Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
1.
AAPS PharmSciTech ; 25(5): 115, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38755324

ABSTRACT

More than 1 billion people worldwide suffer from hypertension; therefore, hypertension management has been categorized as a global health priority. Losartan potassium (LP) is an antihypertensive drug with a limited oral bioavailability of about 33% since it undergoes the initial metabolic cycle. Thus, nasal administration is a unique route to overcome first-pass metabolism. The investigation focused on the potential effects of LP-loaded spanlastic vesicles (SNVs) on LP pharmacodynamics and pharmacokinetic parameters, utilizing a thin-film hydration methodology established on a 3122 full factorial design. Entrapment efficiency (EE%) ranged from 39.8 ± 3.87.8 to 83.8 ± 2.92% for LP-SNVs. Vesicle size (VS) varied from 205.5 ± 6.5.10 to 445.1 ± 13.52 nm, and the percentage of LP released after 8 h (Q8h) ranged from 30.8 ± 3.10 to 68.8 ± 1.45%. LP permeated through the nasal mucosa during 24 h and flocculated from 194.1 ± 4.90 to 435.3 ± 13.53 µg/cm2. After twenty-four hours, the optimal LP-SNVs in-situ gel showed 2.35 times more permeation through the nasal mucosa than the LP solution. It also lowered systolic blood pressure, so it is thought to be better than the reference formulation in terms of pharmacodynamics. The pharmacokinetics studies demonstrated that the intranasal LP-SNVs gel boosted its bioavailability approximately 6.36 times compared to the oral LP solution. Our research showed that intranasal LP-SNVs could be a good nanoplatform because they are well-tolerated and have possible pharmacokinetics and pharmacodynamics.


Subject(s)
Antihypertensive Agents , Gels , Hypertension , Losartan , Losartan/pharmacokinetics , Losartan/administration & dosage , Losartan/pharmacology , Antihypertensive Agents/pharmacokinetics , Antihypertensive Agents/administration & dosage , Antihypertensive Agents/pharmacology , Animals , Hypertension/drug therapy , Male , Rats , Biological Availability , Administration, Intranasal , Nanoparticles/chemistry , Nasal Mucosa/metabolism , Nasal Mucosa/drug effects , Particle Size , Angiotensin II/pharmacokinetics , Angiotensin II/administration & dosage , Angiotensin II/pharmacology , Blood Pressure/drug effects , Rats, Wistar , Chemistry, Pharmaceutical/methods
3.
Crit Care ; 22(1): 69, 2018 Mar 20.
Article in English | MEDLINE | ID: mdl-29558991

ABSTRACT

This article is one of ten reviews selected from the Annual Update in Intensive Care and Emergency Medicine 2018. Other selected articles can be found online at https://www.biomedcentral.com/collections/annualupdate2018 . Further information about the Annual Update in Intensive Care and Emergency Medicine is available from http://www.springer.com/series/8901 .


Subject(s)
Critical Care/methods , Angiotensin II/pharmacokinetics , Angiotensin II/therapeutic use , Angiotensins/pharmacokinetics , Angiotensins/therapeutic use , Critical Care/trends , Humans , Hypotension/drug therapy , Hypotension/etiology , Shock, Septic/complications , Shock, Septic/drug therapy , Vasoconstrictor Agents/pharmacokinetics , Vasoconstrictor Agents/therapeutic use
4.
Ann Pharmacother ; 52(9): 920-927, 2018 09.
Article in English | MEDLINE | ID: mdl-29582666

ABSTRACT

OBJECTIVE: To review and summarize data on angiotensin II (AT-II), approved by the Food and Drug Administration (FDA) in December 2017 to increase blood pressure in adults with septic or other distributive shock. DATA SOURCES: A PubMed/MEDLINE search was conducted using the following terms: (angiotensin ii OR angiotensin 2) AND (shock) from 1966 to February 2018. STUDY SELECTION AND DATA EXTRACTION: A total of 691 citations were reviewed with only relevant clinical data extracted. DATA SYNTHESIS: AT-II is a peptide hormone with a multitude of physiological effects-namely, vasoconstriction of venous and arterial smooth muscle. The priority approval granted by the FDA was secondary to a phase 3 study of patients receiving at least 0.2 µg/kg/min of norepinephrine or equivalent for vasodilatory shock. Compared with placebo, AT-II had a significantly higher rate of response, defined as a mean arterial pressure of 75 mm Hg or an increase of 10 mm Hg. No significant difference was found in death by day 28. CONCLUSIONS: AT-II is a newly available vasoactive agent with a novel mechanism for the treatment of distributive shock. Further research is needed to define its exact role in therapy of shock states, identify patients most likely to benefit, and further study its safety profile in critical illness.


Subject(s)
Angiotensin II/administration & dosage , Shock/drug therapy , Vasoconstrictor Agents/administration & dosage , Angiotensin II/adverse effects , Angiotensin II/pharmacokinetics , Animals , Humans , Shock/metabolism , Treatment Outcome , Vasoconstrictor Agents/adverse effects , Vasoconstrictor Agents/pharmacokinetics
5.
Aging (Albany NY) ; 10(1): 100-114, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29348391

ABSTRACT

The present study was designed to determine the effects of senescence and angiotensin II (Ang II) on expression and processing of amyloid precursor protein (APP) in human brain microvascular endothelial cells (BMECs). Senescence caused a decrease in APP expression thereby resulting in reduced secretion of soluble APPα (sAPPα). In contrast, ß-site APP cleaving enzyme (BACE1) expression and production of amyloid ß (Aß)40 were increased in senescent endothelium. Importantly, in senescent human BMECs, treatment with BACE1 inhibitor IV inhibited Aß generation and increased sAPPα production by enhancing a disintegrin and metalloprotease (ADAM)10 expression. Furthermore, Ang II impaired expression of ADAM10 and significantly reduced generation of sAPPα in senescent human BMECs. This inhibitory effect of Ang II was prevented by treatment with BACE1 inhibitor IV. Our results suggest that impairment of α-processing and shift to amyloidogenic pathway of APP contribute to endothelial dysfunction induced by senescence. Loss of sAPPα in senescent cells treated with Ang II exacerbates detrimental effects of senescence on APP processing. Notably, inhibition of BACE1 has beneficial effects on senescence induced endothelial dysfunction. Reported findings may help to explain contributions of senescent cerebral microvascular endothelium to development of cerebral amyloid angiopathy and Alzheimer's disease (AD) pathology.


Subject(s)
Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Angiotensin II/pharmacokinetics , Aspartic Acid Endopeptidases/metabolism , Endothelial Cells/drug effects , Vasoconstrictor Agents/pharmacology , Aging , Alzheimer Disease/genetics , Amyloid beta-Peptides/genetics , Amyloid beta-Protein Precursor/genetics , Blotting, Western , Brain , Cerebral Amyloid Angiopathy , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation , Humans
6.
J Control Release ; 232: 20-8, 2016 06 28.
Article in English | MEDLINE | ID: mdl-27072027

ABSTRACT

Low efficacy of cationic polymer-based formulations (polyplexes) for systemic gene delivery to tumors remains the crucial concern for their clinical translation. Here we show that modulating the physiological state of a tumor using clinically approved pharmaceuticals can improve delivery of intravenously injected polyplexes to murine melanoma tumors with different characteristics. Direct comparison of drugs with different mechanisms of action has shown that application of nitroglycerin or losartan improved extravasation and tumor uptake of polyplex nanoparticles, whereas angiotensin II had almost no effect on polyplex accumulation and microdistribution in the tumor tissue. Application of nitroglycerin and losartan caused from 2- to 6-fold enhanced efficacy of polyplex-mediated gene delivery depending on the tumor model. The results obtained on polyplex behavior in tumor tissues depending on physiological state of the tumor can be relevant to optimize delivery of polyplexes and other nanomedicines with similar physicochemical properties.


Subject(s)
Drug Delivery Systems , Gene Transfer Techniques , Melanoma, Experimental/therapy , Administration, Intravenous , Angiotensin II/administration & dosage , Angiotensin II/pharmacokinetics , Angiotensin II/pharmacology , Animals , Cell Line, Tumor , Collagen Type I/metabolism , DNA/administration & dosage , Female , Gene Expression Regulation, Neoplastic , Green Fluorescent Proteins/genetics , Losartan/administration & dosage , Losartan/pharmacokinetics , Losartan/pharmacology , Luciferases, Firefly/genetics , Melanoma, Experimental/genetics , Melanoma, Experimental/metabolism , Melanoma, Experimental/physiopathology , Mice, Inbred C57BL , Mice, Inbred DBA , Nanoparticles/administration & dosage , Nitroglycerin/administration & dosage , Nitroglycerin/pharmacokinetics , Nitroglycerin/pharmacology , Oligopeptides/administration & dosage , Oligopeptides/pharmacokinetics , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/pharmacokinetics , Polyethylene Glycols/pharmacology , Polyethyleneimine/administration & dosage , Polyethyleneimine/analogs & derivatives , Polyethyleneimine/pharmacokinetics , Polyethyleneimine/pharmacology , Receptor, Melanocortin, Type 1/metabolism , Regional Blood Flow/drug effects , Vasoconstrictor Agents/administration & dosage , Vasoconstrictor Agents/pharmacokinetics , Vasoconstrictor Agents/pharmacology , Vasodilator Agents/administration & dosage , Vasodilator Agents/pharmacokinetics , Vasodilator Agents/pharmacology
7.
J Am Soc Nephrol ; 26(9): 2129-38, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25556166

ABSTRACT

Recently, the kelch-like protein 3 (KLHL3)-Cullin3 complex was identified as an E3 ubiquitin ligase for with no lysine (WNK) kinases, and the impaired ubiquitination of WNK4 causes pseudohypoaldosteronism type II (PHAII), a hereditary hypertensive disease. However, the involvement of WNK kinase regulation by ubiquitination in situations other than PHAII has not been identified. Previously, we identified the WNK3-STE20/SPS1-related proline/alanine-rich kinase-Na/K/Cl cotransporter isoform 1 phosphorylation cascade in vascular smooth muscle cells and found that it constitutes an important mechanism of vascular constriction by angiotensin II (AngII). In this study, we investigated the involvement of KLHL proteins in AngII-induced WNK3 activation of vascular smooth muscle cells. In the mouse aorta and mouse vascular smooth muscle (MOVAS) cells, KLHL3 was not expressed, but KLHL2, the closest homolog of KLHL3, was expressed. Salt depletion and acute infusion of AngII decreased KLHL2 and increased WNK3 levels in the mouse aorta. Notably, the AngII-induced changes in KLHL2 and WNK3 expression occurred within minutes in MOVAS cells. Results of KLHL2 overexpression and knockdown experiments in MOVAS cells confirmed that KLHL2 is the major regulator of WNK3 protein abundance. The AngII-induced decrease in KLHL2 was not caused by decreased transcription but increased autophagy-mediated degradation. Furthermore, knockdown of sequestosome 1/p62 prevented the decrease in KLHL2, suggesting that the mechanism of KLHL2 autophagy could be selective autophagy mediated by sequestosome 1/p62. Thus, we identified a novel component of signal transduction in AngII-induced vascular contraction that could be a promising drug target.


Subject(s)
Angiotensin II/pharmacokinetics , Microfilament Proteins/metabolism , Muscle Tonus/physiology , Nerve Tissue Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Adaptor Proteins, Signal Transducing/genetics , Angiotensin II/pharmacology , Animals , Aorta , Autophagy/drug effects , Cells, Cultured , Gene Knockdown Techniques , Heat-Shock Proteins/genetics , Mice , Microfilament Proteins/drug effects , Microfilament Proteins/genetics , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Nerve Tissue Proteins/drug effects , Nerve Tissue Proteins/genetics , Protein Serine-Threonine Kinases/drug effects , Sequestosome-1 Protein , Sodium, Dietary/pharmacology , Solute Carrier Family 12, Member 2/metabolism , Vasoconstriction
8.
PLoS One ; 9(8): e105762, 2014.
Article in English | MEDLINE | ID: mdl-25147932

ABSTRACT

The recent identification of a novel binding site for angiotensin (Ang) II as the peptidase neurolysin (E.C. 3.4.24.16) has implications for the renin-angiotensin system (RAS). This report describes the distribution of specific binding of 125I-Sarcosine1, Isoleucine8 Ang II (125I-SI Ang II) in neurolysin knockout mouse brains compared to wild-type mouse brains using quantitative receptor autoradiography. In the presence of p-chloromercuribenzoic acid (PCMB), which unmasks the novel binding site, widespread distribution of specific (3 µM Ang II displaceable) 125I-SI Ang II binding in 32 mouse brain regions was observed. Highest levels of binding >700 fmol/g initial wet weight were seen in hypothalamic, thalamic and septal regions, while the lowest level of binding <300 fmol/g initial wet weight was in the mediolateral medulla. 125I-SI Ang II binding was substantially higher by an average of 85% in wild-type mouse brains compared to neurolysin knockout brains, suggesting the presence of an additional non-AT1, non-AT2, non-neurolysin Ang II binding site in the mouse brain. Binding of 125I-SI Ang II to neurolysin in the presence of PCMB was highest in hypothalamic and ventral cortical brain regions, but broadly distributed across all regions surveyed. Non-AT1, non-AT2, non-neurolysin binding was also highest in the hypothalamus but had a different distribution than neurolysin. There was a significant reduction in AT2 receptor binding in the neurolysin knockout brain and a trend towards decreased AT1 receptor binding. In the neurolysin knockout brains, the size of the lateral ventricles was increased by 56% and the size of the mid forebrain (-2.72 to +1.48 relative to Bregma) was increased by 12%. These results confirm the identity of neurolysin as a novel Ang II binding site, suggesting that neurolysin may play a significant role in opposing the pathophysiological actions of the brain RAS and influencing brain morphology.


Subject(s)
Angiotensin II , Metalloendopeptidases/metabolism , Prosencephalon/metabolism , Renin-Angiotensin System/physiology , Sarcosine , Angiotensin II/pharmacokinetics , Angiotensin II/pharmacology , Animals , Iodine Isotopes/pharmacokinetics , Iodine Isotopes/pharmacology , Metalloendopeptidases/genetics , Mice , Mice, Knockout , Receptor, Angiotensin, Type 2/genetics , Receptor, Angiotensin, Type 2/metabolism , Sarcosine/pharmacokinetics , Sarcosine/pharmacology
9.
J. physiol. biochem ; 70(2): 465-478, jun. 2014.
Article in English | IBECS | ID: ibc-122967

ABSTRACT

The aim of this study was to assess whether endogenous Ang II and oxidative stress produced by acute hypertonic sodium overload may regulate the expression of aquaporin-1 (AQP-1) and aquaporin-2 (AQP-2) in the kidney. Groups of anesthetized male Sprague–Dawley rats were infused with isotonic saline solution (control) or with hypertonic saline solution (Na group, 1 M NaCl), either alone or with losartan (10 mg kg−1) or tempol (0.5 mg min−1 kg−1) during 2 h. Renal function parameters were measured. Groups of unanesthetized animals were injected intraperitoneally with hypertonic saline solution, with or without free access to water intake, Na+W, and Na−W, respectively. The expression of AQP-1, AQP-2, Ang II, eNOS, and NF-kB were evaluated in the kidney by Western blot and immunohistochemistry. AQP-2 distribution was assessed by immunofluorescence. Na group showed increased natriuresis and diuresis, and Ang II and NF-kB expression, but decreased eNOS expression. Losartan or tempol enhanced further the diuresis, and AQP-2 and eNOS expression, as well as decreased Ang II and NF-kB expression. Confocal immunofluorescence imaging revealed labeling of AQP-2 in the apical plasma membrane with less labeling in the intracellular vesicles than the apical membrane in kidney medullary collecting duct principal cells both in C and Na groups. Importantly, our data also show that losartan and tempol induces a predominantly accumulation of AQP-2 in intracellular vesicles. In unanesthetized rats, Na+W group presented increased diuresis, natriuresis, and AQP-2 expression (112 ± 25 vs 64 ± 16; *p < 0.05). Water deprivation increased plasma sodium and diuresis but decreased AQP-2 (46 ± 22 vs 112 ± 25; §p < 0.05) and eNOS expression in the kidney. This study is a novel demonstration that renal endogenous Ang II-oxidative stress, induced in vivo in hypernatremic rats by an acute sodium overload, regulates AQP-2 expression


Subject(s)
Animals , Rats , Hyponatremia/drug therapy , Oxidative Stress , Aquaporins , Angiotensin II/pharmacokinetics , Protective Agents/pharmacokinetics , Disease Models, Animal
10.
Sci Rep ; 3: 2467, 2013.
Article in English | MEDLINE | ID: mdl-23959064

ABSTRACT

The circulating, endocrine renin-angiotensin system (RAS) is important to circulatory homeostasis, while ubiquitous tissue and cellular RAS play diverse roles, including metabolic regulation. Indeed, inhibition of RAS is associated with improved cellular oxidative capacity. Recently it has been suggested that an intra-mitochondrial RAS directly impacts on metabolism. Here we sought to rigorously explore this hypothesis. Radiolabelled ligand-binding and unbiased proteomic approaches were applied to purified mitochondrial sub-fractions from rat liver, and the impact of AngII on mitochondrial function assessed. Whilst high-affinity AngII binding sites were found in the mitochondria-associated membrane (MAM) fraction, no RAS components could be detected in purified mitochondria. Moreover, AngII had no effect on the function of isolated mitochondria at physiologically relevant concentrations. We thus found no evidence of endogenous mitochondrial AngII production, and conclude that the effects of AngII on cellular energy metabolism are not mediated through its direct binding to mitochondrial targets.


Subject(s)
Angiotensin II/pharmacology , Angiotensin II/pharmacokinetics , Mitochondria, Liver/metabolism , Receptor, Angiotensin, Type 1/metabolism , Renin-Angiotensin System/physiology , Animals , Cells, Cultured , Mitochondria, Liver/drug effects , Rats , Renin-Angiotensin System/drug effects
11.
J. physiol. biochem ; 68(3): 353-363, sept. 2012.
Article in English | IBECS | ID: ibc-122324

ABSTRACT

The aim of this study is to assess the effects of losartan and carvedilol on metabolic parameters and renal haemodynamic responses to angiotensin II (Ang II) and adrenergic agonists in the model of fructose-fed rat. Thirty-six Sprague-Dawley rats were fed for 8 weeks either 20% fructose solution (F) or tap water (C) ad libitum. F or C group received either losartan or carvedilol (10 mg/kg p.o.) daily for the last 3 weeks of the study (FL and L) and (FCV and CV), respectively, then in acute studies the renal vasoconstrictor actions of Ang II, noradrenaline (NA), phenylephrine (PE) and methoxamine (ME) were determined. Data, mean ± SEM were analysed using ANOVA with significance at P <0.05. Losartan and carvedilol decreased the area under the glucose tolerance curve of the fructose-fed group. The responses (%) to NA, PE, ME and Ang II in F were lower (P<0.05) than C (F vs. C, 17 ± 2 vs. 38 ± 3; 24 ± 2 vs. 48 ± 2; 12 ± 2 vs. 34 ± 2; 17 ± 2 vs. 26 ± 2), respectively. L had higher (P <0.05) responses to NA and PE while CV had blunted (P <0.05) responses to NA, PE and Ang II compared to C (L, CV vs. C, 47 ± 3, 9 ± 2 vs. 38 ± 3; 61 ± 3, 29 ± 3 vs. 48 ± 2; 16 ± 3, 4 ± 3 vs. 26 ± 2), respectively. FL but not FCV group had enhanced (P <0.05) responses to NA, PE and ME compared to F (FL vs. F, 33 ± 3 vs. 17 ± 2; 45 ± 3 vs. 24 ± 2; 26 ± 3 vs. 12 ± 2), respectively. Losartan and carvedilol had an important ameliorating effect on fructose-induced insulin resistance. Losartan treatment could be an effective tool to restore normal vascular reactivity in the renal circulation of the fructose-fed rat (AU)


Subject(s)
Animals , Rats , Losartan/pharmacokinetics , Fructose/metabolism , Kidney/metabolism , Metabolic Syndrome/drug therapy , Renal Circulation , Antihypertensive Agents/pharmacokinetics , Angiotensin II/pharmacokinetics , Insulin Resistance , Hemodynamics , Protective Agents/pharmacokinetics , Disease Models, Animal
12.
Hypertension ; 59(3): 614-20, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22275533

ABSTRACT

Overactivity of the sympathetic nervous system has long been implicated in the hypertensive response to elevated angiotensin II (Ang II) levels. Although recent studies suggest that high dietary salt may alter cardiovascular responses to Ang II, direct evidence demonstrating chronic activation of sympathetic nerve activity is lacking. The objective of this study was to determine whether a low dose of Ang II, on a background of high salt intake, would result in a chronic increase in renal sympathetic nerve activity (RSNA). Arterial pressure and RSNA were recorded via telemetry. Two groups of rabbits were studied: 1 group drank a 0.9% NaCl solution and received Ang II (20 ng/kg per minute for 21 days, Salt+Ang), and the other drank tap water throughout and was not infused with Ang II (Control). In the Salt+Ang group, mean arterial pressure increased over the first week and remain elevated by 18.5±4.1 mm Hg at day 21. RSNA was not significantly different between groups on day 7 but was significantly elevated in the Salt+Ang group on day 21 (13.5±3.2% compared with 6.8±0.8% in the Control group; P<0.05). Baroreflex control of RSNA showed a rightward shift on day 21, but not day 7, and baroreflex responses indicated that RSNA could not be completely suppressed when arterial pressure was increased. No changes were observed in either mean arterial pressure or RSNA variables in the Control group. Our results support the hypothesis that elevated Ang II levels, in conjunction with a high salt diet, have the ability to chronically increase RSNA and, thus, potentially contribute to the maintenance of hypertension.


Subject(s)
Angiotensin II/pharmacokinetics , Blood Pressure/drug effects , Hypertension/physiopathology , Kidney/innervation , Sodium Chloride, Dietary/pharmacology , Sympathetic Nervous System/drug effects , Telemetry/methods , Action Potentials/drug effects , Animals , Baroreflex/drug effects , Baroreflex/physiology , Blood Pressure/physiology , Chronic Disease , Disease Models, Animal , Female , Follow-Up Studies , Hypertension/blood , Hypertension/chemically induced , Kidney/drug effects , Male , Rabbits , Sympathetic Nervous System/physiopathology
15.
J Neurochem ; 112(5): 1223-34, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20028450

ABSTRACT

Intracerebroventricular (i.c.v.) administration of angiotensin IV (Ang IV) or Leu-Val-Val-haemorphin 7 (LVV-H7) improves memory performance in normal rats and reverses memory deficits in rat models for cognitive impairment. These memory effects were believed to be mediated via the putative 'AT4 receptor'. However, this binding site was identified as insulin-regulated aminopeptidase (IRAP). Correspondingly, Ang IV and LVV-H7 were characterised as IRAP inhibitors. This study investigates whether and how IRAP may be involved in the central effects of Ang IV and LVV-H7. We determined the effects of i.c.v. administration of Ang IV or LVV-H7 on hippocampal neurotransmitter levels using microdialysis in rats. We observed that Ang IV modulates hippocampal acetylcholine levels, whereas LVV-H7 does not. This discrepancy was reflected in the observation that Ang IV binds with micromolar affinity to the AT1 receptor whereas no binding affinity was observed for LVV-H7. Correspondingly, we demonstrated that the AT1 receptor is involved in the effects of Ang IV on hippocampal neurotransmitter levels and on spatial working memory in a plus maze spontaneous alternation task. However, the AT1 receptor was not involved in the spatial memory facilitating effect of LVV-H7. Finally, we demonstrated that Ang IV did not diffuse to the hippocampus following i.c.v. injection, suggesting an extrahippocampal site of action. We propose that AT1 receptors are implicated in the neurochemical and cognitive effects of Ang IV, whereas LVV-H7 may mediate its effects via IRAP.


Subject(s)
Angiotensin II/analogs & derivatives , Hemoglobins/administration & dosage , Hippocampus/drug effects , Memory, Short-Term/drug effects , Neurotransmitter Agents/metabolism , Peptide Fragments/administration & dosage , Receptor, Angiotensin, Type 1/physiology , Angiotensin II/administration & dosage , Angiotensin II/pharmacokinetics , Animals , Dose-Response Relationship, Drug , Hippocampus/diagnostic imaging , Hippocampus/metabolism , Injections, Intraventricular/methods , Iodine Isotopes/pharmacokinetics , Male , Maze Learning/drug effects , Microdialysis/methods , Protein Binding/drug effects , Rats , Rats, Wistar , Space Perception/drug effects , Tissue Distribution/drug effects , Tomography, Emission-Computed, Single-Photon/methods
16.
Exp Eye Res ; 89(1): 109-18, 2009 Jun 15.
Article in English | MEDLINE | ID: mdl-19281810

ABSTRACT

Accumulation of specific deposits and extracellular molecules under the retinal pigment epithelium (RPE) has been previously observed in eyes with age-related macular degeneration (AMD) and may play a role in the pathogenesis of AMD. Even though age is the major determinant for developing AMD, clinical studies have revealed hypertension (HTN) as another systemic risk factor. Angiotensin II (Ang II) is considered the most important hormone associated with HTN. To evaluate the relationship of Ang II to AMD, we studied whether mouse RPE expresses functional Ang II receptor subtypes and whether HTN-induced Ang II regulates expression of these receptors as well as critical ECM molecules (MMP-2 and type IV collagen) involved in ECM turnover in RPE. We used 9-month-old C57BL/6 male mice infused with Ang II alone or Ang II in combination with the AT1 receptor antagonist candesartan or the AT2 receptor antagonist PD123319 for 4 weeks to determine whether HTN-associated Ang II was important for ECM regulation in RPE. We found that mouse RPE expressed both Ang II receptor subtypes at the mRNA and protein levels. Infusion with Ang II induced HTN and elevated plasma and ocular Ang II levels. Ang II also regulated AT1a and AT1b receptor mRNA expression, the intracellular concentration of calcium [Ca(2+)](i), MMP-2 activity, and type IV collagen accumulation. Concurrent administration of Ang II with the AT1 receptor blocker prevented the increase in blood pressure and rise in ocular Ang II levels, as well as the calcium and MMP-2 responses. In contrast, the type IV collagen response to Ang II was prevented by blockade of AT2 receptors, but not AT1 receptors. Plasma Ang II levels were not modified by the AT1 or AT2 receptor blockade. Since the effects of Ang II on MMP-2 and type IV collagen require inhibition of both Ang II receptor subtypes, these receptors may play a role as a potential therapeutic targets to prevent ECM turnover dysregulation in the RPE basement membrane, suggesting a pathogenic mechanism to explain the link between HTN and AMD.


Subject(s)
Angiotensin II/pharmacology , Extracellular Matrix/drug effects , Hypertension/metabolism , Retinal Pigment Epithelium/drug effects , Angiotensin II/pharmacokinetics , Animals , Blood Pressure/drug effects , Calcium Signaling/drug effects , Collagen Type IV/metabolism , Extracellular Matrix/metabolism , Eye/metabolism , Eye Proteins/biosynthesis , Eye Proteins/genetics , Eye Proteins/physiology , Gene Expression Regulation/drug effects , Macular Degeneration/metabolism , Male , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , Receptor, Angiotensin, Type 1/biosynthesis , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/physiology , Receptor, Angiotensin, Type 2/biosynthesis , Receptor, Angiotensin, Type 2/genetics , Receptor, Angiotensin, Type 2/physiology , Retinal Pigment Epithelium/metabolism , Tissue Inhibitor of Metalloproteinase-2/metabolism
17.
Eur J Pharm Biopharm ; 68(3): 724-34, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17618097

ABSTRACT

A transient reduction in the barrier nature of the skin can be a pre-requisite for successful (trans)dermal delivery of some drugs. The aim of this present study was to investigate and effect of a dermal abrading "rotating brush" device on percutaneous absorption and skin integrity. In vitro experiments were conducted using excised human epidermal membrane. The effect of device parameters (bristle type, treatment duration and applied pressure) on skin permeability of model solutes (methyl paraben, butyl paraben, caffeine, acyclovir and angiotensin II) with varying physicochemical properties was examined and compared to established methods of skin penetration enhancement (positive controls). The device parameter which was found to have the most marked effect on permeability of the compounds was bristle type. Profound changes (2- to 100-fold increase) were observed in the epidermal permeability of the hydrophilic penetrants (caffeine, acyclovir and angiotensin II), when the brush device was employed compared to positive controls (ethanol enhancement, delipidisation, iontophoresis and tape-stripping). Findings from this present study support the effectiveness of a rotating brush applied to the skin in enhancing epidermal permeability. Further optimization of operational parameters is required to exploit this simple and effective delivery device.


Subject(s)
Skin/metabolism , Acyclovir/chemistry , Acyclovir/pharmacokinetics , Angiotensin II/chemistry , Angiotensin II/pharmacokinetics , Caffeine/chemistry , Caffeine/pharmacokinetics , Humans , Parabens/chemistry , Parabens/pharmacokinetics , Permeability , Rotation , Technology, Pharmaceutical
18.
Ars pharm ; 49(3): 229-243, 2008. ilus, tab
Article in Es | IBECS | ID: ibc-70447

ABSTRACT

El objetivo de este estudio fue formular comprimidos de rápida disgregación, obtenidos mediante compresión directa,de fármacos con baja solubilidad en agua y diferentes grados de solubilidad, tomando como modelo Valsartány Efavirenz. Se estudió el efecto de diversas concentraciones de diferentes superdisgregantes como crospovidona,croscarmelosa sódica y glicolato sódico de almidón sobre el tiempo de disgregación y la disolución del fármaco invitro. Se observó que el tiempo de disgregación del comprimido con mejor liberación inmediata, de entre todaslas formulaciones probadas, fue de 21,5 ± 1,26 s y 20,16 ± 0,85 s para los comprimidos de Valsartán y Efavirenz,respectivamente que contenían, en ambos casos, un 20% de crospovidona. La liberación del fármaco (tanto en loscomprimidos de Valsartán como Efavirenz) fue más rápida en el caso de las formulaciones con crospovidona encomparación con la otra formulación. El efecto fue más evidente en el caso de Efavirenz, cuya solubilidad en aguaes menor que la de Valsartán. Se observó que era necesario un 20% de crospovidona para obtener una liberacióndel fármaco del 80% en comprimidos de Efavirenz. Los estudios por calorimetría diferencial de barrido no indicaronninguna incompatibilidad fármaco-excipiente. En conclusión, se obtuvieron por compresión directa comprimidosde rápida disgregación de Valsartán y Efavirenz con tiempos de disgregación más cortos y una alta velocidad dedisolución. Además, la crospovidona resultó ser un mejor disgregante tanto para Valsartán como para Efavirenz,de acuerdo con el tiempo de disgregación y los valores T80% obtenidos


The objective of this study was to formulate directly compressible fast disintegrating tablets of poorly water solubledrugs with different extent of drug solubilities, like valsartan and efavirenz, as model drugs. Effect of varying concentrationsof different superdisintegrants such as crospovidone, croscarmellose sodium, and sodium starch glycolate ondisintegration time and in vitro drug dissolution was studied. The disintegration time of the best immediate release tabletformulation among those tested was observed to be 21.5±1.26 sec and 20.16±0.85 sec for valsartan and efavirenz tabletscontaining 20% of Crospovidone, respectively. Drug release (from both valsartan and Efavirenz tablets) was faster fromformulations containing crospovidone compared to the other formulation. The effect was more apparent in Efavirenz,which has lesser aqueous solubility than valsartan. It was observed that 20% crospovidone was required to achieve80% drug release from efavirenz tablets. Differential scanning calorimetric studies did not indicate any drug-excipientincompatibility. In conclusion, directly compressible fast disintegrating tablets of valsartan and efavirenz with shorterdisintegration times and high dissolution rate were obtained and crospovidone seemed to be a better disintegrant forboth valsartan and efavirenz, based on disintegration time and T80% values obtained (AU)


Subject(s)
Drug Delivery Systems/methods , Povidone/chemistry , Povidone/pharmacology , Povidone/pharmacokinetics , Angiotensin II/chemistry , Angiotensin II/pharmacokinetics , Anti-Retroviral Agents/pharmacology , Calorimetry, Differential Scanning/instrumentation , Calorimetry, Differential Scanning/trends , Angiotensin II/chemical synthesis , Angiotensin II/therapeutic use , Anti-Retroviral Agents/pharmacokinetics , Anti-Retroviral Agents/therapeutic use , Anti-Retroviral Agents/chemical synthesis , Calorimetry, Differential Scanning
19.
J Nephrol ; 20(1): 29-35, 2007.
Article in English | MEDLINE | ID: mdl-17347970

ABSTRACT

BACKGROUND: The response of proximal convoluted tubules (PCTs) to angiotensin II is mediated by specific type 1 receptors found on both apical and basolateral surface membrane cells. After ligand association with type 1 receptors, different signaling pathways are triggered and determine changes in fluid absorption (Jv). The presence of AT1 and actin cytoskeleton, which are directly related to Jv, can undergo changes in distribution based on the actions of AngII and losartan. METHODS: Using a microperfusion technique and immunohistochemistry analysis, we investigated the basolateral action in PCTs, of AngII and/or losartan on Jv in rabbits, with regard to AT1 and actin cytoskeleton. RESULTS: AngII increased Jv, while in contrast, losartan and combined AngII + losartan led to its decrease. AngII did not change fluorescence intensity of AT1 receptors on tubular membranes, while losartan and AngII + losartan demonstrated a slight increase after treatment. On the other hand, AngII increased the fluorescence intensity of actin cytoskeleton, while losartan induced a decrease. AngII + losartan led actin cytoskeleton having a higher fluorescence intensity than in the control group. CONCLUSIONS: In the present study, we demonstrated that treatment of the basolateral side of PCT cells with AngII and losartan could lead to changes in absorptive tubular function. Important alterations were detected in AT1 receptor fluorescence on the luminal and basolateral membranes, and changes in F-actin cytoskeleton were verified by fluorescence following these protocols.


Subject(s)
Actins/metabolism , Angiotensin II/pharmacology , Cytoskeleton/metabolism , Receptor, Angiotensin, Type 1/metabolism , Absorption/physiology , Angiotensin II/pharmacokinetics , Angiotensin II Type 1 Receptor Blockers/pharmacokinetics , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Losartan/pharmacokinetics , Losartan/pharmacology , Male , Rabbits
20.
Endocrinology ; 148(1): 317-23, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17053027

ABSTRACT

Endothelium-derived steroidogenic factor (EDSF) is an endothelial peptide that stimulates aldosterone release from bovine adrenal zona glomerulosa (ZG) cells. The regulation of aldosterone release by combinations of EDSF and angiotensin II (AII) or EDSF and ACTH was investigated. Endothelial cells (ECs) and EC-conditioned media (ECCM) increased aldosterone release from ZG cells, an activity attributed to EDSF. AII (10(-12) to 10(-8) M) and ACTH (10(-12) to 10(-9) M) also stimulated the release of aldosterone from ZG cells. The stimulation by AII, but not ACTH, was greatly enhanced when ZG cells were coincubated with ECs. AII was metabolized by ECs to peptides identified by mass spectrometry as angiotensin (1-7) and angiotensin IV. There was very little metabolism of AII by ZG cells. Neither of these two AII metabolites altered aldosterone release from ZG cells, so they could not account for the enhanced response with ECs. AII-induced aldosterone release from ZG cells was enhanced by ECCM but not cell-free conditioned medium. This enhanced response was not due to increased EDSF release from ECs by AII. The synergistic effect of EDSF and AII was apparent when AII was added during or after the generation of ECCM and not observed when the AII component of the enhancement was blocked by the AII antagonist, losartan. These studies indicate that EDSF enhances the steroidogenic effect of AII. In the adrenal gland, ECs are in close anatomical relationship with ZG cells and may sensitize ZG cells to the steroidogenic action of AII by releasing EDSF.


Subject(s)
Aldosterone/metabolism , Angiotensin II/pharmacokinetics , Peptides/metabolism , Vasoconstrictor Agents/pharmacokinetics , Zona Glomerulosa/metabolism , Adrenocorticotropic Hormone/metabolism , Angiotensin I/metabolism , Angiotensin II/analogs & derivatives , Angiotensin II/metabolism , Animals , Cattle , Cells, Cultured , Culture Media, Conditioned/pharmacology , Endothelium/metabolism , Iodine Radioisotopes , Peptide Fragments/metabolism , Zona Glomerulosa/cytology , Zona Glomerulosa/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...