Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(4)2023 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-36835661

RESUMEN

In recent decades, extracellular vesicles have been recognized as "very important particles" (VIPs) associated with aging and age-related disease. During the 1980s, researchers discovered that these vesicle particles released by cells were not debris but signaling molecules carrying cargoes that play key roles in physiological processes and physiopathological modulation. Following the International Society for Extracellular Vesicles (ISEV) recommendation, different vesicle particles (e.g., exosomes, microvesicles, oncosomes) have been named globally extracellular vesicles. These vesicles are essential to maintain body homeostasis owing to their essential and evolutionarily conserved role in cellular communication and interaction with different tissues. Furthermore, recent studies have shown the role of extracellular vesicles in aging and age-associated diseases. This review summarizes the advances in the study of extracellular vesicles, mainly focusing on recently refined methods for their isolation and characterization. In addition, the role of extracellular vesicles in cell signaling and maintenance of homeostasis, as well as their usefulness as new biomarkers and therapeutic agents in aging and age-associated diseases, has also been highlighted.


Asunto(s)
Micropartículas Derivadas de Células , Exosomas , Vesículas Extracelulares , Exosomas/fisiología , Micropartículas Derivadas de Células/fisiología , Transducción de Señal
2.
Arterioscler Thromb Vasc Biol ; 37(7): 1272-1281, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28546219

RESUMEN

OBJECTIVE: ILK (integrin-linked kinase) plays a key role in controlling vasomotor tone and is decreased in atherosclerosis. The objective of this study is to test whether nitric oxide (NO) regulates ILK in vascular remodeling. APPROACH AND RESULTS: We found a striking correlation between increased levels of inducible nitric oxide and decreased ILK levels in human atherosclerosis and in a mouse model of vascular remodeling (carotid artery ligation) comparing with iNOS (inducible NO synthase) knockout mice. iNOS induction produced the same result in mouse aortic endothelial cells, and these effects were mimicked by an NO donor in a time-dependent manner. We found that NO decreased ILK protein stability by promoting the dissociation of the complex ILK/Hsp90 (heat shock protein 90)/eNOS (endothelial NO synthase), leading to eNOS uncoupling. NO also destabilized ILK signaling platform and lead to decreased levels of paxillin and α-parvin. ILK phosphorylation of its downstream target GSK3-ß (glycogen synthase kinase 3 beta) was decreased by NO. Mechanistically, NO increased ILK ubiquitination mediated by the E3 ubiquitin ligase CHIP (C terminus of HSC70-interacting protein), but ILK ubiquitination was not followed by proteasome degradation. Alternatively, NO drove ILK to degradation through the endocytic-lysosomal pathway. ILK colocalized with the lysosome marker LAMP-1 (lysosomal-associated membrane protein 1) in endothelial cells, and inhibition of lysosome activity with chloroquine reversed the effect of NO. Likewise, ILK colocalized with the early endosome marker EEA1 (early endosome antigen 1). ILK endocytosis proceeded via dynamin because a specific inhibitor of dynamin (Dyngo 4a) was able to reverse ILK endocytosis and its lysosome degradation. CONCLUSIONS: Endocytosis regulates ILK signaling in vascular remodeling where there is an overload of inducible NO, and thus its inhibition may represent a novel target to fight atherosclerotic disease.


Asunto(s)
Estenosis Carotídea/enzimología , Endocitosis , Células Endoteliales/enzimología , Lisosomas/enzimología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Anciano , Anciano de 80 o más Años , Animales , Estenosis Carotídea/patología , Estenosis Carotídea/fisiopatología , Células Cultivadas , Modelos Animales de Enfermedad , Células Endoteliales/patología , Femenino , Humanos , Lisosomas/patología , Masculino , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo II/deficiencia , Óxido Nítrico Sintasa de Tipo II/genética , Proteínas Serina-Treonina Quinasas/genética , Estabilidad Proteica , Transporte de Proteínas , Proteolisis , Interferencia de ARN , Transducción de Señal , Factores de Tiempo , Transfección , Ubiquitinación , Remodelación Vascular
3.
Int J Mol Sci ; 19(7)2018 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-29987251

RESUMEN

Atherosclerosis, a chronic inflammatory disease that causes the most heart attacks and strokes in humans, is the leading cause of death in the developing world; its principal clinical manifestation is coronary artery disease. The development of atherosclerosis is attributed to the aging process itself (biological aging) and is also associated with the development of chronic diseases (premature aging). Both aging processes produce an increase in risk factors such as oxidative stress, endothelial dysfunction and proinflammatory cytokines (oxi-inflamm-aging) that might generate endothelial senescence associated with damage in the vascular system. Cellular senescence increases microvesicle release as carriers of molecular information, which contributes to the development and calcification of atherosclerotic plaque, as a final step in advanced atherosclerotic plaque formation. Consequently, this review aims to summarize the information gleaned to date from studies investigating how the senescent extracellular vesicles, by delivering biological signalling, contribute to atherosclerotic calcification.


Asunto(s)
Micropartículas Derivadas de Células/genética , Placa Aterosclerótica/genética , Calcificación Vascular/genética , Envejecimiento/genética , Envejecimiento/metabolismo , Animales , Micropartículas Derivadas de Células/metabolismo , Senescencia Celular , Predisposición Genética a la Enfermedad , Humanos , Placa Aterosclerótica/metabolismo , Factores de Riesgo , Calcificación Vascular/metabolismo
4.
Int J Mol Sci ; 19(5)2018 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-29772765

RESUMEN

Carbamylation is a post-translational modification of proteins that may partake in the oxidative stress-associated cell damage, and its increment has been recently proposed as a "hallmark of aging". The molecular mechanisms associated with aging are related to an increased release of free radicals. We have studied whether carbamylated proteins from the peripheral blood of healthy subjects are related to oxidative damage and aging, taking into account the gender and the immune profile of the subjects. The study was performed in healthy human volunteers. The detection of protein carbamylation and malondialdehyde (MDA) levels was evaluated using commercial kits. The immune profile was calculated using parameters of immune cell function. The results show that the individuals from the elderly group (60⁻79 years old) have increased carbamylated protein and MDA levels. When considered by gender, only men between 60 and 79 years old showed significantly increased carbamylated proteins and MDA levels. When those subjects were classified by their immune profile, the carbamylated protein levels were higher in those with an older immune profile. In conclusion, the carbamylation of proteins in peripheral blood is related to age-associated oxidative damage and to an aging functional immunological signature. Our results suggest that carbamylated proteins may play an important role at the cellular level in the aging process.


Asunto(s)
Envejecimiento/metabolismo , Oxidación-Reducción , Proteínas/metabolismo , Adulto , Factores de Edad , Anciano , Anciano de 80 o más Años , Envejecimiento/sangre , Biomarcadores , Femenino , Humanos , Masculino , Persona de Mediana Edad , Procesamiento Proteico-Postraduccional , Especies Reactivas de Oxígeno/metabolismo , Factores Sexuales , Adulto Joven
5.
J Pathol ; 236(4): 407-20, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25810250

RESUMEN

Inflammation is a main feature of progressive kidney disease. Gremlin binds to bone morphogenetic proteins (BMPs), acting as an antagonist and regulating nephrogenesis and fibrosis among other processes. Gremlin also binds to vascular endothelial growth factor receptor-2 (VEGFR2) in endothelial cells to induce angiogenesis. In renal cells, gremlin regulates proliferation and fibrosis, but there are no data about inflammatory-related events. We have investigated the direct effects of gremlin in the kidney, evaluating whether VEGFR2 is a functional gremlin receptor. Administration of recombinant gremlin to murine kidneys induced rapid and sustained activation of VEGFR2 signalling, located in proximal tubular epithelial cells. Gremlin bound to VEGFR2 in these cells in vitro, activating this signalling pathway independently of its action as an antagonist of BMPs. In vivo, gremlin caused early renal damage, characterized by activation of the nuclear factor (NF)-κB pathway linked to up-regulation of pro-inflammatory factors and infiltration of immune inflammatory cells. VEGFR2 blockade diminished gremlin-induced renal inflammatory responses. The link between gremlin/VEGFR2 and NF-κB/inflammation was confirmed in vitro. Gremlin overexpression was associated with VEGFR2 activation in human renal disease and in the unilateral ureteral obstruction experimental model, where VEGFR2 kinase inhibition diminished renal inflammation. Our data show that a gremlin/VEGFR2 axis participates in renal inflammation and could be a novel target for kidney disease.


Asunto(s)
Mediadores de Inflamación/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Riñón/metabolismo , Nefritis/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Línea Celular Tumoral , Citocinas , Modelos Animales de Enfermedad , Femenino , Humanos , Riñón/patología , Masculino , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Nefritis/etiología , Nefritis/genética , Nefritis/patología , Unión Proteica , Interferencia de ARN , Estudios Retrospectivos , Transducción de Señal , Factores de Tiempo , Transfección , Obstrucción Ureteral/complicaciones , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética
6.
Circulation ; 130(15): 1274-86, 2014 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-25116956

RESUMEN

BACKGROUND: Alternatively spliced tissue factor (asTF) is a novel isoform of full-length tissue factor, which exhibits angiogenic activity. Although asTF has been detected in human plaques, it is unknown whether its expression in atherosclerosis causes increased neovascularization and an advanced plaque phenotype. METHODS AND RESULTS: Carotid (n=10) and coronary (n=8) specimens from patients with stable or unstable angina were classified as complicated or uncomplicated on the basis of plaque morphology. Analysis of asTF expression and cell type-specific expression revealed a strong expression and colocalization of asTF with macrophages and neovessels within complicated, but not uncomplicated, human plaques. Our results showed that the angiogenic activity of asTF is mediated via hypoxia-inducible factor-1α upregulation through integrins and activation of phosphatidylinositol-3-kinase/Akt and mitogen-activated protein kinase pathways. Hypoxia-inducible factor-1α upregulation by asTF also was associated with increased vascular endothelial growth factor expression in primary human endothelial cells, and vascular endothelial growth factor-Trap significantly reduced the angiogenic effect of asTF in vivo. Furthermore, asTF gene transfer significantly increased neointima formation and neovascularization after carotid wire injury in ApoE(-/-) mice. CONCLUSIONS: The results of this study provide strong evidence that asTF promotes neointima formation and angiogenesis in an experimental model of accelerated atherosclerosis. Here, we demonstrate that the angiogenic effect of asTF is mediated via the activation of the hypoxia-inducible factor-1/vascular endothelial growth factor signaling. This mechanism may be relevant to neovascularization and the progression and associated complications of human atherosclerosis as suggested by the increased expression of asTF in complicated versus uncomplicated human carotid and coronary plaques.


Asunto(s)
Empalme Alternativo/fisiología , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Neovascularización Patológica/fisiopatología , Placa Aterosclerótica/fisiopatología , Transducción de Señal/fisiología , Tromboplastina/fisiología , Factor A de Crecimiento Endotelial Vascular/fisiología , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Arterias Carótidas/patología , Arterias Carótidas/fisiopatología , Vasos Coronarios/patología , Vasos Coronarios/fisiopatología , Modelos Animales de Enfermedad , Endotelio Vascular/patología , Endotelio Vascular/fisiopatología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Neointima/fisiopatología , Placa Aterosclerótica/patología , Regulación hacia Arriba/fisiología
7.
Clin Sci (Lond) ; 127(1): 19-31, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24383472

RESUMEN

ILK (integrin-linked kinase) is an intracellular serine/threonine kinase involved in cell-matrix interactions. ILK dysregulation has been described in chronic renal disease and modulates podocyte function and fibrosis, whereas data about its role in inflammation are scarce. AngII (angiotensin II) is a pro-inflammatory cytokine that promotes renal inflammation. AngII blockers are renoprotective and down-regulate ILK in experimental kidney disease, but the involvement of ILK in the actions of AngII in the kidney has not been addressed. Therefore we have investigated whether ILK signalling modulates the kidney response to systemic AngII infusion in wild-type and ILK-conditional knockout mice. In wild-type mice, AngII induced an inflammatory response, characterized by infiltration of monocytes/macrophages and lymphocytes, and up-regulation of pro-inflammatory factors (chemokines, adhesion molecules and cytokines). AngII activated several intracellular signalling mechanisms, such as the NF-κB (nuclear factor κB) transcription factor, Akt and production of ROS (reactive oxygen species). All these responses were prevented in AngII-infused ILK-deficient mice. In vitro studies characterized further the mechanisms regulating the inflammatory response modulated by ILK. In cultured tubular epithelial cells ILK blockade, by siRNA, inhibited AngII-induced NF-κB subunit p65 phosphorylation and its nuclear translocation. Moreover, ILK gene silencing prevented NF-κB-related pro-inflammatory gene up-regulation. The results of the present study demonstrate that ILK plays a key role in the regulation of renal inflammation by modulating the canonical NF-κB pathway, and suggest a potential therapeutic target for inflammatory renal diseases.


Asunto(s)
Angiotensina II/farmacología , Nefritis/enzimología , Proteínas Serina-Treonina Quinasas/fisiología , Animales , Células Cultivadas , Femenino , Silenciador del Gen , Humanos , Mediadores de Inflamación/metabolismo , Túbulos Renales/citología , Túbulos Renales/metabolismo , Ratones Noqueados , FN-kappa B/metabolismo , Nefritis/inducido químicamente , Nefritis/prevención & control , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/fisiología , ARN Interferente Pequeño/genética , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Regulación hacia Arriba/efectos de los fármacos
8.
Neurosci Biobehav Rev ; 163: 105761, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38852847

RESUMEN

The development of new analgesics for neuropathic pain treatment is crucial. The failure of promising drugs in clinical trials may be related to the over-reliance on reflex-based responses (evoked pain) in preclinical drug testing, which may not fully represent clinical neuropathic pain, characterized by spontaneous non-evoked pain (NEP). Hence, strategies for assessing NEP in preclinical studies emerged. This systematic review identified 443 articles evaluating NEP in neuropathic pain models (mainly traumatic nerve injuries in male rodents). An exponential growth in NEP evaluation was observed, which was assessed using 48 different tests classified in 12 NEP-related outcomes: anxiety, exploration/locomotion, paw lifting, depression, conditioned place preference, gait, autotomy, wellbeing, facial grooming, cognitive impairment, facial pain expressions and vocalizations. Although most of these outcomes showed clear limitations, our analysis suggests that conditioning-associated outcomes, pain-related comorbidities, and gait evaluation may be the most effective strategies. Moreover, a minimal part of the studies evaluated standard analgesics. The greater emphasis on evaluating NEP aligning with clinical pain symptoms may enhance analgesic drug development, improving clinical translation.


Asunto(s)
Modelos Animales de Enfermedad , Neuralgia , Animales , Neuralgia/fisiopatología , Neuralgia/tratamiento farmacológico , Dimensión del Dolor/métodos , Humanos , Evaluación Preclínica de Medicamentos , Analgésicos/farmacología
9.
Life Sci ; 351: 122810, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-38871114

RESUMEN

AIMS: Cardiovascular pathology is the main cause of death in chronic kidney disease (CKD) patients. CKD is associated with the accumulation of uremic toxins in the bloodstream, and indoxyl sulfate (IS) is one of the most abundant uremic toxins found in the blood of CKD patients. We conducted an in vitro study to assess the mechanisms underlying the IS-induced endothelial dysfunction that could lead to cardiovascular diseases. We also studied their extracellular vesicles (EVs) owing to their capacity to act as messengers that transmit signals through their cargo. MAIN METHODS: EVs were characterized by nanoparticle tracking analysis, transmission electron microscopy, flow cytometry, and tetraspanin expression. Cell lysates and isolated EVs were analyzed using liquid chromatography coupled with mass spectrometry, followed by Gene Set Enrichment Analysis to identify the altered pathways. KEY FINDINGS: Proteomic analysis of endothelial cells revealed that IS causes an increase in proteins related to adipogenesis, inflammation, and xenobiotic metabolism and a decrease in proliferation. Extracellular matrix elements, as well as proteins associated with myogenesis, response to UV irradiation, and inflammation, were found to be downregulated in IS-treated EVs. Fatty acid metabolism was also found to be increased along with adipogenesis and inflammation observed in cells. SIGNIFICANCE: The treatment of endothelial cells with IS increased the expression of proteins related to adipogenesis, inflammation, and xenobiotic metabolism and was less associated with proliferation. Furthermore, EVs from cells treated with IS may mediate endothelial dysfunction, since they present fewer extracellular matrix elements, myogenesis, inflammatory factors, and proteins downregulated in response to UV radiation.


Asunto(s)
Células Endoteliales , Vesículas Extracelulares , Indicán , Proteómica , Insuficiencia Renal Crónica , Indicán/metabolismo , Vesículas Extracelulares/metabolismo , Humanos , Insuficiencia Renal Crónica/metabolismo , Proteómica/métodos , Células Endoteliales/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Proteoma/metabolismo
10.
Lab Invest ; 93(7): 812-24, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23648563

RESUMEN

Connective tissue growth factor (CTGF/CCN2) is a matricellular protein susceptible to proteolytic degradation. CCN2 levels have been suggested as a potential risk biomarker in several chronic diseases. In body fluids, CCN2 full-length and its degradation fragments can be found; however, their in vivo effects are far from being elucidated. CCN2 was described as a profibrotic mediator, but this concept is changing to a proinflammatory cytokine. In vitro, CCN2 full-length and its C-terminal module IV (CCN2(IV)) exert proinflammatory properties. Emerging evidence suggest that Th17 cells, and its effector cytokine IL-17A, participate in chronic inflammatory diseases. Our aim was to explore whether CCN2(IV) could regulate the Th17 response. In vitro, stimulation of human naive CD4+ T lymphocytes with CCN2(IV) resulted in differentiation to Th17 phenotype. The in vivo effects of CCN2(IV) were studied in C57BL/6 mice. Intraperitoneal administration of recombinant CCN2(IV) did not change serum IL-17A levels, but caused an activation of the Th17 response in the kidney, characterized by interstitial infiltration of Th17 (IL17A+/CD4+) cells and upregulation of proinflammatory mediators. In CCN2(IV)-injected mice, elevated renal levels of Th17-related factors (IL-17A, IL-6, STAT3 and RORγt) were found, whereas Th1/Th2 cytokines or Treg-related factors (TGF-ß and Foxp-3) were not modified. Treatment with an anti-IL-17A neutralizing antibody diminished CCN2(IV)-induced renal inflammation. Our findings unveil that the C-terminal module of CCN2 induces the Th17 differentiation of human Th17 cells and causes a renal Th17 inflammatory response. Furthermore, these data bear out that IL-17A targeting is a promising tool for chronic inflammatory diseases, including renal pathologies.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Interleucina-17/sangre , Riñón/inmunología , Células Th17/fisiología , Animales , Factor de Crecimiento del Tejido Conjuntivo/administración & dosificación , Humanos , Riñón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Factores de Transcripción/metabolismo , Regulación hacia Arriba
11.
Eur Heart J ; 33(2): 264-73, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21606082

RESUMEN

AIMS: The aim of this study was to investigate the effects of liver X receptors (LXRs)-ß preferential activation by LXR-623 (WAY-252623), a novel LXRs agonist, on plaque progression/regression in a rabbit model of advanced atherosclerosis. METHODS AND RESULTS: Advanced atherosclerosis was induced in New Zealand White Rabbits (n= 41). At the end of atherosclerosis induction, animals underwent a baseline magnetic resonance imaging (MRI) and were randomized to receive LXR-623 (1.5, 5, or 15 mg/kg/day), simvastatin (5 mg/kg/day), or placebo. The combination of LXR-1.5/simvastatin was also tested. After a final MRI, animals were euthanized and their aortas processed for further analysis. Simvastatin significantly reduced lesion progression (-25%; P< 0.01) in comparison with the placebo group. A similar effect was observed in the LXR-1.5 and -5 groups. A significant regression (16.5%; P< 0.01) of existing atherosclerosis was observed in the LXR-1.5/simvastatin group. Histological and molecular analysis showed plaque stabilization in the animals treated with the LXR-1.5 and -5, and LXR-1.5/simvastatin. The effects of LXR-623 were observed in the presence of a non-significant effect on total-cholesterol, low-density lipoproteins-cholesterol, and triglyceride levels. CONCLUSION: The results of the present study show that LXR-623 significantly reduces the progression of atherosclerosis and induces plaque regression in combination with simvastatin. These observations could drive future development of novel anti-atherosclerotic therapeutic approaches.


Asunto(s)
Anticolesterolemiantes/farmacología , Aterosclerosis/tratamiento farmacológico , Indazoles/farmacología , Receptores Nucleares Huérfanos/efectos de los fármacos , Placa Aterosclerótica/tratamiento farmacológico , Simvastatina/farmacología , Animales , Aorta Abdominal , Enfermedades de la Aorta/tratamiento farmacológico , Enfermedades de la Aorta/metabolismo , Aterosclerosis/metabolismo , Quimiocina CCL2/metabolismo , Ciclooxigenasa 2/metabolismo , Progresión de la Enfermedad , Combinación de Medicamentos , Sinergismo Farmacológico , Receptores X del Hígado , Angiografía por Resonancia Magnética , Receptores Nucleares Huérfanos/antagonistas & inhibidores , Placa Aterosclerótica/metabolismo , Conejos , Distribución Aleatoria , Tromboplastina/metabolismo , Regulación hacia Arriba
12.
Biomolecules ; 14(1)2023 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-38254635

RESUMEN

This review highlights the significant impact of air quality, specifically particulate matter (PM), on cognitive decline and age-related diseases in the elderly. Despite established links to other pathologies, such as respiratory and cardiovascular illnesses, there is a pressing need for increased attention to the association between air pollution and cognitive aging, given the rising prevalence of neurocognitive disorders. PM sources are from diverse origins, including industrial activities and combustion engines, categorized into PM10, PM2.5, and ultrafine PM (UFPM), and emphasized health risks from both outdoor and indoor exposure. Long-term PM exposure, notably PM2.5, has correlated with declines in cognitive function, with a specific vulnerability observed in women. Recently, extracellular vesicles (EVs) have been explored due to the interplay between them, PM exposure, and human aging, highlighting the crucial role of EVs, especially exosomes, in mediating the complex relationship between PM exposure and chronic diseases, particularly neurological disorders. To sum up, we have compiled the pieces of evidence that show the potential contribution of PM exposure to cognitive aging and the role of EVs in mediating PM-induced cognitive impairment, which presents a promising avenue for future research and development of therapeutic strategies. Finally, this review emphasizes the need for policy changes and increased public awareness to mitigate air pollution, especially among vulnerable populations such as the elderly.


Asunto(s)
Contaminación del Aire , Disfunción Cognitiva , Anciano , Humanos , Femenino , Cognición , Envejecimiento , Contaminación del Aire/efectos adversos , Material Particulado/efectos adversos
13.
Nefrologia (Engl Ed) ; 43(1): 63-80, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37268501

RESUMEN

Chronic kidney disease (CKD) is a pathology with a high worldwide incidence and an upward trend affecting the elderly. When CKD is very advanced, the use of renal replacement therapies is required to prolong its life (dialysis or kidney transplantation). Although dialysis improves many complications of CKD, the disease does not reverse completely. These patients present an increase in oxidative stress, chronic inflammation and the release of extracellular vesicles (EVs), which cause endothelial damage and the development of different cardiovascular diseases (CVD). CKD patients develop premature diseases associated with advanced age, such as CVD. EVs play an essential role in developing CVD in patients with CKD since their number increases in plasma and their content is modified. The EVs of patients with CKD cause endothelial dysfunction, senescence and vascular calcification. In addition, miRNAs free or transported in EVs together with other components carried in these EVs promote endothelial dysfunction, thrombotic and vascular calcification in CKD, among other effects. This review describes the classic factors and focuses on the role of new mechanisms involved in the development of CVD associated with CKD, emphasizing the role of EVs in the development of cardiovascular pathologies in the context of CKD. Moreover, the review summarized the EVs' role as diagnostic and therapeutic tools, acting on EV release or content to avoid the development of CVD in CKD patients.


Asunto(s)
Enfermedades Cardiovasculares , MicroARNs , Insuficiencia Renal Crónica , Calcificación Vascular , Humanos , Anciano , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/epidemiología , Insuficiencia Renal Crónica/complicaciones , Insuficiencia Renal Crónica/terapia , Insuficiencia Renal Crónica/diagnóstico , Calcificación Vascular/etiología , Calcificación Vascular/patología , Inflamación
14.
Nutrients ; 15(6)2023 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-36986078

RESUMEN

As life expectancy increases in many countries, the prevalence of age-related diseases also rises. Among these conditions, chronic kidney disease is predicted to become the second cause of death in some countries before the end of the century. An important problem with kidney diseases is the lack of biomarkers to detect early damage or to predict the progression to renal failure. In addition, current treatments only retard kidney disease progression, and better tools are needed. Preclinical research has shown the involvement of the activation of cellular senescence-related mechanisms in natural aging and kidney injury. Intensive research is searching for novel treatments for kidney diseases as well as for anti-aging therapies. In this sense, many experimental shreds of evidence support that treatment with vitamin D or its analogs can exert pleiotropic protective effects in kidney injury. Moreover, vitamin D deficiency has been described in patients with kidney diseases. Here, we review recent evidence about the relationship between vitamin D and kidney diseases, explaining the underlying mechanisms of the effect of vitamin D actions, with particular attention to the modulation of cellular senescence mechanisms.


Asunto(s)
Enfermedades Renales , Insuficiencia Renal Crónica , Deficiencia de Vitamina D , Humanos , Vitamina D , Insuficiencia Renal Crónica/complicaciones , Riñón , Vitaminas/uso terapéutico , Enfermedades Renales/etiología , Deficiencia de Vitamina D/epidemiología , Senescencia Celular
15.
Clin Kidney J ; 16(8): 1278-1287, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37529650

RESUMEN

Background: Endothelial damage and cardiovascular disease complicate chronic kidney disease (CKD). The increased atherogenicity observed in patients with CKD can be linked to microinflammation and endothelial damage. Circulating endothelial glycocalyx degradation products, such as perlecan and decorin, tend to be elevated in CKD. We aimed to explore the association between the plasma perlecan and decorin levels and this pro-inflammatory and atherogenic state by studying monocyte subpopulations and intracellular adhesion molecule (ICAM)-1 expression in patients with CKD. Methods: We studied 17 healthy controls, 23 patients with advanced CKD, 25 patients on haemodialysis, 23 patients on peritoneal dialysis and 20 patients who underwent kidney transplantation. Perlecan and decorin levels were evaluated using enzyme-linked immunosorbent assays, and the monocyte phenotype was analysed using direct immunofluorescence and flow cytometry. Results: The plasma perlecan levels were higher in patients with CKD than in the healthy controls. These levels were associated with a higher prevalence of ICAM-1+ monocytes. Conversely, patients with advanced CKD (pre-dialysis) had higher plasma decorin levels, which were associated with a reduced ICAM-1 expression per monocyte. Conclusions: Elevated perlecan levels in CKD may be associated with a higher prevalence of ICAM-1+ monocytes and a pro-inflammatory phenotype. Elevated decorin levels may act as a negative regulator of ICAM-1 expression in monocytes. Therefore, perlecan and decorin may be related to inflammation and monocyte activation in CKD and may act as potential markers of endothelial damage.

16.
Front Cardiovasc Med ; 9: 854726, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35498012

RESUMEN

Vascular calcification is an irreversible pathological process associated with a loss of vascular wall function. This process occurs as a result of aging and age-related diseases, such as cardiovascular and chronic kidney diseases, and leads to comorbidities. During these age-related diseases, the endothelium accumulates senescent cells, which stimulate calcification in vascular smooth muscle cells. Currently, vascular calcification is a silent pathology, and there are no early diagnostic tools. Therefore, by the time vascular calcification is diagnosed, it is usually untreatable. Some mediators, such as oxidative stress, inflammation, and extracellular vesicles, are inducers and promoters of vascular calcification. They play a crucial role during vascular generation and the progression of vascular calcification. Extracellular vesicles, mainly derived from injured endothelial cells that have acquired a senescent phenotype, contribute to calcification in a manner mostly dependent on two factors: (1) the number of extracellular vesicles released, and (2) their cargo. In this review, we present state-of-the-art knowledge on the composition and functions of extracellular vesicles involved in the generation and progression of vascular calcification.

17.
Antioxidants (Basel) ; 11(4)2022 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-35453386

RESUMEN

Aging is a risk factor for several diseases, including cardiovascular disease, type 2 diabetes, hypertension, cancer, osteoarthritis, and Alzheimer; oxidative stress is a key player in the development and progression of aging and age-associated diseases [...].

18.
J Pers Med ; 12(2)2022 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-35207703

RESUMEN

Atherosclerosis is probably one of the paradigms of disease linked to aging. Underlying the physiopathology of atherosclerosis are cellular senescence, oxidative stress, and inflammation. These factors are increased in the elderly and from chronic disease patients. Elevated levels of oxidative stress affect cellular function and metabolism, inducing senescence. This senescence modifies the cell phenotype into a senescent secretory phenotype. This phenotype activates immune cells, leading to chronic systemic inflammation. Moreover, due to their secretory phenotype, senescence cells present an increased release of highlighted extracellular vesicles that will change nearby/neighborhood cells and paracrine signaling. For this reason, searching for specific senescent cell biomarkers and therapies against the development/killing of senescent cells has become relevant. Recently, senomorphic and senolityc drugs have become relevant in slowing down or eliminating senescence cells. However, even though they have shown promising results in experimental studies, their clinical use is still yet to be determined.

19.
Am J Physiol Cell Physiol ; 300(4): C907-18, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21209362

RESUMEN

Glomerular diseases are characterized by a sustained synthesis and accumulation of abnormal extracellular matrix proteins, such as collagen type I. The extracellular matrix transmits information to cells through interactions with membrane components, which directly activate many intracellular signaling events. Moreover, accumulating evidence suggests that eicosanoids derived from cyclooxygenase (COX)-2 participate in a number of pathological processes in immune-mediated renal diseases, and it is known that protein kinase B (AKT) may act through different transcription factors in the regulation of the COX-2 promoter. The present results show that progressive accumulation of collagen I in the extracellular medium induces a significant increase of COX-2 expression in human mesangial cells, resulting in an enhancement in PGE(2) production. COX-2 overexpression is due to increased COX-2 mRNA levels. The study of the mechanism implicated in COX-2 upregulation by collagen I showed focal adhesion kinase (FAK) activation. Furthermore, we observed that the activation of the phosphatidylinositol 3-kinase (PI3K)/AKT pathway by collagen I and collagen I-induced COX-2 overexpression was abolished by PI3K and AKT inhibitors. Additionally, we showed that the cAMP response element (CRE) transcription factor is implicated. Finally, we studied COX-2 expression in an animal model, N(G)-nitro-l-arginine methyl ester hypertensive rats. In renal tissue and vascular walls, COX-2 and collagen type I content were upregulated. In summary, our results provide evidence that collagen type I increases COX-2 expression via the FAK/PI3K/AKT/cAMP response element binding protein signaling pathway.


Asunto(s)
Ciclooxigenasa 2/metabolismo , Matriz Extracelular/química , Regulación Enzimológica de la Expresión Génica , Células Mesangiales/enzimología , Animales , Células Cultivadas , Colágeno/química , Colágeno/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Ciclooxigenasa 2/genética , Dinoprostona/metabolismo , Matriz Extracelular/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Humanos , Células Mesangiales/citología , Fosfatidilinositol 3-Quinasas/metabolismo , Regiones Promotoras Genéticas , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Transducción de Señal/fisiología
20.
J Cardiovasc Pharmacol ; 57(3): 325-33, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21164355

RESUMEN

Apolipoprotein A-I (ApoA-I)/high-density lipoprotein (HDL)-raising treatments are effective antiatherosclerotic strategies. We have compared the antiatherogenic effects of human ApoA-I (hApoA-I) overexpression by intraportal and intramuscular gene transfer in atherosclerotic ApoE-knockout mice. Atherosclerotic lesions were induced by atherogenic diet. After atherosclerosis induction, a group of animals was killed and served as atherosclerosis baseline-control group. The remaining animals were randomized into the following groups: (1) atherosclerosis-progression-control, (2) intraportal/vector administration, and (3) intramuscular/vector administration. Aortas and hearts were processed for atherosclerotic quantification by en face Sudan IV and Oil Red-O, respectively. Liver and muscle specimens were processed for protein/gene expression analysis. A sustained increase in hApoA-I/HDL plasma levels was observed in both transduced groups. hApoA-I overexpression abolished plaque progression versus progression-control group. hApoA-I overexpression significantly reduced lesion macrophage, feature indicative of plaque stabilization. Scavenger receptor class-B type I (SR-BI), but not ATP-binding cassette, sub-family A (ABCA), member 1 (ABCA-1), was significantly upregulated in treated groups versus progression-controls. The results of this study show a similar effect of hApoA-I/HDL overexpression on plaque progression/stabilization by 2 different routes of administration. Our results showing similar effects using either intramuscular administration and intraportal route of administration may have significant clinical implications, given the reduced medical risk to patient and cost of intramuscular injections.


Asunto(s)
Aorta/efectos de los fármacos , Apolipoproteína A-I/genética , Apolipoproteína A-I/uso terapéutico , Apolipoproteínas E/genética , Aterosclerosis/tratamiento farmacológico , Dependovirus/metabolismo , Hígado/efectos de los fármacos , Transportador 1 de Casete de Unión a ATP , Transportadoras de Casetes de Unión a ATP/análisis , Transportadoras de Casetes de Unión a ATP/genética , Animales , Aorta/patología , Apolipoproteína A-I/administración & dosificación , Apolipoproteína A-I/sangre , Apolipoproteínas E/metabolismo , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , HDL-Colesterol/sangre , Dependovirus/genética , Dieta Aterogénica , Progresión de la Enfermedad , Evaluación Preclínica de Medicamentos , Vectores Genéticos , Humanos , Inyecciones Intramusculares , Inyecciones Intravenosas , Hígado/anatomía & histología , Hígado/fisiopatología , Ratones , Ratones Noqueados , Terapia Molecular Dirigida , Receptores Depuradores de Clase B/análisis , Receptores Depuradores de Clase B/genética , Factores de Tiempo , Transducción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA