (n=36); and 2) fructose treated (F), with fructose solution (10% w/v) to drink for 6 weeks (n=36). On the day of the experiment, anesthetized male C and F rats were intracerebroventricularly infused with insulin (12 mU/h, n=15) or Ringer's solution as vehicle (n=15) for 2h. Immediately, changes in mean arterial pressure (MAP) in response to an intracerebroventricular subpressor dose of angiotensin II (5 pmol, n=10) or vehicle (n=5) were measured for 10 min. Then, hypothalami were removed and Akt and ERK1/2 phosphorylation levels were determined. In a subset of C (n=10) and F (n=20) animals, PD98059 (p44/42 MAPK inhibitor) or vehicle was administered intracerebroventricularly at a flow rate of 5 µl/min for 1 min. Ten minutes later, insulin (12 mU/h, n=5 for each group) or vehicle (Ringer's solution, only in the F group, n=5) was perfused for 2h at a flow rate of 4 µl/h, and cardiovascular parameters were measured every 15 min. Immediately, changes in MAP and HR in response to a subpressor dose of Ang II (5 pmol/2 µl) were evaluated for 10 min (n=5 for each group). In other subset of animals (n=6 for each group), AT1 and AT2 hypothalamic receptor levels were measured by Western blotting. Intracerebroventricular insulin pre-treatment increased the pressor response to angiotensin II in C rats. In F rats (with or without insulin pretreatment), the pressor response to angiotensin II was higher than that in vehicle pre-treated C animals, but similar to that observed in C after insulin infusion. In C rats phospho-ERK 1/2 hypothalamic levels significantly increased after angiotensin II injection in insulin pretreated animals compared to vehicle pre-treated rats, suggesting that MAPK activation might be involved in insulin potentiation of blood pressure response to angiotensin II in the brain. Phospho-ERK 1/2 hypothalamic levels were significantly increased in vehicle treated F rats compared to C, suggesting that basal MAPK activation might play a role in the enhanced response to angiotensin II observed in these animals. Finally, in F rats, either after vehicle or insulin infusion, angiotensin II injection was associated with a similar increase in phospho-ERK 1/2 hypothalamic levels, comparable to that observed after angiotensin II injection in insulin pre-treated C animals. ERK 1/2 blockade significantly reduced MAP in F rats compared to C. Moreover, ERK 1/2 inhibition completely abolished the Ang II pressor response in F rats and in insulin pre-treated C animals. All these findings suggest that insulin-angiotensin II interaction at hypothalamic level might be involved in the increase in blood pressure observed in the insulin resistant state.


Subject(s)
Angiotensin II/physiology , Blood Pressure , Insulin/physiology , Metabolic Syndrome/physiopathology , Angiotensin II/administration & dosage , Animals , Fructose , Hypothalamus/metabolism , Injections, Intraventricular , Insulin/administration & dosage , Insulin Resistance , MAP Kinase Signaling System , Male , Metabolic Syndrome/chemically induced , Phosphorylation , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 2/metabolism , Vasoconstrictor Agents/administration & dosage , Vasomotor System/physiopathology
14.
Brain Res ; 1486: 62-7, 2012 Nov 27.
Article in English | MEDLINE | ID: mdl-23026079

ABSTRACT

In spite of evidence to the contrary, concern that substances injected into the fourth ventricle (4V) reach forebrain structures challenges the validity of using these injections to evaluate the role of hindbrain structures. Injection of AngII into the lateral ventricle (LV) increases water intake, but a similar response is not observed after injection into the 4V. This alone suggests the requirement of forebrain structures, but the potential for a counteracting, anti-dipsogenic pressor response to hindbrain AngII allows for lingering concern that this competing effect of AngII, rather than lack of forebrain access, underlies the negative result. Here, we used a double cannulation approach (LV and 4V) to evaluate the effect of the AngII receptor antagonist, losartan, on the drinking response to AngII injected into the LV. Injections of losartan into the LV blocked the dipsogenic response to AngII given 5min later into the LV. There was no effect, however, when losartan was injected into 4V, even when we used a dose of losartan that was 25 times greater than needed when injected into the LV. Collectively, these experiments suggest that concerns about diffusion from hindbrain ventricles to forebrain structures are overstated and can be circumvented using proper dose and timing of injections. Moreover, these data provide additional support to the existing literature showing that forebrain structures are key sites in the stimulation of drinking behavior by AngII.


Subject(s)
Angiotensin II/administration & dosage , Cerebral Ventricles/drug effects , Cerebral Ventricles/physiology , Drinking Behavior/drug effects , Drinking Behavior/physiology , Angiotensin II/physiology , Animals , Dose-Response Relationship, Drug , Drinking/drug effects , Drinking/physiology , Drug Evaluation, Preclinical , Injections, Intraventricular , Losartan/administration & dosage , Male , Prosencephalon/drug effects , Prosencephalon/physiology , Rats , Rats, Sprague-Dawley
15.
Lasers Surg Med ; 44(8): 664-74, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22911625

ABSTRACT

BACKGROUND AND OBJECTIVES: Recent in vitro studies by our group indicated that low level laser irradiation (LLLI) modifies cellular processes essential to the progression of abdominal aortic aneurysm (AAA). Using high-frequency ultrasonography (HF-u/s) in the angiotensin-II (Ang-II)-infused, apolipoprotein-E-deficient (Apo-E(-/-) ) mouse model of AAA, we found that LLLI markedly inhibited aneurysm formation and preserved arterial wall elasticity. We now report, using quantitative histopathology, the likely mechanism underlying the preventative effect of LLLI on aneurysm formation in this model. STUDY DESIGN/MATERIALS AND METHODS: This study was performed on 32 Apo-E(-/-) mice of which 10 were Ang-II-infused and LLL-irradiated (780 nm, 2 J/cm(2) , 9-minutes), 12 were Ang-II-infused but not irradiated, and 10 were saline infused. The aortas were excised at 28d, sectioned at 250 µm intervals, and stained with H + E, Movat-pentachrome and picrosirius-red for histomorphometry, and immunostained with Mac-2 and α-actin for detection of macrophages and SMCs, respectively. RESULTS: Transmural disruptions of the aorta occurred with distinct predilection for branch orifices. In the LLLI-treated animals, the frequency of these disruptions was lower (#branches with break points: 17 of 40 vs. 32 of 48, P = 0.023 by Chi-squared), their size smaller (length [mm]: 0.48 ± 0.26 vs. 0.98 ± 1.42, P = 0.044 by ANOVA with FPLSD), and the number of Mac-2-positive macrophages in the intramural areas of these disruptions lower than in the non-treated control (#Macrophages/0.01 mm(2) at break points: 11.6 ± 7.2 vs. 26.0 ± 15.7, P = 0.016 by Kruskal-Wallis). The average size of the medial SMCs was larger reflecting a heightened synthetic state (SMC size [µm(2) ]: 463.9 ± 61.4 vs. 354.9 ± 71.7, P = 0.001 by ANOVA with FPLSD). Furthermore, at sites of transmural disruption, the %area occupied by collagen of the overall area of attempted repair (%Col/WO) was significantly greater in the LLLI-treated animals versus control (%Col/WO: 41 ± 13 vs. 32 ± 16, P = 0.009 by ANOVA with FPLSD). CONCLUSION: Enhanced matrix reinforcement and modification of the inflammatory response at sites of transmural injury are prominent mechanisms by which LLLI reduces AAA progression in this model.


Subject(s)
Adventitia/metabolism , Aortic Aneurysm, Abdominal/radiotherapy , Collagen/metabolism , Low-Level Light Therapy , Adventitia/pathology , Analysis of Variance , Angiotensin II/administration & dosage , Animals , Aorta, Abdominal , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Smooth Muscle/metabolism , Vasoconstrictor Agents/administration & dosage
16.
J Neuroendocrinol ; 24(7): 1078-84, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22375892

ABSTRACT

Nesfatin-1, a post-translational product of the nucleobindin-2 (NucB2) gene, is produced in several brain areas known to be important in neuroendocrine, autonomic and metabolic function, including the hypothalamus and medulla. The hallmark action of the peptide is its ability at picomole doses to inhibit food and water intake in rodents and, indeed, the effect on water intake is more pronounced than that on food intake. In preliminary studies, we observed a decrease in hypothalamic NucB2 expression in response to overnight water deprivation even when food was present, which reversed when water was returned to the animals. We therefore hypothesised that the effect of nesfatin-1 on water drinking was independent of its anorexigenic action. Indeed, rats administered nesfatin-1 i.c.v. consumed significantly less water than controls in response to a subsequent, dipsogenic dose of angiotensin II, or upon return of water bottles after 18 h of fluid restriction (food present), or in response to a hypertonic challenge. Pretreatment with an antisense oligonucleotide against nesfatin-1 significantly reduced levels of immunoreactive nesfatin-1 in the hypothalamic paraventricular nucleus and resulted in exaggerated drinking responses to angiotensin II. The results obtained in the present study suggest that locally produced nesfatin-1 may be an important component of the hypothalamic mechanisms controlling fluid and electrolyte homeostasis.


Subject(s)
Calcium-Binding Proteins/physiology , DNA-Binding Proteins/physiology , Drinking/genetics , Nerve Tissue Proteins/physiology , Angiotensin II/administration & dosage , Angiotensin II/pharmacology , Animals , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/pharmacology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/pharmacology , Dose-Response Relationship, Drug , Drinking/drug effects , Drinking Behavior/drug effects , Drinking Behavior/physiology , Drug Evaluation, Preclinical , Hypothalamus/drug effects , Hypothalamus/metabolism , Hypothalamus/physiology , Injections, Intraventricular , Male , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/pharmacology , Nucleobindins , Rats , Rats, Sprague-Dawley , Thirst/drug effects , Thirst/physiology , Tissue Distribution/drug effects , Water Deprivation/physiology , Water-Electrolyte Balance/drug effects , Water-Electrolyte Balance/genetics
17.
Intensive Care Med ; 38(5): 886-93, 2012 May.
Article in English | MEDLINE | ID: mdl-22302028

ABSTRACT

PURPOSE: To measure renal adenosine triphosphate (ATP) (bioenergetics) during hypotensive sepsis with or without angiotensin II (Ang II) infusion. METHODS: In anaesthetised sheep implanted with a renal artery flow probe and a magnetic resonance coil around one kidney, we induced hypotensive sepsis with intravenous Escherichia coli injection. We measured mean arterial pressure (MAP), heart rate, renal blood flow RBF and renal ATP levels using magnetic resonance spectroscopy. After 2 h of sepsis, we randomly assigned sheep to receive an infusion of Ang II or vehicle intravenously and studied the effect of treatment on the same variables. RESULTS: After E. coli administration, the experimental animals developed hypotensive sepsis (MAP from 92 ± 9 at baseline to 58 ± 4 mmHg at 4 h). Initially, RBF increased, then, after 4 h, it decreased below control levels (from 175 ± 28 at baseline to 138 ± 27 mL/min). Despite decreased RBF and hypotension, renal ATP was unchanged (total ATP to inorganic phosphate ratio from 0.69 ± 0.02 to 0.70 ± 0.02). Ang II infusion restored MAP but caused significant renal vasoconstriction. However, it induced no changes in renal ATP (total ATP to inorganic phosphate ratio from 0.79 ± 0.03 to 0.80 ± 0.02). CONCLUSIONS: During early hypotensive experimental gram-negative sepsis, there was no evidence of renal bioenergetic failure despite decreased RBF. In this setting, the addition of a powerful renal vasoconstrictor does not lead to deterioration in renal bioenergetics.


Subject(s)
Angiotensin II/therapeutic use , Energy Metabolism/physiology , Gram-Negative Aerobic Bacteria/drug effects , Gram-Negative Bacterial Infections/physiopathology , Kidney/metabolism , Sepsis/physiopathology , Vasoconstrictor Agents/therapeutic use , Adenosine Triphosphate/urine , Angiotensin II/administration & dosage , Angiotensin II/pharmacology , Animals , Escherichia coli/pathogenicity , Sepsis/etiology , Sheep , Vasoconstrictor Agents/administration & dosage , Vasoconstrictor Agents/pharmacology
18.
Heart Vessels ; 27(4): 419-23, 2012 Jul.
Article in English | MEDLINE | ID: mdl-21989861

ABSTRACT

The L/N-type calcium channel blocker cilnidipine has been shown to suppress aldosterone production induced by angiotensin II (Ang II) in vitro. In addition, cilnidipine also suppresses the reflex tachycardia induced by its antihypertensive action in vivo. We investigated the effects of cilnidipine on the reflex aldosterone production induced by its antihypertensive action, to identify the differences in the effects of cilnidipine from those of the L-type calcium channel blocker nifedipine. Male SHR/Izm rats were anesthetized by intraperitoneal injection of pentobarbital sodium, and administered an intravenous infusion of saline supplemented or not with Ang II for 30 min. Blood pressure was monitored continuously in the femoral artery. Each of the calcium channel blockers under study was administered intravenously as a bolus through the femoral vein 1 min after the start of the Ang II infusion, and blood samples were collected 30 min after the start of the Ang II infusion. Following administration at nonhypotensive doses, all calcium channel blockers tended to decrease the plasma aldosterone. In particular, cilnidipine significantly suppressed the plasma aldosterone levels. On the other hand, under the condition of Ang II-induced hypertension, administration of a hypotensive dosage of cilnidipine showed no effect on the plasma aldosterone levels, whereas a hypotensive dosage of nifedipine significantly increased the plasma aldosterone levels. Our results suggest that the L/N-type calcium channel blocker cilnidipine reduces the plasma aldosterone level by suppressing the aldosterone production induced by reflex upregulation of the renin-angiotensin-aldosterone system associated with reduction of the blood pressure.


Subject(s)
Aldosterone/blood , Antihypertensive Agents/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/drug effects , Calcium Channels, N-Type/drug effects , Dihydropyridines/pharmacology , Hypertension/drug therapy , Reflex/drug effects , Angiotensin II/administration & dosage , Animals , Antihypertensive Agents/administration & dosage , Blood Pressure/drug effects , Calcium Channel Blockers/administration & dosage , Calcium Channels, L-Type/metabolism , Calcium Channels, N-Type/metabolism , Dihydropyridines/administration & dosage , Disease Models, Animal , Down-Regulation , Hypertension/blood , Hypertension/physiopathology , Injections, Intravenous , Male , Nifedipine/pharmacology , Rats , Rats, Inbred SHR , Renin-Angiotensin System/drug effects
19.
Physiol Behav ; 105(4): 1076-81, 2012 Feb 28.
Article in English | MEDLINE | ID: mdl-22192708

ABSTRACT

Angiotensin II (AngII) plays a key role in maintaining body fluid homeostasis. The physiological and behavioral effects of central AngII include increased blood pressure and fluid intake. In vitro experiments demonstrate that repeated exposure to AngII reduces the efficacy of subsequent AngII, and behavioral studies indicate that prior icv AngII administration reduces the dipsogenic response to AngII administered later. Specifically, rats given a treatment regimen of three icv injections of a large dose of AngII, each separated by 20 min, drink less water in response to a test injection of AngII than do vehicle-treated controls given the same test injection. The present studies were designed to test three potential explanations for the reduced dipsogenic potency of AngII after repeated administration. To this end, we tested for motor impairment caused by repeated injections of AngII, for a possible role of visceral distress or illness, and for differences in the pressor response to the final test injection of AngII. We found that repeated injections of AngII neither affected drinking stimulated by carbachol nor did they produce a conditioned flavor avoidance. Furthermore, we found no evidence that differences in the pressor response to the final test injection of AngII accounted for the difference in intake. In light of these findings, we are able to reject these three explanations for the observed behavioral desensitization, and, we suggest instead that the mechanism for this phenomenon may be at the level of the receptor.


Subject(s)
Angiotensin II/pharmacology , Avoidance Learning/drug effects , Choice Behavior/drug effects , Drinking/drug effects , Drug Tolerance , Vasoconstriction/drug effects , Angiotensin II/administration & dosage , Animals , Carbachol/antagonists & inhibitors , Carbachol/pharmacology , Drug Interactions , Injections, Intraventricular , Male , Rats , Rats, Sprague-Dawley
20.
Endocrinology ; 152(12): 4957-65, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22009728

ABSTRACT

It is generally understood that angiotensin II (AngII) promotes follicle atresia in rats, although recent data suggested that this may not be true in cattle. In this study, we aimed to determine in vivo whether AngII alters follicle development in cattle, using intrafollicular injection of AngII or antagonist into the growing dominant follicle or the second largest subordinate follicle. Injection of saralasin, an AngII antagonist, into the growing dominant follicle inhibited follicular growth, and this inhibitory effect was overcome by systemic FSH supplementation. Injection of AngII into the dominant follicle did not affect follicular growth, whereas injection of AngII into the second largest follicle prevented the expected atresia of this subordinate follicle, and the treated follicle grew at the same rate as the dominant follicle for the next 24 h. Inhibition of AngII action in the dominant follicle decreased estradiol concentrations in follicular fluid and the abundance of mRNA encoding aromatase, 3ß-hydroxysteroid dehydrogenase, LH receptor, and cyclinD2 in granulosa cells, with minimal effects on theca cells. The effect of AngII on aromatase mRNA levels was confirmed using an in vitro granulosa cell culture system. In conclusion, these data suggest that AngII signaling promotes follicle growth in cattle and does so by regulating genes involved in estradiol secretion and granulosa cell proliferation and differentiation.


Subject(s)
Angiotensin II/pharmacology , Ovarian Follicle/growth & development , Signal Transduction , Angiotensin II/administration & dosage , Angiotensin II/physiology , Angiotensin II Type 1 Receptor Blockers , Animals , Aromatase , Cattle , Cell Differentiation , Cell Proliferation , Estradiol , Female , Gene Expression Regulation , Granulosa Cells/cytology , Granulosa Cells/drug effects , Ovarian Follicle/drug effects , Saralasin/administration & dosage , Saralasin/pharmacology , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL