Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 526
Filter
1.
Physiol Res ; 73(2): 227-237, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38710058

ABSTRACT

Nephrotoxicity as a cause of acute kidney injury (AKI) induced by cisplatin (CP), limits its usefulness as an anticancer agent. Diminazene, an angiotensin converting enzyme 2 activator, exhibited renoprotective properties on rat models of kidney diseases. This research aims to investigate the salutary effect of diminazene in comparison with lisinopril or valsartan in CP-induced AKI. The first and second groups of rats received oral vehicle (distilled water) for 9 days, and saline injection or intraperitoneal CP (6 mg/kg) on day 6, respectively. Third, fourth, and fifth groups received intraperitoneal injections of CP on day 6 and diminazene (15 mg/kg/day, orally), lisinopril (10 mg/kg/day, orally), or valsartan (30 mg/kg/day, orally), for 9 days, respectively. 24h after the last day of treatment, blood and kidneys were removed under anesthesia for biochemical and histopathological examination. Urine during the last 24 h before sacrificing the rats was also collected. CP significantly increased plasma urea, creatinine, neutrophil gelatinase-associated lipocalin, calcium, phosphorus, and uric acid. It also increased urinary albumin/creatinine ratio, N-Acetyl-beta-D-Glucosaminidase/creatinine ratio, and reduced creatinine clearance, as well the plasma concentrations of inflammatory cytokines [plasma tumor necrosis factor-alpha, and interleukin-1beta], and significantly reduced antioxidant indices [catalase, glutathione reductase , and superoxide dismutase]. Histopathologically, CP treatment caused necrosis of renal tubules, tubular casts, shrunken glomeruli, and increased renal fibrosis. Diminazine, lisinopril, and valsartan ameliorated CP-induced biochemical and histopathological changes to a similar extent. The salutary effect of the three drugs used is, at least partially, due to their anti-inflammatory and antioxidant effects. Keywords: Cisplatin, Diminazene, ACE2 activator, Lisinopril, Valsartan, Acute kidney injury.


Subject(s)
Acute Kidney Injury , Cisplatin , Diminazene , Lisinopril , Rats, Wistar , Valsartan , Animals , Acute Kidney Injury/chemically induced , Acute Kidney Injury/pathology , Acute Kidney Injury/metabolism , Acute Kidney Injury/prevention & control , Acute Kidney Injury/drug therapy , Lisinopril/pharmacology , Cisplatin/toxicity , Valsartan/pharmacology , Male , Diminazene/analogs & derivatives , Diminazene/pharmacology , Diminazene/therapeutic use , Rats , Antineoplastic Agents/toxicity , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Kidney/drug effects , Kidney/pathology , Kidney/metabolism
2.
Pharmacol Res Perspect ; 12(2): e1189, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38504425

ABSTRACT

Antihypertensive drug therapies have demonstrated their capacity to modulate the inflammatory processes associated with hypertension, leading to improvements in disease progression. Given the prevalent use of polytherapy in treating most hypertensive patients, comprehending the time-dependent effects of combination treatments on inflammation becomes imperative. In this study, spontaneously hypertensive rats (SHR) were divided into seven groups (n = 6): (i) SHR + vehicle, (ii) SHR + nebivolol, (iii) SHR + valsartan, (iv) SHR + lisinopril, (v) SHR + nebivolol-valsartan, (vi) SHR + nebivolol-lisinopril, and (vii) WKY + vehicle. Blood pressure was measured using the tail-cuff method. Temporal alterations in inflammatory cytokines TNF-α, IL-6, and IL-10 were assessed in serum through ELISA and mRNA expression in aortic tissue via qPCR after 1, 2, and 4 weeks of treatment with nebivolol, lisinopril, valsartan, and their respective combinations. Histological alterations in the aorta were assessed. The findings indicated that combined treatments reduced systolic and diastolic blood pressure in SHR. The nebivolol and lisinopril combination demonstrated a significant decrease in IL-6 serum and mRNA expression at both 1 week and 4 weeks into the treatment. Additionally, TNF-α mRNA expression also showed a reduction with this combination at the same time points. Particularly, nebivolol-valsartan significantly decreased TNF-α serum and mRNA expression after one and four weeks of treatment. Furthermore, an elevation in serum IL-10 levels was observed with both combination treatments starting from the second week onwards. This study provides compelling evidence that concurrent administration of nebivolol with lisinopril or valsartan exerts time-dependent effects, reducing proinflammatory cytokines TNF-α and IL-6 while modifying IL-10 levels in an experimental hypertensive model.


Subject(s)
Hypertension , Lisinopril , Humans , Rats , Animals , Nebivolol/pharmacology , Nebivolol/therapeutic use , Rats, Inbred SHR , Lisinopril/pharmacology , Lisinopril/therapeutic use , Interleukin-6/genetics , Tumor Necrosis Factor-alpha/genetics , Antihypertensive Agents/pharmacology , Antihypertensive Agents/therapeutic use , Interleukin-10/genetics , Rats, Inbred WKY , Hypertension/drug therapy , Cytokines , Valsartan/therapeutic use , RNA, Messenger
3.
J Virol ; 98(3): e0180223, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38334329

ABSTRACT

With a high incidence of acute kidney injury among hospitalized COVID-19 patients, considerable attention has been focussed on whether SARS-CoV-2 specifically targets kidney cells to directly impact renal function, or whether renal damage is primarily an indirect outcome. To date, several studies have utilized kidney organoids to understand the pathogenesis of COVID-19, revealing the ability for SARS-CoV-2 to predominantly infect cells of the proximal tubule (PT), with reduced infectivity following administration of soluble ACE2. However, the immaturity of standard human kidney organoids represents a significant hurdle, leaving the preferred SARS-CoV-2 processing pathway, existence of alternate viral receptors, and the effect of common hypertensive medications on the expression of ACE2 in the context of SARS-CoV-2 exposure incompletely understood. Utilizing a novel kidney organoid model with enhanced PT maturity, genetic- and drug-mediated inhibition of viral entry and processing factors confirmed the requirement for ACE2 for SARS-CoV-2 entry but showed that the virus can utilize dual viral spike protein processing pathways downstream of ACE2 receptor binding. These include TMPRSS- and CTSL/CTSB-mediated non-endosomal and endocytic pathways, with TMPRSS10 likely playing a more significant role in the non-endosomal pathway in renal cells than TMPRSS2. Finally, treatment with the antihypertensive ACE inhibitor, lisinopril, showed negligible impact on receptor expression or susceptibility of renal cells to infection. This study represents the first in-depth characterization of viral entry in stem cell-derived human kidney organoids with enhanced PTs, providing deeper insight into the renal implications of the ongoing COVID-19 pandemic. IMPORTANCE: Utilizing a human iPSC-derived kidney organoid model with improved proximal tubule (PT) maturity, we identified the mechanism of SARS-CoV-2 entry in renal cells, confirming ACE2 as the sole receptor and revealing redundancy in downstream cell surface TMPRSS- and endocytic Cathepsin-mediated pathways. In addition, these data address the implications of SARS-CoV-2 exposure in the setting of the commonly prescribed ACE-inhibitor, lisinopril, confirming its negligible impact on infection of kidney cells. Taken together, these results provide valuable insight into the mechanism of viral infection in the human kidney.


Subject(s)
Angiotensin-Converting Enzyme 2 , Kidney , Organoids , SARS-CoV-2 , Virus Internalization , Humans , Angiotensin-Converting Enzyme 2/metabolism , COVID-19/complications , COVID-19/virology , Kidney/cytology , Kidney/drug effects , Kidney/metabolism , Kidney/virology , Lisinopril/pharmacology , Lisinopril/metabolism , Organoids/cytology , Organoids/drug effects , Organoids/metabolism , Organoids/virology , Pandemics , SARS-CoV-2/metabolism , SARS-CoV-2/pathogenicity , Spike Glycoprotein, Coronavirus/metabolism , Virus Internalization/drug effects , Peptidyl-Dipeptidase A/metabolism , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Acute Kidney Injury/etiology , Acute Kidney Injury/metabolism , Acute Kidney Injury/virology , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/virology , Receptors, Coronavirus/metabolism , Models, Biological , Serine Endopeptidases/metabolism , Endosomes/drug effects , Endosomes/metabolism , Endosomes/virology , Gene Expression Regulation/drug effects , Stem Cells/cytology
4.
Expert Rev Clin Pharmacol ; 17(1): 93-100, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38078460

ABSTRACT

BACKGROUND: Although a growing number of observational studies suggest that angiotensin-converting enzyme inhibitors (ACEIs) intake may be a risk factor for psoriasis, evidence is still insufficient to draw definitive conclusions. RESEARCH DESIGN AND METHODS: Drug-targeted Mendelian randomization (DTMR) was used to analyze the causality between genetic proxied ACEIs and psoriasis. Furthermore, we performed a disproportionality analysis based on the FDA adverse event reporting system (FAERS) database to identify more suspicious subclasses of ACEIs. RESULTS: Using two kinds of genetic proxy instruments, the present DTMR research identified genetic proxied ACEIs as risk factors for psoriasis. Furthermore, our disproportionality analysis revealed that ramipril, trandolapril, perindopril, lisinopril, and enalapril were associated with the risk of psoriasis, which validates and refines the findings of the DTMR. CONCLUSIONS: Our integrative study verified that ACEIs, especially ramipril, trandolapril, perindopril, lisinopril, and enalapril, tended to increase the risk of psoriasis statistically.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors , Psoriasis , Humans , Angiotensin-Converting Enzyme Inhibitors/adverse effects , Ramipril/adverse effects , Lisinopril/pharmacology , Perindopril/adverse effects , Pharmacovigilance , Mendelian Randomization Analysis , Enalapril/pharmacology , Psoriasis/drug therapy , Psoriasis/genetics
5.
Pediatr Cardiol ; 45(2): 394-400, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38153545

ABSTRACT

Hypertension after cardiothoracic surgery is common, often requiring pharmacologic management. The recommended first-line antihypertensives in pediatrics are angiotensin converting enzyme inhibitors. Captopril and enalapril are approved for infants and children; however, lisinopril is only approved for > 7 years of age. This study evaluated safety and efficacy of converting from captopril to lisinopril in patients utilizing a pre-defined conversion of 3 mg captopril to 1 mg lisinopril. This was a single center, retrospective study including patients less than 7 years of age admitted for cardiothoracic surgery who received both captopril and lisinopril from 01/01/2017 to 06/01/2022.The primary outcome was mean change in systolic blood pressure (SBP) from baseline for 72 h after conversion of captopril to lisinopril. A total of 99 patients were enrolled. There was a significant decrease in mean SBP (99.12 mmHg vs 94.86 mmHg; p = 0.007) with no difference in DBP (59.23 mmHg vs 61.95 mmHg; p = 0.07) after conversion to lisinopril. Of the 99 patients who were transitioned to lisinopril, 79 (80%) had controlled SBP, 20 (20%) remained hypertensive, 13 (13%) received an increase in their lisinopril dose, and 2 (2%) required an additional antihypertensive agent. There was a low overall rate of AKI (3%) and hyperkalemia (4%) respectively. This study demonstrates that utilizing lisinopril with a conversion rate of 3 mg of captopril to 1 mg of lisinopril was safe and effective for controlling hypertension in pediatric patients following cardiothoracic surgery.


Subject(s)
Hypertension , Lisinopril , Humans , Child , Lisinopril/therapeutic use , Lisinopril/pharmacology , Captopril/therapeutic use , Captopril/pharmacology , Retrospective Studies , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Hypertension/drug therapy , Antihypertensive Agents/therapeutic use , Antihypertensive Agents/pharmacology , Enalapril , Blood Pressure
6.
J Emerg Med ; 64(3): 397-399, 2023 03.
Article in English | MEDLINE | ID: mdl-36925439

ABSTRACT

BACKGROUND: Angiotensin-converting enzyme (ACE) inhibitor overdose is an uncommonly presenting toxicologic emergency. Management is primarily supportive care, but a small body of evidence exists to support naloxone for management of hypotension. CASE REPORT: We present a case of accidental ACE inhibitor overdose. The patient took approximately 300 mg lisinopril over 48 h and presented for evaluation of syncope. He was hypotensive and unresponsive to fluids. We administered naloxone with immediate and sustained resolution in hypotension. The mechanism of action is briefly discussed. WHY SHOULD AN EMERGENCY MEDICINE PHYSICIAN BE AWARE OF THIS?: Naloxone is a rapid, low-risk, low-cost, and effective intervention for hypotension due to ACE inhibitor toxicity. It is supported by basic science research and clinical experience.


Subject(s)
Drug Overdose , Hypotension , Male , Humans , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Naloxone/therapeutic use , Lisinopril/pharmacology , Lisinopril/therapeutic use , Hypotension/drug therapy
7.
Environ Sci Pollut Res Int ; 30(9): 23263-23275, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36319925

ABSTRACT

Sodium fluoride (NaF) is one of the neglected environmental toxicants that has continued to silently cause toxicity to both humans and animals. NaF is universally present in water, soil, and atmosphere. The persistent and alarming rate of increase in cardiovascular and renal diseases caused by chemicals such as NaF in mammalian tissues has led to the use of various drugs for the treatment of these diseases. The present study aimed at evaluating the renoprotective and antihypertensive effects of L-arginine against NaF-induced nephrotoxicity. Thirty male Wistar rats (150-180 g) were used in this study. The rats were randomly divided into five groups of six rats each as follows: Control, NaF (300 ppm), NaF + L-arginine (100 mg/kg), NaF + L-arginine (200 mg/kg), and NaF + lisinopril (10 mg/kg). Histopathological examination and immunohistochemistry of renal angiotensin-converting enzyme (ACE) and mineralocorticoid receptor (MCR) were performed. Markers of renal damage, oxidative stress, antioxidant defense system, and blood pressure parameters were determined. L-arginine and lisinopril significantly (P < 0.05) ameliorated the hypertensive effects of NaF. The systolic, diastolic, and mean arterial blood pressure of the treated groups were significantly (P < 0.05) reduced compared with the hypertensive group. This finding was concurrent with significantly increased serum bioavailability of nitric oxide in the hypertensive rats treated with L-arginine and lisinopril. Also, there was a significant reduction in the level of blood urea nitrogen and creatinine of hypertensive rats treated with L-arginine and lisinopril. There was a significant (P < 0.05) reduction in markers of oxidative stress such as malondialdehyde and protein carbonyl and concurrent increase in the levels of antioxidant enzymes in the kidney of hypertensive rats treated with L-arginine and lisinopril. The results of this study suggest that L-arginine and lisinopril normalized blood pressure, reduced oxidative stress, and the expression of renal ACE and mineralocorticoid receptor, and improved nitric oxide production. Thus, L-arginine holds promise as a potential therapy against hypertension and renal damage.


Subject(s)
Hypertension , Lisinopril , Humans , Rats , Male , Animals , Lisinopril/metabolism , Lisinopril/pharmacology , Lisinopril/therapeutic use , Sodium Fluoride/toxicity , Antioxidants/metabolism , Nitric Oxide/metabolism , Receptors, Mineralocorticoid/metabolism , Receptors, Mineralocorticoid/therapeutic use , Rats, Wistar , Hypertension/chemically induced , Kidney , Blood Pressure , Oxidative Stress , Arginine/metabolism , Arginine/pharmacology , Arginine/therapeutic use , Dietary Supplements , Angiotensins/metabolism , Angiotensins/pharmacology , Angiotensins/therapeutic use , Mammals
8.
Wiad Lek ; 75(10): 2407-2411, 2022.
Article in English | MEDLINE | ID: mdl-36472269

ABSTRACT

OBJECTIVE: The aim: To analyze the dynamics of daily monitoring of blood pressure, intracardiac (according to echocardiography), peripheral hemodynamics (according to ultrasound of the vessels of the lower extremity), the thickness of the intima-media complex (according to carotid sonography) in patients with hypertension the effect of treatment with a combination of lisinopril and amlodipine. PATIENTS AND METHODS: Materials and methods: The study included 40 patients with hypertension with 2 (29 patients) and 3 (11 patients) degrees of hypertension in combination with AOLE with I-III stages of chronic insufficiency of the lower extremity, which revealed hyperkinetic, eukinetic, and hypokinetic types of hypertension with a predominance of the sympathetic nervous system. The groups are comparable in age, sex, duration of hypertension, and medications received in the previous stages. For antihypertensive therapy, the most common drugs for use were selected - lisinopril + amplodipine in fixed doses of 10 and 5 mg, respectively. If after 2 weeks we did not reduce the mean level of SBP and DBP by 10% or more from baseline, we doubled the dose of lisinopril without changing the dose of amlodipine. RESULTS: Results: After 6 months of treatment, in particular, an increase in the pulse index - by 24.8%, a decrease in the resistance index - by 21.1%, an increase in linear and volumetric blood velocity - by 25.6% and 27.4%, respectively, while achieving the target blood pressure. CONCLUSION: Conclusions: It is proved that in the absence of individual contraindications the combination of lisinopril and amlodipine is optimal and universal for effective treatment of patients with hypertension in combination with AOLE in all types of central hemodynamics.


Subject(s)
Arteriosclerosis Obliterans , General Practice , Hypertension , Humans , Amlodipine/therapeutic use , Amlodipine/pharmacology , Lisinopril/therapeutic use , Lisinopril/pharmacology , Antihypertensive Agents/therapeutic use , Antihypertensive Agents/pharmacology , Arteriosclerosis Obliterans/chemically induced , Arteriosclerosis Obliterans/drug therapy , Hypertension/complications , Hypertension/drug therapy , Blood Pressure , Comorbidity
9.
Biopharm Drug Dispos ; 43(6): 233-246, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36299167

ABSTRACT

Lisinopril is an antihypertensive drug with poor intestinal permeability. Enhancement of intestinal absorption depends on a clear understanding of the permeation pathways and absorption mechanisms. Unfortunately, these are not fully elucidated for lisinopril. Accordingly, the aim was to determine lisinopril permeation pathways and obstacles limiting membrane transport with subsequent nomination of appropriate permeation enhancers. This employed an in situ rabbit intestinal perfusion technique, which revealed site-dependent absorptive clearance (PeA/L) from a lisinopril simple solution (5 µg/ml), with paracellular absorption playing a role. Regional drug permeability ranked as colon> duodenum> jejunum> ileum opposing intestinal expression rank of P-glycoprotein (P-gp) efflux transporters. Duodenal and jejunal perfusion of a higher lisinopril concentration (50 µg/ml) reflected saturable absorption, suggesting carrier-mediated transport. The effect of piperine and verapamil as P-gp inhibitors on intestinal absorption of lisinopril was investigated. Coperfusion with either piperine or verapamil significantly enhanced lisinopril absorption, with enhancement being dominant in the ileum segment. This supported the contribution of P-gp transporters to poor lisinopril permeability. On the other hand, coperfusion of lisinopril with zinc acetate dihydrate significantly multiplied lisinopril PeA/L by 2.3- and 6.6-fold in duodenum and ileum segments, respectively, through magnifying intestinal water flux. The study explored the barriers limiting lisinopril intestinal absorption. Moreover, the study exposed clinically relevant lisinopril interactions with common coadministered cargos that should be considered for an appropriate lisinopril regimen. However, this requires further in vivo verification.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1 , Lisinopril , Animals , Rabbits , Lisinopril/pharmacology , Lisinopril/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Intestinal Absorption , Verapamil/pharmacology , Permeability , Intestinal Mucosa/metabolism
10.
Cell Physiol Biochem ; 56(5): 457-483, 2022 Sep 05.
Article in English | MEDLINE | ID: mdl-36057984

ABSTRACT

BACKGROUND/AIMS: Hypertension is treated primarily with angiotensin II (ATII) receptor blockers (ARBs) and angiotensin converting enzyme (ACE) inhibitors (ACEIs). Both ATII and ACEIs can trigger signal transduction via ACE, and a possible correlation between ARB/ACEI therapy and an increased risk of cancer is highly controversial. The question of whether or not ACE as a potential signal transducer affects human melanoma (MV3) cell behavior prompted the present study. METHODS: Expression of ACE, ATII receptor types 1, 2 (AT1R, AT2R), COX2 and MMP2 in MV3 cells was examined by qPCR. AT1R, AT2R and ACE were inhibited with losartan, EMA401 and lisinopril, respectively. Adhesion, migration and invasiveness of MV3 cells seeded on a hepatocyte (Huh7) monolayer or a reconstituted collagen type I matrix were analyzed using video microscopy and Boyden chambers. Integrity of the Huh7 cell layer was confirmed by measuring transepithelial electrical resistance (TEER). ERK1/2 phosphorylation and MMP2 secretion were evaluated by Western blotting. MMP2 activity was inhibited with ARP-100. RESULTS: Losartan, EMA401 and lisinopril stimulated MV3 melanoma cell migration and invasion in a coculture model with Huh7 cells while leaving proliferation and adhesion largely unaffected. The drugs did not interfere with TEER of the hepatocyte monolayer nor with MV3 cell proliferation, but tended to increase the phosphorylation of ERK1/2 and the expression of both COX2 and MMP2. Lisinopril caused a significant increase in MV3 cells' MMP2 secretion and an accelerated MV3 cell-mediated TEER breakdown. The MMP2 inhibitor ARP-100 could antagonize the lisinopril-stimulated invasion of the hepatocyte layer. CONCLUSION: Lisinopril stimulates MV3 cell invasion by increasing the expression and secretion of MMP2.


Subject(s)
Lisinopril , Melanoma , Angiotensin Receptor Antagonists , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Cyclooxygenase 2 , Humans , Lisinopril/pharmacology , Losartan/pharmacology , Matrix Metalloproteinase 2 , Melanoma/drug therapy , Melanoma/metabolism
11.
Fundam Clin Pharmacol ; 36(6): 992-1004, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35697514

ABSTRACT

Low serum concentrations of the amino acid homoarginine (HA) are associated with increased cardiovascular mortality by incompletely understood mechanisms. This study sought to assess the influence of HA on cardiac remodeling in rats undergoing either transaortic banding or inhibition of nitric oxide synthesis by Nω-Nitro-L-arginine methyl ester hydrochloride (L-NAME). Male Wistar rats (n = 136) underwent sham operation (SH) or aortic banding (AB). Both groups were equally divided into 14 subgroups, receiving different doses of HA alone or in combination with lisinopril, spironolactone, or L-NAME for 4 weeks. HA treatment in AB animals resulted in a dose-dependent improvement of cardiac function up to a concentration of 800 mg·kg-1 ·day-1 . Combining 800 mg·kg-1 ·day-1 HA with spironolactone or lisinopril yielded additional effects, showing a positive correlation with LV ejection fraction (+33%, p = 0.0002) and fractional shortening (+41%, p = 0.0014). An inverse association was observed with collagen area fraction (-41%, p < 0.0001), myocyte cross-sectional area (-22%, p < 0.0001) and the molecular markers atrial natriuretic factor (-74%, p = 0.0091), brain natriuretic peptide (-42%, p = 0.0298), beta-myosin heavy chain (-46%, p = 0.0411), and collagen type V alpha 1 chain (-73%, p = 0.0257) compared to placebo-treated AB animals. Co-administration of HA and L-NAME was found to attenuate cardiac remodeling and prevent NO-deficient hypertension following AB. HA treatment has led to a dose-dependent improvement of myocardial function and marked histological and molecular changes in cardiac remodeling following AB. Combining HA with standard heart failure medication resulted in additional beneficial effects boosting its direct impact on heart failure pathophysiology.


Subject(s)
Heart Failure , Hypertension , Rats , Male , Animals , NG-Nitroarginine Methyl Ester/pharmacology , Spironolactone/metabolism , Spironolactone/pharmacology , Spironolactone/therapeutic use , Homoarginine/metabolism , Homoarginine/pharmacology , Homoarginine/therapeutic use , Lisinopril/metabolism , Lisinopril/pharmacology , Lisinopril/therapeutic use , Ventricular Remodeling , Hypertension/drug therapy , Rats, Wistar , Myocardium/metabolism , Heart Failure/drug therapy , Blood Pressure
12.
Geroscience ; 44(6): 2845-2861, 2022 12.
Article in English | MEDLINE | ID: mdl-35767209

ABSTRACT

Diabetes, hypertension, and aging are major contributors to cardiovascular and chronic kidney disease (CKD). Sodium/glucose cotransporter 2 (SGLT2) inhibitors have become a preferred treatment for type II diabetic patients since they have cardiorenal protective effects. However, most elderly diabetic patients also have hypertension, and the effects of SGLT2 inhibitors have not been studied in hypertensive diabetic patients or animal models. The present study examined if controlling hyperglycemia with empagliflozin, or given in combination with lisinopril, slows the progression of renal injury in hypertensive diabetic rats. Studies were performed using hypertensive streptozotocin-induced type 1 diabetic Dahl salt-sensitive (STZ-SS) rats and in deoxycorticosterone-salt hypertensive type 2 diabetic nephropathy (T2DN) rats. Administration of empagliflozin alone or in combination with lisinopril reduced blood glucose, proteinuria, glomerular injury, and renal fibrosis in STZ-SS rats without altering renal blood flow (RBF) or glomerular filtration rate (GFR). Blood pressure and renal hypertrophy were also reduced in rats treated with empagliflozin and lisinopril. Administration of empagliflozin alone or in combination with lisinopril lowered blood glucose, glomerulosclerosis, and renal fibrosis but had no effect on blood pressure, kidney weight, or proteinuria in hypertensive T2DN rats. RBF was not altered in any of the treatment groups, and GFR was elevated in empagliflozin-treated hypertensive T2DN rats. These results indicate that empagliflozin is highly effective in controlling blood glucose levels and slows the progression of renal injury in both hypertensive type 1 and type 2 diabetic rats, especially when given in combination with lisinopril to lower blood pressure.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1 , Diabetes Mellitus, Type 2 , Diabetic Nephropathies , Hypertension , Animals , Rats , Blood Glucose , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/etiology , Fibrosis , Hypertension/complications , Hypertension/drug therapy , Lisinopril/pharmacology , Proteinuria , Rats, Inbred Dahl
13.
J Food Biochem ; 46(11): e14220, 2022 11.
Article in English | MEDLINE | ID: mdl-35561037

ABSTRACT

Studies have shown that some drugs impair spermatogenesis, thereby causing infertility. Thus, this study aims at investigating the effect of Persea americana seed extract on the male reproductive system in cyclosporine-induced rats. Thirty male albino rats were randomly divided into five groups; all groups were induced with cyclosporine except the control group. Group 3 was treated with 10 mg/kg of lisinopril, groups 4 and 5 received 50 and 100 mg/kg of PAE, respectively. The experiment lasted for 7 days. The antioxidant enzymes [superoxide dismutase (SOD) and catalase (CAT)], and arginase activities were evaluated, nitric oxide (NO) and malondialdehyde (MDA) level of the penile tissue homogenate were determined with serum hormonal (follicle stimulating hormone, luteinizing hormone and testosterone) concentration and sperm quality. This study showed that cyclosporine induction caused a significant (p < .05) decrease in the SOD, CAT, sperm quality, NO, and hormonal level as compared to the control group, with a simultaneous increase in arginase activity and MDA level. However, treatment with PAE and lisinopril significantly (p < .05) increase antioxidant enzyme activities, sperm quality, NO, and serum hormonal level, with a decrease MDA level and arginase activity when compared with cyclosporine-induced group. This study showed that P. americana seed extract could be useful in the management of hormonal disruption resulting from oxidative stress in male folks. PRACTICAL APPLICATIONS: Some pharmaco-agents have been reported to alter spermatogenesis, thereby causing infertility. Plants represent natural resources use in the management of several human diseases from time immemorial. Persea americana seed is a part of the fruit that most people do throw away after consuming the edible portion of the fruit. However, the therapeutic and pharmacological activities of P. americana seed have been reported. Therefore, this study sought to investigate the effects of P. americana seed extract on cyclosporine-induced reprotoxicity.


Subject(s)
Infertility , Persea , Plant Extracts , Animals , Male , Rats , Antioxidants/metabolism , Arginase , Cyclosporine/pharmacology , Infertility/drug therapy , Lisinopril/pharmacology , Persea/chemistry , Plant Extracts/pharmacology , Rats, Wistar , Reproduction , Seeds/chemistry , Spermatozoa , Superoxide Dismutase/metabolism
14.
Curr Med Chem ; 29(42): 6422-6432, 2022.
Article in English | MEDLINE | ID: mdl-35125079

ABSTRACT

BACKGROUND: Bradykinin-potentiating peptides (BPPs) are snake venom peptides inhibiting the angiotensin-converting enzyme (ACE). ACE plays an important role in the regulation of blood pressure. BPPs lead to the development of ACE inhibitors for the treatment of hypertension. OBJECTIVE: The objective of the present work was to carry out a comprehensive comparative study of four synthesised snake venom BPPs in vivo. METHODS: Four synthesised snake venom BPPs were administered to rats via the intraperitoneal route for 15 days at a fixed dose. Lisinopril was used as a comparative standard. Thirty male albino rats were divided into six groups: A, B, C, D, E (lisinopril), and F (control). Group F was maintained as the control group and given only saline. After 15 days, blood samples and tissues were removed for the study of selective biochemical parameters and histomorphometric analysis. Statistical evaluation of all results was also performed. RESULTS: The results indicated that peptide I, with the sequence ZSAPGNEAIPP, was highly toxic and adversely affected all the biochemical and histological parameters studied in this work. Peptide II (ZNWPHPQIPP) and peptide IV (ZQWAQGRAPHPP) showed lower toxicity. None of the BPPs raised the serum creatinine level and exhibited nephroprotective effects. Although lisinopril raised the creatinine level, it showed a protective role towards the pancreas and lungs in parallel. CONCLUSION: The present work shows that although there is a high sequence similarity between the four BPPs, their in vivo activity varies. The sequences of peptide II and peptide IV can be used to improve the design of current ACE inhibitors used for hypertension treatment.


Subject(s)
Antihypertensive Agents , Bradykinin , Animals , Male , Amino Acid Sequence , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Angiotensins , Antihypertensive Agents/pharmacology , Antihypertensive Agents/therapeutic use , Bradykinin/pharmacology , Creatinine , Lisinopril/pharmacology , Lisinopril/therapeutic use , Peptides/pharmacology , Peptides/therapeutic use , Peptides/analysis , Pharmaceutical Preparations , Snake Venoms , Rats
15.
J Med Chem ; 65(4): 3371-3387, 2022 02 24.
Article in English | MEDLINE | ID: mdl-35113565

ABSTRACT

Selective inhibition of the angiotensin-converting enzyme C-domain (cACE) and neprilysin (NEP), leaving the ACE N-domain (nACE) free to degrade bradykinin and other peptides, has the potential to provide the potent antihypertensive and cardioprotective benefits observed for nonselective dual ACE/NEP inhibitors, such as omapatrilat, without the increased risk of adverse effects. We have synthesized three 1-carboxy-3-phenylpropyl dipeptide inhibitors with nanomolar potency based on the previously reported C-domain selective ACE inhibitor lisinopril-tryptophan (LisW) to probe the structural requirements for potent dual cACE/NEP inhibition. Here we report the synthesis, enzyme kinetic data, and high-resolution crystal structures of these inhibitors bound to nACE and cACE, providing valuable insight into the factors driving potency and selectivity. Overall, these results highlight the importance of the interplay between the S1' and S2' subsites for ACE domain selectivity, providing guidance for future chemistry efforts toward the development of dual cACE/NEP inhibitors.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacology , Neprilysin/pharmacology , Peptidyl-Dipeptidase A/drug effects , Angiotensin-Converting Enzyme Inhibitors/chemical synthesis , Binding Sites/drug effects , Bradykinin/metabolism , Computer Simulation , Crystallography, X-Ray , Humans , Kinetics , Lisinopril/pharmacology , Peptidyl-Dipeptidase A/chemistry , Pyridines/pharmacology , Thiazepines/pharmacology
16.
Genes (Basel) ; 13(1)2022 01 14.
Article in English | MEDLINE | ID: mdl-35052483

ABSTRACT

Despite impressive results in restoring physical performance in rodent models, treatment with renin-angiotensin system (RAS) inhibitors, such as Lisinopril, have highly mixed results in humans, likely, in part, due to genetic variation in human populations. To date, the genetic determinants of responses to drugs, such as RAS inhibitors, remain unknown. Given the complexity of the relationship between physical traits and genetic background, genomic studies which predict genotype- and age-specific responses to drug treatments in humans or vertebrate animals are difficult. Here, using 126 genetically distinct lines of Drosophila melanogaster, we tested the effects of Lisinopril on age-specific climbing speed and endurance. Our data show that functional response and sensitivity to Lisinopril treatment ranges from significant protection against physical decline to increased weakness depending on genotype and age. Furthermore, genome-wide analyses led to identification of evolutionarily conserved genes in the WNT signaling pathway as being significantly associated with variations in physical performance traits and sensitivity to Lisinopril treatment. Genetic knockdown of genes in the WNT signaling pathway, Axin, frizzled, nemo, and wingless, diminished or abolished the effects of Lisinopril treatment on climbing speed traits. Our results implicate these genes as contributors to the genotype- and age-specific effects of Lisinopril treatment and because they have orthologs in humans, they are potential therapeutic targets for improvement of resiliency. Our approach should be widely applicable for identifying genomic variants that predict age- and sex-dependent responses to any type of pharmaceutical treatment.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacology , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Gene Expression Regulation, Developmental/drug effects , Genome-Wide Association Study , Lisinopril/pharmacology , Physical Functional Performance , Age Factors , Animals , Drosophila Proteins/genetics , Drosophila melanogaster/drug effects , Drosophila melanogaster/growth & development , Female , Male , Muscle, Skeletal/cytology , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism
17.
Int J Radiat Oncol Biol Phys ; 113(1): 177-191, 2022 05 01.
Article in English | MEDLINE | ID: mdl-35093482

ABSTRACT

PURPOSE: Radiation-induced lung injury is a major dose-limiting toxicity for thoracic radiation therapy patients. In experimental models, treatment with angiotensin converting enzyme (ACE) inhibitors mitigates radiation pneumonitis; however, the mechanism of action is not well understood. Here, we evaluate the direct role of ACE inhibition on lung immune cells. METHODS AND MATERIALS: ACE expression and activity were determined in the lung immune cell compartment of irradiated adult rats after either high dose fractionated radiation therapy to the right lung (5 fractions × 9 Gy) or a single dose of 13.5 Gy partial body irradiation. Mitigation of radiation-induced pneumonitis with the ACE-inhibitor lisinopril was evaluated in the 13.5 Gy rat partial body irradiation model. During pneumonitis, we characterized inflammation and immune cell content in the lungs and bronchoalveolar lavage fluid. In vitro mechanistic studies were performed using primary human monocytes and the human monocytic THP-1 cell line. RESULTS: In both the partial body irradiation and fractionated radiation therapy models, radiation increased ACE activity in lung immune cells. Treatment with lisinopril improved survival during radiation pneumonitis (P = .0004). Lisinopril abrogated radiation-induced increases in bronchoalveolar lavage fluid monocyte chemoattractant protein 1 (chemokine ligand 2) and MIP-1a cytokine levels (P < .0001). Treatment with lisinopril reduced both ACE expression (P = .006) and frequency of CD45+ CD11b+ lung myeloid cells (P = .004). In vitro, radiation injury acutely increased ACE activity (P = .045) and reactive oxygen species (ROS) generation (P = .004) in human monocytes, whereas treatment with lisinopril blocked radiation-induced increases in both ACE and ROS. Radiation-induced ROS generation was blocked by pharmacologic inhibition of either NADPH oxidase 2 (P = .012) or the type 1 angiotensin receptor (P = .013). CONCLUSIONS: These data demonstrate radiation-induced ACE activation within the immune compartment promotes the pathogenesis of radiation pneumonitis, while ACE inhibition suppresses activation of proinflammatory immune cell subsets. Mechanistically, our in vitro data demonstrate radiation directly activates the ACE/type 1 angiotensin receptor pathway in immune cells and promotes generation of ROS via NADPH oxidase 2.


Subject(s)
Radiation Injuries , Radiation Pneumonitis , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Animals , Humans , Lisinopril/pharmacology , Lisinopril/therapeutic use , Lung/radiation effects , Monocytes , NADPH Oxidase 2/metabolism , Peptidyl-Dipeptidase A/metabolism , Peptidyl-Dipeptidase A/therapeutic use , Radiation Injuries/pathology , Radiation Pneumonitis/drug therapy , Radiation Pneumonitis/etiology , Radiation Pneumonitis/prevention & control , Rats , Reactive Oxygen Species/metabolism , Receptors, Angiotensin/metabolism , Receptors, Angiotensin/therapeutic use
18.
Curr Mol Pharmacol ; 15(3): 582-588, 2022.
Article in English | MEDLINE | ID: mdl-34077352

ABSTRACT

BACKGROUND: We previously tested two angiotensin-converting enzyme (ACE) inhibitors and two dipeptidyl peptidase-4 (DPP-4) inhibitors for dual enzyme inhibitory effect. Only two DPP-4 inhibitors, linagliptin and sitagliptin, were able to inhibit ACE. OBJECTIVE: In the present study, we investigated if other inhibitors of ACE or DPP-4 could simultaneously inhibit the activities of both DPP-4 and ACE. METHODS: Forty Sprague Dawley rats were used. The control group received only saline. The other three groups were treated with anagliptin, ramipril, or lisinopril. Two different doses were tested separated with a 6-day drug-free interval. Angiotensin II (Ang II) levels and the activities of ACE and DPP-4 were measured from blood samples at baseline and days 1, 10, and 14. After the oral glucose challenge test, levels of the active form of glucagon-like peptide-1 (GLP-1) were measured. RESULTS: Regardless of the dose, anagliptin did not show any inhibitory effect on the activity of ACE or Ang II levels. Concerning ramipril and lisinopril, only a high dose of lisinopril was able to produce a modest reduction of the DPP-4 activity but not enough to inhibit the inactivation of GLP-1. CONCLUSION: It seems that while most of the ACE inhibitors cannot affect DPP-4 activity, inhibitors of DPP-4 vary in their effects on ACE activity. The selection of DPP-4 inhibitors under different clinical situations should take into account the action of these drugs on ACE.


Subject(s)
Dipeptidyl-Peptidase IV Inhibitors , Lisinopril , Angiotensin II , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Glucagon-Like Peptide 1 , Lisinopril/pharmacology , Pyrimidines , Ramipril/pharmacology , Rats , Rats, Sprague-Dawley
19.
F1000Res ; 10: 15, 2021.
Article in English | MEDLINE | ID: mdl-34707860

ABSTRACT

Background: Angiotensin-converting enzyme (ACE) inhibitors have been shown to promote endothelial progenitor cell (EPC) function. However, the efficacies of different ACE inhibitors in improving the migratory capabilities of ECPs in coronary artery disease (CAD) patients is unclear. This study compared the effectiveness of captopril, lisinopril, and ramipril toward the migration capability of impaired EPCs from CAD patients. Methods: We isolated peripheral blood mononuclear cells (PBMCs), separated EPCs from PBMCs, and divided them into an untreated group (control) and treated groups of captopril, lisinopril, and ramipril at doses of 1mM, 10mM, and 100mM. EPC migration was evaluated using the Boyden chamber assay. Analysis of variance (ANOVA) was performed using SPSS 25.0. Results: This study showed that treatment with captopril, lisinopril, and ramipril starting at the lowest dose (1 mM) increased EPC migration (65,250 ± 6,750 cells; 60,750± 5,030 cells; and 49,500 ± 8,400 cells, respectively) compared to control (43,714 ± 7,216 cells). Increased migration of EPCs was observed by increasing the treatment dose to 10 mM with captopril, lisinopril, and ramipril (90,000 ± 16,837 cells; 79,071 ± 2,043 cells; and 64,285 ± 11,824 cells, respectively). The highest EPC migration was shown for lisinopril 100 mM (150,750 ± 16,380 cells), compared to captopril and ramipril at the same dose (105,750 ± 8112 cells and 86,625 ± 5,845 cells, respectively). Conclusions: Captopril, ramipril, and lisinopril were shown to increase EPC migration in a dose-dependent manner. Low-dose (1 mM) and medium-dose (10 mM) captopril had a larger effect on ECP migration than lisinopril and ramipril. Meanwhile, high-dose lisinopril (100mM) had the highest migration effect, suggesting it may be preferable for promoting EPC migration in CAD patients.


Subject(s)
Captopril , Endothelial Progenitor Cells , Captopril/pharmacology , Humans , Leukocytes, Mononuclear , Lisinopril/pharmacology , Ramipril/pharmacology
20.
Int J Biol Macromol ; 190: 989-998, 2021 Nov 01.
Article in English | MEDLINE | ID: mdl-34537299

ABSTRACT

The present study aimed to investigate the thermal- and pH-dependent gelation behavior of gelatin/HPMCP blends using ultraviolet (UV) spectrophotometry, viscoelasticity, and dynamic light scattering (DLS). We found that the release of lisinopril from gelatin/HPMCP gels can be inhibited at low pH. UV spectrophotometric analysis showed that pH had a significant effect on the transparency of aqueous HPMCP systems and gelatin/HPMCP gels. The viscoelastic patterns of gelatin/HPMCP at pH 4.6 considerably differed from those of gelatin/HPMCP at pH 5.2 and 6.0. DLS measurements showed that HPMCP molecules in low concentrations underwent strong aggregation below pH 4.8. Such HPMCP aggregation induces a physical barrier in the matrix structures of the gelatin/HPMCP gels, which inhibits the drug release at pH 1.2. This hydrogel delivery system using polymer blends of gelatin/HPMCP can be used in oral gel formulations with pH-responsive properties.


Subject(s)
Drug Liberation , Gelatin/chemistry , Methylcellulose/analogs & derivatives , Dynamic Light Scattering , Elastic Modulus , Hydrogen-Ion Concentration , Lisinopril/pharmacology , Methylcellulose/chemistry , Polymers/chemistry , Rheology , Static Electricity
SELECTION OF CITATIONS
SEARCH DETAIL
...