Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
Int J Mol Sci ; 25(1)2024 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-38203807

RESUMEN

Increased body weight (BW) induces inappropriate renin-angiotensin system (RAS) activation. The activation of the intrarenal RAS is associated with increased urinary angiotensinogen (uAGT), blood pressure (BP), and kidney damage. Here, we examined uAGT excretion levels in young non-diabetic human subjects with overweight (OW) and non-diabetic mice with high-fat diet (HFD)-induced OW. Human subjects (women and men; 20-28 years old) included two groups: (a) overweight (OW, n = 17, BMI ≥ 25); and (b) controls (normal weight (NW; n = 26, BMI ≤ 25). In these subjects, we measured BP, albuminuria, and protein levels of uAGT by ELISA adjusted by urinary creatinine (expressed by uAGT/uCrea). Mice (female and male C57BL/6J mice, 8 ± 2 weeks of age) also included two groups: HFD or normal fat diet (NFD) fed for 8 weeks. We measured BW, fasting blood glucose (FBG), BP by telemetry, albuminuria, and uAGT by ELISA. In humans: (i) no significant changes were observed in BP, albuminuria, and FBG when comparing NW and OW subjects; (ii) multivariate logistic regression analysis of independent predictors related to uAGT/uCrea levels demonstrated a strong association between uAGT and overweight; (iii) urinary reactive oxygen species (ROS) were augmented in men and women with OW; (iv) the uAGT/uCrea ratio was higher in men with OW. However, the uAGT/uCrea values were lower in women even with OW. In mice: (i) males fed an HFD for 8 weeks became OW while females did not; (ii) no changes were observed either in FBG, BP, or albuminuria; (iii) kidney ROS were augmented in OW male mice after 28 weeks but not in females; (iv) OW male mice showed augmented excretion of uAGT but this was undetectable in females fed either NFD or HFD. In humans and mice who are OW, the urinary excretion of AGT differs between males and females and overcomes overt albuminuria.


Asunto(s)
Angiotensinógeno , Sobrepeso , Sistema Renina-Angiotensina , Caracteres Sexuales , Adulto , Animales , Femenino , Humanos , Masculino , Ratones , Adulto Joven , Albuminuria , Angiotensinógeno/orina , Ratones Endogámicos C57BL , Especies Reactivas de Oxígeno
2.
Antioxidants (Basel) ; 12(6)2023 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-37372017

RESUMEN

OBJECTIVES: Homozygous familial hypercholesteremia (HoFH) is a rare, life-threatening metabolic disease, mainly caused by a mutation in the LDL receptor. If untreated, HoFH causes premature death from acute coronary syndrome. Lomitapide is approved by the FDA as a therapy to lower lipid levels in adult patients with HoFH. Nevertheless, the beneficial effect of lomitapide in HoFH models remains to be defined. In this study, we investigated the effect of lomitapide on cardiovascular function using LDL receptor-knockout mice (LDLr-/-). METHODS: Six-week-old LDLr-/- mice were fed a standard diet (SD) or a high-fat diet (HFD) for 12 weeks. Lomitapide (1 mg/Kg/Day) was given by oral gavage for the last 2 weeks in the HFD group. Body weight and composition, lipid profile, blood glucose, and atherosclerotic plaques were measured. Vascular reactivity and markers for endothelial function were determined in conductance arteries (thoracic aorta) and resistance arteries (mesenteric resistance arteries (MRA)). Cytokine levels were measured by using the Mesoscale discovery V-Plex assays. RESULTS: Body weight (47.5 ± 1.5 vs. 40.3 ± 1.8 g), % of fat mass (41.6 ± 1.9% vs. 31.8 ± 1.7%), blood glucose (215.5 ± 21.9 vs. 142.3 ± 7.7 mg/dL), and lipid levels (cholesterol: 600.9 ± 23.6 vs. 451.7 ± 33.4 mg/dL; LDL/VLDL: 250.6 ± 28.9 vs. 161.1 ± 12.24 mg/dL; TG: 299.5 ± 24.1 vs. 194.1 ± 28.1 mg/dL) were significantly decreased, and the % of lean mass (56.5 ± 1.8% vs. 65.2 ± 2.1%) was significantly increased in the HFD group after lomitapide treatment. The atherosclerotic plaque area also decreased in the thoracic aorta (7.9 ± 0.5% vs. 5.7 ± 0.1%). After treatment with lomitapide, the endothelium function of the thoracic aorta (47.7 ± 6.3% vs. 80.7 ± 3.1%) and mesenteric resistance artery (66.4 ± 4.3% vs. 79.5 ± 4.6%) was improved in the group of LDLr-/- mice on HFD. This was correlated with diminished vascular endoplasmic (ER) reticulum stress, oxidative stress, and inflammation. CONCLUSIONS: Treatment with lomitapide improves cardiovascular function and lipid profile and reduces body weight and inflammatory markers in LDLr-/- mice on HFD.

3.
Environ Toxicol Pharmacol ; 100: 104160, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37236494

RESUMEN

This work studied the distribution, reactivity, and biological effects of pentavalent or trivalent antimony (Sb(V), Sb(III)) and N-methylglucamine antimonate (NMG-Sb(V)) in Wistar Rats. The expression of fibrosis genes such as α - SMA, PAI-1, and CTGF were determined in Liver, and Kidney tissues. Wistar rats were treated with different concentrations of Sb(V), Sb(III), As(V) and As(III), and MA via intra-peritoneal injections. The results indicated a noteworthy elevation in mRNA levels of plasminogen activator 1 (PAI-1) in the kidneys of rats that were injected. The main accumulation site for Sb(V) was observed to be the liver, from which it is primarily excreted in its reduced form (Sb(III)) through the urine. The generation of Sb(III) in the kidneys has been found to induce damage through the expression of α-SMA and CTGF, and also lead to a higher creatinine clearance compared to As(III).


Asunto(s)
Antimonio , Inhibidor 1 de Activador Plasminogénico , Ratas , Animales , Antimonio/toxicidad , Antimonio/metabolismo , Ratas Wistar , Antimoniato de Meglumina
4.
Antioxidants (Basel) ; 12(3)2023 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-36978977

RESUMEN

Chronic diabetes mellitus (DM) can lead to kidney damage associated with increased reactive oxygen species (ROS), proteinuria, and tubular damage. Altered protein expression levels of transforming growth factor-beta 1 (TGF-ß1), fibronectin, and renal NADPH oxidase (NOX-4) are associated with the profibrotic phenotype in renal tubular cells. NOX-4 is one of the primary sources of ROS in the diabetic kidney and responsible for the induction of profibrotic factors in collecting duct (CD) cells. The renal medulla is predominantly composed of CDs; in DM, these CD cells are exposed to high glucose (HG) load. Currently there is no published literature describing the expression of these markers in the renal medulla in male and female mice during the early phase of DM, or the role of NOX-4-induced ROS. Our aim was to evaluate changes in transcripts and protein abundances of TGF-ß1, fibronectin, and NOX-4 along with ROS levels in renal medullary tissues from male and female mice during a short period of streptozotocin (STZ)-induced type 1 DM and the effect of HG in cultured CD cells. CF-1 mice were injected with or without a single dose of STZ (200 mg/kg) and euthanized at day 6. STZ females showed higher expression of fibronectin and TGF-ß1 when compared to control mice of either gender. Interestingly, STZ female mice showed a >30-fold increase on mRNA levels and a 3-fold increase in protein levels of kidney medullary NOX-4. Both male and female STZ mice showed increased intrarenal ROS. In primary cultures of inner medullary CD cells exposed to HG over 48 h, the expression of TGF-ß1, fibronectin, and NOX-4 were augmented. M-1 CD cells exposed to HG showed increased ROS, fibronectin, and TGF-ß1; this effect was prevented by NOX-4 inhibition. Our data suggest that at as early as 6 days of STZ-induced DM, the expression of profibrotic markers TGF-ß1 and fibronectin increases in renal medullary CD cells. Antioxidants mechanisms in male and female in renal medullary tissues seems to be differentially regulated by the actions of NOX-4.

5.
Antioxidants (Basel) ; 12(1)2023 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-36671022

RESUMEN

OBJECTIVES: Short-chain fatty acids (SCFAs), the main metabolites released from the gut microbiota, are altered during hypertension and obesity. SCFAs play a beneficial role in the cardiovascular system. However, the effect of SCFAs on cerebrovascular endothelial cells is yet to be uncovered. In this study, we use brain endothelial cells to investigate the in vitro effect of SCFAs on heme oxygenase 2 (HO-2) and mitochondrial function after angiotensin II (Ang-II) treatment. METHODS: Brain human microvascular endothelial cells were treated with Ang-II (500 nM for 24 h) in the presence and absence of an SCFAs cocktail (1 µM; acetate, propionate, and butyrate) and/or HO-2 inhibitor (SnPP 5 µM). At the end of the treatment, HO-2, endothelial markers (p-eNOS and NO production), inflammatory markers (TNFα, NFκB-p50, and -p65), calcium homeostasis, mitochondrial membrane potential, mitochondrial ROS and H2O2, and mitochondrial respiration were determined in all groups of treated cells. KEY RESULTS: Our data showed that SCFAs rescued HO-2 after Ang-II treatment. Additionally, SCFAs rescued Ang-II-induced eNOS reduction and mitochondrial membrane potential impairment and mitochondrial respiration damage. On the other hand, SCFAs reduced Ang-II-induced inflammation, calcium dysregulation, mitochondrial ROS, and H2O2. All of the beneficial effects of SCFAs on endothelial cells and mitochondrial function occurred through HO-2. CONCLUSIONS: SCFAs treatment restored endothelial cells and mitochondrial function following Ang-II-induced oxidative stress. SCFAs exert these beneficial effects by acting on HO-2. Our results are opening the door for more studies to investigate the effect the of SCFAs/HO-2 axis on hypertension and obesity-induced cerebrovascular diseases.

6.
J Med Primatol ; 52(2): 131-134, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36377612

RESUMEN

Increases of soluble urokinase plasminogen activator receptor (suPAR) were measured in both urine and plasma of a Chlorocebus aethiops (African green monkey; AGM) mucosal infected with SARS-CoV-2. The data indicate that elevated suPAR may be associated with renal dysfunction and pathology in the context of COVID-19.


Asunto(s)
COVID-19 , Enfermedades Renales , Animales , Chlorocebus aethiops , COVID-19/complicaciones , Receptores del Activador de Plasminógeno Tipo Uroquinasa , SARS-CoV-2 , Biomarcadores
7.
Curr Hypertens Rev ; 18(2): 91-100, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35170417

RESUMEN

The production of renin by the principal cells of the collecting duct has widened our understanding of the regulation of intrarenal angiotensin II (Ang II) generation and blood pressure. In the collecting duct, Ang II increases the synthesis and secretion of renin by mechanisms involving the activation of Ang II type 1 receptor (AT1R) via stimulation of the PKCα, Ca2+, and cAMP/PKA/CREB pathways. Additionally, paracrine mediators, including vasopressin (AVP), prostaglandins, bradykinin (BK), and atrial natriuretic peptide (ANP), regulate renin in principal cells. During Ang II-dependent hypertension, despite plasma renin activity suppression, renin and prorenin receptor (RPR) are upregulated in the collecting duct and promote de novo formation of intratubular Ang II. Furthermore, activation of PRR by its natural agonists, prorenin and renin, may contribute to the stimulation of profibrotic factors independent of Ang II. Thus, the interactions of RAS components with paracrine hormones within the collecting duct enable tubular compartmentalization of the RAS to orchestrate complex mechanisms that increase intrarenal Ang II, Na+ reabsorption, and blood pressure.


Asunto(s)
Hipertensión , Renina , Humanos , Angiotensina II , Presión Sanguínea , Receptor de Prorenina , Receptores de Superficie Celular/metabolismo , Sistema Renina-Angiotensina
8.
Cureus ; 13(9): e18194, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34589374

RESUMEN

Anticancer drugs play an important role in reducing mortality rates and increasing life expectancy in cancer patients. Treatments include monotherapy and/or a combination of radiation therapy, chemotherapy, hormone therapy, or immunotherapy. Despite great advances in drug development, some of these treatments have been shown to induce cardiotoxicity directly affecting heart function and structure, as well as accelerating the development of cardiovascular disease. Such side effects restrict treatment options and can negatively affect disease management. Consequently, when managing cancer patients, it is vital to understand the mechanisms causing cardiotoxicity to better monitor heart function, develop preventative measures against cardiotoxicity, and treat heart failure when it occurs in this patient population. This review discusses the role and mechanism of major chemotherapy agents with principal cardiovascular complications in cancer patients.

9.
Sci Rep ; 11(1): 13815, 2021 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-34226610

RESUMEN

Growing evidence indicates that prorenin receptor (PRR) is upregulated in collecting duct (CD) of diabetic kidney. Prorenin is secreted by the principal CD cells, and is the natural ligand of the PRR. PRR activation stimulates fibrotic factors, including fibronectin, collagen, and transforming growth factor-ß (TGF-ß) contributing to tubular fibrosis. However, whether high glucose (HG) contributes to this effect is unknown. We tested the hypothesis that HG increases the abundance of PRR at the plasma membrane of the CD cells, thus contributing to the stimulation of downstream fibrotic factors, including TGF-ß, collagen I, and fibronectin. We used streptozotocin (STZ) male Sprague-Dawley rats to induce hyperglycemia for 7 days. At the end of the study, STZ-induced rats showed increased prorenin, renin, and angiotensin (Ang) II in the renal inner medulla and urine, along with augmented downstream fibrotic factors TGF-ß, collagen I, and fibronectin. STZ rats showed upregulation of PRR in the renal medulla and preferential distribution of PRR on the apical aspect of the CD cells. Cultured CD M-1 cells treated with HG (25 mM for 1 h) showed increased PRR in plasma membrane fractions compared to cells treated with normal glucose (5 mM). Increased apical PRR was accompanied by upregulation of TGF-ß, collagen I, and fibronectin, while PRR knockdown prevented these effects. Fluorescence resonance energy transfer experiments in M-1 cells demonstrated augmented prorenin activity during HG conditions. The data indicate HG stimulates profibrotic factors by inducing PRR translocation to the plasma membrane in CD cells, which in perspective, might be a novel mechanism underlying the development of tubulointerstitial fibrosis in diabetes mellitus.


Asunto(s)
Diabetes Mellitus Experimental/genética , Nefropatías Diabéticas/genética , Glucosa/metabolismo , Túbulos Renales Colectores/metabolismo , Receptores de Superficie Celular/genética , Animales , Colágeno/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/patología , Modelos Animales de Enfermedad , Fibronectinas/genética , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Hiperglucemia/genética , Hiperglucemia/metabolismo , Hiperglucemia/patología , Túbulos Renales Colectores/patología , Ratas , Factor de Crecimiento Transformador beta/genética , Receptor de Prorenina
10.
Front Cardiovasc Med ; 8: 644797, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34179130

RESUMEN

Diabetes mellitus (DM) causes high glucose (HG) levels in the plasma and urine. The (pro)renin receptor (PRR) is a key regulator of renal Na+ handling. PRR is expressed in intercalated (IC) cells of the collecting duct (CD) and binds renin to promote angiotensin (Ang) II formation, thereby contributing to Na+ reabsorption. In DM, the Kreb's cycle is in a state of suppression in most tissues. However, in the CD, expression of glucose transporters is augmented, boosting the Kreb's cycle and consequently causing α-ketoglutarate (αKG) accumulation. The αKG receptor 1 (OXGR1) is a Gq-coupled receptor expressed on the apical membrane of IC cells of the CD. We hypothesize that HG causes αKG secretion and activation of OXGR1, which increases PRR expression in CD cells. This effect then promotes intratubular AngII formation and Na+ reabsorption. To test this hypothesis, streptozotocin (STZ)-induced diabetic mice were treated with or without montelukast (ML), an OXGR1 antagonist, for 6 days. STZ mice had higher urinary αKG and PRR expression along with augmented urinary AngII levels and Na+ retention. Treatment with ML prevented all these effects. Similarly, primary cultured inner medullary CD cells treated with HG showed increased PRR expression, while OXGR1 antagonist prevented this effect. αKG increases PRR expression, while treatments with ML, PKC inhibition, or intracellular Ca2+ depletion impair this effect. In silico analysis suggested that αKG binds to mouse OXGR1. These results indicate that HG conditions promote increased levels of intratubular αKG and OXGR1-dependent PRR upregulation, which impact AngII formation and Na+ reabsorption.

11.
Nat Rev Nephrol ; 17(7): 481-492, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33824491

RESUMEN

The intrarenal renin-angiotensin system is critical for the regulation of tubule sodium reabsorption, renal haemodynamics and blood pressure. The excretion of renin in urine can result from its increased filtration, the inhibition of renin reabsorption by megalin in the proximal tubule, or its secretion by the principal cells of the collecting duct. Modest increases in circulating or intrarenal angiotensin II (ANGII) stimulate the synthesis and secretion of angiotensinogen in the proximal tubule, which provides sufficient substrate for collecting duct-derived renin to form angiotensin I (ANGI). In models of ANGII-dependent hypertension, ANGII suppresses plasma renin, suggesting that urinary renin is not likely to be the result of increased filtered load. In the collecting duct, ANGII stimulates the synthesis and secretion of prorenin and renin through the activation of ANGII type 1 receptor (AT1R) expressed primarily by principal cells. The stimulation of collecting duct-derived renin is enhanced by paracrine factors including vasopressin, prostaglandin E2 and bradykinin. Furthermore, binding of prorenin and renin to the prorenin receptor in the collecting duct evokes a number of responses, including the non-proteolytic enzymatic activation of prorenin to produce ANGI from proximal tubule-derived angiotensinogen, which is then converted into ANGII by luminal angiotensin-converting enzyme; stimulation of the epithelial sodium channel (ENaC) in principal cells; and activation of intracellular pathways linked to the upregulation of cyclooxygenase 2 and profibrotic genes. These findings suggest that dysregulation of the renin-angiotensin system in the collecting duct contributes to the development of hypertension by enhancing sodium reabsorption and the progression of kidney injury.


Asunto(s)
Hipertensión/fisiopatología , Túbulos Renales Colectores/fisiopatología , Sistema Renina-Angiotensina/fisiología , Angiotensina II/metabolismo , Humanos , Receptores de Superficie Celular/fisiología , Renina/metabolismo , Receptor de Prorenina
12.
Sci Rep ; 10(1): 21154, 2020 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-33273645

RESUMEN

Obesity has been firmly established as a major risk factor for common disease states including hypertension, type 2 diabetes mellitus, and chronic kidney disease. Increased body mass index (BMI) contributes to the activation of both the systemic and intra-tubular renin angiotensin systems (RAS), which are in turn associated with increased blood pressure (BP) and kidney damage. In this cross-sectional study, 43 subjects of normal or increased body weight were examined in order to determine the correlation of BMI or body fat mass (BFM) with blood pressure, fasting blood glucose (FBG), and urinary kidney injury markers such as interleukin-18 (IL-18), connective tissue growth factor (CTGF), neutrophil gelatinase-associated lipocalin, and kidney injury molecule-1 (KIM-1). Our results showed that: (1) subjects with increased body weight showed significantly higher BP, BFM, total body water and metabolic age; (2) BMI was positively correlated to both systolic (R2 = 0.1384, P = 0.01) and diastolic BP (R2 = 0.2437, P = 0.0008); (3) BFM was positively correlated to DBP (R2 = 0.1232, P = 0.02) and partially correlated to urine protein (R2 = 0.047, P = 0.12) and FBG (R2 = 0.07, P = 0.06); (4) overweight young adults had higher urinary mRNA levels of renin, angiotensinogen, IL-18 and CTGF. These suggest that BMI directly affects BP, kidney injury markers, and the activation of the intra-tubular RAS even in normotensive young adults. Given that BMI measurements and urine analyses are non-invasive, our findings may pave the way to developing a new and simple method of screening for the risk of chronic kidney disease in adults.


Asunto(s)
Biomarcadores/orina , Riñón/lesiones , Riñón/metabolismo , Sobrepeso/genética , Sobrepeso/orina , Sistema Renina-Angiotensina/genética , Tejido Adiposo , Adiposidad , Adolescente , Angiotensinógeno/metabolismo , Glucemia/metabolismo , Presión Sanguínea , Índice de Masa Corporal , Factor de Crecimiento del Tejido Conjuntivo/genética , Factor de Crecimiento del Tejido Conjuntivo/orina , Ayuno/sangre , Femenino , Humanos , Interleucina-18/genética , Interleucina-18/orina , Riñón/fisiopatología , Modelos Lineales , Masculino , ARN Mensajero/genética , ARN Mensajero/metabolismo , Renina/metabolismo , Adulto Joven
13.
Front Physiol ; 11: 559341, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33281610

RESUMEN

In the kidney, the stimulation of renin production by the collecting duct (CD-renin) contributes to the development of hypertension. The CD is a major nephron segment for the synthesis of nitric oxide (NO), and low NO bioavailability in the renal medulla is associated with hypertension. However, it is unknown whether NO regulates renin production in the CD. To test the hypothesis that low intrarenal NO levels stimulate the production of CD-renin, we first examined renin expression in the distal nephron segments of CD-eNOS deficient mice. In these mice, specific CD-renin immunoreactivity was increased compared to wild-type littermates; however, juxtaglomerular (JG) renin was not altered. To further assess the intracellular mechanisms involved, we then treated M-1 cells with either 1 mM L-NAME (L-arginine analog), an inhibitor of NO synthase activity, or 1 mM NONOate, a NO donor. Both treatments increased intracellular renin protein levels in M-1 cells. However, only the inhibition of NOS with L-NAME stimulated renin synthesis and secretion as reflected by the increase in Ren1C transcript and renin protein levels in the extracellular media, respectively. In addition, NONOate induced a fast mobilization of cGMP and intracellular renin accumulation. These response was partially prevented by guanylyl cyclase inhibition with ODQ (1H-[1,2,4] oxadiazolo[4,3-a]quinoxalin-1]. Accumulation of intracellular renin was blocked by protein kinase G (PKG) and protein kinase C (PKC) inhibitors. Our data indicate that low NO bioavailability increases CD-renin synthesis and secretion, which may contribute to the activation of intrarenal renin angiotensin system.

14.
Cardiovasc Diabetol ; 19(1): 136, 2020 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-32907629

RESUMEN

The endothelium plays a pivotal role in maintaining vascular health. Obesity is a global epidemic that has seen dramatic increases in both adult and pediatric populations. Obesity perturbs the integrity of normal endothelium, leading to endothelial dysfunction which predisposes the patient to cardiovascular diseases. MicroRNAs (miRNAs) are short, single-stranded, non-coding RNA molecules that play important roles in a variety of cellular processes such as differentiation, proliferation, apoptosis, and stress response; their alteration contributes to the development of many pathologies including obesity. Mediators of obesity-induced endothelial dysfunction include altered endothelial nitric oxide synthase (eNOS), Sirtuin 1 (SIRT1), oxidative stress, autophagy machinery and endoplasmic reticulum (ER) stress. All of these factors have been shown to be either directly or indirectly caused by gene regulatory mechanisms of miRNAs. In this review, we aim to provide a comprehensive description of the therapeutic potential of miRNAs to treat obesity-induced endothelial dysfunction. This may lead to the identification of new targets for interventions that may prevent or delay the development of obesity-related cardiovascular disease.


Asunto(s)
Endotelio/fisiopatología , MicroARNs/genética , Obesidad/fisiopatología , Antagomirs , Autofagia/genética , Estrés del Retículo Endoplásmico/genética , Regulación de la Expresión Génica , Humanos , MicroARNs/antagonistas & inhibidores , MicroARNs/uso terapéutico , Imitación Molecular , Terapia Molecular Dirigida , Óxido Nítrico Sintasa de Tipo III/genética , Obesidad/genética , Estrés Oxidativo/genética , Tratamiento con ARN de Interferencia , Sirtuina 1/genética
15.
Adv Physiol Educ ; 44(3): 314-322, 2020 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-32568005

RESUMEN

The integrated mechanisms of heart contraction are some of the most complex processes for undergraduate biomedical students to understand. Visual models have the potential to enhance learning environments by providing visual representations of complex mechanisms. Despite their benefits, the use of visual models in undergraduate classrooms is still limited. For this study, we tested the effect of a learning sequence of activities related to the cardiac cycle using an augmented reality (AR) application for smartphones and tablets. We were interested in understanding the ability of students to draw and label figures reflecting cardiac function after experiencing the learning sequence using AR. Undergraduate students of the biomedical sciences (control n = 43, experimental n = 58) were enrolled in the course, and their drawings were evaluated using multiple levels of complexity (1 = basic to 5 = complex) through a pre-/posttest structure that included a learning sequence based on AR in the experimental group and regular lecture-based activities in the control group. The complexity of students' drawings was evaluated on the anatomical, physiological, and molecular aspects of heart contraction. We used Cohen's kappa index for interrater reliability when determining the complexity of drawings. Control and experimental groups showed no differences in baseline knowledge (preexamination quiz). The students who experienced the AR activities showed an increase in the complexity of representation levels in posttest results and also showed a significant difference in scores for the final exam in the heart physiology course. Our results indicate that using AR enhances the comprehension of anatomical and physiological concepts of the cardiac cycle for undergraduate biomedical students.


Asunto(s)
Anatomía , Realidad Aumentada , Educación de Pregrado en Medicina , Fisiología , Anatomía/educación , Comprensión , Curriculum , Evaluación Educacional , Humanos , Aprendizaje , Fisiología/educación , Reproducibilidad de los Resultados , Estudiantes
16.
Vitam Horm ; 113: 217-238, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32138949

RESUMEN

Vasopressin, also named antidiuretic hormone (ADH), arginine vasopressin (AVP) is the main hormone responsible for water maintenance in the body through the antidiuretic actions in the kidney. The posterior pituitary into the blood releases vasopressin formed in the hypothalamus. Hypothalamic osmotic neurons are responsible to initiate the cascade for AVP actions. The effects of AVP peptide includes activation of V2 receptors which stimulate the formation of cyclic AMP (cAMP) and phosphorylation of water channels aquaporin 2 (AQP2) in the collecting duct. AVP also has vasoconstrictor effects through V1a receptors in the vasculature, while V1b is found in the nervous system. V1a and b receptors increases intracellular Ca2+ while activation of V2 receptors of signaling pathways are related to cAMP-dependent phosphorylation in kidney collecting ducts acting in coordination to stimulate water and electrolyte homeostasis. AVP potentiate formation of intratubular angiotensin II (Ang II) through V2 receptors-dependent distal tubular renin formation, contributing to Na+ reabsorption. On the same way, Ang II receptors are able to potentiate the effects of V2-dependent stimulation of AQP2 abundance in the plasma membrane. The role of AVP in hypertension and renal disease has been demonstrated in pathological states with the involvement of V2 receptors in the progression of kidney damage in diabetes and also on the stimulation of intracellular pathways linked to the development of polycystic kidney.


Asunto(s)
Arginina Vasopresina/metabolismo , Hipertensión/fisiopatología , Riñón/metabolismo , Insuficiencia Renal Crónica/fisiopatología , Sistema Renina-Angiotensina/fisiología , Humanos , Hipertensión/metabolismo , Insuficiencia Renal Crónica/metabolismo
17.
Front Pharmacol ; 10: 1314, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31803050

RESUMEN

Chronic kidney disease (CKD) is characterized by renal dysfunction, which is a common feature of other major diseases, such as hypertension and diabetes. Unilateral ureteral obstruction (UUO) has been used as a model of CKD in experimental animals and consists of total obstruction of one kidney ureter. The UUO decreases renal blood flow, which promotes the synthesis of renin in the juxtaglomerular apparatus, the first step in renin-angiotensin system (RAS) cascade. RAS induces inflammation and remodeling, along with reduced renal function. However, it remains unknown whether intrarenal RAS (iRAS) is activated in early stages of CKD. Our objective was to characterize different iRAS components in the renal cortex and in the medulla in an early phase of UUO. Male C57BL/6 mice (8-12 weeks old) were subjected to UUO in the left kidney, or to sham surgery, and were euthanized after 7 days (n = 5/group). Renal function, renal inflammatory/remodeling processes, and iRAS expression were evaluated. UUO increased plasma creatinine, right renal hypertrophy (9.08 ± 0.31, P < 0.05 vs. Sham), and tubular dilatation in the left kidney cortex (42.42 ± 8.19µm, P < 0.05 vs. Sham). This correlated with the increased mRNA of IL-1ß (1.73 ± 0.14, P < 0.01 vs. Sham, a pro-inflammatory cytokine) and TGF-ß1 (1.76 ± 0.10, P < 0.001 vs. Sham, a pro-fibrotic marker). In the renal cortex of the left kidney, UUO increased the mRNA and protein levels of renin (in 35% and 28%, respectively, P < 0.05 vs. Sham). UUO decreased mRNA and protein levels for the (pro)renin receptor in the renal medulla (0.67 ± 0.036 and 0.88 ± 0.028, respectively, P < 0.05 vs. Sham). Our results suggest that modulation of iRAS components depends on renal localization and occurs in parallel with remodeling and pro-inflammatory/pro-fibrotic mechanisms.

18.
Front Pharmacol ; 10: 1212, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31680980

RESUMEN

In angiotensin II (Ang II)-dependent hypertensive rats there is an increased expression of proximal tubule angiotensinogen (AGT), collecting duct renin and angiotensin converting enzyme (ACE), which contributes to intratubular Ang II formation. Ang II acts on Ang II type 1 receptors promoting sodium retention and vasoconstriction. However concurrently, the ACE2-Ang-(1-7) axis and the expression of kallikrein and medullary prostaglandins counteract the effects of Ang II, promoting natriuresis and vasodilation. Human studies demonstrate that dietary potassium (K+) intake lowers blood pressure. In this report we evaluate the expression of AGT, ACE, medullary prorenin/renin, ACE2, kallikrein and cyclooxygenase-2 (COX-2) in Ang II-infused rats fed with high K+ diet (2%) for 14 days. Dietary K+ enhances diuresis in non-infused and in Ang II-infused rats. The rise in systolic blood pressure in Ang II-infused rats was attenuated by dietary K+. Ang II-infused rats showed increased renal protein levels of AGT, ACE and medullary prorenin and renin. This effect was attenuated in the Ang II + K+ group. Ang II infusion decreased ACE2 compared to the control group; however, K+ diet prevented this effect in the renal medulla. Furthermore, medullary COX-2 was dramatically induced by K+ diet in non-infused and in Ang II infused rats. Dietary K+ greatly increased kallikrein immunostaining in normotensive rats and in Ang II-hypertensive rats. These results indicate that a high K+ diet attenuates Ang II-dependent hypertension by preventing the induction of ACE, AGT and collecting duct renin and by enhancing medullary COX-2 and ACE2 protein expression in the kidney.

19.
Front Pharmacol ; 10: 803, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31396082

RESUMEN

The binding of prorenin to the (pro)renin receptor (PRR) triggers the activation of MAPK/ERK1/2 pathway, induction of cyclooxygenase-2 (COX-2), NOX-4-dependent production of reactive oxygen species (ROS), and the induction of transforming growth factor ß (TGF-ß) and profibrotic factors connecting tissue growth factor (CTGF) and plasminogen activator inhibitor (PAI-I) in collecting duct (CD) cells. However, the role of COX-2 and the intracellular pathways involved are not clear. We hypothesized that the PRR activation increases profibrotic factors through COX-2-mediated PGE2 activation of E prostanoid receptor 4 (EP4), upregulation of NOX-4/ROS production, and activation of Smad pathway in mouse CD cells. Recombinant prorenin increased ROS production and protein levels of CTGF, PAI-I, and TGF-ß in M-1 CD cell line. Inhibition of MAPK, NOX-4, and COX-2 prevented this effect. Inhibition of MEK, COX-2, and EP4 also prevented the upregulation of NOX-4. Because TGF-ß activates Smad pathway, we evaluate the phosphorylation of Smad2 and 3. COX-2 inhibition or EP4 antagonism significantly prevented phosphorylation of Smad 2/3. Mice that were infused with recombinant prorenin showed an induction in the expression of CTGF, PAI-I, TGF-ß, fibronectin, and collagen I in isolated collecting ducts as well as the expression of alpha smooth muscle actin (α-SMA) in renal tissues. COX-2 inhibition prevented this induction. These results indicate that the induction of TGF-ß, CTGF, PAI-I, and ROS occurs through PRR-dependent activation of MAPK and NOX-4; however, this mechanism depends on COX-2-derived PGE2 production and the activation of EP4 and Smad pathway.

20.
Obesity (Silver Spring) ; 27(7): 1050-1058, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30938942

RESUMEN

Over the past three decades, the increasing rates of obesity have led to an alarming obesity epidemic worldwide. Obesity is associated with an increased risk of cardiovascular diseases; thus, it is essential to define the molecular mechanisms by which obesity affects heart function. Individuals with obesity and overweight have shown changes in cardiac structure and function, leading to cardiomyopathy, hypertrophy, atrial fibrillation, and arrhythmia. Autophagy is a highly conserved recycling mechanism that delivers proteins and damaged organelles to lysosomes for degradation. In the hearts of patients and mouse models with obesity, this process is impaired. Furthermore, it has been shown that autophagy flux restoration in obesity models improves cardiac function. Therefore, autophagy may play an important role in mitigating the adverse effects of obesity on the heart. Throughout this review, we will discuss the benefits of autophagy on the heart in obesity and how regulating autophagy might be a therapeutic tool to reduce the risk of obesity-associated cardiovascular diseases.


Asunto(s)
Autofagia , Enfermedades Cardiovasculares/fisiopatología , Cardiopatías/fisiopatología , Obesidad/complicaciones , Humanos , Obesidad/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...