Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
1.
Curr Hypertens Rep ; 25(6): 91-106, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37052810

RESUMEN

PURPOSE OF REVIEW: To address the mechanistic pathways focusing on mitochondria dysfunction, oxidative stress, sirtuins imbalance, and other contributors in patient with metabolic syndrome and cardiovascular disease. Sodium glucose co-transporter type 2 (SGLT-2) inhibitors deeply influence these mechanisms. Recent randomized clinical trials have shown impressive results in improving cardiac function and reducing cardiovascular and renal events. These unexpected results generate the need to deepen our understanding of the molecular mechanisms able to generate these effects to help explain such significant clinical outcomes. RECENT FINDINGS: Cardiovascular disease is highly prevalent among individuals with metabolic syndrome and diabetes. Furthermore, mitochondrial dysfunction is a principal player in its development and persistence, including the consequent cardiac remodeling and events. Another central protagonist is the renin-angiotensin system; the high angiotensin II (Ang II) activity fuel oxidative stress and local inflammatory responses. Additionally, sirtuins decline plays a pivotal role in the process; they enhance oxidative stress by regulating adaptive responses to the cellular environment and interacting with Ang II in many circumstances, including cardiac and vascular remodeling, inflammation, and fibrosis. Fasting and lower mitochondrial energy generation are conditions that substantially reduce most of the mentioned cardiometabolic syndrome disarrangements. In addition, it increases sirtuins levels, and adenosine monophosphate-activated protein kinase (AMPK) signaling stimulates hypoxia-inducible factor-1ß (HIF-1 beta) and favors ketosis. All these effects favor autophagy and mitophagy, clean the cardiac cells with damaged organelles, and reduce oxidative stress and inflammatory response, giving cardiac tissue protection. In this sense, SGLT-2 inhibitors enhance the level of at least four sirtuins, some located in the mitochondria. Moreover, late evidence shows that SLGT-2 inhibitors mimic this protective process, improving mitochondria function, oxidative stress, and inflammation. Considering the previously described protection at the cardiovascular level is necessary to go deeper in the knowledge of the effects of SGLT-2 inhibitors on the mitochondria function. Various of the protective effects these drugs clearly had shown in the trials, and we briefly describe it could depend on sirtuins enhance activity, oxidative stress reduction, inflammatory process attenuation, less interstitial fibrosis, and a consequent better cardiac function. This information could encourage investigating new therapeutic strategies for metabolic syndrome, diabetes, heart and renal failure, and other diseases.


Asunto(s)
Enfermedades Cardiovasculares , Diabetes Mellitus , Hipertensión , Síndrome Metabólico , Sirtuinas , Inhibidores del Cotransportador de Sodio-Glucosa 2 , Humanos , Inhibidores del Cotransportador de Sodio-Glucosa 2/uso terapéutico , Síndrome Metabólico/tratamiento farmacológico , Sirtuinas/metabolismo , Sirtuinas/farmacología , Enfermedades Cardiovasculares/tratamiento farmacológico , Remodelación Ventricular , Hipertensión/tratamiento farmacológico , Estrés Oxidativo/fisiología , Angiotensina II/metabolismo , Fibrosis
2.
Curr Hypertens Rep ; 24(11): 547-562, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35796869

RESUMEN

PURPOSE OF REVIEW: This review summarizes the involvement of inflammaging in vascular damage with focus on the epigenetic mechanisms by which inflammaging-induced hypertension is triggered. RECENT FINDINGS: Inflammaging in hypertension is a complex condition associated with the production of inflammatory mediators by the immune cells, enhancement of oxidative stress, and tissue remodeling in vascular smooth muscle cells and endothelial cells. Cellular processes are numerous, including inflammasome assembly and cell senescence which may involve mitochondrial dysfunction, autophagy, DNA damage response, dysbiosis, and many others. More recently, a series of noncoding RNAs, mainly microRNAs, have been described as possessing epigenetic actions on the regulation of inflammasome-related hypertension, emerging as a promising therapeutic strategy. Although there are a variety of pharmacological agents that effectively regulate inflammaging-related hypertension, a deeper understanding of the epigenetic events behind the control of vessel deterioration is needed for the treatment or even to prevent the disease onset.


Asunto(s)
Hipertensión , MicroARNs , Envejecimiento , Células Endoteliales , Epigénesis Genética , Humanos , Hipertensión/genética , Inflamasomas , Inflamación , Mediadores de Inflamación , MicroARNs/genética
3.
Am J Physiol Lung Cell Mol Physiol ; 319(6): L941-L948, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32996774

RESUMEN

The COVID-19 pandemic has reached most of the countries worldwide causing death, which often results from an inflammatory storm associated with severe acute respiratory syndrome (SARS). This has prompted researchers to seek specific novel and definitive treatments urgently. In this context, it is interesting to evaluate the preventive and therapeutic effects of existing pharmacological agents that could be useful. In this regard, vitamin D supplementation, particularly in individuals likely to be deficient, may be a promising option. Vitamin D is a hormone that modulates many of the same inflammatory and oxidative signaling pathways triggered during COVID-19. For example, vitamin D suppresses the actions of the renin-angiotensin system, which has a determining role in the pathophysiology of the inflammatory response related to COVID-19. This paper analyzes the evidence that vitamin D supplementation might be a valuable preventive/therapeutic measure in groups at risk for or infected with COVID-19. It also discusses how clinical studies could be best designed to evaluate the possible advantages of vitamin D supplementation for the benefit of public health during the pandemic.


Asunto(s)
COVID-19/prevención & control , Suplementos Dietéticos , Pandemias/prevención & control , Síndrome de Dificultad Respiratoria/prevención & control , SARS-CoV-2/metabolismo , Vitamina D/uso terapéutico , Animales , COVID-19/epidemiología , COVID-19/metabolismo , COVID-19/patología , Humanos , Estrés Oxidativo/efectos de los fármacos , Sistema Renina-Angiotensina/efectos de los fármacos , Síndrome de Dificultad Respiratoria/epidemiología , Síndrome de Dificultad Respiratoria/metabolismo , Síndrome de Dificultad Respiratoria/patología
4.
Curr Hypertens Rep ; 22(1): 6, 2020 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-31925548

RESUMEN

PURPOSE OF REVIEW: Several studies have attributed garlic's beneficial properties to its high content of organosulfur compounds (OSCs). Here, we summarized recent studies published and some own findings regarding OSCs and its effects on cardiovascular disease, inflammation, and obesity. RECENT FINDING: The analysis of the multiple actions produced by OSCs suggests that many of its bioactivities interfere against inflammation, oxidative stress, obesogenic effects, and mitochondrial dysfunction. Accumulating evidence from in vitro, animal, and human studies reinforce the notion that OSCs modify signaling pathways that trigger chronic diseases, and to highlight, actions over these pathways are related to the treatment of disorders addressed in this review. Garlic's bioactive OSCs behave like a nutraceutical panacea because they cover a broad spectrum of applications with promising impact for the prevention and treatment of prevalent chronic pathologies associated with low-grade inflammation.


Asunto(s)
Enfermedades Cardiovasculares , Ajo , Hipertensión , Obesidad , Extractos Vegetales , Animales , Enfermedades Cardiovasculares/terapia , Humanos , Hipertensión/terapia , Inflamación , Obesidad/terapia , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico
5.
Stress ; 22(2): 169-181, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30547701

RESUMEN

Although a large number of available treatments and strategies, the prevalence of cardiovascular diseases continues to grow worldwide. Emerging evidence supports the notion of counteracting stress as a critical component of a comprehensive therapeutic strategy for cardiovascular disease. Indeed, an unhealthy lifestyle is a burden to biological variables such as plasma glucose, lipid profile, and blood pressure control. Recent findings identify allostatic load as a new paradigm for an integrated understanding of the importance of psychosocial stress and its impact on the development and maintenance of cardiovascular disease. Allostasis complement homeostasis and integrates behavioral and physiological mechanisms by which genes, early experiences, environment, lifestyle, diet, sleep, and physical exercise can modulate and adapt biological responses at the cellular level. For example, variability is a physiological characteristic of blood pressure necessary for survival and the allostatic load in hypertension can contribute to its related cardiovascular morbidity and mortality. Therefore, the current review will focus on the mechanisms that link hypertension to allostatic load, which includes psychosocial stress, inflammation, and mitochondrial dysfunction. We will describe and discuss new insights on neuroendocrine-immune effects linked to allostatic load and its impact on the cellular and molecular responses; the links between allostatic load, inflammation, and endothelial dysfunction; the epidemiological evidence supporting the pathophysiological origins of hypertension; and the biological embedding of allostatic load and hypertension with an emphasis on mitochondrial dysfunction.


Asunto(s)
Alostasis/fisiología , Hipertensión/fisiopatología , Inflamación/fisiopatología , Mitocondrias/metabolismo , Estrés Psicológico/fisiopatología , Presión Sanguínea/fisiología , Homeostasis/fisiología , Humanos
6.
Curr Hypertens Rep ; 20(5): 45, 2018 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-29744660

RESUMEN

PURPOSE OF REVIEW: Here, we review the known relations between hypertension and obesity to inflammation and postulate the endogenous protective effect of melatonin and its potential as a therapeutic agent. We will describe the multiple effects of melatonin on blood pressure, adiposity, body weight, and focus on mitochondrial-related anti-inflammatory and antioxidant protective effects. RECENT FINDINGS: Hypertension and obesity are usually associated with systemic and tissular inflammation. The progressive affection of target-organs involves multiple mediators of inflammation, most of them redundant, which make anti-inflammatory strategies ineffective. Melatonin reduces blood pressure, body weight, and inflammation. The mechanisms of action of this ancient molecule of protection involve multiple levels of action, from subcellular to intercellular. Mitochondria is a key inflammatory element in vascular and adipose tissue and a potential pharmacological target. Melatonin protects against mitochondrial dysfunction. Melatonin reduces blood pressure and adipose tissue dysfunction by multiple anti-inflammatory/antioxidant actions and provides potent protection against mitochondria-mediated injury in hypertension and obesity. This inexpensive and multitarget molecule has great therapeutic potential against both epidemic diseases.


Asunto(s)
Antiinflamatorios/uso terapéutico , Hipertensión/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Melatonina/uso terapéutico , Obesidad/tratamiento farmacológico , Animales , Antioxidantes/uso terapéutico , Presión Sanguínea/efectos de los fármacos , Humanos , Hipertensión/complicaciones , Hipertensión/fisiopatología , Inflamación/metabolismo , Inflamación/fisiopatología , Mitocondrias/metabolismo , Obesidad/complicaciones , Obesidad/fisiopatología , Estrés Oxidativo/efectos de los fármacos
7.
J Pineal Res ; 65(4): e12513, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29851143

RESUMEN

Lethal ventricular arrhythmias increase in patients with chronic kidney disease that suffer an acute coronary event. Chronic kidney disease induces myocardial remodeling, oxidative stress, and arrhythmogenesis. A manifestation of the relationship between kidney and heart is the concomitant reduction in vitamin D receptor (VDR) and the increase in angiotensin II receptor type 1 (AT1 ). Melatonin has renal and cardiac protective actions. One potential mechanism is the increase in the heat shock protein 70 (Hsp70)-an antioxidant factor. We aim to determine the mechanisms involved in melatonin (Mel) prevention of kidney damage and arrhythmogenic heart remodeling. Unilateral ureteral-obstruction (UUO) and sham-operated rats were treated with either melatonin (4 mg/kg/day) or vehicle for 15 days. Hearts and kidneys from obstructed rats showed a reduction in VDR and Hsp70. Associated with AT1 up-regulation in the kidneys and the heart of UUO rats also increased oxidative stress, fibrosis, apoptosis, mitochondrial edema, and dilated crests. Melatonin prevented these changes and ventricular fibrillation during reperfusion. The action potential lengthened and hyperpolarized in melatonin-treated rats throughout the experiment. We conclude that melatonin prevents renal damage and arrhythmogenic myocardial remodeling during unilateral ureteral obstruction due to a decrease in oxidative stress/fibrosis/apoptosis associated with AT1 reduction and Hsp70-VDR increase.


Asunto(s)
Proteínas HSP70 de Choque Térmico/metabolismo , Melatonina/uso terapéutico , Receptor de Angiotensina Tipo 1/metabolismo , Receptores de Calcitriol/metabolismo , Taquicardia Ventricular/tratamiento farmacológico , Taquicardia Ventricular/metabolismo , Actinas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Fibrosis/metabolismo , Proteínas HSP70 de Choque Térmico/genética , Etiquetado Corte-Fin in Situ , Técnicas In Vitro , Riñón/metabolismo , Masculino , Microscopía Electrónica , Microscopía Fluorescente , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Miocardio/metabolismo , NADPH Oxidasas/metabolismo , Ratas , Ratas Endogámicas WKY , Receptor de Angiotensina Tipo 1/genética , Receptores de Calcitriol/genética
8.
Curr Hypertens Rep ; 19(2): 11, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28233236

RESUMEN

Mitochondria are essential for the maintenance of normal physiological function of tissue cells. Mitochondria are subject to dynamic processes in order to establish a control system related to survival or cell death and adaptation to changes in the metabolic environment of cells. Mitochondrial dynamics includes fusion and fission processes, biogenesis, and mitophagy. Modifications of mitochondrial dynamics in organs involved in energy metabolism such as the pancreas, liver, skeletal muscle, and white adipose tissue could be of relevance for the development of insulin resistance, obesity, and type 2 diabetes. Mitochondrial dynamics and the factors involved in its regulation are also critical for neuronal development, survival, and function. Modifications in mitochondrial dynamics in either agouti-related peptide (AgRP) or pro-opiomelanocortin (POMC), circuits which regulates feeding behavior, are related to changes of food intake, energy balance, and obesity development. Activation of the sympathetic nervous system has been considered as a crucial point in the pathogenesis of hypertension among obese individuals and it also plays a key role in cardiac remodeling. Hypertension-related cardiac hypertrophy is associated with changes in metabolic substrate utilization, dysfunction of the electron transport chain, and ATP synthesis. Alterations in both mitochondrial dynamics and ROS production have been associated with endothelial dysfunction, development of hypertension, and cardiac hypertrophy. Finally, it might be postulated that alterations of mitochondrial dynamics in white adipose tissue could contribute to the development and maintenance of hypertension in obesity situations through leptin overproduction. Leptin, together with insulin, will induce activation of sympathetic nervous system with consequences at renal, vascular, and cardiac levels, driving to sodium retention, hypertension, and left ventricular hypertrophy. Moreover, both leptin and insulin will induce mitochondrial alterations into arcuate nucleus leading to signals driving to increased food intake and reduced energy expenditure. This, in turn would perpetuate white adipose tissue excess and its well-known metabolic and cardiovascular consequences.


Asunto(s)
Hipertensión/fisiopatología , Mitocondrias/metabolismo , Enfermedades Mitocondriales/complicaciones , Obesidad/fisiopatología , Animales , Metabolismo Energético , Humanos , Hipertensión/complicaciones , Resistencia a la Insulina , Obesidad/complicaciones , Sistema Nervioso Simpático/metabolismo
9.
Curr Hypertens Rep ; 18(1): 1, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26694820

RESUMEN

Insulin resistance (IR) is present in pathologies such as diabetes, obesity, metabolic syndrome, impaired glucose tolerance, hypertension, inflammation, cardiac disease, and dyslipidemias. Population studies show that IR is multifactorial and has genetic components, such as defects in the insulin-signaling pathway (as serine phosphorylation on insulin substrate or decreased activation of signaling molecules) and RAS/MAPK-dependent pathways. IR is connected to mitochondrial dysfunction, overproduction of oxidants, accumulation of fat, and an over-activation of the renin-angiotensin system linked to the NADPH oxidase activity. In addition, nitric oxide (NO), synthesized by nitric oxide synthases (endothelial and inducible), is also associated with IR when both impaired release and reduced bioavailability of all which lead to inflammation and hypertension. However, increased NO may promote vasculoprotection. Moreover, reduced NO release induces heat shock protein 70 kDa (HSP70) expression in IR and diabetes, mediating beneficial effects against oxidative stress injury, inflammation and apoptosis. HSP70 may be used as biomarker of the chronicity of diabetes. Hsp72 (inducible protein) is linked to vascular complications with a high-fat diet by blocking inflammation signaling (cytoprotective and anti-cytotoxicity intracellular role). Elucidating the IR signaling pathways and the roles of NO and HSPs is relevant to the application of new treatments, such as heat shock and thermal therapy, nitrosylated drugs, chemical chaperones or exercise training.


Asunto(s)
Proteínas de Choque Térmico/metabolismo , Resistencia a la Insulina , Óxido Nítrico/metabolismo , Animales , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Estrés Oxidativo , Vitamina D/metabolismo
10.
BMC Nephrol ; 17: 34, 2016 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-27009470

RESUMEN

BACKGROUND: Hypertension is a public health problem with mostly unknown causes, and where strong hereditary genetic alterations have not been fully elucidated. However, the use of experimental models has provided valuable information. Recent evidences suggest that alterations in key nephrogenic factors, such as Wilms' tumor 1 transcription factor (WT-1), could contribute to the development of hypertension. The aim of this paper is to evaluate the expression of WT-1 and related genes in the nephrogenic process in connection with the development of hypertension as well as the corresponding anatomical and functional correlation. METHODS: Male spontaneously hypertensive and control rats were evaluated weekly from birth until week 8 of life. Their blood pressure was taken weekly using the tail-cuff blood pressure system. Weekly, 5 rats per group were sacrificed with a lethal injection of pentobarbital, and their kidneys were removed, decapsulated and weighed. The serum was collected for measuring biochemical parameters. The results were assessed using one-way analysis of variance for comparisons between groups. RESULTS: The relationship between renal weight/total body weights was established, without significantly different values. These data were compared with apoptosis, fibrosis, number and size of the glomeruli. The elevation of systolic blood pressure was significant since week 6. Biochemical values differed slightly. Histology showed a slight increase in deposits of collagen fibers since week 4. Additionally, in kidney cortices, the expression of WT-1, heat shock protein 70 (Hsp70) and vitamin D receptors (VDR) decreased since week 4. Finally, we demonstrated ultrastructural damage to mitochondria since week 4. CONCLUSIONS: Our results would suggest an unprecedented link, possibly a regulatory mechanism, between WT-1 on nephrogenic alteration processes and their relationship with hypertension. Moreover, and previous to the increase in blood pressure, we demonstrated low expressions of WT-1, VDR and Hsp70 in kidneys from neonatal SHRs. If so, this may suggest that deregulation in the expression of WT-1 and its impact on nephrogenesis induction could be crucial in understanding the development and maintenance of hypertension.


Asunto(s)
Hipertensión/metabolismo , Riñón/metabolismo , Mitocondrias/ultraestructura , Proteínas WT1/metabolismo , Animales , Animales Recién Nacidos , Apoptosis , Presión Sanguínea , Peso Corporal , Fibrosis , Proteínas HSP70 de Choque Térmico/metabolismo , Hipertensión/patología , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Riñón/patología , Corteza Renal/metabolismo , Masculino , Microscopía Confocal , Microscopía Electrónica , Microscopía Fluorescente , Tamaño de los Órganos , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Receptores de Calcitriol/metabolismo
11.
Am J Physiol Heart Circ Physiol ; 309(1): H15-44, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25934099

RESUMEN

Caloric restriction (CR), renin angiotensin system blockade (RAS-bl), and rapamycin-mediated mechanistic target of rapamycin (mTOR) inhibition increase survival and retard aging across species. Previously, we have summarized CR and RAS-bl's converging effects, and the mitochondrial function changes associated with their physiological benefits. mTOR inhibition and enhanced sirtuin and KLOTHO signaling contribute to the benefits of CR in aging. mTORC1/mTORC2 complexes contribute to cell growth and metabolic regulation. Prolonged mTORC1 activation may lead to age-related disease progression; thus, rapamycin-mediated mTOR inhibition and CR may extend lifespan and retard aging through mTORC1 interference. Sirtuins by deacetylating histone and transcription-related proteins modulate signaling and survival pathways and mitochondrial functioning. CR regulates several mammalian sirtuins favoring their role in aging regulation. KLOTHO/fibroblast growth factor 23 (FGF23) contribute to control Ca(2+), phosphate, and vitamin D metabolism, and their dysregulation may participate in age-related disease. Here we review how mTOR inhibition extends lifespan, how KLOTHO functions as an aging suppressor, how sirtuins mediate longevity, how vitamin D loss may contribute to age-related disease, and how they relate to mitochondrial function. Also, we discuss how RAS-bl downregulates mTOR and upregulates KLOTHO, sirtuin, and vitamin D receptor expression, suggesting that at least some of RAS-bl benefits in aging are mediated through the modulation of mTOR, KLOTHO, and sirtuin expression and vitamin D signaling, paralleling CR actions in age retardation. Concluding, the available evidence endorses the idea that RAS-bl is among the interventions that may turn out to provide relief to the spreading issue of age-associated chronic disease.


Asunto(s)
Envejecimiento/metabolismo , Angiotensina II/metabolismo , Restricción Calórica , Longevidad , Mitocondrias/metabolismo , Sistema Renina-Angiotensina , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/metabolismo , Glucuronidasa/metabolismo , Humanos , Proteínas Klotho , Diana Mecanicista del Complejo 1 de la Rapamicina , Complejos Multiproteicos/metabolismo , Receptores de Calcitriol/metabolismo , Transducción de Señal , Sirtuinas/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Vitamina D/metabolismo
12.
Curr Hypertens Rep ; 17(1): 504, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25432896

RESUMEN

Mitochondria are the primary generators of cellular reactive oxygen species (ROS); their pathophysiological roles in hypertension and insulin resistance are but imperfectly understood. Mitochondrial dysfunction has been linked to the etiologies of many complex diseases, but many other factors, including the upregulation of the renin-angiotensin system (RAS) and vitamin D deficiency, have also been implicated in hypertension pathogenesis. Hypertension resulting from the disruption of the RAS contributes to the risk of cardiovascular disease. Likewise, experimental and clinical evidence indicate that RAS stimulation and low vitamin D levels are inversely related and represent risk factors associated with the pathogenesis of hypertension. Furthermore, RAS activation induces insulin resistance, resulting in increases in ROS levels. High levels of ROS are harmful to cells, having the potential to trigger both mitochondrial-mediated apoptosis and the degradation of the mitochondrial DNA. Diabetes risk is also associated with high levels of oxidative stress; taking vitamin D, however, may reduce that risk. The finding that mitochondria possess both a functional RAS and vitamin D receptors is the starting point for improving our understanding of the interaction of mitochondria and chronic disease states, which understanding should lead to decreases in the chronic disease burden attributable to hypertension, diabetes, or both.


Asunto(s)
Hipertensión/metabolismo , Resistencia a la Insulina , Mitocondrias/metabolismo , Estrés Oxidativo/fisiología , Humanos
13.
Kidney Blood Press Res ; 39(4): 340-52, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25300759

RESUMEN

BACKGROUND/AIMS: The search for new therapies providing cardiorenal protection in chronic kidney disease (CKD) has led to treatments that combine conventional renin-angiotensin-aldosterone-system inhibitors with other drugs that exhibit potential in disease management. METHODS: In rats made uremic by renal ablation, we examined the effects of addition of the endothelin-A receptor antagonist atrasentan to a previously examined combination of enalapril (angiotensin converting enzyme inhibitor) and paricalcitol (vitamin D receptor activator) on cardiac and renal parameters. The effects of the individual and combined drugs were examined after a 3-month treatment. RESULTS: A decrease in systolic blood pressure, serum creatinine and proteinuria, and improvement of renal histology in uremic rats were attributed to enalapril and/or paricalcitol treatment; atrasentan alone had no effect. In heart tissue, individual treatment with the drugs blunted the increase in cardiomyocyte size, and combined treatment additively decreased cardiomyocyte size to normal levels. Perivascular fibrosis was blunted in uremic control rats with atrasentan or enalapril treatment. CONCLUSIONS: We found distinct cardiac and renal effects of atrasentan. Combination treatment with atrasentan, enalapril and paricalcitol provided positive effects on cardiac remodeling in uremic rats, whereas combination treatment did not offer further protective effects on blood pressure, proteinuria or renal histology.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Conservadores de la Densidad Ósea/uso terapéutico , Enalapril/uso terapéutico , Antagonistas de los Receptores de Endotelina/uso terapéutico , Ergocalciferoles/uso terapéutico , Corazón/efectos de los fármacos , Riñón/efectos de los fármacos , Pirrolidinas/uso terapéutico , Uremia/tratamiento farmacológico , Animales , Atrasentán , Quimioterapia Combinada , Femenino , Glomeruloesclerosis Focal y Segmentaria/patología , Corazón/fisiopatología , Riñón/fisiopatología , Pruebas de Función Renal , Miocardio/patología , Nefrectomía , Nefritis/patología , Ratas , Ratas Sprague-Dawley , Análisis de Supervivencia , Uremia/fisiopatología
14.
Physiol Rep ; 12(12): e16094, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38924381

RESUMEN

The renin-angiotensin system (RAS)-a classical blood pressure regulator-largely contributes to healthy organ development and function. Besides, RAS activation promotes age-related changes and age-associated diseases, which are attenuated/abolished by RAS-blockade in several mammalian species. RAS-blockers also increase rodent lifespan. In previous work, we discussed how RAS-blockade downregulates mTOR and growth hormone/IGF-1 signaling, and stimulates AMPK activity (together with klotho, sirtuin, and vitamin D-receptor upregulation), and proposed that at least some of RAS-blockade's aging benefits are mediated through regulation of these intermediaries and their signaling to mitochondria. Here, we included RAS-blockade's impact on other aging regulatory pathways, that is, TGF-ß, NF-kB, PI3K, MAPK, PKC, Notch, and Wnt, all of which affect mitochondria. No direct evidence is available on RAS/RAS-blockade-aging regulatory pathway-mitochondria interactions. However, existing results allow to conjecture that RAS-blockers neutralize mitochondrial dysfunction by acting on the discussed pathways. The reviewed evidence led us to propose that the foundation is laid for conducting clinical trials aimed at testing whether angiotensin-converting enzyme inhibitors (ACEi) or angiotensin receptor blockers (ARB)-even at subclinical doses-offer the possibility to live longer and in better health. As ACEi and ARB are low cost and well-tolerated anti-hypertension therapies in use for over 35 years, investigating their administration to attenuate/prevent aging effects seems simple to implement.


Asunto(s)
Envejecimiento , Inhibidores de la Enzima Convertidora de Angiotensina , Sistema Renina-Angiotensina , Humanos , Sistema Renina-Angiotensina/efectos de los fármacos , Envejecimiento/efectos de los fármacos , Envejecimiento/metabolismo , Envejecimiento/fisiología , Animales , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Transducción de Señal/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/efectos de los fármacos , Antagonistas de Receptores de Angiotensina/farmacología , Antagonistas de Receptores de Angiotensina/uso terapéutico
15.
World J Exp Med ; 14(2): 91519, 2024 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-38948421

RESUMEN

Mitochondrial dysfunction is a key driver of cardiovascular disease (CVD) in metabolic syndrome and diabetes. This dysfunction promotes the production of reactive oxygen species (ROS), which cause oxidative stress and inflammation. Angiotensin II, the main mediator of the renin-angiotensin-aldosterone system, also contributes to CVD by promoting ROS production. Reduced activity of sirtuins (SIRTs), a family of proteins that regulate cellular metabolism, also worsens oxidative stress. Reduction of energy production by mitochondria is a common feature of all metabolic disorders. High SIRT levels and 5' adenosine monophosphate-activated protein kinase signaling stimulate hypoxia-inducible factor 1 beta, which promotes ketosis. Ketosis, in turn, increases autophagy and mitophagy, processes that clear cells of debris and protect against damage. Sodium-glucose cotransporter-2 inhibitors (SGLT2i), a class of drugs used to treat type 2 diabetes, have a beneficial effect on these mechanisms. Randomized clinical trials have shown that SGLT2i improves cardiac function and reduces the rate of cardiovascular and renal events. SGLT2i also increase mitochondrial efficiency, reduce oxidative stress and inflammation, and strengthen tissues. These findings suggest that SGLT2i hold great potential for the treatment of CVD. Furthermore, they are proposed as anti-aging drugs; however, rigorous research is needed to validate these preliminary findings.

16.
Curr Pharm Des ; 30(13): 969-974, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38551044

RESUMEN

In metabolic syndrome and diabetes, compromised mitochondrial function emerges as a critical driver of cardiovascular disease, fueling its development and persistence, culminating in cardiac remodeling and adverse events. In this context, angiotensin II - the main interlocutor of the renin-angiotensin-aldosterone system - promotes local and systemic oxidative inflammatory processes. To highlight, the low activity/expression of proteins called sirtuins negatively participates in these processes, allowing more significant oxidative imbalance, which impacts cellular and tissue responses, causing tissue damage, inflammation, and cardiac and vascular remodeling. The reduction in energy production of mitochondria has been widely described as a significant element in all types of metabolic disorders. Additionally, high sirtuin levels and AMPK signaling stimulate hypoxia- inducible factor 1 beta and promote ketonemia. Consequently, enhanced autophagy and mitophagy advance through cardiac cells, sweeping away debris and silencing the orchestra of oxidative stress and inflammation, ultimately protecting vulnerable tissue from damage. To highlight and of particular interest, SGLT2 inhibitors (SGLT2i) profoundly influence all these mechanisms. Randomized clinical trials have evidenced a compelling picture of SGLT2i emerging as game-changers, wielding their power to demonstrably improve cardiac function and slash the rates of cardiovascular and renal events. Furthermore, driven by recent evidence, SGLT2i emerge as cellular supermolecules, exerting their beneficial actions to increase mitochondrial efficiency, alleviate oxidative stress, and curb severe inflammation. Its actions strengthen tissues and create a resilient defense against disease. In conclusion, like a treasure chest brimming with untold riches, the influence of SGLT2i on mitochondrial function holds untold potential for cardiovascular health. Unlocking these secrets, like a map guiding adventurers to hidden riches, promises to pave the way for even more potent therapeutic strategies.


Asunto(s)
Mitocondrias , Inhibidores del Cotransportador de Sodio-Glucosa 2 , Humanos , Inhibidores del Cotransportador de Sodio-Glucosa 2/farmacología , Mitocondrias/metabolismo , Mitocondrias/efectos de los fármacos , Animales , Enfermedades Cardiovasculares/tratamiento farmacológico , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/prevención & control
17.
Am J Physiol Cell Physiol ; 304(11): C1027-39, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23364265

RESUMEN

This review attempts to show that there may be a relationship between inflammatory processes induced by chronic overstimulation of the renin-angiotensin system (RAS) and the worldwide deficiency of vitamin D (VitD) and that both disorders are probably associated with environmental factors. Low VitD levels represent a risk factor for several apparently different diseases, such as infectious, autoimmune, neurodegenerative, and cardiovascular diseases, as well as diabetes, osteoporosis, and cancer. Moreover, VitD insufficiency seems to predispose to hypertension, metabolic syndrome, left ventricular hypertrophy, heart failure, and chronic vascular inflammation. On the other hand, inappropriate stimulation of the RAS has also been associated with the pathogenesis of hypertension, heart attack, stroke, and hypertrophy of the left ventricle and vascular smooth muscle cells. Because VitD receptors (VDRs) and RAS receptors are almost distributed in the same tissues, a possible link between VitD and the RAS is even more plausible. Furthermore, from an evolutionary point of view, both systems were developed simultaneously, actively participating in the regulation of inflammatory and immunological mechanisms. Changes in RAS activity and activation of the VDR seem to be inversely related; thus any changes in one of these systems would have a completely opposite effect on the other, making it possible to speculate that the two systems could have a feedback relationship. In fact, the pandemic of VitD deficiency could be the other face of increased RAS activity, which probably causes lower activity or lower levels of VitD. Finally, from a therapeutic point of view, the combination of RAS blockade and VDR stimulation appears to be more effective than either RAS blockade or VDR stimulation individually.


Asunto(s)
Inflamación/complicaciones , Pandemias , Sistema Renina-Angiotensina/fisiología , Deficiencia de Vitamina D/epidemiología , Deficiencia de Vitamina D/etiología , Animales , Salud Global , Humanos , Inflamación/fisiopatología , Deficiencia de Vitamina D/fisiopatología
18.
Kidney Int ; 84(6): 1145-53, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24107846

RESUMEN

The role of hyperphosphatemia in the pathogenesis of secondary hyperparathyroidism, cardiovascular disease, and progression of renal failure is widely known. Here we studied effects of dietary phosphate restriction on mortality and vascular calcification in uremic rats. Control and uremic rats were fed a high-phosphate diet and at 3 months a portion of rats of each group were killed. Serum phosphate and the calcium phosphate product increased in uremic rats, as did aortic calcium. Of the rats, 56% had positive aortic staining for calcium (von Kossa), RUNX2, and osteopontin. The remaining uremic rats were continued on diets containing high phosphate without and with sevelamer, or low phosphate, and after 3 more months they were killed. Serum phosphate was highest in uremic rats on high phosphate. Serum PTH and FGF-23 were markedly lower in rats on low phosphate. Mortality on high phosphate was 71.4%, with sevelamer reducing this to 37.5% and phosphate restriction to 5.9%. Positive aortic staining for von Kossa, RUNX2, and osteopontin was increased, but phosphate restriction inhibited this. Kidneys from low-phosphate and sevelamer-treated uremic rats had less interstitial fibrosis, glomerulosclerosis, and inflammation than those of uremic rats on high phosphate. Importantly, kidneys from rats on low phosphate showed improvement over kidneys from high-phosphate rats at 3 months. Left ventricles from rats on low phosphate had less perivascular fibrosis and smaller cardiomyocyte size compared to rats on high phosphate. Thus, intensive phosphate restriction significantly reduces mortality in uremic rats with severe vascular calcification.


Asunto(s)
Aorta/metabolismo , Dieta , Hiperfosfatemia/terapia , Riñón/metabolismo , Fosfatos/deficiencia , Uremia/terapia , Calcificación Vascular/metabolismo , Calcificación Vascular/terapia , Animales , Aorta/efectos de los fármacos , Aorta/patología , Biomarcadores/sangre , Calcio/metabolismo , Quelantes/farmacología , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Modelos Animales de Enfermedad , Femenino , Factores de Crecimiento de Fibroblastos/sangre , Fibrosis , Hiperfosfatemia/etiología , Hiperfosfatemia/metabolismo , Hiperfosfatemia/patología , Riñón/efectos de los fármacos , Riñón/patología , Miocardio/metabolismo , Miocardio/patología , Osteopontina/metabolismo , Hormona Paratiroidea/sangre , Fosfatos/administración & dosificación , Fosfatos/sangre , Poliaminas/farmacología , Ratas , Ratas Sprague-Dawley , Sevelamer , Factores de Tiempo , Uremia/etiología , Uremia/metabolismo , Uremia/patología , Calcificación Vascular/etiología , Calcificación Vascular/patología
19.
Am J Physiol Renal Physiol ; 302(12): F1595-605, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22492946

RESUMEN

Vitamin D slows the progression of chronic kidney disease. Furthermore, activators of vitamin D receptors (VDR) have suppressant effects on the renin-angiotensin system, as well as anti-inflammatory and antifibrotic actions. This study aimed to evaluate the cytoprotective effects of paricalcitol, a VDR activator, at the mitochondrial level using an obstructive nephropathy model [unilateral ureteral obstruction (UUO)]. Rats subjected to UUO and controls were treated daily with vehicle or paricalcitol. The control group underwent a sham surgery. The treatment was done for 15 days (30 ng/kg). The following were determined: biochemical parameters; fibrosis; apoptosis; mitochondrial morphology; VDR, AT(1) receptor, and NADPH oxidase 4 expression; and NADPH oxidase activity (in total and in mitochondrial fractions from the renal cortex). VDR activation prevented fibrosis (20 ± 5 vs. 60 ± 10%) and the number of TUNEL-positive apoptotic cells (10 ± 3 vs. 25 ± 4) in UUO. Biochemical, histological, and molecular studies suggest mitochondrial injury. Electron microscopy revealed in UUO electronically luminous material in the nucleus. Some mitochondria were increased in size and contained dilated crests and larger than normal spaces in their interiors. These changes were not present with paricalcitol treatment. Additionally, high AT(1)-receptor mRNA and NADPH activity was reverted in mitochondrial fractions from obstructed paricalcitol-treated animals (0.58 ± 0.06 vs. 0.95 ± 0.05 relative densitometry units and 9,000 ± 800 vs. 15,000 ± 1,000 relative fluorescence units·µg protein(-1)·min(-1), respectively). These changes were consistent with an improvement in VDR expression (0.75 ± 0.05 vs. 0.35 ± 0.04 relative densitometry units). These results suggest that paricalcitol confers a protective effect and reveal, as well, a possible AT(1) receptor-dependent protective effect that occurs at the mitochondrial level.


Asunto(s)
Citoprotección/efectos de los fármacos , Ergocalciferoles/farmacología , Enfermedades Renales/patología , Riñón/patología , Mitocondrias/patología , Obstrucción Ureteral/patología , Animales , Apoptosis/efectos de los fármacos , Ergocalciferoles/uso terapéutico , Fibrosis , Riñón/efectos de los fármacos , Riñón/metabolismo , Enfermedades Renales/tratamiento farmacológico , Enfermedades Renales/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Ratas , Ratas Endogámicas WKY , Sistema Renina-Angiotensina , Obstrucción Ureteral/metabolismo
20.
Am J Physiol Renal Physiol ; 302(1): F141-9, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-21957179

RESUMEN

Angiotensin-converting enzyme (ACE) inhibitors ameliorate the progression of renal disease. In combination with vitamin D receptor activators, they provide additional benefits. In the present study, uremic (U) rats were treated as follows: U+vehicle (UC), U+enalapril (UE; 25 mg/l in drinking water), U+paricalcitol (UP; 0.8 µg/kg ip, 3 × wk), or U+enalapril+paricalcitol (UEP). Despite hypertension in UP rats, proteinuria decreased by 32% vs. UC rats. Enalapril alone, or in combination with paricalcitol, further decreased proteinuria (≈70%). Glomerulosclerosis and interstitial infiltration increased in UC rats. Paricalcitol and enalapril inhibited this. The increase in cardiac atrial natriuretic peptide (ANP) seen in UC rats was significantly decreased by paricalcitol. Enalapril produced a more dramatic reduction in ANP. Renal oxidative stress plays a critical role in inflammation and progression of sclerosis. The marked increase in p22(phox), a subunit of NADPH oxidase, and decrease in endothelial nitric oxide synthase were inhibited in all treated groups. Cotreatment with both compounds inhibited the uremia-induced increase in proinflammatory inducible nitric oxide synthase (iNOS) and glutathione peroxidase activity better than either compound alone. Glutathione reductase was also increased in UE and UP rats vs. UC. Kidney 4-hydroxynonenal was significantly increased in the UC group compared with the normal group. Combined treatment with both compounds significantly blunted this increase, P < 0.05, while either compound alone had no effect. Additionally, the expression of Mn-SOD was increased and CuZn-SOD decreased by uremia. This was ameliorated in all treatment groups. Cotreatment with enalapril and paricalcitol had an additive effect in increasing CuZn-SOD expression. In conclusion, like enalapril, paricalcitol alone can improve proteinuria, glomerulosclerosis, and interstitial infiltration and reduce renal oxidative stress. The effects of paricalcitol may be amplified when an ACE inhibitor is added since cotreatment with both compounds seems to have an additive effect on ameliorating uremia-induced changes in iNOS and CuZn-SOD expression, peroxidase activity, and renal histomorphometry.


Asunto(s)
Enalapril/uso terapéutico , Ergocalciferoles/uso terapéutico , Glomerulonefritis/tratamiento farmacológico , Proteinuria/tratamiento farmacológico , Receptores de Calcitriol/efectos de los fármacos , Aldehídos/metabolismo , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Animales , Factor Natriurético Atrial/efectos de los fármacos , Femenino , Riñón/efectos de los fármacos , NADPH Oxidasas/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Estrés Oxidativo/efectos de los fármacos , Proteinuria/metabolismo , Ratas , Superóxido Dismutasa/metabolismo , Uremia/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda