Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Microvasc Res ; 153: 104653, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38220030

RESUMEN

The use of e-cigarettes or vapes is increasingly popular amongst a range of different demographics however the research in this area is surprisingly sparse. Clinical reports of e-cigarette- or vaping use-associated lung injury (EVALI) and vascular disruption, in both nicotine-containing and nicotine-free e-cigarette smokers, prompts the need for further research with a focus on the pulmonary endothelium. Using a common brand of e-cigarette (eVape) and an in vitro model of the human lung microvasculature, we investigated the effect of nicotine-free eVape fluid on pulmonary endothelial barrier integrity, oxidative stress and inflammation profile. Findings demonstrate reactive oxygen species-dependent breakdown of the pulmonary endothelium and release of inflammatory cytokines. These phenotypic changes, following exposure to nicotine-free eVape fluid, were accompanied by dysregulation of a number of adheren junctions-related genes of which ARF6 was most abundantly overexpressed. Further investigation of ARF6 identified it as a key regulator in eVape-induced barrier disruption and ROS accumulation. This study demonstrates, for the first time, the barrier disruptive effect of nicotine-free e-cigarette fluid on the pulmonary microvasculature and the ARF6 and ROS-dependent molecular mechanisms underlying this damage. Whilst these studies focus on a human in vitro model of the pulmonary microvasculature, the results support clinical case studies on EVALI and demonstrate a need for further investigation of the impact of nicotine-free e-cigarettes on the lung.


Asunto(s)
Sistemas Electrónicos de Liberación de Nicotina , Lesión Pulmonar , Vapeo , Humanos , Vapeo/efectos adversos , Especies Reactivas de Oxígeno/metabolismo , Pulmón/metabolismo , Nicotina/toxicidad , Endotelio/metabolismo
2.
Int J Mol Sci ; 22(10)2021 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-34063332

RESUMEN

Artificial sweeteners (AS) are synthetic sugar substitutes that are commonly consumed in the diet. Recent studies have indicated considerable health risks which links the consumption of AS with metabolic derangements and gut microbiota perturbations. Despite these studies, there is still limited data on how AS impacts the commensal microbiota to cause pathogenicity. The present study sought to investigate the role of commonly consumed AS on gut bacterial pathogenicity and gut epithelium-microbiota interactions, using models of microbiota (Escherichia coli NCTC10418 and Enterococcus faecalis ATCC19433) and the intestinal epithelium (Caco-2 cells). Model gut bacteria were exposed to different concentrations of the AS saccharin, sucralose, and aspartame, and their pathogenicity and changes in interactions with Caco-2 cells were measured using in vitro studies. Findings show that sweeteners differentially increase the ability of bacteria to form a biofilm. Co-culture with human intestinal epithelial cells shows an increase in the ability of model gut bacteria to adhere to, invade and kill the host epithelium. The pan-sweet taste inhibitor, zinc sulphate, effectively blocked these negative impacts. Since AS consumption in the diet continues to increase, understanding how this food additive affects gut microbiota and how these damaging effects can be ameliorated is vital.


Asunto(s)
Enterococcus faecalis/efectos de los fármacos , Escherichia coli/efectos de los fármacos , Microbioma Gastrointestinal/efectos de los fármacos , Edulcorantes/farmacología , Aspartame/administración & dosificación , Aspartame/farmacología , Adhesión Bacteriana/efectos de los fármacos , Biopelículas/efectos de los fármacos , Células CACO-2 , Relación Dosis-Respuesta a Droga , Enterococcus faecalis/patogenicidad , Escherichia coli/patogenicidad , Microbioma Gastrointestinal/fisiología , Hemólisis/efectos de los fármacos , Humanos , Sacarina/administración & dosificación , Sacarina/farmacología , Sacarosa/administración & dosificación , Sacarosa/análogos & derivados , Sacarosa/farmacología , Edulcorantes/administración & dosificación
3.
Eur Respir J ; 55(3)2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31949110

RESUMEN

Endothelial dysfunction is a hallmark of pulmonary arterial hypertension (PAH) but there are no established methods to study pulmonary artery endothelial cells (PAECs) from living patients. We sought to culture PAECs from pulmonary artery catheter (PAC) balloons used during right-heart catheterisation (RHC) to characterise successful culture attempts and to describe PAEC behaviour.PAECs were grown in primary culture to confluence and endothelial cell phenotype was confirmed. Standard assays for apoptosis, migration and tube formation were performed between passages three to eight. We collected 49 PAC tips from 45 subjects with successful PAEC culture from 19 balloons (39%).There were no differences in subject demographic details or RHC procedural details in successful versus unsuccessful attempts. However, for subjects who met haemodynamic criteria for PAH, there was a higher but nonsignificant (p=0.10) proportion amongst successful attempts (10 out of 19, 53%) versus unsuccessful attempts (nine out of 30, 30%). A successful culture was more likely in subjects with a lower cardiac index (p=0.03) and higher pulmonary vascular resistance (p=0.04). PAECs from a subject with idiopathic PAH were apoptosis resistant compared to commercial PAECs (p=0.04) and had reduced migration compared to PAECs from a subject with portopulmonary hypertension with high cardiac output (p=0.01). PAECs from a subject with HIV-associated PAH formed fewer (p=0.01) and shorter (p=0.02) vessel networks compared to commercial PAECs.Sustained culture and characterisation of PAECs from RHC balloons is feasible, especially in PAH with high haemodynamic burden. This technique may provide insight into endothelial dysfunction during PAH pathogenesis.


Asunto(s)
Arteria Pulmonar , Enfermedades Vasculares , Catéteres , Células Cultivadas , Células Endoteliales , Humanos , Pulmón
4.
Exp Eye Res ; 197: 108104, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32522479

RESUMEN

Age-related macular degeneration (AMD) and proliferative diabetic retinopathy (DR) are two of the most common and severe causes of vision loss in the population. Both conditions are associated with excessive levels of vascular endothelial growth factor (VEGF) in the eye which results in an increase in the formation of new blood vessels through a process called neovascularisation. As such, anti-VEGF therapies are currently utilised as a treatment for patients with AMD however they are associated with painful administration of injections and potential degeneration of healthy endothelium. There is therefore growing interest in alternate treatment options to reduce neovascularisation in the eye. The use of carotenoids, lutein (L) and zeaxanthin (Z), has been shown to improve vision loss parameters in patients with AMD, however the underlying mechanisms are not well-understood. We studied the impact of these compounds on neovascularisation processes using an in vitro cell model of the retinal microvascular endothelium. Our findings show that L and Z reduced VEGF-induced tube formation whilst, in combination (5:1 ratio), the compounds significantly blocked VEGF-induced neovascularisation. The carotenoids, individually and in combination, reduced VEGF-induced oxidative stress concomitant with increased activity of the NADPH oxidase, Nox4. We further demonstrated that the Nox4 inhibitor, GLX7013114, attenuated the protective effect of L and Z. Taken together, these findings indicate the protective effect of the carotenoids, L and Z, in reducing VEGF-mediated neovascularisation via a Nox4-dependent pathway. These studies implicate the potential for these compounds to be used as a therapeutic approach for patients suffering from AMD and proliferative DR.


Asunto(s)
Células Endoteliales/metabolismo , Luteína/farmacología , Degeneración Macular/tratamiento farmacológico , NADPH Oxidasa 4/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Zeaxantinas/farmacología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Humanos , Degeneración Macular/metabolismo , Degeneración Macular/patología , Microvasos/efectos de los fármacos , Microvasos/metabolismo , Microvasos/patología , Especies Reactivas de Oxígeno/metabolismo , Vasos Retinianos/efectos de los fármacos , Vasos Retinianos/metabolismo , Vasos Retinianos/patología
5.
Graefes Arch Clin Exp Ophthalmol ; 257(1): 71-81, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30353220

RESUMEN

BACKGROUND: One of the most prevalent microvascular complications for patients with diabetes is diabetic retinopathy (DR) associated with increased retinal endothelial blood vessel formation. Treatments to reduce vascularisation in the retinal endothelium are linked to improved sight in patients with DR. Recently, we have demonstrated the novel protective role of the artificial sweetener, sucralose, and the sweet taste receptor, T1R3, in the pulmonary endothelium to reduce vascular leak. In the present study, we examined the role of sucralose and sweet taste receptors on vasculogenic processes (proliferation, migration, adhesion and tube formation) in a cell model of the retinal endothelium. METHODS: We exposed human retinal microvascular endothelial cells (RMVEC) to VEGF as an in vitro model of DR in the presence and absence of T1R3 agonist sucralose. RESULTS: In RMVEC, we observed increased VEGF-induced cell proliferation, migration, adhesion and tube formation, which was significantly attenuated by exposure to the artificial sweetener sucralose. Following siRNA knockdown of the sweet taste receptor, T1R3, but not T1R2, the protective effect of sucralose on VEGF-induced RMVEC vasculogenic processes was blocked. We further demonstrate that sucralose attenuates VEGF-induced Akt phosphorylation to protect the retinal microvasculature. CONCLUSION: These studies are the first to demonstrate a protective effect of an artificial sweetener, through the sweet taste receptor T1R3, on VEGF-induced vasculogenesis in a retinal microvascular endothelial cell line.


Asunto(s)
Retinopatía Diabética/complicaciones , Endotelio Vascular/metabolismo , Regulación de la Expresión Génica , Receptores Acoplados a Proteínas G/genética , Neovascularización Retiniana/genética , Vasos Retinianos/patología , Sacarosa/análogos & derivados , Western Blotting , Línea Celular , Movimiento Celular , Proliferación Celular , Retinopatía Diabética/genética , Retinopatía Diabética/patología , Endotelio Vascular/patología , Ensayo de Inmunoadsorción Enzimática , Humanos , Microvasos/metabolismo , Microvasos/patología , ARN/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Acoplados a Proteínas G/biosíntesis , Receptores Acoplados a Proteínas G/efectos de los fármacos , Neovascularización Retiniana/etiología , Neovascularización Retiniana/patología , Vasos Retinianos/metabolismo , Sacarosa/farmacología
6.
Am J Physiol Lung Cell Mol Physiol ; 314(1): L165-L176, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-28971978

RESUMEN

A hallmark of acute respiratory distress syndrome (ARDS) is pulmonary vascular permeability. In these settings, loss of barrier integrity is mediated by cell-contact disassembly and actin remodeling. Studies into molecular mechanisms responsible for improving microvascular barrier function are therefore vital in the development of therapeutic targets for reducing vascular permeability in ARDS. The sweet taste receptor T1R3 is a G protein-coupled receptor, activated following exposure to sweet molecules, to trigger a gustducin-dependent signal cascade. In recent years, extraoral locations for T1R3 have been identified; however, no studies have focused on T1R3 within the vasculature. We hypothesize that activation of T1R3, in the pulmonary vasculature, plays a role in regulating endothelial barrier function in settings of ARDS. Our study demonstrated expression of T1R3 within the pulmonary vasculature, with a drop in expression levels following exposure to barrier-disruptive agents. Exposure of lung microvascular endothelial cells to the intensely sweet molecule sucralose attenuated LPS- and thrombin-induced endothelial barrier dysfunction. Likewise, sucralose exposure attenuated bacteria-induced lung edema formation in vivo. Inhibition of sweet taste signaling, through zinc sulfate, T1R3, or G-protein siRNA, blunted the protective effects of sucralose on the endothelium. Sucralose significantly reduced LPS-induced increased expression or phosphorylation of the key signaling molecules Src, p21-activated kinase (PAK), myosin light chain-2 (MLC2), heat shock protein 27 (HSP27), and p110α phosphatidylinositol 3-kinase (p110αPI3K). Activation of T1R3 by sucralose protects the pulmonary endothelium from edemagenic agent-induced barrier disruption, potentially through abrogation of Src/PAK/p110αPI3K-mediated cell-contact disassembly and Src/MLC2/HSP27-mediated actin remodeling. Identification of sweet taste sensing in the pulmonary vasculature may represent a novel therapeutic target to protect the endothelium in settings of ARDS.


Asunto(s)
Endotelio Vascular/efectos de los fármacos , Pulmón/efectos de los fármacos , Infecciones por Pseudomonas/microbiología , Receptores Acoplados a Proteínas G/metabolismo , Sacarosa/análogos & derivados , Edulcorantes/farmacología , Gusto/efectos de los fármacos , Animales , Permeabilidad Capilar , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Pulmón/citología , Pulmón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación , Pseudomonas aeruginosa/aislamiento & purificación , Transducción de Señal , Sacarosa/farmacología
7.
Am J Physiol Lung Cell Mol Physiol ; 312(5): L748-L759, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28258105

RESUMEN

Right ventricular (RV) dysfunction is associated with numerous smoking-related illnesses, including chronic obstructive pulmonary disease (COPD), in which it is present even in the absence of pulmonary hypertension. It is unknown whether exposure to cigarette smoke (CS) has direct effects on RV function and cardiac fibroblast (CF) proliferation or collagen synthesis. In this study, we evaluated cardiac function and fibrosis in mice exposed to CS and determined mechanisms of smoke-induced changes in CF signaling and fibrosis. AKR mice were exposed to CS for 6 wk followed by echocardiography and evaluation of cardiac hypertrophy, collagen content, and pulmonary muscularization. Proliferation and collagen content were evaluated in primary isolated rat CFs exposed to CS extract (CSE) or nicotine. Markers of cell proliferation, fibrosis, and proliferative signaling were determined by immunoblot or Sircol collagen assay. Mice exposed to CS had significantly decreased RV function, as determined by tricuspid annular plane systolic excursion. There were no changes in left ventricular parameters. RV collagen content was significantly elevated, but there was no change in RV hypertrophy or pulmonary vascular muscularization. CSE directly increased CF proliferation and collagen content in CF. Nicotine alone reproduced these effects. CSE and nicotine-induced fibroblast proliferation and collagen content were mediated through α7 nicotinic acetylcholine receptors and were dependent on PKC-α, PKC-δ, and reduced p38-MAPK phosphorylation. CS and nicotine have direct effects on CFs to induce proliferation and fibrosis, which may negatively affect right heart function.


Asunto(s)
Fibroblastos/metabolismo , Fibroblastos/patología , Ventrículos Cardíacos/patología , Miocardio/patología , Fumar/efectos adversos , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo , Animales , Proliferación Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/fisiopatología , Hemodinámica/efectos de los fármacos , Hipertrofia Ventricular Derecha/complicaciones , Hipertrofia Ventricular Derecha/diagnóstico por imagen , Hipertrofia Ventricular Derecha/patología , Hipertrofia Ventricular Derecha/fisiopatología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones Endogámicos AKR , Nicotina/farmacología , Fosforilación/efectos de los fármacos , Proteína Quinasa C-alfa/metabolismo , Proteína Quinasa C-delta/metabolismo , Ratas Sprague-Dawley , Remodelación Vascular/efectos de los fármacos , Disfunción Ventricular Derecha/complicaciones , Disfunción Ventricular Derecha/diagnóstico por imagen , Disfunción Ventricular Derecha/patología , Disfunción Ventricular Derecha/fisiopatología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
8.
Am J Respir Cell Mol Biol ; 54(6): 769-81, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26551054

RESUMEN

Pulmonary edema occurs in settings of acute lung injury, in diseases, such as pneumonia, and in acute respiratory distress syndrome. The lung interendothelial junctions are maintained in part by vascular endothelial (VE)-cadherin, an adherens junction protein, and its surface expression is regulated by endocytic trafficking. The Rab family of small GTPases are regulators of endocytic trafficking. The key trafficking pathways are regulated by Rab4, -7, and -9. Rab4 regulates the recycling of endosomes to the cell surface through a rapid-shuttle process, whereas Rab7 and -9 regulate trafficking to the late endosome/lysosome for degradation or from the trans-Golgi network to the late endosome, respectively. We recently demonstrated a role for the endosomal adaptor protein, p18, in regulation of the pulmonary endothelium through enhanced recycling of VE-cadherin to adherens junction. Thus, we hypothesized that Rab4, -7, and -9 regulate pulmonary endothelial barrier function through modulating trafficking of VE-cadherin-positive endosomes. We used Rab mutants with varying activities and associations to the endosome to study endothelial barrier function in vitro and in vivo. Our study demonstrates a key role for Rab4 activation and Rab9 inhibition in regulation of vascular permeability through enhanced VE-cadherin expression at the interendothelial junction. We further showed that endothelial barrier function mediated through Rab4 is dependent on extracellular signal-regulated kinase phosphorylation and activity. Thus, we demonstrate that Rab4 and -9 regulate VE-cadherin levels at the cell surface to modulate the pulmonary endothelium through extracellular signal-regulated kinase-dependent and -independent pathways, respectively. We propose that regulating select Rab GTPases represents novel therapeutic strategies for patients suffering with acute respiratory distress syndrome.


Asunto(s)
Antígenos CD/metabolismo , Cadherinas/metabolismo , Endosomas/metabolismo , Endotelio/metabolismo , Pulmón/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Animales , Permeabilidad de la Membrana Celular/efectos de los fármacos , Endocitosis/efectos de los fármacos , Endosomas/efectos de los fármacos , Endotelio/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Lipopolisacáridos/farmacología , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Transporte de Proteínas/efectos de los fármacos , Pseudomonas aeruginosa/efectos de los fármacos , Ratas
9.
FASEB J ; 29(3): 868-81, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25404710

RESUMEN

Vascular permeability is a hallmark of several disease states including acute lung injury (ALI). Endocytosis of VE-cadherin, away from the interendothelial junction (IEJ), causes acute endothelial barrier permeability. A novel protein, p18, anchors to the endosome membrane and plays a role in late endosomal signaling via MAPK and mammalian target of rapamycin. However, the fate of the VE-cadherin-positive endosome has yet to be elucidated. We sought to elucidate a role for p18 in VE-cadherin trafficking and thus endothelial barrier function, in settings of ALI. Endothelial cell (EC) resistance, whole-cell ELISA, and filtration coefficient were studied in mice or lung ECs overexpressing wild-type or nonendosomal-binding mutant p18, using green fluorescent protein as a control. We demonstrate a protective role for the endocytic protein p18 in endothelial barrier function in settings of ALI in vitro and in vivo, through enhanced recycling of VE-cadherin-positive early endosomes to the IEJ. In settings of LPS-induced ALI, we show that Src tethered to the endosome tyrosine phosphorylates p18 concomitantly with VE-cadherin internalization and pulmonary edema formation. We conclude that p18 regulates pulmonary endothelial barrier function in vitro and in vivo, by enhancing recycling of VE-cadherin-positive endosomes to the IEJ.


Asunto(s)
Lesión Pulmonar Aguda/patología , Antígenos CD/metabolismo , Cadherinas/metabolismo , Permeabilidad Capilar/efectos de los fármacos , Endocitosis/efectos de los fármacos , Endotelio Vascular/patología , Nucleósido Difosfato Quinasas NM23/metabolismo , Edema Pulmonar/patología , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/metabolismo , Uniones Adherentes , Animales , Western Blotting , Células Cultivadas , Endosomas/efectos de los fármacos , Endosomas/metabolismo , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Ensayo de Inmunoadsorción Enzimática , Técnicas para Inmunoenzimas , Inmunoprecipitación , Lipopolisacáridos/toxicidad , Ratones , Ratones Endogámicos C57BL , Fosforilación/efectos de los fármacos , Edema Pulmonar/inducido químicamente , Edema Pulmonar/metabolismo , Transducción de Señal , Tirosina/metabolismo , Familia-src Quinasas/metabolismo
10.
Exp Physiol ; 101(6): 731-42, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27164183

RESUMEN

What is the central question of this study? Although SGLT2 inhibitors represent a promising treatment for patients suffering from diabetic nephropathy, the influence of metabolic disruption on the expression and function of glucose transporters is largely unknown. What is the main finding and its importance? In vivo models of metabolic disruption (Goto-Kakizaki type II diabetic rat and junk-food diet) demonstrate increased expression of SGLT1, SGLT2 and GLUT2 in the proximal tubule brush border. In the type II diabetic model, this is accompanied by increased SGLT- and GLUT-mediated glucose uptake. A fasted model of metabolic disruption (high-fat diet) demonstrated increased GLUT2 expression only. The differential alterations of glucose transporters in response to varying metabolic stress offer insight into the therapeutic value of inhibitors. SGLT2 inhibitors are now in clinical use to reduce hyperglycaemia in type II diabetes. However, renal glucose reabsorption across the brush border membrane (BBM) is not completely understood in diabetes. Increased consumption of a Western diet is strongly linked to type II diabetes. This study aimed to investigate the adaptations that occur in renal glucose transporters in response to experimental models of diet-induced insulin resistance. The study used Goto-Kakizaki type II diabetic rats and normal rats rendered insulin resistant using junk-food or high-fat diets. Levels of protein kinase C-ßI (PKC-ßI), GLUT2, SGLT1 and SGLT2 were determined by Western blotting of purified renal BBM. GLUT- and SGLT-mediated d-[(3) H]glucose uptake by BBM vesicles was measured in the presence and absence of the SGLT inhibitor phlorizin. GLUT- and SGLT-mediated glucose transport was elevated in type II diabetic rats, accompanied by increased expression of GLUT2, its upstream regulator PKC-ßI and SGLT1 protein. Junk-food and high-fat diet feeding also caused higher membrane expression of GLUT2 and its upstream regulator PKC-ßI. However, the junk-food diet also increased SGLT1 and SGLT2 levels at the proximal tubule BBM. Glucose reabsorption across the proximal tubule BBM, via GLUT2, SGLT1 and SGLT2, is not solely dependent on glycaemic status, but is also influenced by diet-induced changes in glucose metabolism. We conclude that different metabolic disturbances result in complex adaptations in renal glucose transporter protein levels and function.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Túbulos Renales Proximales/metabolismo , Membranas/metabolismo , Animales , Glucemia/metabolismo , Dieta Alta en Grasa/efectos adversos , Glucosa/metabolismo , Transportador de Glucosa de Tipo 2/metabolismo , Hiperglucemia/metabolismo , Resistencia a la Insulina/fisiología , Riñón/metabolismo , Masculino , Proteína Quinasa C/metabolismo , Ratas , Ratas Sprague-Dawley , Transportador 1 de Sodio-Glucosa/metabolismo , Transportador 2 de Sodio-Glucosa/metabolismo
11.
Am J Respir Cell Mol Biol ; 52(6): 695-707, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25317600

RESUMEN

Enhanced protein tyrosine phosphorylation is associated with changes in vascular permeability through formation and dissolution of adherens junctions and regulation of stress fiber formation. Inhibition of the protein tyrosine phosphorylase SH2 domain-containing protein tyrosine phosphatase 2 (SHP2) increases tyrosine phosphorylation of vascular endothelial cadherin and ß-catenin, resulting in disruption of the endothelial monolayer and edema formation in the pulmonary endothelium. Vascular permeability is a hallmark of acute lung injury (ALI); thus, enhanced SHP2 activity offers potential therapeutic value for the pulmonary vasculature in diseases such as ALI, but this has not been characterized. To assess whether SHP2 activity mediates protection against edema in the endothelium, we assessed the effect of molecular activation of SHP2 on lung endothelial barrier function in response to the edemagenic agents LPS and thrombin. Both LPS and thrombin reduced SHP2 activity, correlated with decreased focal adhesion kinase (FAK) phosphorylation (Y(397) and Y(925)) and diminished SHP2 protein-protein associations with FAK. Overexpression of constitutively active SHP2 (SHP2(D61A)) enhanced baseline endothelial monolayer resistance and completely blocked LPS- and thrombin-induced permeability in vitro and significantly blunted pulmonary edema formation induced by either endotoxin (LPS) or Pseudomonas aeruginosa exposure in vivo. Chemical inhibition of FAK decreased SHP2 protein-protein interactions with FAK concomitant with increased permeability; however, overexpression of SHP2(D61A) rescued the endothelium and maintained FAK activity and FAK-SHP2 protein interactions. Our data suggest that SHP2 activation offers the pulmonary endothelium protection against barrier permeability mediators downstream of the FAK signaling pathway. We postulate that further studies into the promotion of SHP2 activation in the pulmonary endothelium may offer a therapeutic approach for patients suffering from ALI.


Asunto(s)
Células Endoteliales/enzimología , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Lesión Pulmonar Aguda/enzimología , Lesión Pulmonar Aguda/inmunología , Animales , Permeabilidad Capilar , Células Cultivadas , Células Endoteliales/inmunología , Endotelio Vascular/patología , Lipopolisacáridos/farmacología , Pulmón/irrigación sanguínea , Ratones Endogámicos C57BL , Microvasos/patología , Fosforilación , Mapas de Interacción de Proteínas , Procesamiento Proteico-Postraduccional , Ratas , Flujo Sanguíneo Regional
12.
Am J Physiol Lung Cell Mol Physiol ; 309(7): L700-9, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26254426

RESUMEN

Neovascularization, the formation of new blood vessels, requires multiple processes including vascular leak, migration, and adhesion. Endosomal proteins, such as Rabs, regulate trafficking of key signaling proteins involved in neovascularization. The novel endosome protein, p18, enhances vascular endothelial (VE)-cadherin recycling from early endosome to cell junction to improve pulmonary endothelial barrier function. Since endothelial barrier integrity is vital in neovascularization, we sought to elucidate the role for endosome proteins p18 and Rab4, Rab7, and Rab9 in the process of vessel formation within the pulmonary vasculature. Overexpression of wild-type p18 (p18(wt)), but not the nonendosomal-binding mutant (p18(N39)), significantly increased lung microvascular endothelial cell migration, adhesion, and both in vitro and in vivo tube formation. Chemical inhibition of mTOR or p38 attenuated the proneovascularization role of p18(wt). Similar to the effect of p18(wt), overexpression of prorecycling wild-type (Rab4(WT)) and endosome-anchored (Rab4(Q67L)) Rab4 enhanced neovascularization processes, whereas molecular inhibition of Rab4, by using the nonendosomal-binding mutant (Rab4(S22N)) attenuated VEGF-induced neovascularization. Unlike p18, Rab4-induced neovascularization was independent of mTOR or p38 inhibition but was dependent on p18 expression. This study shows for the first time that neovascularization within the pulmonary vasculature is dependent on the prorecycling endocytic proteins Rab4 and p18.


Asunto(s)
Proteínas Portadoras/metabolismo , Endosomas/metabolismo , Endotelio/metabolismo , Pulmón/metabolismo , Neovascularización Fisiológica/fisiología , Proteínas de Unión al GTP rab4/metabolismo , Animales , Proteínas Portadoras/genética , Endosomas/genética , Endotelio/citología , Mutación , Ratas , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteínas de Unión al GTP rab4/genética
13.
Am J Physiol Lung Cell Mol Physiol ; 308(8): L827-36, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25659900

RESUMEN

Pulmonary hypertension (PH) eventually leads to right ventricular (RV) fibrosis and dysfunction that is associated with increased morbidity and mortality. Although angiotensin II plays an important role in RV remodeling associated with hypoxic PH, the molecular mechanisms underlying RV fibrosis in PH largely remain unresolved. We hypothesized that PKC-p38 signaling is involved in RV collagen accumulation in PH and in response to angiotensin II stimulation. Adult male Sprague-Dawley rats were exposed to 3 wk of normoxia or hypoxia (10% FiO2 ) as a model of PH. Hypoxic rats developed RV hypertrophy and fibrosis associated with an increase in PKC ßII and δ protein expression and p38 dephosphorylation in freshly isolated RV cardiac fibroblasts. Further mechanistic studies were performed in cultured primary cardiac fibroblasts stimulated with angiotensin II, a key activator of ventricular fibrosis in PH. Angiotensin II induced a reduction in p38 phosphorylation that was attenuated following chemical inhibition of PKC ßII and δ. Molecular and chemical inhibition of PKC ßII and δ abrogated angiotensin II-induced cardiac fibroblast proliferation and collagen deposition in vitro. The effects of PKC inhibition on proliferation and fibrosis were reversed by chemical inhibition of p38. Conversely, constitutive activation of p38 attenuated angiotensin II-induced increase of cardiac fibroblast proliferation and collagen accumulation. PKC ßII- and δ-dependent inactivation of p38 regulates cardiac fibroblast proliferation and collagen deposition in response to angiotensin II, which suggests that the PKC-p38 signaling in cardiac fibroblasts may be involved and important in the pathophysiology of RV fibrosis in PH.


Asunto(s)
Angiotensina II/fisiología , Hipertensión Pulmonar/enzimología , Hipertrofia Ventricular Derecha/enzimología , Proteína Quinasa C beta/fisiología , Proteína Quinasa C-delta/fisiología , Animales , Hipoxia de la Célula , Proliferación Celular , Células Cultivadas , Colágeno/metabolismo , Activación Enzimática , Fibroblastos/enzimología , Fibrosis , Ventrículos Cardíacos/patología , Hipertensión Pulmonar/complicaciones , Masculino , Ratas Sprague-Dawley , Disfunción Ventricular Derecha/enzimología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
14.
Front Nutr ; 11: 1366409, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38721028

RESUMEN

Introduction: Recent studies have indicated considerable health risks associated with the consumption of artificial sweeteners. Neotame is a relatively new sweetener in the global market however there is still limited data on the impact of neotame on the intestinal epithelium or the commensal microbiota. Methods: In the present study, we use a model of the intestinal epithelium (Caco-2) and microbiota (Escherichia coli and Enterococcus faecalis) to investigate how physiologically-relevant exposure of neotame impacts intestinal epithelial cell function, gut bacterial metabolism and pathogenicity, and gut epithelium-microbiota interactions. Results: Our findings show that neotame causes intestinal epithelial cell apoptosis and death with siRNA knockdown of T1R3 expression significantly attenuating the neotame-induced loss to cell viability. Similarly, neotame exposure results in barrier disruption with enhanced monolayer leak and reduced claudin-3 cell surface expression through a T1R3-dependent pathway. Using the gut bacteria models, E. coli and E. faecalis, neotame significantly increased biofilm formation and metabolites of E. coli, but not E. faecalis, reduced Caco-2 cell viability. In co-culture studies, neotame exposure increased adhesion capacity of E. coli and E. faecalis onto Caco-2 cells and invasion capacity of E. coli. Neotame-induced biofilm formation, E.coli-specific Caco-2 cell death, adhesion and invasion was identified to be meditated through a taste-dependent pathway. Discussion: Our study identifies novel pathogenic effects of neotame on the intestinal epithelium or bacteria alone, and in co-cultures to mimic the gut microbiome. These findings demonstrate the need to better understand food additives common in the global market and the molecular mechanisms underlying potential negative health impacts.

15.
Nephrol Dial Transplant ; 28(4): 839-45, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23222417

RESUMEN

BACKGROUND: Proteases can increase the activity of the epithelial sodium channel (ENaC) by cleaving its α- or γ-subunit. However, evidence so far comes only from studies in vitro in either heterologous expression systems or isolated nephron segments. The present study has tested whether exposure to a luminal protease can alter sodium reabsorption along the rat collecting duct in vivo. METHODS: Rats on normal laboratory chow were prepared for renal micropuncture. Late distal tubules of superficial nephrons were microinjected and perfused twice (3 nL min(-1) for 3-6 min) with a solution similar to native tubular fluid, but containing (14)[C]inulin and (22)Na. The first perfusion was either a control solution or solution containing amiloride 1 mM or hydrochlorothiazide (HCTZ ) 1 mM; the second perfusion was either a control solution (time control) or a solution containing chymotrypsin 2 µg mL(-1) ± aprotinin 100 µg mL(-1) or amiloride 1 mM or HCTZ 1 mM. Urinary recoveries of (14)[C]inulin and (22)Na were recorded. RESULTS: In time controls, the Na/In ratio did not change significantly (32.2 ± 3.4% versus 34.5 ± 3.1%). In contrast, chymotrypsin reduced the ratio from 33.3 ± 3.8% to 25.5 ± 2.5% (P < 0.05), indicating an increase in sodium reabsorption. When co-injected with chymotrypsin, the protease inhibitor aprotinin abolished the stimulatory effect of chymotrypsin on sodium reabsorption (31.7 ± 3.4% versus 32.1 ± 2.1%), while aprotinin alone had no effect. When chymotrypsin was co-injected with HCTZ, the Na/In ratio decreased from 36.8 ± 2.3% to 28.0 ± 3.4% (P < 0.05), whereas when given with amiloride, there was no change in the ratio (45.8 ± 3.4% versus 45.5 ± 2.3%), indicating that stimulation of sodium reabsorption by chymotrypsin was ENaC-dependent. CONCLUSIONS: These findings demonstrate proteolytic activation of ENaC in vivo, and suggest that changes in protease activity of the glomerular filtrate and tubular fluid in health or disease could affect net renal sodium excretion.


Asunto(s)
Canales Epiteliales de Sodio/metabolismo , Túbulos Renales Colectores/efectos de los fármacos , Túbulos Renales Colectores/metabolismo , Péptido Hidrolasas/metabolismo , Sodio/metabolismo , Amilorida/farmacología , Animales , Bloqueadores del Canal de Sodio Epitelial/farmacología , Canales Epiteliales de Sodio/química , Transporte Iónico , Masculino , Nefronas/efectos de los fármacos , Nefronas/metabolismo , Ratas , Ratas Sprague-Dawley , Sodio/administración & dosificación
16.
PLoS One ; 18(12): e0295272, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38091359

RESUMEN

BACKGROUND: As with many countries around the world, the incidence of diabetes in Bangladesh is increasing significantly. Whilst there is controversy in the field regarding the health impact of artificial sweeteners in Western communities, the link between sweetener consumption and awareness in Bangladesh has not been established. METHODS: In the present study, 260 diabetic patients completed a questionnaire survey to investigate the use and awareness of sweeteners and how this links to demographics and potential co-morbidities. RESULTS: Findings show that daily artificial sweetener consumption is significantly associated with hypertension but not other co-morbidities such as kidney disease or obesity. We further demonstrate that there is limited checking of artificial sweeteners in food or drink products by participants. the rurality of diabetic participants was found to significantly correlates with lower awareness of any health impact of artificial sweeteners. CONCLUSIONS: The findings from this study demonstrate that there is a need to increase the awareness of artificial sweetener use in diabetic patients in Bangladesh. Combined with a more robust understanding of the health impact of artificial sweeteners, these findings suggest that there is potential to improve outcomes for diabetic patients by improving this awareness.


Asunto(s)
Diabetes Mellitus , Edulcorantes , Humanos , Edulcorantes/efectos adversos , Bangladesh/epidemiología , Diabetes Mellitus/epidemiología , Diabetes Mellitus/inducido químicamente , Obesidad/epidemiología , Encuestas y Cuestionarios
17.
Pharmaceutics ; 16(1)2023 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-38258076

RESUMEN

Zinc diethyldithiocarbamate (Zn (DDC)2), a disulfiram metabolite (anti-alcoholism drug), has shown a strong anti-cancer activity in vitro. However, its application was limited by its low aqueous solubility and rapid metabolism. In this study, the solubility enhancement of Zn (DDC)2 is investigated by forming inclusion complexes with cyclodextrins. The inclusion complexes were prepared using two different types of beta-cyclodextrins, SBE-CD and HP-CD. Phase solubility diagrams for the resulting solutions were assessed; subsequently, the solutions were freeze-dried for further characterisation studies using DSC, TGA, XRD, and FTIR. The cytotoxic activity of the produced inclusion complexes was evaluated on human lung carcinoma cells using the MTT assay. The solubility of Zn (DDC)2 increased significantly upon adding beta-cyclodextrins, reaching approximately 4 mg/mL for 20% w/w CD solutions. The phase solubility diagram of Zn (DDC)2 was of the Ap-type according to the Higuchi and Connors model. Characterisation studies confirmed the inclusion of the amorphous drug in the CD-Zn (DDC)2 complexes. The cytotoxicity of Zn (DDC)2 was enhanced 10-fold by the inclusion complexes compared to the free drug. Overall, the resulting CD-Zn (DDC)2 inclusion complexes have a potential for treatment against lung cancer.

18.
Nutrients ; 15(2)2023 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-36678293

RESUMEN

There is current scientific interest in naturally sourced phenolic compounds and their potential benefits to health, as well as the effective role polyphenols may provide in an exercise setting. This study investigated the chronic effects of supplementation with a biodynamic and organic olive fruit water phytocomplex (OliPhenolia® [OliP]), rich in hydroxytyrosol (HT), on submaximal and exhaustive exercise performance and respiratory markers of recovery. Twenty-nine recreationally active participants (42 ± 2 yrs; 71.1 ± 2.1 kg; 1.76 ± 0.02 m) consumed 2 × 28 mL∙d−1 of OliP or a taste- and appearance-matched placebo (PL) over 16 consecutive days. Participants completed a demanding, aerobic exercise protocol at ~75% maximal oxygen uptake (V˙O2max) for 65 min 24 h before sub- and maximal performance exercise tests prior to and following the 16-day consumption period. OliP reduced the time constant (τ) (p = 0.005) at the onset of exercise, running economy (p = 0.015) at lactate threshold 1 (LT1), as well as the rating of perceived exertion (p = 0.003) at lactate turnpoint (LT2). Additionally, OliP led to modest improvements in acute recovery based upon a shorter time to achieve 50% of the end of exercise V˙O2 value (p = 0.02). Whilst OliP increased time to exhaustion (+4.1 ± 1.8%), this was not significantly different to PL (p > 0.05). Phenolic compounds present in OliP, including HT and related metabolites, may provide benefits for aerobic exercise and acute recovery in recreationally active individuals. Further research is needed to determine whether dose-response or adjunct use of OliP alongside longer-term training programs can further modulate exercise-associated adaptations in recreationally active individuals, or indeed support athletic performance.


Asunto(s)
Olea , Humanos , Ejercicio Físico/fisiología , Prueba de Esfuerzo , Ácido Láctico , Consumo de Oxígeno/fisiología
19.
Front Nutr ; 10: 1262044, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38144428

RESUMEN

Dietary protein is crucial for optimising physical training adaptations such as muscular strength and mass, which are key aims for athletic populations, including British Army recruits. New recruits fail to meet the recommended protein intake during basic training (BT), with negligible amounts consumed in the evening. This study assessed the influence of a daily bolus of protein prior to sleep on performance adaptations, body composition and recovery in British Army recruits. 99 men and 23 women [mean ± standard deviation (SD): age: 21.3 ± 3.5 years, height: 174.8 ± 8.4 cm, body mass 75.4 ± 12.2 kg] were randomised into a dietary control (CON), carbohydrate placebo (PLA), moderate (20 g) protein (MOD) or high (60 g) protein (HIGH) supplementation group. Supplements were isocaloric and were consumed on weekday evenings between 2000 and 2100 for 12 weeks during BT. Performance tests (mid-thigh pull, medicine ball throw, 2 km run time, maximal push-up, and maximal vertical jump) and body composition were assessed at the start and end of BT. Dietary intake, energy expenditure, salivary hormones, urinary nitrogen balance, perceived muscle soreness, rating of perceived exertion, mood, and fatigue were assessed at the start, middle and end of BT. Protein supplementation increased protein intake in HIGH (2.16 ± 0.50 g⸱kg-1⸱day-1) and MOD (1.71 ± 0.48 g⸱kg-1⸱day-1) compared to CON (1.17 ± 0.24 g⸱kg-1⸱day-1) and PLA (1.31 ± 0.29 g⸱kg-1⸱day-1; p < 0.001). Despite this, there was no impact of supplementation on mid-thigh pull performance (CON = 7 ± 19%, PLA = 7 ± 19%, MOD = 0 ± 16%, and HIGH = 4 ± 14%; p = 0.554) or any other performance measures (p > 0.05). Fat-free mass changes were also similar between groups (CON = 4 ± 3%, PLA = 4 ± 4%, MOD = 3 ± 3%, HIGH = 5 ± 4%, p = 0.959). There was no impact of protein supplementation on any other body composition or recovery measure. We conclude no benefits of pre-bed protein supplementation to improve performance, body composition and recovery during BT. It is possible the training stimulus was great enough, limiting the impact of protein supplementation. However, the high degree of inter-participant variability suggests an individualised use of protein supplementation should be explored, particularly in those who consume sub-optimal (<1.6 g⸱kg-1⸱day-1) habitual amounts of protein. Clinical trial registration: The study was registered with ClinicalTrials.gov, U.S. national institutes (identifier: NCT05998590).

20.
Front Nutr ; 10: 1264042, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38130446

RESUMEN

British Army basic training (BT) is physically demanding with new recruits completing multiple bouts of physical activity each day with limited recovery. Load carriage is one of the most physically demanding BT activities and has been shown to induce acute exercise-induced muscle damage (EIMD) and impair muscle function. Protein supplementation can accelerate muscle recovery by attenuating EIMD and muscle function loss. This study investigated the impact of an additional daily bolus of protein prior to sleep throughout training on acute muscle recovery following a load carriage test in British Army recruits. Ninety nine men and 23 women (mean ± SD: age: 21.3 ± 3.5 yrs., height: 174.8 ± 8.4 cm, body mass 75.4 ± 12.2 kg) were randomized to dietary control (CON), carbohydrate placebo (PLA), moderate (20 g; MOD) or high (60 g; HIGH) protein supplementation. Muscle function (maximal jump height), perceived muscle soreness and urinary markers of muscle damage were assessed before (PRE), immediately post (POST), 24-h post (24 h-POST) and 40-h post (40 h-POST) a load carriage test. There was no impact of supplementation on muscle function at POST (p = 0.752) or 40 h-POST (p = 0.989) load carriage but jump height was greater in PLA compared to HIGH at 24 h-POST (p = 0.037). There was no impact of protein supplementation on muscle soreness POST (p = 0.605), 24 h-POST (p = 0.182) or 40 h-POST (p = 0.333). All groups had increased concentrations of urinary myoglobin and 3-methylhistidine, but there was no statistical difference between groups at any timepoint (p > 0.05). We conclude that pre-sleep protein supplementation does not accelerate acute muscle recovery following load carriage in British Army recruits during basic training. The data suggests that consuming additional energy in the form of CHO or protein was beneficial at attenuating EIMD, although it is acknowledged there were no statistical differences between groups. Although EIMD did occur as indicated by elevated urinary muscle damage markers, it is likely that the load carriage test was not arduous enough to reduce muscle function, limiting the impact of protein supplementation. Practically, protein supplementation above protein intakes of 1.2 g⸱kg-1⸱day-1 following load carriage over similar distances (4 km) and carrying similar loads (15-20 kg) does not appear to be warranted.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA