Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Amino Acids ; 53(5): 653-664, 2021 May.
Article in English | MEDLINE | ID: mdl-33791863

ABSTRACT

The renin-angiotensin system (RAS) has a central role in renal and cardiovascular homeostasis. Angiotensin-(1-7) (Ang1-7), one of the RAS active peptides, exerts beneficial effects through different mechanisms. These biological actions suggest that Ang1-7 is an effective therapeutic agent for treating various diseases associated with activated RAS. However, its short half-life and poor pharmacokinetics restrict its therapeutic utility. Our laboratory has successfully synthesized and characterized an Ang1-7 conjugate (Ang Conj.) with a prolonged half-life and improved pharmacokinetics profile. The Ang Conj. has been prepared by PEGylation of Ang1-7 and conjugation with a bisphosphonate using solid-phase peptide synthesis and characterized by HPLC and mass spectrometer. The compound's stability has been tested in different storage conditions. The bone binding capacity was evaluated using a hydroxyapatite assay. Pharmacokinetic and tissue distribution studies were performed using iodinated peptides in rats. Ang Conj. was synthesized with > 90% purity. Bone mineral affinity testing showed Ang Conj. exhibited significantly higher bone mineral affinity than Ang1-7. The Ang Conj. remained stable for more than a month using all tested storage conditions. The Ang Conj. demonstrated higher affinity to bone, a longer half-life, and better bioavailability when compared with the native peptide. These results support that conjugation of Ang1-7 with bisphosphonate enables it to utilize bone as a reservoir for the sustained delivery of Ang1-7 to maintain therapeutic plasma levels. High chemical stability and about five to tenfold prolongation of Ang Conj. plasma half-life after administrations into rats proves the effectiveness of our approach.


Subject(s)
Angiotensin I/chemistry , Angiotensin I/pharmacokinetics , Bone and Bones/metabolism , Diphosphonates/chemistry , Peptide Fragments/chemistry , Peptide Fragments/pharmacokinetics , Angiotensin I/blood , Angiotensin I/chemical synthesis , Animals , Biological Availability , Drug Delivery Systems , Drug Stability , Male , Peptide Fragments/blood , Peptide Fragments/chemical synthesis , Rats , Rats, Wistar , Solid-Phase Synthesis Techniques , Tissue Distribution
2.
Kidney Int ; 96(4): 906-917, 2019 10.
Article in English | MEDLINE | ID: mdl-31307778

ABSTRACT

The Renin-Angiotensin System (RAS) possesses a counter-regulatory axis composed of angiotensin converting enzyme (ACE)2, angiotensin-(1-7) [Ang-(1-7)] and the Mas receptor, which opposes many AT1-receptor-mediated effects of ligand angiotensin II. Ang-(1-7), as a ligand of the Mas receptor, has inhibitory effects on renal inflammation and fibrosis in experimental diabetes. However, Ang-(1-7) has a short half-life in plasma, which may render it unsuitable for use in clinics. Here, we investigated the effects of the lanthionine-stabilized Ang-(1-7), cyclic (c)Ang-(1-7), a lanthipeptide that is more peptidase-resistant than the linear peptide, in BTBR ob/ob mice with type 2 diabetic nephropathy. BTBR ob/ob mice received vehicle, cAng-(1-7), or the ACE inhibitor lisinopril. The treatment started at ten weeks of age, when the animals had already developed albuminuria, and ended at 19-20 weeks of age. cAng-(1-7) limited albuminuria progression, and limited podocyte dysfunction similarly to lisinopril. cAng-(1-7), unlike lisinopril, reduced glomerular fibrosis and inflammation, and counteracted glomerular capillary rarefaction. Furthermore, when cAng-(1-7) was combined with lisinopril, a superior antiproteinuric effect than with lisinopril alone was found, in association with better preservation of podocyte proteins and amelioration of capillary density. Thus, adding cAng-(1-7) to ACE-inhibitor therapy could benefit those diabetic patients who do not respond completely to ACE-inhibitor therapy.


Subject(s)
Angiotensin I/administration & dosage , Angiotensin-Converting Enzyme Inhibitors/administration & dosage , Diabetes Mellitus, Type 2/complications , Diabetic Nephropathies/drug therapy , Peptide Fragments/administration & dosage , Proteinuria/drug therapy , Alanine/administration & dosage , Alanine/analogs & derivatives , Alanine/chemistry , Alanine/pharmacokinetics , Angiotensin I/chemistry , Angiotensin I/pharmacokinetics , Animals , Diabetes Mellitus, Type 2/genetics , Diabetic Nephropathies/diagnosis , Diabetic Nephropathies/etiology , Diabetic Nephropathies/pathology , Disease Models, Animal , Drug Therapy, Combination/methods , Half-Life , Humans , Kidney Glomerulus/blood supply , Kidney Glomerulus/drug effects , Kidney Glomerulus/pathology , Kidney Glomerulus/ultrastructure , Lisinopril/administration & dosage , Male , Mice , Mice, Transgenic , Microscopy, Electron, Transmission , Peptide Fragments/chemistry , Peptide Fragments/pharmacokinetics , Peptides, Cyclic/administration & dosage , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacokinetics , Proteinuria/diagnosis , Proteinuria/etiology , Proteinuria/pathology , Sulfides/administration & dosage , Sulfides/chemistry , Sulfides/pharmacokinetics
3.
J Pharmacol Exp Ther ; 369(1): 9-25, 2019 04.
Article in English | MEDLINE | ID: mdl-30709867

ABSTRACT

Increasing evidence indicates that decreased brain blood flow, increased reactive oxygen species (ROS) production, and proinflammatory mechanisms accelerate neurodegenerative disease progression such as that seen in vascular contributions to cognitive impairment and dementia (VCID) and Alzheimer's disease and related dementias. There is a critical clinical need for safe and effective therapies for the treatment and prevention of cognitive impairment known to occur in patients with VCID and chronic inflammatory diseases such as heart failure (HF), hypertension, and diabetes. This study used our mouse model of VCID/HF to test our novel glycosylated angiotensin-(1-7) peptide Ang-1-6-O-Ser-Glc-NH2 (PNA5) as a therapy to treat VCID and to investigate circulating inflammatory biomarkers that may be involved. We demonstrate that PNA5 has greater brain penetration compared with the native angiotensin-(1-7) peptide. Moreover, after treatment with 1.0/mg/kg, s.c., for 21 days, PNA5 exhibits up to 10 days of sustained cognitive protective effects in our VCID/HF mice that last beyond the peptide half-life. PNA5 reversed object recognition impairment in VCID/HF mice and rescued spatial memory impairment. PNA5 activation of the Mas receptor results in a dose-dependent inhibition of ROS in human endothelial cells. Last, PNA5 treatment decreased VCID/HF-induced activation of brain microglia/macrophages and inhibited circulating tumor necrosis factor α, interleukin (IL)-7, and granulocyte cell-stimulating factor serum levels while increasing that of the anti-inflammatory cytokine IL-10. These results suggest that PNA5 is an excellent candidate and "first-in-class" therapy for treating VCID and other inflammation-related brain diseases.


Subject(s)
Angiotensin I/chemistry , Angiotensin I/pharmacology , Cognitive Dysfunction/complications , Cognitive Dysfunction/drug therapy , Dementia, Vascular/complications , Memory/drug effects , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Proto-Oncogene Proteins/agonists , Receptors, G-Protein-Coupled/agonists , Angiotensin I/pharmacokinetics , Angiotensin I/therapeutic use , Animals , Behavior, Animal/drug effects , Biomarkers/metabolism , Brain/drug effects , Brain/metabolism , Brain/pathology , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/physiopathology , Electrocardiography , Glycosylation , Half-Life , Heart Failure/complications , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Inflammation/physiopathology , Male , Maze Learning/drug effects , Mice , Peptide Fragments/pharmacokinetics , Peptide Fragments/therapeutic use , Proto-Oncogene Mas , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Spatial Memory/drug effects , Ventricular Remodeling/drug effects
4.
Physiol Rep ; 9(7): e14812, 2021 04.
Article in English | MEDLINE | ID: mdl-33904655

ABSTRACT

Cardiovascular diseases are the principal cause of death worldwide, with hypertension being the most common cardiovascular disease risk factor. High blood pressure (BP) is also associated with an increased risk of poor cognitive performance and dementia including Alzheimer's disease. Angiotensin 1-7 (Ang 1-7), a product of the renin-angiotensin system (RAS), exhibits central and peripheral actions to reduce BP. Recent data from our lab reveals that the addition of a non-radioactive iodine molecule to the tyrosine in position 4 of Ang 1-7 (iodoAng 1-7) makes it ~1000-fold more potent than Ang 1-7 in competing for the 125 I-Ang 1-7 binding site (Stoyell-Conti et al., 2020). Moreover, the addition of the non-radioactive iodine molecule increases (~4-fold) iodoAng 1-7's ability to bind to the AT1 receptor (AT1R), the primary receptor for Ang II. Preliminary data indicates that iodoAng 1-7 can also compete for the 125 I-Ang IV binding site with a low micromolar IC50. Thus, our aims were to compare the effects of chronic treatment of the Spontaneously Hypertensive Rat (SHR) with iodoAng 1-7 (non-radioactive iodine isotope) and Ang 1-7 on arterial pressure, heart rate, and cognitive function. For this study, male SHRs were divided into three groups and treated with Saline, Ang 1-7, or iodoAng 1-7 administrated subcutaneously using a 28-day osmotic mini pump. Systolic BP was measured non-invasively by the tail-cuff technique. Cognitive function was assessed by Y-Maze test and novel object recognition (NOR) test. We have demonstrated in SHRs that subcutaneous administration of high doses of iodoAng 1-7 prevented the increase in heart rate with age, while Ang 1-7 showed a trend toward preventing the increase in heart rate, possibly by improving baroreflex control of the heart. Conversely, neither Ang 1-7 nor iodoAng 1-7 administered subcutaneously affected BP nor cognitive function.


Subject(s)
Angiotensin I/therapeutic use , Blood Pressure , Cognition , Heart Rate , Hypertension/drug therapy , Peptide Fragments/therapeutic use , Angiotensin I/administration & dosage , Angiotensin I/pharmacokinetics , Animals , Iodine Radioisotopes , Male , Peptide Fragments/administration & dosage , Peptide Fragments/pharmacokinetics , Protein Binding , Rats , Rats, Inbred SHR , Receptor, Angiotensin, Type 1/metabolism
5.
Eur J Pharmacol ; 585(2-3): 303-12, 2008 May 13.
Article in English | MEDLINE | ID: mdl-18417117

ABSTRACT

Therapeutic modulation of the renin-angiotensin system is not complete without taking into consideration the beneficial effects of angiotensin-(1-7) in cardiovascular pathology. Various pharmacological pathways are already exploited to involve this heptapeptide in therapy as both inhibitors of angiotensin-converting enzyme and angiotensin II type 1 receptor blockers increase its levels. These drugs and administered angiotensin-(1-7) elicit various common effects, and some effects of the drugs are partially mediated by angiotensin-(1-7). The pharmacodynamic profile of angiotensin-(1-7) is rather complex, and in vitro and in vivo studies demonstrated a wide palette of effects for angiotensin-(1-7), some of them potentially beneficial for cardiovascular disease. Using various animal models to study cardiovascular physiology and disease it was shown that angiotensin-(1-7) has antihypertensive, antihypertrophic, antifibrotic and antithrombotic properties, all properties that may prove beneficial in a clinical setting. We also observed a novel action of angiotensin-(1-7), namely its capacity to stimulate the proliferation of endothelial progenitor cells. Access of angiotensin-(1-7) to the clinic, however, is restricted due to its unfavorable pharmacokinetic properties. In order to benefit of the therapeutic potential of angiotensin-(1-7) it is crucial to increase its half-life, either by using more stable analogues, which are now under development, or specific delivery methods. We here review the pharmacological characteristics and therapeutic potential of angiotensin-(1-7), implementing the experimental strategies taken to exploit the pharmacological mechanism of this heptapeptide in a clinical setting, and present our contribution to this field of research.


Subject(s)
Angiotensin I/pharmacology , Angiotensin I/therapeutic use , Peptide Fragments/pharmacology , Peptide Fragments/therapeutic use , Angiotensin I/pharmacokinetics , Animals , Antihypertensive Agents/pharmacology , Antihypertensive Agents/therapeutic use , Bone Marrow Cells/drug effects , Diabetes Mellitus/drug therapy , Heart Diseases/drug therapy , Humans , Hypertension/drug therapy , Peptide Fragments/pharmacokinetics , Renin-Angiotensin System/drug effects , Ventricular Remodeling/drug effects
6.
Drugs R D ; 18(1): 51-54, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29168127

ABSTRACT

In an earlier single-dose escalation study to evaluate the safety and pharmacokinetics of orally administered des-aspartate-angiotensin I (DAA-I) in healthy subjects, the plasma level of DAA-I could not be determined because DAA-I is rapidly degraded in the circulation. The present study investigated the oral bioavailability of DAA-I by measuring the prostaglandin E2 metabolite (PGEM) in the plasma samples of the same trial. PGEM is a stable derivative of PGE2, which has been shown to be a biomarker of DAA-I. The data show that plasma from two of the three subjects who were orally administered the efficacious preclinical dose of 0.70Ā mg/kg DAA-I exhibited a significant PGEM peak at 5-6Ā h postdose. Plasma of subjects who were administered 0.08 and 1.5Ā mg/kg DAA-I, the subefficacious and two-times efficacious dose, respectively, did not exhibit a similar PGEM peak. This observation is concordant with the known in vivo actions of DAA-I, especially its hypoglycemic action where maximum efficacy occurred at a dose of 0.7Ā mg/kg, and decreased to nil at the two-times efficacious dose. The onset of the PGEM peak at 5-6Ā h postdose was closed to the 4-h onset of absorption of [C14]DAA-I seen in preclinical rat studies, albeit the absorption kinetics between rodents and humans are not identical. The occurrence of polymorphism of enzymes involved in the formation and degradation of PGE2 is common, and this has been attributed to contributing to the variation in response, onset and peak PGEM observed among the three subjects who were administered the efficacious dose.


Subject(s)
Angiotensin I/analogs & derivatives , Dinoprostone/analogs & derivatives , Angiotensin I/administration & dosage , Angiotensin I/pharmacokinetics , Biological Availability , Dinoprostone/blood , Dose-Response Relationship, Drug , Humans
7.
Regul Pept ; 138(2-3): 59-65, 2007 Feb 01.
Article in English | MEDLINE | ID: mdl-17187874

ABSTRACT

This review presents a novel experimental approach for investigating the chronic actions of short-lived peptides in specific sites of the brain. This method combines the advantages of three different techniques: liposome encapsulation, site-specific microinjection and telemetry. First, liposomes can be designed to remain located at the injection site for a long period of time, where they protect encapsulated peptide from rapid degradation and act as a sustained-release system. Secondly, microinjection allows the administration of peptides in specific sites of the brain with minimal side effects. Finally, using telemetry, it is possible to register physiological parameters and their circadian variations in undisturbed free-moving animals for several days. Angiotensin-(1-7) and angiotensin II were used as peptide models, in order to validate the proposed method. Following the unilateral microinjection of the liposome-encapsulated peptides into the rostral ventrolateral medulla (RVLM) of Wistar rats, long-lasting cardiovascular actions were elicited, for several days. Importantly, new physiological actions of angiotensin-(1-7) at the RVLM were unmasked: modulation of the circadian rhythms of blood pressure and heart rate. It is felt that this method can be applied to a wide variety of short-lived bioactive peptides and should encounter numerous applications in the field of neurosciences.


Subject(s)
Brain/metabolism , Nanotechnology/methods , Peptides/pharmacology , Angiotensin I/chemistry , Angiotensin I/pharmacokinetics , Angiotensin I/pharmacology , Animals , Blood Pressure/drug effects , Heart Rate/drug effects , Liposomes/chemistry , Peptide Fragments/chemistry , Peptide Fragments/pharmacokinetics , Peptide Fragments/pharmacology , Peptides/chemistry , Peptides/pharmacokinetics , Rats
8.
Cancer Chemother Pharmacol ; 57(5): 559-68, 2006 May.
Article in English | MEDLINE | ID: mdl-16096787

ABSTRACT

PURPOSE: Multilineage cytopenias occur following myelosuppressive chemotherapy. Most hematopoietic agents differentiate along a single lineage and fail to prevent progressive cytopenias. Angiotensin 1-7 [A(1-7)] is a hematopoietic agent that stimulates the proliferation of multipotential and differentiated progenitor cells in cultured bone marrow and human cord blood. The purpose of this study was to determine the optimal biologic dose and the maximum tolerated dose of A(1-7). EXPERIMENTAL DESIGN: This study determined the safety and activity of A(1-7) following chemotherapy in patients with breast cancer. Toxicity was assessed by administering A(1-7) daily for 7 days followed by a 7-day washout prior to the first cycle of chemotherapy. Beginning 2 days after chemotherapy and continuing daily for at least 10 days, fifteen patients received five different A(1-7) doses and five patients received filgrastim as a comparator group over three cycles of chemotherapy. RESULTS: No dose-limiting toxicity was observed following A(1-7). The frequency of adverse events was slightly lower in A(1-7) than in filgrastim patients. No patient required a chemotherapy modification due to hematologic toxicity. There was an apparent differential dose-response sensitivity of the various lineages to A(1-7). At a dose of 100 microg/kg, A(1-7) reduced the frequency of grade 2-4 thrombocytopenia, anemia, and grade 3-4 lymphopenia as compared to filgrastim. CONCLUSION: These data suggest that A(1-7) may be beneficial in attenuating multilineage cytopenias following chemotherapy at a dose of 100 mug/kg per day.


Subject(s)
Angiotensin I/administration & dosage , Antihypertensive Agents/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Peptide Fragments/administration & dosage , Adult , Aged , Angiotensin I/pharmacokinetics , Antihypertensive Agents/pharmacokinetics , Breast Neoplasms/diagnosis , Cyclophosphamide/administration & dosage , Dose-Response Relationship, Drug , Doxorubicin/administration & dosage , Female , Humans , Maximum Tolerated Dose , Middle Aged , Peptide Fragments/pharmacokinetics , Prospective Studies
9.
Drugs R D ; 16(4): 317-326, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27681888

ABSTRACT

Des-aspartate-angiotensin I (DAA-I) is an endogenous angiotensin peptide and a prototype angiotensin receptor agonist (ARA). It acts on the angiotensin AT1 receptor and antagonises the deleterious actions of angiotensin II. DAA-I attenuates animal models of human disease in which angiotensin II has been implicated, such as cardiac hypertrophy, neointima formation, arteriosclerosis, renal failure, post-infarction injuries, diabetes, viral infection, chemical-induced inflammation, heat stroke, cancer, and gamma radiation lethality. DAA-I crosses Caco-2 cells and is effective at sub-nanomolar concentrations. These two properties are responsible for its oral efficacy. A single dose-escalation study was conducted to evaluate the safety, tolerability and pharmacokinetics of orally administered DAA-I in 18 healthy subjects. DAA-I was safe and well tolerated by the subjects, who were administered either 0.08, 0.70 or 1.50Ā mg/kg of the compound. The heart rate and systolic and diastolic blood pressures determined at each post-dose measurement remained within the clinically acceptable range. Across all cohorts, DAA-I had no substantial effect on blood pressures compared with placebo. Electrocardiographs (ECGs) were normal, and none of the subjects complained of chest discomfort. All clinical laboratory tests obtained before and after DAA-I and placebo treatment were normal. Pharmacokinetic analysis over a 12-h period following DAA-I administration did not show any increase of its level beyond basal concentration. This is in line with studies showing that intravenously administered DAA-I is rapidly metabolized and has a short half-life. We postulate that, during its short systemic sojourn, DAA-I exerts its actions via biased agonism on the angiotensin AT1 receptor.The ClinicalTrial.gov assignment number for this study is NCT02666196.


Subject(s)
Angiotensin I/analogs & derivatives , Administration, Oral , Adult , Angiotensin I/administration & dosage , Angiotensin I/pharmacokinetics , Dose-Response Relationship, Drug , Female , Humans , Male , Middle Aged , Young Adult
10.
Cardiovasc Res ; 23(4): 279-85, 1989 Apr.
Article in English | MEDLINE | ID: mdl-2556215

ABSTRACT

Since certain non-vascular angiotensin II (AII) receptors may be activated by angiotensin I (AI), and since sustained increase in AI levels accompanies chronic treatment with converting enzyme inhibitors (CEI) which block conversion of AI to AII, the question of whether AI has significant biological effects is of clinical relevance. We therefore sought to develop an in vitro culture system in which effects of angiotensin I, independently of its conversion to AII, could be studied in cloned aortic vascular endothelial cells (VEC). This was complicated by peptide degradation during the period of observation, both by angiotensin converting enzyme (ACE) on the surface of VEC and by angiotensinases in either the serum component of culture media or associated with the cell monolayer. Accordingly, we examined the half life of AI under relevant cell culture conditions, with and without confluent fetal bovine aortic endothelial cells (FBAEC). Factors assessed included (1) fetal calf serum: commercial source, concentration in culture media, effects of converting enzyme inhibitor (CEI: MK422) and/or heat inactivation (superimposed on the commercially performed process); and (2) effect of FBAEC in monolayer culture, with and without CEI. Results showed that (1) in the absence of cells, loss of AI in culture media, when present, was solely due to the presence of fetal calf serum (FCS) and showed a dose dependent response; (2) FCS from differing sources may vary dramatically in capacity for AI breakdown; and (3) serum related AI disappearance included a heat resistant ACE like component (inhibitable by CEI) and a heat sensitive/CEI resistant component dominant at concentrations of FCS exceeding 5%.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Angiotensin I/pharmacology , Endothelium, Vascular/drug effects , Angiotensin I/pharmacokinetics , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Cattle , Cells, Cultured , Culture Media , Dose-Response Relationship, Drug , Enalaprilat/pharmacology , Endothelium, Vascular/metabolism , Half-Life , Hot Temperature
11.
Hypertension ; 29(6): 1240-51, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9180624

ABSTRACT

We used a modification of the isolated perfused rat heart, in which coronary effluent and interstitial transudate were separately collected, to investigate the uptake and clearance of exogenous renin, angiotensinogen, and angiotensin I (Ang I) as well as the cardiac production of Ang I. The levels of these compounds in interstitial transudate were considered to be representative of the levels in the cardiac interstitial fluid. During perfusion with renin or angiotensinogen, the steady-state levels (mean +/- SD) in interstitial transudate were 64 +/- 34% (P < .05 for difference from the arterial level, n = 8) and 108 +/- 42% (n = 6) of the arterial level, respectively; the levels in coronary effluent were not significantly different from those in interstitial transudate. Ang I was not detectable in interstitial transudate during perfusion with Tyrode's buffer or angiotensinogen. It was very low in interstitial transudate during perfusion with renin and rose to much higher levels during combined renin and angiotensinogen perfusion. The total production rate of Ang I present in interstitial fluid could be largely explained by the renin-angiotensinogen reaction in the fluid phase of the interstitial compartment. In contrast, the total production rate of Ang I present in coronary effluent and the net ejection rate of Ang I via coronary effluent were, respectively, 4.6 +/- 2.2 and 2.8 +/- 1.3 (P < .01 and P < .05 for difference from 1.0, n = 6) times higher than could be explained by Ang I formation in the fluid phase of the intravascular compartment. Ang I from the interstitial fluid contributed little to the Ang I in the intravascular fluid and vice versa. These data reveal two tissue sites of Ang I production, ie, the interstitial fluid and a site closer to the blood compartment, possibly vascular surface-bound renin. There was no evidence that the release of locally produced Ang I into coronary effluent and interstitial transudate occurred independently of blood-derived renin or angiotensinogen.


Subject(s)
Angiotensin I/pharmacokinetics , Myocardium/metabolism , Renin/pharmacokinetics , Angiotensin I/biosynthesis , Angiotensin I/pharmacology , Angiotensinogen/metabolism , Angiotensinogen/pharmacokinetics , Animals , Extracellular Space/metabolism , Heart/drug effects , Male , Models, Cardiovascular , Rats , Rats, Wistar , Renin/metabolism , Renin/pharmacology , Swine
12.
Auton Autacoid Pharmacol ; 23(5-6): 297-305, 2003.
Article in English | MEDLINE | ID: mdl-15255814

ABSTRACT

1. The regulation by angiotensin II (Ang II) formed locally on nerve-stimulated purinergic and adrenergic components of double-peaked vasoconstrictions in the canine splenic artery and Ang II receptor subtypes involved were investigated. 2. The perfusion of the precursor angiotensin I (Ang I, 0.1-1 nm) did not affect the vasoconstrictor responses to noradrenaline (NA, 0.03-1 nmol) and adenosine 5'-triphosphate (ATP, 0.03-1 micromol). The second component vasoconstrictor response to nerve stimulation was dose dependently potentiated by Ang I (0.1-1 nm). The first peaked constriction was slightly, but insignificantly increased. The potentiating effects of Ang I were abolished by KRH-594 (10 nm), a selective AT(1) receptor antagonist, but not by PD 123319 (1-10 nm), an AT(2) receptor antagonist. KRH-594 (10 nm) or PD 123319 (10 nm) never affected the vasoconstrictions to either NA or ATP. 3. The treatment with KRH-594 (1-10 nm) produced a greater inhibition on the second peaked response than the first one, although both of them were dose dependently inhibited. PD 123319 (1-10 nm) did not affect the vasoconstrictor responses induced by nerve stimulation. 4. Inhibition of angiotensin-converting enzyme with 10 nm enalaprilat reduced the second peaked response, having no significant inhibition on the first peaked response. A higher dose of enalaprilat (100 nm) produced a greater inhibition of the second peak than the first one. It reduced the second peak by approximately 65%, while the first peak was decreased approximately 35%. After treatment with enalaprilat, Ang I (1 nm) failed to enhance the neuronal vascular response. Enalaprilat at doses used did not affect the vasoconstrictions to either NA or ATP. 5. The present results indicate that endogenously generated Ang II may produce a more marked potentiation of adrenergic transmission than purinergic transmission via activation of prejunctional AT(1) receptors.


Subject(s)
Myocytes, Smooth Muscle/cytology , Neurotransmitter Agents/metabolism , Receptor, Angiotensin, Type 1/physiology , Splenic Artery/cytology , Adenosine Triphosphate/administration & dosage , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/pharmacokinetics , Angiotensin I/administration & dosage , Angiotensin I/antagonists & inhibitors , Angiotensin I/pharmacokinetics , Angiotensin II/metabolism , Angiotensin II/pharmacology , Animals , Dogs , Dose-Response Relationship, Drug , Drug Synergism , Electric Stimulation , Enalaprilat/pharmacology , Female , Imidazoles/administration & dosage , Male , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/metabolism , Norepinephrine/administration & dosage , Norepinephrine/pharmacokinetics , Perfusion/methods , Pyridines/administration & dosage , Receptor, Angiotensin, Type 1/classification , Receptor, Angiotensin, Type 1/drug effects , Receptor, Angiotensin, Type 2/physiology , Splenic Artery/metabolism , Tetrazoles/pharmacology , Thiadiazoles/pharmacology , Vasoconstriction/drug effects , Vasoconstriction/physiology
13.
Peptides ; 51: 65-73, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24262271

ABSTRACT

Low angiotensin-(1-7) (Ang-(1-7)) concentration is observed in some cardiovascular diseases and exercise training seems to restore its concentration in the heart. Recently, a novel formulation of an orally active Ang-(1-7) included in hydroxy-propyl-beta-cyclodextrin (HPB-CD) was developed and chronically administered in experimental models of cardiovascular diseases. The present study examined whether chronic administration of HPB-CD/Ang-(1-7) produces beneficial cardiovascular effects in spontaneously hypertensive rats (SHR), as well as to compare the results obtained with those produced by exercise training. Male SHR (15-week old) were divided in control (tap water) or treated with HPB-CD/Ang-(1-7) (corresponding to 30Āµgkg(-1)day(-1) of Ang-(1-7)) by gavage, concomitantly or not to exercise training (treadmill, 10 weeks). After chronic treatment, hemodynamic, morphometric and molecular analysis in the heart were performed. Chronic HPB-CD/Ang-(1-7) decreased arterial blood pressure (BP) and heart rate in SHR. The inclusion compound significantly improved left ventricular (LV) end-diastolic pressure, restored the maximum and minimum derivatives (dP/dT) and decreased cardiac hypertrophy index in SHR. Chronic treatment improved autonomic control by attenuating sympathetic modulation on heart and vessels and the SAP variability, as well as increasing parasympathetic modulation and HR variability. Overall results were similar to those obtained with exercise training. These results show that chronic treatment with the HPB-CD/Ang-(1-7) inclusion compound produced beneficial effects in SHR resembling the ones produced by exercise training. This observation reinforces the potential cardiovascular therapeutic effect of this novel peptide formulation.


Subject(s)
Angiotensin I/administration & dosage , Antihypertensive Agents/administration & dosage , Excipients/administration & dosage , Hypertension/therapy , Peptide Fragments/administration & dosage , beta-Cyclodextrins/administration & dosage , 2-Hydroxypropyl-beta-cyclodextrin , Administration, Oral , Angiotensin I/pharmacokinetics , Animals , Antihypertensive Agents/pharmacokinetics , Blood Pressure , Combined Modality Therapy , Drug Evaluation, Preclinical , Exercise Therapy , Heart Rate , Hypertension/physiopathology , Male , Myocardium/metabolism , Myocardium/pathology , Peptide Fragments/pharmacokinetics , Physical Conditioning, Animal , Rats , Rats, Inbred SHR , Ventricular Pressure
14.
J Pharm Sci ; 101(1): 373-80, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21858825

ABSTRACT

The objectives of this study were to determine the potential systemic and local toxicity, as well as evaluate the toxicokinetic (TK) profile of angiotensin (1-7) [A(1-7)] when administered daily via subcutaneous injection for 28 days to Sprague-Dawley rats and Beagle dogs. A(1-7) is a member of the renin-angiotensin system and has undergone clinical evaluation for the treatment of chemotherapy-induced myelosuppression. In this present study, A(1-7) was given at 10 mg/(kg day) for 28 days to rats and canines. At day 27, blood was harvested to evaluate the TK parameters. On day 28, systemic toxicology was evaluated. Following A(1-7) administration for 27 days, no plasma A(1-7) accumulation was detected in canines; however, increased A(1-7) plasma concentrations were detected in rats. Despite the accumulation observed in rats, no detectable toxicity was found following A(1-7) administration for 28 days. The TK analysis of A(1-7) revealed a plasma half-life of 20-30 min in both rats and canines. The time to maximum plasma concentration was found to be 15 and 26.25 min in rats and canines, respectively. This study shows that subcutaneous administration of A(1-7) at 10 mg/(kg day) for 28 days did not lead to any detectable toxicities in either rats or canines.


Subject(s)
Angiotensin I/pharmacokinetics , Angiotensin I/toxicity , Peptide Fragments/pharmacokinetics , Peptide Fragments/toxicity , Angiotensin I/blood , Animals , Dogs , Female , Half-Life , Injections, Intravenous , Male , Peptide Fragments/blood , Pharmacokinetics , Rats , Rats, Sprague-Dawley
15.
Peptides ; 31(5): 893-8, 2010 May.
Article in English | MEDLINE | ID: mdl-20206220

ABSTRACT

Instability and proteolytic degradation limit the delivery options and in vivo efficacy of many therapeutic peptides. We previously generated a thioether stabilized angiotensin-(1-7) analog, cAng-(1-7), which is resistant against proteolytic degradation in the circulation. We here investigated oral and pulmonary delivery of this compound. In a first step we investigated the in vitro stability of the peptide under conditions that mimic those that will be met after oral administration. We demonstrated that cAng-(1-7) is stable at pH 2.0, a pH value close to that of the stomach, has enhanced resistance to breakdown by proteases from pancreas at pH 7.4, and is resistant to breakdown by proteases from liver at the lysosomal pH 5.0. We subsequently demonstrated that, in the absence of any delivery system or formulation, cAng-(1-7) can be delivered orally and via the lung, with bioavailabilities of 0.28+/-0.05% and 28+/-5%, whereas drug uptake was maximal after subcutaneous administration (bioavailability of 98+/-6%). Therapeutic concentrations could be reached via all three routes of administration. The data prove that introduction of a thioether bridge in peptides opens novel delivery options for medically important peptides.


Subject(s)
Administration, Oral , Angiotensin I/administration & dosage , Angiotensin I/pharmacokinetics , Peptide Fragments/administration & dosage , Peptide Fragments/pharmacokinetics , Administration, Cutaneous , Animals , Chromatography, High Pressure Liquid , Chromatography, Liquid , Drug Administration Routes , Male , Mass Spectrometry , Rats , Rats, Sprague-Dawley , Trachea/metabolism
16.
Clin Cancer Res ; 15(23): 7398-404, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19920106

ABSTRACT

PURPOSE: Angiotensin-(1-7) [Ang-(1-7)] is an endogenous peptide hormone of the renin-angiotensin system with antiproliferative and antiangiogenic properties. The primary objective of this study was to establish the recommended phase II dose of Ang-(1-7) for treating patients with advanced cancer. Secondary objectives were to assess toxicities, pharmacokinetics, clinical activity, and plasma biomarkers. EXPERIMENTAL DESIGN: Patients with advanced solid tumors refractory to standard therapy were treated with escalating doses of Ang-(1-7) in cohorts of three patients. Ang-(1-7) was administered by s.c. injection once daily for 5 days on a 3-week cycle. Tumor measurements were done every two cycles and treatment was continued until disease progression or unacceptable toxicity. RESULTS: Eighteen patients were enrolled. Dose-limiting toxicities encountered at the 700 microg/kg dose included stroke (grade 4) and reversible cranial neuropathy (grade 3). Other toxicities were generally mild. One patient developed a 19% reduction in tumor measurements. Three additional patients showed clinical benefit with stabilization of disease lasting more than 3 months. On day 1, Ang-(1-7) administration led to a decrease in plasma placental growth factor (PlGF) levels in patients with clinical benefit (P = 0.04) but not in patients without clinical benefit (P = 0.25). On day 5, PlGF levels remained lower in patients with clinical benefit compared with patients without clinical benefit (P = 0.04). CONCLUSIONS: Ang-(1-7) is a first-in-class antiangiogenic drug with activity for treating cancer that is linked to reduction of plasma PlGF levels. The recommended phase II dose is 400 microg/kg for this administration schedule.


Subject(s)
Angiogenesis Inhibitors/pharmacokinetics , Angiotensin I/pharmacokinetics , Neoplasms/blood , Neoplasms/drug therapy , Peptide Fragments/pharmacokinetics , Adult , Aged , Aged, 80 and over , Angiogenesis Inhibitors/therapeutic use , Angiotensin I/therapeutic use , Biomarkers, Tumor/metabolism , Cohort Studies , Dose-Response Relationship, Drug , Female , Humans , Male , Middle Aged , Models, Biological , Peptide Fragments/therapeutic use , Peptides/chemistry
17.
J Chromatogr B Biomed Appl ; 655(1): 127-31, 1994 Apr 22.
Article in English | MEDLINE | ID: mdl-8061820

ABSTRACT

A protocol to separate ten peptides of the angiotensin family by capillary electrophoresis was described. The experiment was carried out using the Waters Quanta 4000 Electrophoresis system and the steps taken to determine the optimum electrophoretic conditions include (i) the use of different electrolytes, (ii) variation of ionic composition and pH of the electrolytes, (iii) variation of applied voltage and the wavelength of ultraviolet detection. Successful separation of the ten angiotensin peptides was obtained using a voltage of 10 kV, 0.1 M phosphoric acid (pH 1.95) as electrolyte and ultraviolet detection at 185 nm. The protocol was then used to follow the metabolism of exogenous angiotensin I (ANG I) in rat lung homogenate and the separation and identification of an angiotensin peptide in human plasma. In addition to these two applications, the protocol can be used to separate and identify angiotensins and other peptides for which specific antibodies have yet to be developed.


Subject(s)
Angiotensins/analysis , Angiotensin I/pharmacokinetics , Angiotensins/blood , Angiotensins/isolation & purification , Animals , Electrophoresis , Humans , Lung/metabolism , Male , Rats , Rats, Sprague-Dawley , Spectrophotometry, Ultraviolet
18.
Circulation ; 94(8): 1886-93, 1996 Oct 15.
Article in English | MEDLINE | ID: mdl-8873664

ABSTRACT

BACKGROUND: The presence of mRNA for the essential components of the renin-angiotensin system (RAS) has been found in animal and human hearts. The present study was designed to provide evidence for the existence of a (functional) cardiac RAS. METHODS AND RESULTS: Twenty-four patients with atypical chest pain undergoing coronary angiography for diagnostic purposes were investigated. The cardiac production rate of angiotensins was estimated by measurement of the cardiac extraction of 125I-angiotensin I and 125I-angiotensin II associated with the determination of endogenous angiotensins in aortic and coronary sinus blood in normal, low, or high sodium diets. In a normal sodium diet, angiotensin I and II aorta-coronary sinus gradients were tendentially negative (-1.8 +/- 2.5 and -0.9 +/- 1.7 pg/mL, respectively), and the amounts of angiotensin I and II added by cardiac tissues were 6.5 +/- 3.1 and 2.7 +/- 1.3 pg/mL, respectively. The low sodium diet caused a significant increase in both plasma renin activity (PRA) and angiotensin I concentration in aortic but not in coronary sinus blood, resulting in a more negative aorta-coronary sinus gradient (-9.7 +/- 3.1 pg/mL, P < .01). Angiotensin formation by PRA in blood during transcardiac passage increased (P < .001), whereas angiotensin I formed by cardiac tissues decreased dramatically. Accordingly, in the low sodium diet, 125I-angiotensin II extraction did not change, the cardiac fractional conversion rate of 125I-angiotensin I to 125I-angiotensin II notably decreased (P < .01), and angiotensin II formation by cardiac tissues was undetectable. The high sodium diet caused a decrease in PRA and no changes in cardiac extraction of radiolabeled angiotensins; conversely, angiotensin I formed by cardiac tissues, cardiac Ang I fractional conversion rate, and angiotensin II formed during transcardiac passage significantly (P < .01 for all) increased. CONCLUSIONS: These results provide evidence for the existence of a functional cardiac RAS independent of but related to the circulating RAS.


Subject(s)
Myocardium/metabolism , Renin-Angiotensin System/physiology , Adult , Angiotensin I/blood , Angiotensin I/pharmacokinetics , Angiotensin II/blood , Angiotensin II/pharmacokinetics , Angiotensins/analysis , Chromatography, High Pressure Liquid , Coronary Circulation , Diet, Sodium-Restricted , Female , Humans , Male , Middle Aged , Myocardium/chemistry , Peptide Fragments/analysis , Tissue Distribution
19.
Biochem J ; 248(3): 691-6, 1987 Dec 15.
Article in English | MEDLINE | ID: mdl-3325040

ABSTRACT

A kinetic analysis of the tyrosine-specific protein kinase of pp60c-src from the C1300 mouse neuroblastoma cell line Neuro-2A and pp60c-src expressed in fibroblasts was carried out to determine the nature of the increased specific activity of the neuroblastoma enzyme. In immune-complex kinase assays with ATP-Mn2+ and the tyrosine-containing peptide angiotensin I as phosphoacceptor substrate, pp60c-src from the neuroblastoma cell line was characterized by a maximum velocity (Vmax.) that was 7-15-fold greater than the Vmax. of pp60c-src from fibroblasts. The neuroblastoma enzyme exhibited Km values for ATP (16 +/- 3 microM) and angiotensin I (6.8 +/- 2.6 mM) that were similar to Km values for ATP (25 +/- 3 microM) and angiotensin I (6.5 +/- 1.7 mM) of pp60c-src from fibroblasts. pp60v-src expressed in Rous-sarcoma-virus-transformed cells exhibited an ATP Km value (25 +/- 4 microM) and an angiotensin I Km value (6.6 +/- 0.5 mM) that approximated the values determined for pp60c-src in neuroblastoma cells and fibroblasts. These results indicate that the pp60c-src kinase from neuroblastoma cells has a higher turnover number than pp60c-src kinase from fibroblasts, and that the neural form of the enzyme would be expected to exhibit increased catalytic activity at the saturating concentrations of ATP that are found intracellularly.


Subject(s)
Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Adenosine Triphosphate/pharmacokinetics , Angiotensin I/pharmacokinetics , Animals , Cell Line , Enzyme Activation , Fibroblasts/enzymology , Immunoelectrophoresis , Kinetics , Mice , Neuroblastoma/metabolism , Tumor Cells, Cultured/metabolism
20.
Anal Chem ; 74(15): 3877-86, 2002 Aug 01.
Article in English | MEDLINE | ID: mdl-12175179

ABSTRACT

Two new applications using multiple ion isolations in the cell of a Fourier transform-ion cyclotron resonance mass spectrometer equipped with an electrospray ionization source are described. A procedure that uses multiple ion isolations of an analyte and calibrants for internal calibration at each stage in a MSn experiment, under high-resolution exact-mass conditions, for structural characterization/elucidation of angiotensin I and rapamycin is illustrated. Fragment ion mass accuracies < 1.0 ppm are demonstrated and routinely achieved. Purification of a mixture is illustrated by isolating multiple charge states of a protein-drug complex from residual protein for further MSn studies to elucidate the site of covalent drug bonding using IRMPD for a mixture of epidermal growth factor receptor (EGFr) protein and EGFr-drug complex. The procedure developed for multiple ion isolations is referred to as multi-CHEF, multiple correlated harmonic excitation fields, in which tailored waveforms are used to notch out multiple mass regions of a spectrum with minimal off-resonance excitation.


Subject(s)
Pharmaceutical Preparations/analysis , Proteins/metabolism , Angiotensin I/analysis , Angiotensin I/pharmacokinetics , Animals , Binding Sites , Bradykinin/analysis , Bradykinin/metabolism , Calibration , Cyclotrons , ErbB Receptors/analysis , ErbB Receptors/metabolism , Fourier Analysis , Humans , Ions , Mass Spectrometry , Melitten/analysis , Melitten/metabolism , Molecular Weight , Pharmacokinetics , Proteins/analysis , Sirolimus/analysis , Sirolimus/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL