Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
Front Pharmacol ; 10: 715, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31417401

RESUMEN

Epithelial-to-mesenchymal transition (EMT) is a self-regulated physiological process required for tissue repair that, in non-controled conditions may lead to fibrosis, angiogenesis, loss of normal organ function or cancer. Although several molecular pathways involved in EMT regulation have been described, this process does not have any specific treatment. This article introduces a systematic review of effective natural plant compounds and their extract that modulates the pathological EMT or its deleterious effects, through acting on different cellular signal transduction pathways both in vivo and in vitro. Thereby, cryptotanshinone, resveratrol, oxymatrine, ligustrazine, osthole, codonolactone, betanin, tannic acid, gentiopicroside, curcumin, genistein, paeoniflorin, gambogic acid and Cinnamomum cassia extracts inhibit EMT acting on transforming growth factor-ß (TGF-ß)/Smads signaling pathways. Gedunin, carnosol, celastrol, black rice anthocyanins, Duchesnea indica, cordycepin and Celastrus orbiculatus extract downregulate vimectin, fibronectin and N-cadherin. Sulforaphane, luteolin, celastrol, curcumin, arctigenin inhibit ß-catenin signaling pathways. Salvianolic acid-A and plumbagin block oxidative stress, while honokiol, gallic acid, piperlongumine, brusatol and paeoniflorin inhibit EMT transcription factors such as SNAIL, TWIST and ZEB. Plectranthoic acid, resveratrol, genistein, baicalin, polyphyllin I, cairicoside E, luteolin, berberine, nimbolide, curcumin, withaferin-A, jatrophone, ginsenoside-Rb1, honokiol, parthenolide, phoyunnanin-E, epicatechin-3-gallate, gigantol, eupatolide, baicalin and baicalein and nitidine chloride inhibit EMT acting on other signaling pathways (SIRT1, p38 MAPK, NFAT1, SMAD, IL-6, STAT3, AQP5, notch 1, PI3K/Akt, Wnt/ß-catenin, NF-κB, FAK/AKT, Hh). Despite the huge amount of preclinical data regarding EMT modulation by the natural compounds of plant, clinical translation is poor. Additionally, this review highlights some relevant examples of clinical trials using natural plant compounds to modulate EMT and its deleterious effects. Overall, this opens up new therapeutic alternatives in cancer, inflammatory and fibrosing diseases through the control of EMT process.

2.
Front Physiol ; 10: 630, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31191339

RESUMEN

Background: Appetite disorders are frequent and scantly studied in peritoneal dialysis (PD) patients and are associated with malnutrition and cardiovascular complications. Objective: We investigated the relationship between uremic insulin resistance, pro-inflammatory cytokines, and appetite-related peptides release (ARPr) with eating-behavior disorders in PD patients. Methods: We included 42 PD patients (12 suffering anorexia, 12 obese with high food-intake, and 18 asymptomatic) and 10 controls. We measured blood levels of ARPr including orexigens [neuropeptide-Y (NPY), ghrelin, and nitric-oxide], anorexigens [cholecystokinin, insulin, corticotropin-releasing factor, leptin, and adiponectin (Ad)], and cytokines (TNF-α, sTNFα-R2, and IL-6) both at baseline and after administering a standard-food stimulus (SFS). We also measured the expression of TNF-α, leptin and Ad-encoding mRNAs in abdominal adipose tissue. We compared these markers with eating motivation measured by a Visual Analog Scale (VAS). Results: Anorexics showed both little appetite, measured by a VAS, and low levels of orexigens that remained constant after SFS, coupled with high levels of anorexigens at baseline and after SFS. Obeses showed higher appetite, increased baseline levels of orexigens, lower baseline levels of anorexigens and cytokines and two peaks of NPY after SFS. The different patterns of ARPr and cytokines pointed to a close relationship with uremic insulin resistance. In fact, the euglycemic-hyperglycemic clamp reproduced these disorders. In anorexics, TNF-α fat expression was increased. In obese patients, leptin expression in fat tissue was down-regulated and showed correlation with the appetite. Conclusion: In PD, appetite is governed by substances that are altered at baseline and abnormally released. Such modulators are controlled by insulin metabolism and cytokines and, while anorexics display inflammatory predominance, obese patients predominantly display insulin resistance.

4.
Sci Rep ; 7: 44941, 2017 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-28327551

RESUMEN

Peritoneal dialysis (PD) is an effective renal replacement therapy, but a significant proportion of patients suffer PD-related complications, which limit the treatment duration. Mesothelial-to-mesenchymal transition (MMT) contributes to the PD-related peritoneal dysfunction. We analyzed the genetic reprograming of MMT to identify new biomarkers that may be tested in PD-patients. Microarray analysis revealed a partial overlapping between MMT induced in vitro and ex vivo in effluent-derived mesothelial cells, and that MMT is mainly a repression process being higher the number of genes that are down-regulated than those that are induced. Cellular morphology and number of altered genes showed that MMT ex vivo could be subdivided into two stages: early/epithelioid and advanced/non-epithelioid. RT-PCR array analysis demonstrated that a number of genes differentially expressed in effluent-derived non-epithelioid cells also showed significant differential expression when comparing standard versus low-GDP PD fluids. Thrombospondin-1 (TSP1), collagen-13 (COL13), vascular endothelial growth factor A (VEGFA), and gremlin-1 (GREM1) were measured in PD effluents, and except GREM1, showed significant differences between early and advanced stages of MMT, and their expression was associated with a high peritoneal transport status. The results establish a proof of concept about the feasibility of measuring MMT-associated secreted protein levels as potential biomarkers in PD.


Asunto(s)
Reprogramación Celular/genética , Células Epiteliales/metabolismo , Células Epiteliales/patología , Transición Epitelial-Mesenquimal/genética , Genómica , Diálisis Peritoneal , Biomarcadores , Soluciones para Diálisis/química , Perfilación de la Expresión Génica , Genómica/métodos , Glucólisis , Humanos , Diálisis Peritoneal/efectos adversos , Transcriptoma
5.
J Am Soc Nephrol ; 27(12): 3561-3576, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27151919

RESUMEN

Patients with ESRD undergoing peritoneal dialysis develop progressive peritoneal fibrosis, which may lead to technique failure. Recent data point to Th17-mediated inflammation as a key contributor in peritoneal damage. The leukocyte antigen CD69 modulates the setting and progression of autoimmune and inflammatory diseases by controlling the balance between Th17 and regulatory T cells (Tregs). However, the relevance of CD69 in tissue fibrosis remains largely unknown. Thus, we explored the role of CD69 in fibroproliferative responses using a mouse model of peritoneal fibrosis induced by dialysis fluid exposure under either normal or uremic status. We found that cd69-/- mice compared with wild-type (WT) mice showed enhanced fibrosis, mesothelial to mesenchymal transition, IL-17 production, and Th17 cell infiltration in response to dialysis fluid treatment. Uremia contributed partially to peritoneal inflammatory and fibrotic responses. Additionally, antibody-mediated CD69 blockade in WT mice mimicked the fibrotic response of cd69-/- mice. Finally, IL-17 blockade in cd69-/- mice decreased peritoneal fibrosis to the WT levels, and mixed bone marrow from cd69-/- and Rag2-/-γc-/- mice transplanted into WT mice reproduced the severity of the response to dialysis fluid observed in cd69-/- mice, showing that CD69 exerts its regulatory function within the lymphocyte compartment. Overall, our results indicate that CD69 controls tissue fibrosis by regulating Th17-mediated inflammation.


Asunto(s)
Antígenos CD/inmunología , Antígenos de Diferenciación de Linfocitos T/inmunología , Lectinas Tipo C/inmunología , Fibrosis Peritoneal/inmunología , Animales , Antígenos CD/fisiología , Antígenos de Diferenciación de Linfocitos T/fisiología , Femenino , Lectinas Tipo C/deficiencia , Lectinas Tipo C/fisiología , Ratones , Células Th17/fisiología
6.
Nefrologia ; 36(4): 368-75, 2016.
Artículo en Inglés, Español | MEDLINE | ID: mdl-27118192

RESUMEN

Cardiovascular factors are one of the main causes of morbidity and mortality in patients with chronic kidney disease. Bone mineral metabolism disorders and inflammation are pathological conditions that involve increased cardiovascular risk in chronic kidney disease. The cardiovascular risk involvement of bone mineral metabolism classical biochemical parameters such as phosphorus, calcium, vitamin D and PTH is well known. The newest markers, FGF23 and klotho, could also be implicated in cardiovascular disease.


Asunto(s)
Trastorno Mineral y Óseo Asociado a la Enfermedad Renal Crónica , Animales , Biomarcadores , Huesos/metabolismo , Calcineurina/fisiología , Enfermedades Cardiovasculares/etiología , Trastorno Mineral y Óseo Asociado a la Enfermedad Renal Crónica/complicaciones , Trastorno Mineral y Óseo Asociado a la Enfermedad Renal Crónica/metabolismo , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/fisiología , Glucuronidasa/fisiología , Humanos , Proteínas Klotho , Ratones , Minerales/metabolismo , Modelos Biológicos , Hormona Paratiroidea/fisiología , Fósforo/metabolismo , Insuficiencia Renal Crónica/complicaciones , Insuficiencia Renal Crónica/metabolismo , Factores de Riesgo , Calcificación Vascular/etiología , Calcificación Vascular/prevención & control , Vitamina D/metabolismo
7.
Oncotarget ; 7(21): 30133-46, 2016 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-27102153

RESUMEN

UNLABELLED: Peritoneal dialysis (PD) is a form of renal replacement treatment, which employs the peritoneal membrane (PM) to eliminate toxins that cannot be removed by the kidney. The procedure itself, however, contributes to the loss of the PM ultrafiltration capacity (UFC), leading consequently to the technique malfunction. ß-blockers have been considered deleterious for PM due to their association with loss of UFC and induction of fibrosis. Herein we analyzed the effects of Nebivolol, a new generation of ß1-blocker, on PM alterations induced by PD fluids (PDF).In vitro: We found that mesothelial cells (MCs) express ß1-adrenergic receptor. MCs were treated with TGF-ß to induce mesothelial-to-mesenchymal transition (MMT) and co-treated with Nebivolol. Nebivolol reversed the TGF-ß effects, decreasing extracellular matrix synthesis, and improved the fibrinolytic capacity, decreasing plasminogen activator inhibitor-1 (PAI-1) and increasing tissue-type plasminogen activator (tPA) supernatant levels. Moreover, Nebivolol partially inhibited MMT and decreased vascular endothelial growth factor (VEGF) and IL-6 levels in supernatants.In vivo: Twenty-one C57BL/6 mice were divided into 3 groups. Control group carried a catheter without PDF infusion. Study group received intraperitoneally PDF and oral Nebivolol during 30 days. PDF group received PDF alone. Nebivolol maintained the UFC and reduced PM thickness, MMT and angiogenesis promoted by PDF. It also improved the fibrinolytic capacity in PD effluents decreasing PAI-1 and IL-8 and increased tPA levels. CONCLUSION: Nebivolol protects PM from PDF-induced damage, promoting anti-fibrotic, anti-angiogenic, anti-inflammatory and pro-fibrinolytic effects.


Asunto(s)
Agonistas de Receptores Adrenérgicos beta 1/farmacología , Soluciones para Diálisis/efectos adversos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Nebivolol/farmacología , Diálisis Peritoneal/efectos adversos , Peritoneo/efectos de los fármacos , Peritoneo/patología , Agonistas de Receptores Adrenérgicos beta 1/uso terapéutico , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Femenino , Fibrinólisis/efectos de los fármacos , Fibrosis , Humanos , Interleucina-8/metabolismo , Ratones , Ratones Endogámicos C57BL , Nebivolol/uso terapéutico , Neovascularización Patológica/inducido químicamente , Neovascularización Patológica/tratamiento farmacológico , Peritoneo/citología , Inhibidor 1 de Activador Plasminogénico/metabolismo , Receptores Adrenérgicos beta 1/metabolismo , Serpina E2/metabolismo , Activador de Tejido Plasminógeno/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Factor A de Crecimiento Endotelial Vascular/metabolismo
8.
Biomed Res Int ; 2015: 989560, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26688823

RESUMEN

Preservation of peritoneal membrane (PM) is essential for long-term survival in peritoneal dialysis (PD). Continuous presence of PD fluids (PDF) in the peritoneal cavity generates chronic inflammation and promotes changes of the PM, such as fibrosis, angiogenesis, and lymphangiogenesis. Mesothelial-to-mesenchymal transition (MMT) and endothelial-to-mesenchymal transition (Endo-MT) seem to play a central role in this pathogenesis. We speculated that Rapamycin, a potent immunosuppressor, could be beneficial by regulating blood and lymphatic vessels proliferation. We demonstrate that mice undergoing a combined PD and Rapamycin treatment (PDF + Rapa group) presented a reduced PM thickness and lower number of submesothelial blood and lymphatic vessels, as well as decreased MMT and Endo-MT, comparing with their counterparts exposed to PD alone (PDF group). Peritoneal water transport in the PDF + Rapa group remained at control level, whereas PD effluent levels of VEGF, TGF-ß, and TNF-α were lower than in the PDF group. Moreover, the treatment of mesothelial cells with Rapamycin in vitro significantly decreased VEGF synthesis and selectively inhibited the VEGF-C and VEGF-D release when compared with control cells. Thus, Rapamycin has a protective effect on PM in PD through an antifibrotic and antiproliferative effect on blood and lymphatic vessels. Moreover, it inhibits Endo-MT and, at least partially, MMT.


Asunto(s)
Transición Epitelial-Mesenquimal/efectos de los fármacos , Linfangiogénesis/efectos de los fármacos , Membranas Artificiales , Neovascularización Fisiológica/efectos de los fármacos , Diálisis Peritoneal/efectos adversos , Sirolimus/farmacología , Animales , Citocinas/sangre , Femenino , Ratones
9.
PLoS One ; 8(4): e61165, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23637793

RESUMEN

Mesothelial-to-mesenchymal transition (MMT) is an auto-regulated physiological process of tissue repair that in uncontrolled conditions such as peritoneal dialysis (PD) can lead to peritoneal fibrosis. The maximum expression of peritoneal fibrosis induced by PD fluids and other peritoneal processes is the encapsulating peritoneal sclerosis (EPS) for which no specific treatment exists. Tamoxifen, a synthetic estrogen, has successfully been used to treat retroperitoneal fibrosis and EPS associated with PD. Hence, we used in vitro and animal model approaches to evaluate the efficacy of Tamoxifen to inhibit the MMT as a trigger of peritoneal fibrosis. In vitro studies were carried out using omentum-derived mesothelial cells (MCs) and effluent-derived MCs. Tamoxifen blocked the MMT induced by transforming growth factor (TGF)-ß1, as it preserved the expression of E-cadherin and reduced the expression of mesenchymal-associated molecules such as snail, fibronectin, collagen-I, α-smooth muscle actin, and matrix metalloproteinse-2. Tamoxifen-treatment preserved the fibrinolytic capacity of MCs treated with TGF-ß1 and decreased their migration capacity. Tamoxifen did not reverse the MMT of non-epitheliod MCs from effluents, but it reduced the expression of some mesenchymal molecules. In mice PD model, we demonstrated that MMT progressed in parallel with peritoneal membrane thickness. In addition, we observed that Tamoxifen significantly reduced peritoneal thickness, angiogenesis, invasion of the compact zone by mesenchymal MCs and improved peritoneal function. Tamoxifen also reduced the effluent levels of vascular endothelial growth factor and leptin. These results demonstrate that Tamoxifen is a therapeutic option to treat peritoneal fibrosis, and that its protective effect is mediated via modulation of the MMT process.


Asunto(s)
Transición Epitelial-Mesenquimal/efectos de los fármacos , Diálisis Peritoneal/efectos adversos , Peritoneo/citología , Peritoneo/efectos de los fármacos , Tamoxifeno/farmacología , Animales , Femenino , Fibrinólisis/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica/tratamiento farmacológico , Peritoneo/irrigación sanguínea , Peritoneo/fisiología , Fenotipo , Tamoxifeno/uso terapéutico , Factor de Crecimiento Transformador beta1/farmacología
10.
PLoS One ; 8(4): e60776, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23585849

RESUMEN

Vascular endothelial growth factor (VEGF) is up-regulated during mesothelial to mesenchymal transition (MMT) and has been associated with peritoneal membrane dysfunction in peritoneal dialysis (PD) patients. It has been shown that normal and malignant mesothelial cells (MCs) express VEGF receptors (VEGFRs) and co-receptors and that VEGF is an autocrine growth factor for mesothelioma. Hence, we evaluated the expression patterns and the functional relevance of the VEGF/VEGFRs/co-receptors axis during the mesenchymal conversion of MCs induced by peritoneal dialysis. Omentum-derived MCs treated with TGF-ß1 plus IL-1ß (in vitro MMT) and PD effluent-derived MCs with non-epithelioid phenotype (ex vivo MMT) showed down-regulated expression of the two main receptors Flt-1/VEGFR-1 and KDR/VEGFR-2, whereas the co-receptor neuropilin-1 (Nrp-1) was up-regulated. The expression of the Nrp-1 ligand semaphorin-3A (Sema-3A), a functional VEGF competitor, was repressed throughout the MMT process. These expression pattern changes were accompanied by a reduction of the proliferation capacity and by a parallel induction of the invasive capacity of MCs that had undergone an in vitro or ex vivo MMT. Treatment with neutralizing anti-VEGF or anti-Nrp-1 antibodies showed that these molecules played a relevant role in cellular proliferation only in naïve omentum-derived MCs. Conversely, treatment with these blocking antibodies, as well as with recombinant Sema-3A, indicated that the switched VEGF/VEGFRs/co-receptors axis drove the enhanced invasion capacity of MCs undergoing MMT. In conclusion, the expression patterns of VEGFRs and co-receptors change in MCs during MMT, which in turn would determine their behaviour in terms of proliferation and invasion in response to VEGF.


Asunto(s)
Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal , Neuropilina-1/genética , Epiplón/metabolismo , Diálisis Peritoneal , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Adulto , Anciano , Anticuerpos Neutralizantes/farmacología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Femenino , Regulación de la Expresión Génica , Humanos , Interleucina-1beta/farmacología , Masculino , Persona de Mediana Edad , Neuropilina-1/antagonistas & inhibidores , Neuropilina-1/metabolismo , Epiplón/efectos de los fármacos , Epiplón/patología , Cultivo Primario de Células , Semaforina-3A/genética , Semaforina-3A/metabolismo , Semaforina-3A/farmacología , Transducción de Señal , Factor de Crecimiento Transformador beta1/farmacología , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/farmacología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
11.
J Ren Nutr ; 23(5): e89-95, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23046736

RESUMEN

OBJECTIVE: Testosterone deficiency is a common finding in men with chronic kidney disease (CKD). Testosterone is thought to play an important anabolic role in muscle synthesis, and muscle wasting is an important and deleterious characteristic of protein-energy wasting (PEW) in CKD. It is presently unknown if reduced endogenous testosterone associates with features of muscle wasting in men with CKD. METHODS: This was a cross-sectional observational study of 267 men with CKD stages 2-4 (mean ± standard deviation age 67 ± 13 years, estimated glomerular filtration rate 42.9 [interquartile range 30.2-56.7] mL/min/1.73 m²) with measurements of endogenous testosterone and surrogates of PEW such as albumin, prealbumin, high-sensitivity C-reactive protein (CRP) and normalized protein nitrogen appearance (nPNA). Fat-free mass was estimated by bioelectrical impedance vector analysis (BIVA) and muscle strength by handgrip dynamometry. RESULTS: Across decreasing thirds of testosterone distribution, patients were incrementally older and CRP levels rose significantly. Prealbumin, hemoglobin, nPNA, handgrip strength, and BIVA estimated surrogates of muscle mass and nutritional status (fat-free mass, body cell mass, and phase angle) were progressively reduced (P < .05 for all). In multivariate regression analyses including age, renal function, and other important confounders, testosterone significantly and independently contributed to explain the variances of handgrip strength and fat-free mass (P < .05 for all). CONCLUSIONS: Endogenous testosterone independently associates with muscle strength and fat-free mass in men with moderate CKD. It is plausible that the reduction in testosterone levels that accompanies CKD may further contribute to the procatabolic environment leading to muscle wasting.


Asunto(s)
Índice de Masa Corporal , Fuerza de la Mano/fisiología , Insuficiencia Renal Crónica/sangre , Testosterona/sangre , Anciano , Anciano de 80 o más Años , Peso Corporal , Proteína C-Reactiva/metabolismo , Estudios Transversales , Impedancia Eléctrica , Tasa de Filtración Glomerular , Humanos , Masculino , Persona de Mediana Edad , Análisis Multivariante , Estado Nutricional , Fenotipo , Estudios Prospectivos , Análisis de Regresión , Insuficiencia Renal Crónica/fisiopatología , Factores de Riesgo , Testosterona/deficiencia
12.
Perit Dial Int ; 32(3): 292-304, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22215656

RESUMEN

BACKGROUND: Peritoneal membrane damage induced by peritoneal dialysis (PD) is largely associated with epithelial-to-mesenchymal transition (EMT) of mesothelial cells (MCs), which is believed to be a result mainly of the glucose degradation products (GDPs) present in PD solutions. OBJECTIVES: This study investigated the impact of bicarbonate-buffered, low-GDP PD solution (BicaVera: Fresenius Medical Care, Bad Homburg, Germany) on EMT of MCs in vitro and ex vivo. IN VITRO STUDIES: Omentum-derived MCs were incubated with lactate-buffered standard PD fluid or BicaVera fluid diluted 1:1 with culture medium. Ex vivo studies: From 31 patients randomly distributed to either standard or BicaVera solution and followed for 24 months, effluents were collected every 6 months for determination of EMT markers in effluent MCs. RESULTS: Culturing of MCs with standard fluid in vitro resulted in morphology change to a non-epithelioid shape, with downregulation of E-cadherin (indicative of EMT) and strong induction of vascular endothelial growth factor (VEGF) expression. By contrast, in vitro exposure of MCs to bicarbonate/low-GDP solution had less impact on both EMT parameters. Ex vivo studies partially confirmed the foregoing results. The BicaVera group, with a higher prevalence of the non-epithelioid MC phenotype at baseline (for unknown reasons), showed a clear and significant trend to gain and maintain an epithelioid phenotype at medium- and longer-term and to show fewer fibrogenic characteristics. By contrast, the standard solution group demonstrated a progressive and significantly higher presence of the non-epithelioid phenotype. Compared with effluent MCs having an epithelioid phenotype, MCs with non-epithelioid morphology showed significantly lower levels of E-cadherin and greater levels of fibronectin and VEGF. In comparing the BicaVera and standard solution groups, MCs from the standard solution group showed significantly higher secretion of interleukin 8 and lower secretion of collagen I, but no differences in the levels of other EMT-associated molecules, including fibronectin, VEGF, E-cadherin, and transforming growth factor ß1. Peritonitis incidence was similar in both groups. Functionally, the use of BicaVera fluid was associated with higher transport of small molecules and lower ultrafiltration capacity. CONCLUSIONS: Effluent MCs grown ex vivo from patients treated with bicarbonate/low-GDP BicaVera fluid showed a trend to acquire an epithelial phenotype, with lower production of proinflammatory cytokines and chemokines (such as interleukin 8) than was seen with MCs from patients treated with a lactate-buffered standard PD solution.


Asunto(s)
Bicarbonatos/farmacocinética , Soluciones para Diálisis/farmacología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Glucosa/metabolismo , Glucosa/farmacología , Diálisis Peritoneal , Bicarbonatos/análisis , Células Cultivadas , Soluciones para Diálisis/química , Glucosa/análisis , Humanos
13.
J Am Soc Nephrol ; 22(9): 1682-95, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21742730

RESUMEN

During peritoneal dialysis (PD), mesothelial cells undergo mesothelial-to-mesenchymal transition (MMT), a process associated with peritoneal-membrane dysfunction. Because TGF-ß1 can induce MMT, we evaluated the efficacy of TGF-ß1-blocking peptides in modulating MMT and ameliorating peritoneal damage in a mouse model of PD. Exposure of the peritoneum to PD fluid induced fibrosis, angiogenesis, functional impairment, and the accumulation of fibroblasts. In addition to expressing fibroblast-specific protein-1 (FSP-1), some fibroblasts co-expressed cytokeratin, indicating their mesothelial origin. These intermediate-phenotype (Cyto(+)/FSP-1(+)) fibroblasts had features of myofibroblasts with fibrogenic capacity. PD fluid treatment triggered the appearance of CD31(+)/FSP-1(+) and CD45(+)/FSP-1(+) cells, suggesting that fibroblasts also originate from endothelial cells and from cells recruited from bone marrow. Administration of blocking peptides significantly ameliorated fibrosis and angiogenesis, improved peritoneal function, and reduced the number of FSP-1(+) cells, especially in the Cyto(+)/FSP-1(+) subpopulation. Conversely, overexpression of TGF-ß1 in the peritoneum by adenovirus-mediated gene transfer led to a marked accumulation of fibroblasts, most of which derived from the mesothelium. Taken together, these results demonstrate that TGF-ß1 drives the peritoneal deterioration induced by dialysis fluid and highlights a role of TGF-ß1-mediated MMT in the pathophysiology of peritoneal-membrane dysfunction.


Asunto(s)
Transdiferenciación Celular/efectos de los fármacos , Diálisis Peritoneal/efectos adversos , Fibrosis Peritoneal/etiología , Peritoneo/patología , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Biomarcadores/metabolismo , Células Cultivadas , Soluciones para Diálisis/efectos adversos , Femenino , Inyecciones Intraperitoneales , Queratinas/metabolismo , Ratones , Ratones Endogámicos C57BL , Miofibroblastos/metabolismo , Miofibroblastos/patología , Fragmentos de Péptidos/farmacología , Fragmentos de Péptidos/uso terapéutico , Péptidos/farmacología , Péptidos/uso terapéutico , Fibrosis Peritoneal/metabolismo , Fibrosis Peritoneal/patología , Fibrosis Peritoneal/prevención & control , Fenotipo , Receptores de Factores de Crecimiento Transformadores beta/uso terapéutico , Proteína de Unión al Calcio S100A4 , Proteínas S100/metabolismo , Factor de Crecimiento Transformador beta1/antagonistas & inhibidores
14.
Nephrol Dial Transplant ; 26(1): 282-91, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20571097

RESUMEN

BACKGROUND: Peritoneal membrane deterioration during peritoneal dialysis (PD) is associated with epithelial-to-mesenchymal transition (EMT) of mesothelial cells (MC), which is believed to be mainly due to glucose degradation products (GDPs) present in PD solutions. Here we investigate the impact of GDPs in PD solutions on the EMT of MC in vitro and ex vivo. METHODS: For in vitro studies, omentum-derived MC were incubated with standard PD fluid or low-GDP solution diluted 1:1 with culture medium. For ex vivo studies, 33 patients, who were distributed at random to either the 'standard' or the 'low GDP' groups, were followed over 24 months. Effluents were collected every 6 months to determine EMT markers in effluent MC. RESULTS: Exposure of MC to standard fluid in vitro resulted in morphological change into a non-epitheloid shape, down-regulation of E-cadherin, indicative of EMT, and in a strong induction of vascular endothelial growth factor (VEGF) expression. In contrast, in vitro exposure of MC to low-GDP solution did not lead to these phenotype changes. This could be confirmed ex vivo, as the prevalence of non-epitheloid phenotype of MC in the standard group was significantly higher with increasing PD duration and MC isolated from this group showed significantly higher levels of EMT-associated molecules including fibronectin, collagen I, VEGF, IL-8 and TGF-ß levels when compared with the low-GDP group. Over time, the expression of E-cadherin also decreased in the standard but increased in the low-GDP group. In addition, the levels of EMT-associated molecules (fibronectin, VEGF and IL-8) increased in the standard but decreased in the low-GDP group. A similar trend was also observed for collagen I and for TGF-ß (for the first year), but did not reach global statistical significance. Accordingly, effluent MC with non-epitheloid morphology showed significantly lower levels of E-cadherin and greater levels of fibronectin, collagen I, VEGF and IL 8 when compared with MC with epitheloid phenotype. The incidence of peritonitis did not significantly influence these results. Drop-out due to technique failure was less in the 'balance' group. The functional, renal and peritoneal evaluation of patients being treated with either standard or 'balance' fluid did not show any significant difference over time. CONCLUSIONS: MC from PD effluent of patients treated with a PD fluid containing low GDP levels show fewer signs of EMT and the respective molecules than MC from patients treated with standard fluid, indicating a better preservation of the peritoneal membrane structure and a favourable outcome in patients using low-GDP fluid. It also confirms the hypothesis that the protection of EMT by GDP-reduced fluids is also present in vivo.


Asunto(s)
Soluciones para Diálisis/farmacología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Glucosa/metabolismo , Mesodermo/citología , Mesodermo/efectos de los fármacos , Diálisis Peritoneal , Western Blotting , Ensayo de Inmunoadsorción Enzimática , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Pronóstico , Estudios Prospectivos , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor A de Crecimiento Endotelial Vascular/metabolismo
15.
Lab Invest ; 90(10): 1517-32, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20531289

RESUMEN

Exposure to non-physiological solutions during peritoneal dialysis (PD) produces structural alterations to the peritoneal membrane and ultrafiltration dysfunction. The high concentration of glucose and glucose degradation products in standard PD fluids induce a local diabetic environment, which leads to the formation of advanced glycation end products (AGEs) that have an important role in peritoneal membrane deterioration. Peroxisome proliferator-activated receptor γ (PPAR-γ) agonists are used to treat type II diabetes and they have beneficial effects on inflammation, fibrosis, and angiogenesis. Hence, we evaluated the efficacy of the PPAR-γ agonist rosiglitazone (RSG) in ameliorating peritoneal membrane damage in a mouse PD model, and we analyzed the mechanisms underlying the protection offered by RSG. Exposure of the peritoneum to PD fluid resulted in AGEs accumulation, an inflammatory response, the loss of mesothelial cell monolayer and invasion of the compact zone by mesothelial cells, fibrosis, angiogenesis, and functional impairment of the peritoneum. Administration of RSG diminished the accumulation of AGEs, preserved the mesothelial monolayer, decreased the number of invading mesothelial cells, reduced fibrosis and angiogenesis, and improved peritoneal function. Interestingly, instead of reducing the leukocyte recruitment, RSG administration enhanced this process and specifically, the recruitment of CD3+ lymphocytes. Furthermore, RSG treatment augmented the levels of the anti-inflammatory cytokine interleukin (IL)-10 and increased the recruitment of CD4+ CD25+ FoxP3+ cells, suggesting that regulatory T cells mediated the protection of the peritoneal membrane. In cell-culture experiments, RSG did not prevent or reverse the mesothelial to mesenchymal transition, although it decreased mesothelial cells apoptosis. Accordingly, RSG appears to produce pleiotropic protective effects on the peritoneal membrane by reducing the accumulation of AGEs and inflammation, and by preserving the mesothelial cells monolayer, highlighting the potential of PPAR-γ activation to ameliorate peritoneal deterioration in PD patients.


Asunto(s)
Soluciones para Diálisis/efectos adversos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , PPAR gamma/agonistas , Diálisis Peritoneal/efectos adversos , Peritoneo/efectos de los fármacos , Peritoneo/patología , Tiazolidinedionas/farmacología , Animales , Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal/efectos de los fármacos , Epitelio/efectos de los fármacos , Epitelio/patología , Fibrosis , Glucosa/metabolismo , Productos Finales de Glicación Avanzada/metabolismo , Inmunidad Celular/efectos de los fármacos , Inflamación , Ratones , PPAR gamma/metabolismo , Peritoneo/inmunología , Peritoneo/metabolismo , Rosiglitazona , Linfocitos T/inmunología
16.
Nephrol Dial Transplant ; 25(4): 1098-108, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20067910

RESUMEN

BACKGROUND: During peritoneal dialysis (PD), mesothelial cells (MC) undergo an epithelial-to-mesenchymal transition (EMT), and this process is associated with peritoneal membrane (PM) damage. Bone morphogenic protein-7 (BMP-7) antagonizes transforming growth factor (TGF)-beta1, modulates EMT and protects against fibrosis. Herein, we analysed the modulating role of BMP-7 on EMT of MC in vitro and its protective effects in a rat PD model. METHODS: Epitheliod or non-epitheliod MC were analysed for the expression of BMP-7, TGF-beta1, activated Smads, epithelial cadherin (E-cadherin), collagen I, alpha smooth muscle cell actin (alpha-SMA) and vascular endothelial growth factor (VEGF) using standard procedures. Rats were daily instilled with PD fluid with or without BMP-7 during 5 weeks. Histological analyses were carried out in parietal peritoneum. Fibrosis was quantified with van Gieson or Masson's trichrome staining. Vasculature, activated macrophages and invading MC were quantified by immunofluorescence analysis. Quantification of infiltrating leukocytes and MC density in liver imprints was performed by May-Grünwald-Giemsa staining. Hyaluronic acid levels were determined by ELISA. RESULTS: MC constitutively expressed BMP-7, and its expression was downregulated during EMT. Treatment with recombinant BMP-7 resulted in blockade of TGF-beta1-induced EMT of MC. We provide evidence of a Smad-dependent mechanism for the blockade of EMT. Exposure of rat peritoneum to PD fluid resulted in inflammatory and regenerative responses, invasion of the compact zone by MC, fibrosis and angiogenesis. Administration of BMP-7 decreased the number of invading MC and reduced fibrosis and angiogenesis. In contrast, BMP-7 had no effect on inflammatory and regenerative responses, suggesting that these are EMT-independent, and probably upstream, processes. CONCLUSIONS: Data point to a balance between BMP-7 and TGF-beta1 in the control of EMT and indicate that blockade of EMT may be a therapeutic approach to ameliorate peritoneal membrane damage during PD.


Asunto(s)
Proteína Morfogenética Ósea 7/fisiología , Epitelio/metabolismo , Mesodermo/metabolismo , Diálisis Peritoneal , Peritoneo/metabolismo , Actinas/metabolismo , Animales , Western Blotting , Cadherinas/metabolismo , Diferenciación Celular , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Fibrosis , Humanos , Técnicas para Inmunoenzimas , Masculino , Ratas , Ratas Wistar , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta1/antagonistas & inhibidores
17.
Adv Perit Dial ; 26: 130-6, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21348395

RESUMEN

Bowel bacterial overgrowth syndrome (BBOS) is an important cause of gastrointestinal (GI) abnormalities. Proinflammatory cytokines (PICs) are excessively produced and accumulate because of kidney failure in dialysis patients who experience chronic infections such as BBOS. We explored the association between GL function, BBOS, and the malnutrition, inflammation, and atherosclerosis (MIA) syndrome. We studied GI malabsorption and maldigestion by analyzing fecal starch, sugar, fat, and nitrogen; intestinal protein permeability (alpha1-antitrypsin fecal clearance); and fecal chymotrypsin. We evaluated BBOS by breath hydrogen test (BHT) after a 3-day fat-and-carbohydrate-overload diet. Positive BHT was present in 10 patients, showing a high prevalence of GI macronutrient malabsorption and maldigestion, and compared with the other patients, the highest plasma levels of tumor necrosis factor alpha and interleukin 6 and lower levels of albumin and prealbumin. Those 10 patients were treated with a combination of several antibiotics, including neomycin, amoxicillin-clavulanate, and quinolones. Between 2 and 3 months later, the BHT, markers of nutrition, and PIC were re-tested. All treated patients showed an improvement in nutrition status and a lesser inflammatory pattern. The BBOS infectious process is found frequently in dialysis patients in association with GI malabsorption and maldigestion, malnutrition, and systemic inflammation. Hyperproduction of PIC because of BBOS induces MIA through a double pathway: GI disorders and deleterious systemic effects.


Asunto(s)
Aterosclerosis/etiología , Síndrome del Asa Ciega/complicaciones , Enfermedades Gastrointestinales/complicaciones , Desnutrición/etiología , Diálisis Peritoneal , Adulto , Anciano , Antibacterianos/uso terapéutico , Síndrome del Asa Ciega/diagnóstico , Síndrome del Asa Ciega/tratamiento farmacológico , Pruebas Respiratorias , Proteína C-Reactiva/análisis , Femenino , Enfermedades Gastrointestinales/diagnóstico , Enfermedades Gastrointestinales/tratamiento farmacológico , Humanos , Inflamación/etiología , Interleucina-6/sangre , Absorción Intestinal , Masculino , Persona de Mediana Edad , Estado Nutricional , Diálisis Peritoneal/efectos adversos , Factor de Necrosis Tumoral alfa/sangre
18.
J Am Soc Nephrol ; 20(3): 582-92, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19158357

RESUMEN

During peritoneal dialysis (PD), exposure of the peritoneal membrane to nonphysiologic solutions causes inflammation, ultimately leading to altered structure and function. Myofibroblasts, one of the cell types that contribute to dysfunction of the peritoneal membrane, can originate from mesothelial cells (MCs) by epithelial-to-mesenchymal transition (EMT), a process that has been associated with an increased rate of peritoneal transport. Because cyclooxygenase-2 (COX-2) is induced by inflammation, we studied the role of COX-2 in the deterioration of the peritoneal membrane. We observed that nonepithelioid MCs found in peritoneal effluent expressed higher levels of COX-2 than epithelioid MCs. The mass transfer coefficient for creatinine correlated with MC phenotype and with COX-2 levels. Although COX-2 was upregulated during EMT of MCs in vitro, COX-2 inhibition did not prevent EMT. In a mouse model of PD, however, COX-2 inhibition with Celecoxib resulted in reduced fibrosis and in partial recovery of ultrafiltration, outcomes that were associated with a reduction of inflammatory cells. Furthermore, PD fluid with a low content of glucose degradation products did not induce EMT or COX-2; the peritoneal membranes of mice treated with this fluid showed less worsening than mice exposed to standard fluid. In conclusion, upregulation of COX-2 during EMT may mediate peritoneal inflammation, suggesting COX-2 inhibition as a potential strategy to ameliorate peritoneal deterioration in PD patients.


Asunto(s)
Ciclooxigenasa 2/metabolismo , Soluciones para Diálisis/efectos adversos , Diálisis Peritoneal/efectos adversos , Peritoneo/enzimología , Adulto , Anciano , Anciano de 80 o más Años , Animales , Transporte Biológico Activo , Celecoxib , Células Cultivadas , Ciclooxigenasa 2/genética , Inhibidores de la Ciclooxigenasa 2/farmacología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/enzimología , Células Epiteliales/patología , Epitelio/efectos de los fármacos , Epitelio/enzimología , Epitelio/patología , Femenino , Humanos , Inflamación/prevención & control , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Modelos Animales , Peritoneo/efectos de los fármacos , Peritoneo/patología , Peritoneo/fisiopatología , Pirazoles/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Transcripción de la Familia Snail , Sulfonamidas/farmacología , Factores de Transcripción/genética , Regulación hacia Arriba , Adulto Joven
19.
Pathol Res Pract ; 204(8): 563-7, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18467038

RESUMEN

Uremia is considered capable of inducing structural anomalies of the peritoneum, including hyalinizing vasculopathy (HV). To further elucidate the contribution of uremia to the severity of HV, we performed an autopsy study of peritoneal dialysis (PD) patients with severe peritoneal HV lesions. Uremia is a systemic condition and, if capable of inducing HV, it will be expected to be detected outside the peritoneum. Seven autopsy cases of PD patients showing prominent peritoneal HV lesions were selected. Histological slides from the peritoneum, abdominal organs, heart and pericardium, lungs, visceral pleura, and central nervous system were reviewed. Peritoneal lesions were intense in all patients with prominent HV, fibrosis, and a variable presence of inflammation, fibrin, and calcification. Except for focal HV lesions in the intestinal submucosa of one diabetic patient, HV lesions were limited to the peritoneal membrane. None of the other extraperitoneal tissues showed such lesions. In conclusion, extraperitoneal vessels of PD patients show no relevant HV lesions when compared to peritoneal ones. This observation suggests that PD-related factors are the main contributors to the severity of vasculopathy. Uremia may participate in the development of the lesion but it does not seem to be responsible for its severity.


Asunto(s)
Calcinosis/patología , Hialina/metabolismo , Diálisis Peritoneal/efectos adversos , Peritoneo/irrigación sanguínea , Enfermedades Vasculares/patología , Anciano , Autopsia , Calcinosis/etiología , Femenino , Fibrosis , Humanos , Masculino , Persona de Mediana Edad , Factores de Riesgo , Índice de Severidad de la Enfermedad , España , Uremia/complicaciones , Uremia/patología , Enfermedades Vasculares/etiología , Enfermedades Vasculares/metabolismo
20.
J Ren Nutr ; 18(1): 107-13, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18089455

RESUMEN

Patients with chronic kidney disease frequently experience loss of appetite (anorexia), which increases in severity during the progression of the disease and may lead to protein-energy wasting, morbidity, and mortality. Anorexia represents a multiple, complex, and multifactorial disorder that may have its origin in renal failure (contemplating not only retention of uremic toxins but also peptides and cytokines) but that later on also involves metabolic abnormalities not yet corrected by dialysis therapy. This paper reviews current knowledge about the clinical signs of uremic anorexia as well as mechanisms involved. Based on megestrol acetate interventions and the recent observation that sex may modulate uremic appetite behavior, the potential role of sex hormones in treating chronic kidney disease anorexia needs to be further explored.


Asunto(s)
Anorexia/epidemiología , Trastornos de Alimentación y de la Ingestión de Alimentos/epidemiología , Uremia/complicaciones , Apetito , Citocinas/sangre , Trastornos de Alimentación y de la Ingestión de Alimentos/fisiopatología , Homeostasis , Humanos , Absorción Intestinal , Leptina/fisiología , Óxido Nítrico/fisiología , Uremia/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA