Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39.396
Filter
1.
Sci Rep ; 14(1): 10789, 2024 05 11.
Article in English | MEDLINE | ID: mdl-38734719

ABSTRACT

Brown adipocytes are potential therapeutic targets for the prevention of obesity-associated metabolic diseases because they consume circulating glucose and fatty acids for heat production. Angiotensin II (Ang II) peptide is involved in the pathogenesis of obesity- and cold-induced hypertension; however, the mechanism underlying the direct effects of Ang II on human brown adipocytes remains unclear. Our transcriptome analysis of chemical compound-induced brown adipocytes (ciBAs) showed that the Ang II type 1 receptor (AGTR1), but not AGTR2 and MAS1 receptors, was expressed. The Ang II/AGTR1 axis downregulated the expression of mitochondrial uncoupling protein 1 (UCP1). The simultaneous treatment with ß-adrenergic receptor agonists and Ang II attenuated UCP1 expression, triglyceride lipolysis, and cAMP levels, although cAMP response element-binding protein (CREB) phosphorylation was enhanced by Ang II mainly through the protein kinase C pathway. Despite reduced lipolysis, both coupled and uncoupled mitochondrial respiration was enhanced in Ang II-treated ciBAs. Instead, glycolysis and glucose uptake were robustly activated upon treatment with Ang II without a comprehensive transcriptional change in glucose metabolic genes. Elevated mitochondrial energy status induced by Ang II was likely associated with UCP1 repression. Our findings suggest that the Ang II/AGTR1 axis participates in mitochondrial thermogenic functions via glycolysis.


Subject(s)
Adipocytes, Brown , Angiotensin II , Glycolysis , Mitochondria , Thermogenesis , Uncoupling Protein 1 , Humans , Adipocytes, Brown/metabolism , Adipocytes, Brown/drug effects , Glycolysis/drug effects , Angiotensin II/pharmacology , Angiotensin II/metabolism , Mitochondria/metabolism , Mitochondria/drug effects , Thermogenesis/drug effects , Uncoupling Protein 1/metabolism , Uncoupling Protein 1/genetics , Lipolysis/drug effects , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 1/genetics , Glucose/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism
2.
J Physiol Pharmacol ; 75(2): 185-194, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38736265

ABSTRACT

We have previously described local aldosterone synthesis in mouse colon. In the renin-angiotensin-aldosterone system (RAAS), angiotensin II (Ang II) peptide is the physiological factor which stimulates aldosterone synthesis in the adrenal glands. We have recently demonstrated that Ang II stimulates aldosterone synthesis also in mouse colon. Here, we conducted a 75-min ex vivo incubation of murine colonic tissue and evaluated the effects of three other Ang peptides, Ang I (1 µM), Ang III (0.1 µM) and Ang (1-7) (0.1 µM) on aldosterone synthesis. As a possible mechanism, their effects on tissue levels of the rate-limiting enzyme, aldosterone synthase (CYP11B2) were measured by ELISA and Western blot. Ang III significantly elevated the amount of tissue CYP11B2 protein in colon. The values of released aldosterone in colon tissue incubation were increased over the control in the presence of Ang I, II or III, however, being statistically non-significant. In Western blot analysis, the values of tissue CYP11B2 protein content were elevated by Ang I and II. Ang (1-7) alone in colon did not influence CYP11B2 protein levels in the incubation experiment but showed higher aldosterone release without statistical significance. Ang (1-7) showed an antagonistic effect towards Ang II in release of aldosterone in adrenal gland. An overall estimation of a single peptide (three measured variables), the results were always in an increasing direction. The responses of aldosterone synthesis to high levels of glucose (44 mM) and potassium (18.8 mM) as physiological stimulators in vivo were investigated in the colon incubation. Glucose, equal to four times the concentration of the control buffer in the incubation, showed higher values of aldosterone release in colon than control without statistical significance similarly to the effect seen in adrenal glands. Increasing the concentration of potassium in the incubation buffer exerted no effect on colonic aldosterone production. Intriguingly, no correlation was found between aldosterone release and the tissue CYP11B2 protein content in colon. In summary, the response of colonic aldosterone synthesis to different Ang peptides resembles, but is not identical to, the situation in the adrenal glands.


Subject(s)
Aldosterone , Colon , Cytochrome P-450 CYP11B2 , Glucose , Potassium , Animals , Male , Mice , Aldosterone/metabolism , Angiotensin I/physiology , Angiotensin II/physiology , Angiotensin III/physiology , Colon/metabolism , Colon/drug effects , Cytochrome P-450 CYP11B2/metabolism , Glucose/metabolism , Peptide Fragments/physiology , Potassium/metabolism
3.
Int J Mol Sci ; 25(9)2024 May 03.
Article in English | MEDLINE | ID: mdl-38732210

ABSTRACT

Investigating the role of podocytes in proteinuric disease is imperative to address the increasing global burden of chronic kidney disease (CKD). Studies strongly implicate increased levels of monocyte chemoattractant protein-1 (MCP-1/CCL2) in proteinuric CKD. Since podocytes express the receptor for MCP-1 (i.e., CCR2), we hypothesized that podocyte-specific MCP-1 production in response to stimuli could activate its receptor in an autocrine manner, leading to further podocyte injury. To test this hypothesis, we generated podocyte-specific MCP-1 knockout mice (Podo-Mcp-1fl/fl) and exposed them to proteinuric injury induced by either angiotensin II (Ang II; 1.5 mg/kg/d, osmotic minipump) or Adriamycin (Adr; 18 mg/kg, intravenous bolus). At baseline, there were no between-group differences in body weight, histology, albuminuria, and podocyte markers. After 28 days, there were no between-group differences in survival, change in body weight, albuminuria, kidney function, glomerular injury, and tubulointerstitial fibrosis. The lack of protection in the knockout mice suggests that podocyte-specific MCP-1 production is not a major contributor to either Ang II- or Adr-induced glomerular disease, implicating that another cell type is the source of pathogenic MCP-1 production in CKD.


Subject(s)
Angiotensin II , Chemokine CCL2 , Doxorubicin , Mice, Knockout , Podocytes , Animals , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Podocytes/metabolism , Podocytes/pathology , Podocytes/drug effects , Doxorubicin/adverse effects , Mice , Male , Renal Insufficiency, Chronic/chemically induced , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology , Gene Deletion , Disease Models, Animal
4.
Clin Sci (Lond) ; 138(10): 573-597, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38718356

ABSTRACT

The three striatins (STRN, STRN3, STRN4) form the core of STRiatin-Interacting Phosphatase and Kinase (STRIPAK) complexes. These place protein phosphatase 2A (PP2A) in proximity to protein kinases thereby restraining kinase activity and regulating key cellular processes. Our aim was to establish if striatins play a significant role in cardiac remodelling associated with cardiac hypertrophy and heart failure. All striatins were expressed in control human hearts, with up-regulation of STRN and STRN3 in failing hearts. We used mice with global heterozygote gene deletion to assess the roles of STRN and STRN3 in cardiac remodelling induced by angiotensin II (AngII; 7 days). Using echocardiography, we detected no differences in baseline cardiac function or dimensions in STRN+/- or STRN3+/- male mice (8 weeks) compared with wild-type littermates. Heterozygous gene deletion did not affect cardiac function in mice treated with AngII, but the increase in left ventricle mass induced by AngII was inhibited in STRN+/- (but not STRN3+/-) mice. Histological staining indicated that cardiomyocyte hypertrophy was inhibited. To assess the role of STRN in cardiomyocytes, we converted the STRN knockout line for inducible cardiomyocyte-specific gene deletion. There was no effect of cardiomyocyte STRN knockout on cardiac function or dimensions, but the increase in left ventricle mass induced by AngII was inhibited. This resulted from inhibition of cardiomyocyte hypertrophy and cardiac fibrosis. The data indicate that cardiomyocyte striatin is required for early remodelling of the heart by AngII and identify the striatin-based STRIPAK system as a signalling paradigm in the development of pathological cardiac hypertrophy.


Subject(s)
Angiotensin II , Cardiomegaly , Mice, Knockout , Myocytes, Cardiac , Animals , Angiotensin II/pharmacology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Cardiomegaly/genetics , Cardiomegaly/pathology , Cardiomegaly/metabolism , Cardiomegaly/physiopathology , Male , Humans , Muscle Proteins/metabolism , Muscle Proteins/genetics , Ventricular Remodeling , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Calmodulin-Binding Proteins , Nerve Tissue Proteins
5.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167224, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38723872

ABSTRACT

BACKGROUND: Pentamethylquercetin (PMQ) is a natural polymethyl flavonoid that possesses anti-apoptotic and other biological properties. Abdominal aortic aneurysm (AAA), a fatal vascular disease with a high risk of rupture, is associated with phenotypic switching and apoptosis of medial vascular smooth muscle cells (VSMCs). This study aimed to investigate the protective effects of PMQ on the development of AAA and the underlying mechanism. METHODS: ApoE-/- mice were continuously infused with angiotensin II (Ang II) for 4 weeks to develop the AAA model. Intragastric administration of PMQ was initiated 5 days before Ang II infusion and continued for 4 weeks. In vitro, VSMCs were cultured and pretreated with PMQ, stimulated with Ang II. Real-time PCR, western blotting, and immunofluorescence staining were used to examine the roles and mechanisms of PMQ on the phenotypic switching and apoptosis of VSMCs. RESULTS: PMQ dose-dependently reduced the incidence of Ang II-induced AAA, aneurysm diameter enlargement, elastin degradation, VSMCs phenotypic switching and apoptosis. Furthermore, PMQ also inhibited phenotypic switching and apoptosis in Ang II-stimulated VSMCs. PMQ exerted protective effects by regulating the C/EBPß/PTEN/AKT/GSK-3ß axis. AAV-mediated overexpression of PTEN reduced the therapeutic effects of PMQ in the AAA model mice, suggesting that the effects of PMQ on Ang II-mediated AAA formation were related to the PTEN/AKT/GSK-3ß axis. PMQ inhibited VSMCs phenotypic switching and apoptosis by bounding to C/EBPß at Lys253 with hydrogen bond to regulate C/EBPß nuclear translocation and PTEN/AKT/GSK-3ß axis, thereby inhibiting Ang II-induced AAA formation. CONCLUSIONS: Pentamethylquercetin inhibits angiotensin II-induced abdominal aortic aneurysm formation by bounding to C/EBPß at Lys253. Therefore, PMQ prevents the formation of AAA and reduces the incidence of AAA.


Subject(s)
Angiotensin II , Aortic Aneurysm, Abdominal , Apoptosis , Muscle, Smooth, Vascular , Quercetin , Animals , Aortic Aneurysm, Abdominal/metabolism , Aortic Aneurysm, Abdominal/pathology , Aortic Aneurysm, Abdominal/prevention & control , Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/drug therapy , Angiotensin II/pharmacology , Mice , Quercetin/analogs & derivatives , Quercetin/pharmacology , Apoptosis/drug effects , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Male , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Disease Models, Animal , PTEN Phosphohydrolase/metabolism , PTEN Phosphohydrolase/genetics , Proto-Oncogene Proteins c-akt/metabolism , Mice, Inbred C57BL , Glycogen Synthase Kinase 3 beta/metabolism , Signal Transduction/drug effects , Cells, Cultured , Cell Nucleus/metabolism , Cell Nucleus/drug effects
6.
Arch Virol ; 169(6): 121, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38753119

ABSTRACT

Previous studies have demonstrated the relevance of several soluble molecules in the pathogenesis of dengue. In this regard, a possible role for angiotensin II (Ang II) in the pathophysiology of dengue has been suggested by the observation of a blockade of Ang II in patients with dengue, increased expression of molecules related to Ang II production in the plasma of dengue patients, increased expression of circulating cytokines and soluble molecules related to the action of Ang II, and an apparent relationship between DENV, Ang II effects, and miRNAs. In addition, in ex vivo experiments, the blockade of Ang II AT1 receptor and ACE-1 (angiotensin converting enzyme 1), both of which are involved in Ang II production and its function, inhibits infection of macrophages by DENV, suggesting a role of Ang II in viral entry or in intracellular viral replication of the virus. Here, we discuss the possible mechanisms of Ang II in the entry and replication of DENV. Ang II has the functions of increasing the expression of DENV entry receptors, creation of clathrin-coated vesicles, and increasing phagocytosis, all of which are involved in DENV entry. This hormone also modulates the expression of the Rab5 and Rab7 proteins, which are important in the endosomal processing of DENV during viral replication. This review summarizes the data related to the possible involvement of Ang II in the entry of DENV into cells and its replication.


Subject(s)
Angiotensin II , Dengue Virus , Virus Internalization , Virus Replication , Angiotensin II/metabolism , Humans , Dengue Virus/physiology , Dengue Virus/genetics , Animals , Dengue/virology , Dengue/metabolism
7.
J Am Heart Assoc ; 13(10): e033998, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38726925

ABSTRACT

BACKGROUND: The vasoconstrictor effects of angiotensin II via type 1 angiotensin II receptors in vascular smooth muscle cells are well established, but the direct effects of angiotensin II on vascular endothelial cells (VECs) in vivo and the mechanisms how VECs may mitigate angiotensin II-mediated vasoconstriction are not fully understood. The present study aimed to explore the molecular mechanisms and pathophysiological relevance of the direct actions of angiotensin II on VECs in kidney and brain microvessels in vivo. METHODS AND RESULTS: Changes in VEC intracellular calcium ([Ca2+]i) and nitric oxide (NO) production were visualized by intravital multiphoton microscopy of cadherin 5-Salsa6f mice or the endothelial uptake of NO-sensitive dye 4-amino-5-methylamino-2',7'-difluorofluorescein diacetate, respectively. Kidney fibrosis by unilateral ureteral obstruction and Ready-to-use adeno-associated virus expressing Mouse Renin 1 gene (Ren1-AAV) hypertension were used as disease models. Acute systemic angiotensin II injections triggered >4-fold increases in VEC [Ca2+]i in brain and kidney resistance arterioles and capillaries that were blocked by pretreatment with the type 1 angiotensin II receptor inhibitor losartan, but not by the type 2 angiotensin II receptor inhibitor PD123319. VEC responded to acute angiotensin II by increased NO production as indicated by >1.5-fold increase in 4-amino-5-methylamino-2',7'-difluorofluorescein diacetate fluorescence intensity. In mice with kidney fibrosis or hypertension, the angiotensin II-induced VEC [Ca2+]i and NO responses were significantly reduced, which was associated with more robust vasoconstrictions, VEC shedding, and microthrombi formation. CONCLUSIONS: The present study directly visualized angiotensin II-induced increases in VEC [Ca2+]i and NO production that serve to counterbalance agonist-induced vasoconstriction and maintain residual organ blood flow. These direct and endothelium-specific angiotensin II effects were blunted in disease conditions and linked to endothelial dysfunction and the development of vascular pathologies.


Subject(s)
Angiotensin II , Brain , Calcium , Hypertension , Kidney , Microvessels , Nitric Oxide , Vasoconstriction , Animals , Nitric Oxide/metabolism , Angiotensin II/pharmacology , Hypertension/metabolism , Hypertension/physiopathology , Hypertension/drug therapy , Kidney/blood supply , Kidney/metabolism , Calcium/metabolism , Vasoconstriction/drug effects , Microvessels/metabolism , Microvessels/drug effects , Microvessels/pathology , Brain/metabolism , Brain/blood supply , Mice , Disease Models, Animal , Male , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Mice, Inbred C57BL , Calcium Signaling/drug effects
8.
J Hypertens ; 42(6): 1101-1104, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38690908

ABSTRACT

Isolated nocturnal hypertension (INHT), defined as nighttime elevated blood pressure (BP) with normal daytime BP assessed by ambulatory BP monitoring, is associated with higher cardiovascular morbidity and mortality. We hypothesized that an alteration in the circulating renin-angiotensin system (RAS) contributes to INHT development. We examined circulating levels of angiotensin (Ang) (1-7) and Ang II and ACE2 activity in 26 patients that met the INHT criteria, out of 50 that were referred for BP evaluation (62% women, 45 ±â€Š16 years old). Those with INHT were older, had a higher BMI, lower circulating Ang-(1-7) (P = 0.002) and Ang II levels (P = 0.02) and no change in ACE2 activity compared to those normotensives. Nighttime DBP was significantly correlated with Ang-(1-7) and Ang II levels. Logistic regression showed significant association in Ang-(1-7) and Ang II levels with INHT. Our study reveals differences in circulating RAS in individuals with INHT.


Subject(s)
Angiotensin II , Angiotensin I , Hypertension , Peptide Fragments , Humans , Angiotensin I/blood , Female , Male , Middle Aged , Peptide Fragments/blood , Hypertension/blood , Hypertension/physiopathology , Adult , Angiotensin II/blood , Renin-Angiotensin System/physiology , Circadian Rhythm , Blood Pressure , Angiotensin-Converting Enzyme 2/blood , Blood Pressure Monitoring, Ambulatory , Peptidyl-Dipeptidase A/blood
9.
J Cell Mol Med ; 28(9): e18321, 2024 May.
Article in English | MEDLINE | ID: mdl-38712979

ABSTRACT

As a main extraction compound from Scutellaria baicalensis Georgi, Baicalin exhibits various biological activities. However, the underlying mechanism of Baicalin on hypertension-induced heart injury remains unclear. In vivo, mice were infused with angiotensin II (Ang II; 500 ng/kg/min) or saline using osmotic pumps, followed by intragastrically administrated with Baicalin (5 mg/kg/day) for 4 weeks. In vitro, H9C2 cells were stimulated with Ang II (1 µM) and treated with Baicalin (12.5, 25 and 50 µM). Baicalin treatment significantly attenuated the decrease in left ventricular ejection fraction and left ventricular fractional shortening, increase in left ventricular mass, left ventricular systolic volume and left ventricular diastolic volume of Ang II infused mice. Moreover, Baicalin treatment reversed 314 differentially expressed transcripts in the cardiac tissues of Ang II infused mice, and enriched multiple enriched signalling pathways (including apoptosis, autophagy, AMPK/mTOR signalling pathway). Consistently, Baicalin treatment significantly alleviated Ang II-induced cell apoptosis in vivo and in vitro. Baicalin treatment reversed the up-regulation of Bax, cleaved-caspase 3, cleaved-caspase 9, and the down-regulation of Bcl-2. Meanwhile, Baicalin treatment alleviated Ang II-induced increase of autophagosomes, restored autophagic flux, and down-regulated LC3II, Beclin 1, as well as up-regulated SQSTM1/p62 expression. Furthermore, autophagy inhibitor 3-methyladenine treatment alleviated the increase of autophagosomes and the up-regulation of Beclin 1, LC3II, Bax, cleaved-caspase 3, cleaved-caspase 9, down-regulation of SQSTM1/p62 and Bcl-2 expression after Ang II treated, which similar to co-treatment with Baicalin. Baicalin treatment reduced the ratio of p-AMPK/AMPK, while increased the ratio of p-mTOR/mTOR. Baicalin alleviated Ang II-induced cardiomyocyte apoptosis and autophagy, which might be related to the inhibition of the AMPK/mTOR pathway.


Subject(s)
AMP-Activated Protein Kinases , Angiotensin II , Apoptosis , Autophagy , Flavonoids , Myocytes, Cardiac , Signal Transduction , TOR Serine-Threonine Kinases , Flavonoids/pharmacology , Animals , Autophagy/drug effects , Apoptosis/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , TOR Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Mice , AMP-Activated Protein Kinases/metabolism , Male , Mice, Inbred C57BL , Cell Line , Rats
10.
Cells ; 13(9)2024 May 03.
Article in English | MEDLINE | ID: mdl-38727319

ABSTRACT

In our previous studies, we showed that the generation of ovarian tumors in NSG mice (immune-compromised) resulted in the induction of muscle and cardiac cachexia, and treatment with withaferin A (WFA; a steroidal lactone) attenuated both muscle and cardiac cachexia. However, our studies could not address if these restorations by WFA were mediated by its anti-tumorigenic properties that might, in turn, reduce the tumor burden or WFA's direct, inherent anti-cachectic properties. To address this important issue, in our present study, we used a cachectic model induced by the continuous infusion of Ang II by implanting osmotic pumps in immunocompetent C57BL/6 mice. The continuous infusion of Ang II resulted in the loss of the normal functions of the left ventricle (LV) (both systolic and diastolic), including a significant reduction in fractional shortening, an increase in heart weight and LV wall thickness, and the development of cardiac hypertrophy. The infusion of Ang II also resulted in the development of cardiac fibrosis, and significant increases in the expression levels of genes (ANP, BNP, and MHCß) associated with cardiac hypertrophy and the chemical staining of the collagen abundance as an indication of fibrosis. In addition, Ang II caused a significant increase in expression levels of inflammatory cytokines (IL-6, IL-17, MIP-2, and IFNγ), NLRP3 inflammasomes, AT1 receptor, and a decrease in AT2 receptor. Treatment with WFA rescued the LV functions and heart hypertrophy and fibrosis. Our results demonstrated, for the first time, that, while WFA has anti-tumorigenic properties, it also ameliorates the cardiac dysfunction induced by Ang II, suggesting that it could be an anticachectic agent that induces direct effects on cardiac muscles.


Subject(s)
Angiotensin II , Cachexia , Mice, Inbred C57BL , Withanolides , Withanolides/pharmacology , Withanolides/therapeutic use , Animals , Cachexia/drug therapy , Cachexia/pathology , Mice , Cardiomegaly/drug therapy , Cardiomegaly/pathology , Cytokines/metabolism , Myocardium/pathology , Myocardium/metabolism , Fibrosis , Female
11.
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
12.
FASEB J ; 38(9): e23654, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38717442

ABSTRACT

Heart failure and cardiac remodeling are both characterized by mitochondrial dysfunction. Healthy mitochondria are required for adequate contractile activity and appropriate regulation of cell survival. In the mammalian heart, enhancement of the mitochondrial unfolded protein response (UPRmt) is cardioprotective under pressure overload conditions. We explored the UPRmt and the underlying regulatory mechanism in terms of hypertension-induced cardiac remodeling and the cardioprotective effect of metformin. Male spontaneously hypertensive rats and angiotensin II-treated neonatal rat cardiomyocytes were used to induce cardiac hypertrophy. The results showed that hypertension induced the formation of aberrant mitochondria, characterized by a reduced mtDNA/nDNA ratio and swelling, as well as lower levels of mitochondrial complexes I to V and inhibition of the expression of one protein subunit of each of complexes I to IV. Such changes eventually enlarged cardiomyocytes and increased cardiac fibrosis. Metformin treatment increased the mtDNA/nDNA ratio and regulated the UPRmt, as indicated by increased expression of activating transcription factor 5, Lon protease 1, and heat shock protein 60, and decreased expression of C/EBP homologous protein. Thus, metformin improved mitochondrial ultrastructure and function in spontaneously hypertensive rats. In vitro analyses revealed that metformin reduced the high levels of angiotensin II-induced mitochondrial reactive oxygen species in such animals and stimulated nuclear translocation of heat shock factor 1 (HSF1). Moreover, HSF1 small-interfering RNA reduced the metformin-mediated improvements in mitochondrial morphology and the UPRmt by suppressing hypertrophic signals and cardiomyocyte apoptosis. These results suggest that HSF1/UPRmt signaling contributes to the beneficial effects of metformin. Metformin-mediated targeting of mitochondrial protein homeostasis and modulation of HSF1 levels have potential therapeutic implications in terms of cardiac remodeling.


Subject(s)
Heat Shock Transcription Factors , Metformin , Myocytes, Cardiac , Rats, Inbred SHR , Unfolded Protein Response , Animals , Metformin/pharmacology , Unfolded Protein Response/drug effects , Male , Rats , Heat Shock Transcription Factors/metabolism , Heat Shock Transcription Factors/genetics , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Hypertension/metabolism , Hypertension/drug therapy , Ventricular Remodeling/drug effects , Mitochondria, Heart/metabolism , Mitochondria, Heart/drug effects , Angiotensin II/pharmacology , Cardiomegaly/metabolism , Cardiomegaly/drug therapy , Cardiomegaly/pathology , Transcription Factors/metabolism , Transcription Factors/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Rats, Inbred WKY
13.
Cells ; 13(9)2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38727271

ABSTRACT

Vascular smooth muscle cells (VSMCs) play a key role in aortic aneurysm formation. Bone morphogenetic proteins (BMPs) have been implicated as important regulators of VSMC phenotype, and dysregulation of the BMP pathway has been shown to be associated with vascular diseases. The aim of this study was to investigate for the first time the effects of BMP-4 on the VSMC phenotype and to understand its role in the development of thoracic aortic aneurysms (TAAs). Using the angiotensin II (AngII) osmotic pump model in mice, aortas from mice with VSMC-specific BMP-4 deficiency showed changes similar to AngII-infused aortas, characterised by a loss of contractile markers, increased fibrosis, and activation of matrix metalloproteinase 9. When BMP-4 deficiency was combined with AngII infusion, there was a significantly higher rate of apoptosis and aortic dilatation. In vitro, VSMCs with mRNA silencing of BMP-4 displayed a dedifferentiated phenotype with activated canonical BMP signalling. In contrast, BMP-2-deficient VSMCs exhibited the opposite phenotype. The compensatory regulation between BMP-2 and BMP-4, with BMP-4 promoting the contractile phenotype, appeared to be independent of the canonical signalling pathway. Taken together, these results demonstrate the impact of VSMC-specific BMP-4 deficiency on TAA development.


Subject(s)
Angiotensin II , Aortic Aneurysm, Thoracic , Bone Morphogenetic Protein 2 , Bone Morphogenetic Protein 4 , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Phenotype , Animals , Bone Morphogenetic Protein 4/metabolism , Aortic Aneurysm, Thoracic/metabolism , Aortic Aneurysm, Thoracic/pathology , Aortic Aneurysm, Thoracic/genetics , Mice , Bone Morphogenetic Protein 2/metabolism , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Angiotensin II/pharmacology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Signal Transduction , Mice, Inbred C57BL , Male , Apoptosis/drug effects , Disease Models, Animal
14.
Am J Physiol Cell Physiol ; 326(4): C1203-C1211, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38581656

ABSTRACT

Cardiometabolic diseases are often associated with heightened levels of angiotensin II (Ang II), which accounts for the observed oxidative stress, inflammation, and fibrosis. Accumulating evidence indicates a parallel upregulation of dipeptidyl dipeptidase 4 (DPP4) activity in cardiometabolic diseases, with its inhibition shown to mitigate oxidative stress, inflammation, and fibrosis. These findings highlight an overlap between the pathophysiological mechanisms used by Ang II and DPP4. Recent evidence demonstrates that targeted inhibition of DPP4 prevents the rise in Ang II and its associated molecules in experimental models of cardiometabolic diseases. Similarly, inhibitors of the angiotensin I-converting enzyme (ACE) or Ang II type 1 receptor (AT1R) blockers downregulate DPP4 activity, establishing a bidirectional relationship between DPP4 and Ang II. Here, we discuss the current evidence supporting the cross talk between Ang II and DPP4, along with the potential mechanisms promoting this cross regulation. A comprehensive analysis of this bidirectional relationship across tissues will advance our understanding of how DPP4 and Ang II collectively promote the development and progression of cardiometabolic diseases.


Subject(s)
Angiotensin II , Cardiovascular Diseases , Humans , Dipeptidyl Peptidase 4 , Peptidyl-Dipeptidase A , Receptor, Angiotensin, Type 1 , Inflammation , Fibrosis , Angiotensin I
15.
Biol Pharm Bull ; 47(4): 809-817, 2024.
Article in English | MEDLINE | ID: mdl-38583954

ABSTRACT

Several reports assume that myocardial necroptotic cell death is induced during the development of chronic heart failure. Although it is well accepted that angiotensin II induces apoptotic cell death of cardiac myocytes, the involvement of angiotensin II in the induction of myocardial necroptosis during the development of heart failure is still unknown. Therefore, we examined the role of angiotensin II in myocardial necroptosis using rat failing hearts following myocardial infarction and cultured cardiomyocytes. We found that administration of azilsartan, an angiotensin II AT1 receptor blocker, or trandolapril, an angiotensin-converting enzyme inhibitor, to rats from the 2nd to the 8th week after myocardial infarction resulted in preservation of cardiac function and attenuation of mixed lineage kinase domain-like (MLKL) activation. Furthermore, the ratio of necroptotic cell death was increased in neonatal rat ventricular cardiomyocytes cultured with conditioned medium from rat cardiac fibroblasts in the presence of angiotensin II. This increase in necroptotic cells was attenuated by pretreatment with azilsartan. Furthermore, activated MLKL was increased in cardiomyocytes cultured in conditioned medium. Pretreatment with azilsartan also prevented the conditioned medium-induced increase in activated MLKL. These results suggest that angiotensin II contributes to the induction of myocardial necroptosis during the development of heart failure.


Subject(s)
Heart Failure , Myocardial Infarction , Animals , Rats , Angiotensin II/pharmacology , Angiotensin II/metabolism , Angiotensin Receptor Antagonists , Culture Media, Conditioned/metabolism , Heart Failure/metabolism , Myocardial Infarction/metabolism , Myocytes, Cardiac , Protein Kinases/metabolism
16.
Mol Biol Rep ; 51(1): 506, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38622341

ABSTRACT

BACKGROUND: Atrial Fibrillation (AF), a prevalent arrhythmic condition, is intricately associated with atrial fibrosis, a major pathological contributor. Central to the development of atrial fibrosis is myocardial inflammation. This study focuses on Atrial Natriuretic Peptide (ANP) and its role in mitigating atrial fibrosis, aiming to elucidate the specific mechanisms by which ANP exerts its effects, with an emphasis on fibroblast dynamics. METHODS AND RESULTS: The study involved forty Sprague-Dawley rats, divided into four groups: control, Angiotensin II (Ang II), Ang II + ANP, and ANP only. The administration of 1 µg/kg/min Ang II was given to Ang II and Ang II + ANP groups, while both Ang II + ANP and ANP groups received 0.1 µg/kg/min ANP intravenously for a duration of 14 days. Cardiac fibroblasts were used for in vitro validation of the proposed mechanisms. The study observed that rats in the Ang II and Ang II + ANP groups showed an increase in blood pressure and a decrease in body weight, more pronounced in the Ang II group. Diastolic dysfunction, a characteristic of the Ang II group, was alleviated by ANP. Additionally, ANP significantly reduced Ang II-induced atrial fibrosis, myofibroblast proliferation, collagen overexpression, macrophage infiltration, and the elevated expression of Interleukin 6 (IL-6) and Tenascin-C (TN-C). Transcriptomic sequencing indicated enhanced PI3K/Akt signaling in the Ang II group. Furthermore, in vitro studies showed that ANP, along with the PI3K inhibitor LY294002, effectively reduced PI3K/Akt pathway activation and the expression of TN-C, collagen-I, and collagen-III, which were induced by Ang II. CONCLUSIONS: The study demonstrates ANP's potential in inhibiting myocardial inflammation and reducing atrial fibrosis. Notably, ANP's effect in countering atrial fibrosis seems to be mediated through the suppression of the Ang II-induced PI3K/Akt-Tenascin-C signaling pathway. These insights enhance our understanding of AF pathogenesis and position ANP as a potential therapeutic agent for treating atrial fibrosis.


Subject(s)
Atrial Fibrillation , Atrial Natriuretic Factor , Rats , Animals , Rats, Sprague-Dawley , Atrial Natriuretic Factor/pharmacology , Atrial Natriuretic Factor/metabolism , Proto-Oncogene Proteins c-akt , Phosphatidylinositol 3-Kinases , Tenascin , Atrial Fibrillation/drug therapy , Angiotensin II/pharmacology , Inflammation/drug therapy , Collagen , Fibrosis
17.
Physiol Rep ; 12(8): e16011, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38627219

ABSTRACT

Renin-angiotensin system activation contributes to skeletal muscle atrophy in aging individuals with chronic diseases. We aimed to explore the effects of cholecalciferol (VD3) and calcitriol (1,25VD3) on signaling of muscle proteolysis and oxidative stress in myotubes challenged with angiotensin II (AII). The mouse C2C12 myotubes were assigned to vehicle, AII, AII + VD3, AII + 1,25VD3, and AII + losartan groups. The expression levels of muscle-specific E3 ubiquitin ligase proteins, autophagy-related proteins, and oxidative stress markers were investigated. We demonstrated the diverse effects of VD3 and 1,25VD3 on AII-induced myotube atrophy. The myotube diameter was preserved by treatment with 100 nM VD3 and losartan, while 1 and 10 nM 1,25VD3 increased levels of FoxO3a, MuRF1, and atrogin-1 protein expression in myotubes exposed to AII. Treatment with AII + 10 nM 1,25VD3 resulted in the upregulation of LC3B-II, LC3B-II/LC3B-I, and mature cathepsin L, which are autophagic marker proteins. The p62/SQSTM1 protein was downregulated and vitamin D receptor was upregulated after treatment with AII + 10 nM 1,25VD3. A cellular redox imbalance was observed as AII + 10 nM 1,25VD3-induced reactive oxygen species and NADPH oxidase-2 overproduction, and these changes were associated with an inadequate response of antioxidant superoxide dismutase-1 and catalase proteins. Collectively, these findings provide a translational perspective on the role of vitamin D3 in alleviating muscle atrophy related to high levels of AII.


Subject(s)
Angiotensin II , Calcitriol , Mice , Animals , Calcitriol/adverse effects , Calcitriol/metabolism , Angiotensin II/pharmacology , Angiotensin II/metabolism , Proteolysis , Cholecalciferol/adverse effects , Losartan/pharmacology , Muscle Fibers, Skeletal/metabolism , Muscular Atrophy/metabolism , Oxidative Stress , Muscle, Skeletal/metabolism
18.
Anal Chem ; 96(16): 6106-6111, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38594830

ABSTRACT

This study explores the innovative field of pulsed direct current arc-induced nanoelectrospray ionization mass spectrometry (DCAI-nano-ESI-MS), which utilizes a low-temperature direct current (DC) arc to induce ESI during MS analyses. By employing a 15 kV output voltage, the DCAI-nano-ESI source effectively identifies various biological molecules, including angiotensin II, bradykinin, cytochrome C, and soybean lecithin, showcasing impressive analyte signals and facilitating multicharge MS in positive- and negative-ion modes. Notably, results show that the oxidation of fatty acids using a DC arc produces [M + O - H]- ions, which aid in identifying the location of C═C bonds in unsaturated fatty acids and distinguishing between isomers based on diagnostic ions observed during collision-induced dissociation tandem MS. This study presents an approach for identifying the sn-1 and sn-2 positions in phosphatidylcholine using phosphatidylcholine and nitrate adduct ions, accurately determining phosphatidylcholine molecular configurations via the Paternò-Büchi reaction. With all the advantages above, DCAI-nano-ESI holds significant promise for future analytical and bioanalytical applications.


Subject(s)
Nanotechnology , Spectrometry, Mass, Electrospray Ionization , Spectrometry, Mass, Electrospray Ionization/methods , Cytochromes c/chemistry , Cytochromes c/analysis , Bradykinin/chemistry , Bradykinin/analysis , Angiotensin II/chemistry , Angiotensin II/analysis , Phosphatidylcholines/chemistry , Phosphatidylcholines/analysis , Glycine max/chemistry
19.
Hypertension ; 81(6): 1308-1319, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38563153

ABSTRACT

BACKGROUND: Abnormalities of resistance arteries may play essential roles in the pathophysiology of aging and hypertension. Deficiency of the vascular extracellular matrix protein MFAP4 (microfibrillar-associated protein 4) has previously been observed as protective against aberrant arterial remodeling. We hypothesized that MFAP4-deficiency would reduce age- and hypertension-dependent arterial changes in extracellular matrix composition and stiffening. METHODS: Mesenteric arteries were isolated from old (20-23 months) littermate Mfap4+/+ and Mfap4-/- mice, and 2-photon excitation microscopy imaging was used to quantify elastin and collagen volumes and dimensions in the vascular wall. Ten-week-old littermate Mfap4+/+ and Mfap4-/- mice were subjected to 20 days of continuous Ang II (angiotensin II) infusion and hypertension was monitored using invasive blood pressure measurements. Arterial stiffness, responses to vascular constrictors, and myogenic tone were monitored using wire- or pressure-myography. Collagen contents were assessed by Western blotting. RESULTS: MFAP4-deficiency significantly increased collagen volume and elastin fragmentation in aged mesenteric arteries without affecting arterial stiffness. MFAP4-deficient mice exhibited reduced diastolic pressure in Ang II-induced hypertension. There was no significant effect of MFAP4-deficiency on mesenteric artery structural remodeling or myogenic tone, although collagen content in mesenteric arteries was tendentially increased in hypertensive Mfap4+/+ mice relative to Mfap4-/- mice. Increased efficacy of vasoconstrictors (phenylephrine, thromboxane) and reduced stiffness were observed in Ang II-treated Mfap4-/- mouse mesenteric arteries in ex vivo myography recordings. CONCLUSIONS: MFAP4-deficiency reduces the elastin/collagen ratio in the aging resistance artery without affecting arterial stiffness. In contrast, MFAP4-deficiency reduces the stiffness of resistance arteries and ameliorates Ang II-induced hypertension.


Subject(s)
Aging , Angiotensin II , Hypertension , Mesenteric Arteries , Vascular Resistance , Vascular Stiffness , Animals , Hypertension/physiopathology , Hypertension/metabolism , Hypertension/genetics , Mice , Mesenteric Arteries/physiopathology , Mesenteric Arteries/drug effects , Mesenteric Arteries/metabolism , Vascular Stiffness/physiology , Vascular Stiffness/drug effects , Vascular Resistance/physiology , Aging/physiology , Angiotensin II/pharmacology , Elastin/metabolism , Blood Pressure/physiology , Extracellular Matrix Proteins/metabolism , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/deficiency , Mice, Knockout , Disease Models, Animal , Male , Collagen/metabolism
20.
Biochem Biophys Res Commun ; 715: 149997, 2024 Jun 30.
Article in English | MEDLINE | ID: mdl-38678782

ABSTRACT

The immune system is involved in hypertension development with different immune cells reported to have either pro or anti-hypertensive effects. In hypertension, immune cells have been thought to infiltrate blood pressure-regulating organs, resulting in either elevation or reduction of blood pressure. There is controversy over whether macrophages play a detrimental or beneficial role in the development of hypertension, and the few existing studies have yielded conflicting results. This study aimed to determine the effects of angiotensin II (Ang II) salt-induced hypertension on renal immune cells and to determine whether renal macrophages are involved in the induction of hypertension. Hypertension was induced by administration of Ang II and saline for two weeks. The effects of hypertension on kidney immune cells were assessed using flow cytometry. Macrophage infiltration in the kidney was assessed by immunohistochemistry and kidney fibrosis was assessed using trichrome stain and kidney real time-qPCR. Liposome encapsulated clodronate was used to deplete macrophages in C57BL/6J mice and investigate the direct role of macrophages in hypertension induction. Ang II saline mice group developed hypertension, had increased renal macrophages, and had increased expression of Acta2 and Col1a1 and kidney fibrotic areas. Macrophage depletion blunted hypertension development and reduced the expression of Acta2 and Col1a1 in the kidney and kidney fibrotic areas in Ang II saline group. The results of this study demonstrate that macrophages infiltrate the kidneys and increase kidney fibrosis in Ang II salt-induced hypertension, and depletion of macrophages suppresses the development of hypertension and decreases kidney fibrosis. This indicates that macrophages play a direct role in hypertension development. Hence macrophages have a potential to be considered as therapeutic target in hypertension management.


Subject(s)
Angiotensin II , Disease Models, Animal , Fibrosis , Hypertension , Kidney , Macrophages , Mice, Inbred C57BL , Animals , Angiotensin II/pharmacology , Macrophages/metabolism , Macrophages/pathology , Macrophages/drug effects , Hypertension/chemically induced , Hypertension/pathology , Hypertension/metabolism , Kidney/pathology , Kidney/metabolism , Kidney/drug effects , Mice , Male , Sodium Chloride, Dietary/adverse effects , Kidney Diseases/chemically induced , Kidney Diseases/pathology , Kidney Diseases/metabolism , Kidney Diseases/etiology , Blood Pressure/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...