Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Artículo en Inglés | MEDLINE | ID: mdl-39197425

RESUMEN

INTRODUCTION: Peritoneal Ultrafiltration (PUF) has been proposed as an additional therapeutic option for Refractory Congestive Heart Failure (RCHF) patients. Despite promising observational studies and/or case report results, limited clinical trials data exist, and so far, PUF solutions remain only indicated for chronic kidney diseases (CKD). In this article, we describe a multicenter, randomized, controlled, unblinded, adaptive design clinical trial, about to start, investigating the effects of PolyCore™, an innovative PUF solution, in the treatment of RCHF patients. METHODS: The Peritoneal Ultrafiltration in Cardiorenal Syndrome (PURE) study is a Phase II, multicenter, randomized, controlled, unblinded, adaptive design clinical trial that aims to evaluate the safety and efficacy of PUF, using PolyCore™ as the investigational solution, in the treatment of RCHF patients who present with prominent right ventricular failure due to afterload mismatch, functional tricuspid regurgitation and enlarged cava vein consequent to intravascular fluid overload. Approximately 84 patients will be randomized 1:1 either to continue with their prescribed guidelines-directed medical therapy or to add the PUF treatment on top of it. The primary objective is to evaluate if PUF treatment has an impact on the composite endpoint of the patient's mortality or worsening of the patient's condition such as hospitalization for cardiovascular causes, increasing the initial daily dose of loop diuretic or worsening of renal function. Statistical analysis for the primary endpoint will be standard survival analysis to estimate the failure rate at month 7 for each group via Kaplan-Meier curves. Sensitivity analysis and various secondary analyses, including a multiple events analysis, will be conducted to evaluate the robustness of the primary endpoint results. Safety will be evaluated for up to 12 months. CONCLUSION: The PURE Study was designed to evaluate the safety and efficacy of peritoneal ultrafiltration with PolyCore™ on top of guidelines-directed medical therapy in patients with RCHF, assuming a combined clinical endpoint of mortality or worsening patients' condition. If successful, the treatment should allow for an improvement of the RCHF symptoms, decreasing hospitalization rate of patients. CLINICALTRIALS: gov Identifier: NCT03994874.

2.
Int J Mol Sci ; 25(6)2024 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-38542505

RESUMEN

Peritoneal dialysis (PD) is a home-based efficacious modality for the replacement of renal function in end-stage kidney failure patients, but it is still under-prescribed. A major limitation is the durability of the dialytic technique. Continuous exposure of the peritoneum to bioincompatible conventional glucose-based solutions is thought to be the main cause of the long-term morpho-functional peritoneal changes that eventually result in ultrafiltration failure. Poor PD solution biocompatibility is primarily related to the high glucose content, which is not only detrimental to the peritoneal membrane but has many potential metabolic side effects. To improve the clinical outcome and prolong the survival of the treatment, PD-related bioincompatibility urgently needs to be overcome. However, combining dialytic and osmotic efficacy with a satisfactory biocompatible profile is proving to be quite difficult. New approaches targeting the composition of the PD solution include the replacement of glucose with other osmotic agents, and the addition of cytoprotective or osmo-metabolic compounds. Other strategies include the infusion of mesenchymal cells or the administration of orally active agents. In the present article, we review the current evidence on efforts to improve the biocompatible and functional performance of PD, focusing on studies performed in vivo (animal models of PD, human subjects on PD).


Asunto(s)
Diálisis Peritoneal , Diálisis Renal , Animales , Humanos , Diálisis Peritoneal/efectos adversos , Soluciones para Diálisis/efectos adversos , Peritoneo , Glucosa/uso terapéutico
3.
Int J Mol Sci ; 23(9)2022 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-35563220

RESUMEN

Peritoneal dialysis (PD) is an efficient renal replacement therapy for patients with end-stage renal disease. Even if it ensures an outcome equivalent to hemodialysis and a better quality of life, in the long-term, PD is associated with the development of peritoneal fibrosis and the consequents patient morbidity and PD technique failure. This unfavorable effect is mostly due to the bio-incompatibility of PD solution (mainly based on high glucose concentration). In the present review, we described the mechanisms and the signaling pathway that governs peritoneal fibrosis, epithelial to mesenchymal transition of mesothelial cells, and angiogenesis. Lastly, we summarize the present and future strategies for developing more biocompatible PD solutions.


Asunto(s)
Diálisis Peritoneal , Fibrosis Peritoneal , Soluciones para Diálisis/metabolismo , Transición Epitelial-Mesenquimal , Humanos , Diálisis Peritoneal/efectos adversos , Fibrosis Peritoneal/etiología , Fibrosis Peritoneal/metabolismo , Fibrosis Peritoneal/terapia , Peritoneo/patología , Calidad de Vida
4.
Semin Cancer Biol ; 62: 86-98, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31348993

RESUMEN

Heparanase (HPSE) is an endoglycosidase that catalyses the cutting of the side chains of heparan-sulphate proteoglycans (HS), thus determining the remodelling of the extracellular matrix and basement membranes, as well as promoting the release of different HS-related molecules as growth factors, cytokines and enzymes. Ever since the HPSE was identified in the late 1980s, several experimental studies have shown that its overexpression was instrumental in increasing tumor growth, metastatic dissemination, angiogenesis and inflammation. More recently, HPSE involvment has also been demonstrated in mediating tumor-host crosstalk, in inducing gene transcription, in the activation of signaling pathways and in the formation of exosomes and in autophagy. All of these activities (enzymatic and non-enzymatic) together make heparanase a multifunctional molecule that increases the aggressiveness and chemo-resistance of tumor cells. Conversely, heparanase gene-silencing or tumor treatment with compounds that inhibit heparanase activity have been shown to significantly attenuate tumor progression in different animal models of tumorigenesis, further emphasizing the therapeutic potential of anti-heparanase therapy for several types of neoplasms. This review focuses on present knowledge and recent development in the study of heparanase in cancer progression as well as on novel mechanisms by which heparanase regulates tumor metastasis and chemo-resistance. Moreover, recent advances in strategies for its inhibition as a potential therapeutic option will be discussed.


Asunto(s)
Glucuronidasa/genética , Glucuronidasa/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Animales , Autofagia , Coagulación Sanguínea , Manejo de la Enfermedad , Progresión de la Enfermedad , Susceptibilidad a Enfermedades , Resistencia a Antineoplásicos , Exosomas/metabolismo , Matriz Extracelular/metabolismo , Glucuronidasa/antagonistas & inhibidores , Humanos , Inflamación/complicaciones , Inflamación/etiología , Inflamación/metabolismo , Inflamación/patología , Espacio Intracelular/metabolismo , Terapia Molecular Dirigida , Neoplasias/patología , Neoplasias/terapia , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Microambiente Tumoral
5.
Int J Mol Sci ; 22(6)2021 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-33804258

RESUMEN

The endothelial glycocalyx, the gel layer covering the endothelium, is composed of glycosaminoglycans, proteoglycans, and adsorbed plasma proteins. This structure modulates vessels' mechanotransduction, vascular permeability, and leukocyte adhesion. Thus, it regulates several physiological and pathological events. In the present review, we described the mechanisms that disturb glycocalyx stability such as reactive oxygen species, matrix metalloproteinases, and heparanase. We then focused our attention on the role of glycocalyx degradation in the induction of profibrotic events and on the possible pharmacological strategies to preserve this delicate structure.


Asunto(s)
Endotelio/química , Fibrosis/genética , Glicocálix/química , Mecanotransducción Celular/genética , Proteínas Sanguíneas/química , Proteínas Sanguíneas/genética , Permeabilidad Capilar/genética , Endotelio/ultraestructura , Fibrosis/patología , Glucuronidasa/efectos adversos , Glicocálix/genética , Glicocálix/ultraestructura , Glicosaminoglicanos/química , Glicosaminoglicanos/genética , Humanos , Metaloproteinasas de la Matriz/efectos adversos , Proteoglicanos/química , Proteoglicanos/genética , Especies Reactivas de Oxígeno/efectos adversos
6.
Int J Mol Sci ; 22(15)2021 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-34360717

RESUMEN

Peritoneal dialysis (PD) is an important, if underprescribed, modality for the treatment of patients with end-stage kidney disease. Among the barriers to its wider use are the deleterious effects of currently commercially available glucose-based PD solutions on the morphological integrity and function of the peritoneal membrane due to fibrosis. This is primarily driven by hyperglycaemia due to its effects, through multiple cytokine and transcription factor signalling-and their metabolic sequelae-on the synthesis of collagen and other extracellular membrane components. In this review, we outline these interactions and explore how novel PD solution formulations are aimed at utilizing this knowledge to minimise the complications associated with fibrosis, while maintaining adequate rates of ultrafiltration across the peritoneal membrane and preservation of patient urinary volumes. We discuss the development of a new generation of reduced-glucose PD solutions that employ a variety of osmotically active constituents and highlight the biochemical rationale underlying optimization of oxidative metabolism within the peritoneal membrane. They are aimed at achieving optimal clinical outcomes and improving the whole-body metabolic profile of patients, particularly those who are glucose-intolerant, insulin-resistant, or diabetic, and for whom daily exposure to high doses of glucose is contraindicated.


Asunto(s)
Diabetes Mellitus/terapia , Soluciones para Diálisis/uso terapéutico , Intolerancia a la Glucosa/terapia , Resistencia a la Insulina , Fallo Renal Crónico/terapia , Diálisis Peritoneal , Soluciones para Diálisis/efectos adversos , Glucosa/efectos adversos , Glucosa/uso terapéutico , Humanos , Peritoneo
7.
Adv Exp Med Biol ; 1221: 669-684, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32274731

RESUMEN

Organ fibrosis is defined as a deregulated wound-healing process characterized by a progressive accumulation of fibrous tissue and by reduced remodeling that can lead to the loss of functionality of the affected organ. This pathological process is quite common in several parenchymal organs such as kidneys, liver, and lungs and represents a real health emergency in developed western countries since a real anti-fibrotic therapy is not yet available in most cases. Heparanase (HPSE), which is the enzyme that cuts off the side chains of heparan sulfate (HS) proteoglycan, appears to be involved in the aetiopathogenesis of fibrosis in all these organs, even if with different mechanisms. Here we discuss how the interplay between HPSE and components of the immune and inflammatory responses can activate recruitment, proliferation, and activation of myofibroblasts which represent the main cell type responsible for the deposition of fibrous matrix. Finally, bearing in mind that once the activity of HPSE is inhibited no other molecule is able to perform the same function, it is desirable that this enzyme could prove to be a suitable pharmacological target in anti-fibrotic therapy.


Asunto(s)
Fibrosis/enzimología , Fibrosis/patología , Glucuronidasa/metabolismo , Fibrosis/metabolismo , Proteoglicanos de Heparán Sulfato , Humanos , Miofibroblastos , Especificidad de Órganos , Cicatrización de Heridas
8.
J Transl Med ; 17(1): 12, 2019 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-30616602

RESUMEN

BACKGROUND: The epithelial to mesenchymal transition (EMT) is a multi-factorial biological mechanism involved in renal and hepatic fibrosis and the IL-1 beta has been assumed as a mediator of this process although data are not exhaustive. Therefore, the aim of our study was to evaluate the role of this cytokine in the EMT of renal proximal tubular epithelial cells (HK-2) and stellate cells (LX-2) and the protective/anti-fibrotic effect of its inhibition by Canakinumab (a specific human monoclonal antibody targeted against IL-1beta). METHODS: Both cell types were treated with IL-1 beta (10 ng/ml) for 6 and 24 h with and without Canakinumab (5 µg/ml). As control we used TGF-beta (10 ng/ml). Expression of EMT markers (vimentin, alpha-SMA, fibronectin) were evaluated through western blotting and immunofluorescence. Genes expression for matrix metalloproteinases (MMP)-2 was measured by Real-Time PCR and enzymatic activity by zymography. Cellular motility was assessed by scratch assay. RESULTS: IL-1 beta induced a significant up-regulation of EMT markers in both cell types and increased the MMP-2 protein expression and enzymatic activity, similarly to TGF-beta. Moreover, IL-1 beta induced a higher rate of motility in HK-2. Canakinumab prevented all these modifications in both cell types. CONCLUSIONS: Our results clearly demonstrate the role of IL-1 beta in the EMT of renal/stellate cells and it underlines, for the first time, the therapeutic potential of its specific inhibition on the prevention/minimization of organ fibrosis.


Asunto(s)
Transición Epitelial-Mesenquimal/efectos de los fármacos , Células Estrelladas Hepáticas/patología , Interleucina-1beta/farmacología , Túbulos Renales/patología , Anticuerpos Monoclonales Humanizados/farmacología , Biomarcadores/metabolismo , Línea Celular , Movimiento Celular/efectos de los fármacos , Células Estrelladas Hepáticas/efectos de los fármacos , Células Estrelladas Hepáticas/enzimología , Humanos , Túbulos Renales/efectos de los fármacos , Túbulos Renales/enzimología , Metaloproteinasa 2 de la Matriz/metabolismo , Factor de Crecimiento Transformador beta1/farmacología
9.
FASEB J ; 32(2): 742-756, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28970256

RESUMEN

Heparanase (HPSE) is part of the biologic network triggered by ischemia/reperfusion (I/R) injury, a complication of renal transplantation and acute kidney injury. During this period, the kidney or graft undergoes a process of macrophages recruitment and activation. HPSE may therefore control these biologic effects. We measured the ability of HPSE and its inhibitor, SST0001, to regulate macrophage polarization and the crosstalk between macrophages and HK-2 renal tubular cells during in vitro hypoxia/reoxygenation (H/R). Furthermore, we evaluated in vivo renal inflammation, macrophage polarization, and histologic changes in mice subjected to monolateral I/R and treated with SST0001 for 2 or 7 d. The in vitro experiments showed that HPSE sustained M1 macrophage polarization and modulated apoptosis, the release of damage associated molecular patterns in post-H/R tubular cells, the synthesis of proinflammatory cytokines, and the up-regulation of TLRs on both epithelial cells and macrophages. HPSE also regulated M1 polarization induced by H/R-injured tubular cells and the partial epithelial-mesenchymal transition of these epithelial cells by M1 macrophages. All these effects were prevented by inhibiting HPSE. Furthermore, the inhibition of HPSE in vivo reduced inflammation and M1 polarization in mice undergoing I/R injury, partially restored renal function and normal histology, and reduced apoptosis. These results show for the first time that HPSE regulates macrophage polarization as well as renal damage and repair after I/R. HPSE inhibitors could therefore provide a new pharmacologic approach to minimize acute kidney injury and to prevent the chronic profibrotic damages induced by I/R.-Masola, V., Zaza, G., Bellin, G., Dall'Olmo, L., Granata, S., Vischini, G., Secchi, M. F., Lupo, A., Gambaro, G., Onisto, M. Heparanase regulates the M1 polarization of renal macrophages and their crosstalk with renal epithelial tubular cells after ischemia/reperfusion injury.


Asunto(s)
Células Epiteliales/enzimología , Glucuronidasa/metabolismo , Enfermedades Renales/enzimología , Túbulos Renales/enzimología , Macrófagos/enzimología , Daño por Reperfusión/enzimología , Animales , Células Epiteliales/patología , Enfermedades Renales/patología , Túbulos Renales/lesiones , Túbulos Renales/patología , Macrófagos/patología , Ratones , Daño por Reperfusión/patología
10.
Int J Mol Sci ; 19(4)2018 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-29677166

RESUMEN

The administration of Everolimus (EVE), a mTOR inhibitor used in transplantation and cancer, is often associated with adverse effects including pulmonary fibrosis. Although the underlying mechanism is not fully clarified, this condition could be in part caused by epithelial to mesenchymal transition (EMT) of airway cells. To improve our knowledge, primary bronchial epithelial cells (BE63/3) were treated with EVE (5 and 100 nM) for 24 h. EMT markers (α-SMA, vimentin, fibronectin) were measured by RT-PCR. Transepithelial resistance was measured by Millicell-ERS ohmmeter. mRNA and microRNA profiling were performed by Illumina and Agilent kit, respectively. Only high dose EVE increased EMT markers and reduced the transepithelial resistance of BE63/3. Bioinformatics showed 125 de-regulated genes that, according to enrichment analysis, were implicated in collagen synthesis/metabolism. Connective tissue growth factor (CTGF) was one of the higher up-regulated mRNA. Five nM EVE was ineffective on the pro-fibrotic machinery. Additionally, 3 miRNAs resulted hyper-expressed after 100 nM EVE and able to regulate 31 of the genes selected by the transcriptomic analysis (including CTGF). RT-PCR and western blot for MMP12 and CTGF validated high-throughput results. Our results revealed a complex biological network implicated in EVE-related pulmonary fibrosis and underlined new potential disease biomarkers and therapeutic targets.


Asunto(s)
Antineoplásicos/efectos adversos , Everolimus/efectos adversos , MicroARNs/genética , Fibrosis Pulmonar/metabolismo , Transcriptoma/genética , Actinas/metabolismo , Animales , Línea Celular , Células Cultivadas , Biología Computacional , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Transición Epitelial-Mesenquimal/efectos de los fármacos , Fibronectinas/metabolismo , Humanos , Metaloproteinasa 12 de la Matriz/metabolismo , Ratones , Células 3T3 NIH , Fibrosis Pulmonar/genética , ARN Mensajero/metabolismo
11.
Nephrol Dial Transplant ; 32(7): 1145-1154, 2017 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-28064160

RESUMEN

BACKGROUND: Epithelial-to-mesenchymal transition (EMT) of peritoneal mesothelial cells induced by high glucose (HG) levels is a major biological mechanism leading to myofibroblast accumulation in the omentum of patients on peritoneal dialysis (PD). Heparanase (HPSE), an endoglycosidase that cleaves heparan sulfate chains, is involved in the EMT of several cell lines, and may have a major role in this pro-fibrotic process potentially responsible for the failure of dialysis. Its specific inhibition may therefore plausibly minimize this pathological condition. METHODS: An in vitro study employing several biomolecular strategies was conducted to assess the role of HPSE in the HG-induced mesothelial EMT process, and to measure the effects of its specific inhibition by SST0001, a N-acetylated glycol-split heparin with a strong anti-HPSE activity. Rat mesothelial cells were grown for 6 days in HG (200 mM) culture medium with or without SST0001. Then EMT markers (VIM, α-SMA, TGF-ß) and vascular endothelial growth factor (VEGF) (a factor involved in neoangiogenesis) were measured by real-time PCR and immunofluorescence/western blotting. As a functional analysis, trans-epithelial resistance (TER) and permeability to albumin were also measured in our in vitro model using a Millicell-ERS ohmmeter and a spectrophotometer, respectively. RESULTS: Our results showed that 200 mM of glucose induced a significant gene and protein up-regulation of VEGF and all EMT markers after 6 days of culture. Intriguingly, adding SST0001 on day 3 reversed these biological and cellular effects. HPSE inhibition also restored the normal TER and permeability lost during the HG treatment. CONCLUSION: Taken together, our data confirm that HG can induce EMT of mesothelial cells, and that HPSE plays a central part in this process. Our findings also suggest that pharmacological HPSE inhibition could prove a valuable therapeutic tool for minimizing fibrosis and avoiding a rapid decline in the efficacy of dialysis in patients on PD, though clinical studies and/or trials would be needed to confirm the clinical utility of this treatment.


Asunto(s)
Transición Epitelial-Mesenquimal/efectos de los fármacos , Epitelio/efectos de los fármacos , Fibrosis/fisiopatología , Glucosa/farmacología , Glucuronidasa/antagonistas & inhibidores , Heparina/análogos & derivados , Peritoneo/efectos de los fármacos , Animales , Células Cultivadas , Epitelio/metabolismo , Epitelio/patología , Heparina/farmacología , Peritoneo/metabolismo , Peritoneo/patología , Ratas , Factor de Crecimiento Transformador beta/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
12.
J Pathol ; 238(1): 7-9, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26387558

RESUMEN

Vitamin D deficiency is inevitable in chronic kidney diseases. Clinical and experimental therapies with vitamin D supplements or analogues have demonstrated nephroprotective effects, which vitamin D exerts partly by controlling the renin-angiotensin-aldosterone system, but also by modulating other signalling pathways. In recent work published in the Journal of Pathology, Garsen and colleagues identified heparanase as a novel target of vitamin D and its antiproteinuric activity. Heparanase is an endoglycosidase with a role in remodelling the extracellular matrix through its ability to degrade heparan sulphate, and is involved in the pathogenesis of several proteinuric and fibrotic renal diseases. The new evidence that vitamin D inhibits heparanase expression sets the stage for a better understanding of the vitamin's kidney-protecting effects and its possible application to proteinuric and non-proteinuric chronic kidney diseases.


Asunto(s)
Calcitriol/farmacología , Glucuronidasa/metabolismo , Podocitos/enzimología , Proteinuria/metabolismo , Animales
13.
Biochim Biophys Acta ; 1843(9): 2122-8, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24937189

RESUMEN

Epithelial-mesenchymal transition (EMT) of tubular cells is one of the mechanisms which contribute to renal fibrosis and transforming growth factor-ß (TGF-ß) is one of the main triggers. Heparanase (HPSE) is an endo-ß-D-glucuronidase that cleaves heparan-sulfate thus regulating the bioavailability of growth factors (FGF-2, TGF-ß). HPSE controls FGF-2-induced EMT in tubular cells and is necessary for the development of diabetic nephropathy in mice. The aim of this study was to investigate whether HPSE can modulate the expression and the effects of TGF-ß in tubular cells. First we proved that the lack of HPSE or its inhibition prevents the increased synthesis of TGF-ß by tubular cells in response to pro-fibrotic stimuli such as FGF-2, advanced glycosylation end products (AGE) and albumin overload. Second, since TGF-ß may derive from sources different from tubular cells, we investigated whether HPSE modulates tubular cell response to exogenous TGF-ß. HPSE does not prevent EMT induced by TGF-ß although it slows its onset; indeed in HPSE-silenced cells the acquisition of a mesenchymal phenotype does not develop as quickly as in wt cells. Additionally, TGF-ß induces an autocrine loop to sustain its signal, whereas the lack of HPSE partially interferes with this autocrine loop. Overall these data confirm that HPSE is a key player in renal fibrosis since it interacts with the regulation and the effects of TGF-ß. HPSE is needed for pathological TGF-ß overexpression in response to pro-fibrotic factors. Furthermore, HPSE modulates TGF-ß-induced EMT: the lack of HPSE delays tubular cell transdifferentiation, and impairs the TGF-ß autocrine loop.


Asunto(s)
Glucuronidasa/metabolismo , Riñón/metabolismo , Riñón/patología , Factor de Crecimiento Transformador beta/genética , Actinas/genética , Actinas/metabolismo , Animales , Comunicación Autocrina , Biomarcadores/metabolismo , Línea Celular , Fibronectinas/genética , Fibronectinas/metabolismo , Fibrosis , Regulación de la Expresión Génica , Glucuronidasa/genética , Humanos , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Mesodermo/metabolismo , Mesodermo/patología , Ratones , Factor de Crecimiento Transformador beta/metabolismo , Vimentina/genética , Vimentina/metabolismo
14.
J Transl Med ; 13: 181, 2015 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-26040666

RESUMEN

Tubulo-interstitial fibrosis has been recognized as the hallmark of progression of chronic kidney disease, but, despite intensive research studies, there are currently no biomarkers or effective treatments for this condition. In this context, a promising candidate could be heparanase-1 (HPSE), an endoglycosidase that cleaves heparan sulfate chains and thus takes part in extracellular matrix remodeling. As largely described, it has a central role in the pathogenesis of cancer and inflammation, and it participates in the complex biological machinery involved in the onset of different renal proteinuric diseases (e.g., diabetic nephropathy, glomerulonephritis). Additionally, HPSE may significantly influence the progression of chronic kidney damage trough its major role in the biological pathway of renal fibrogenesis. Here, we briefly summarize data supporting the role of HPSE in renal damage, focusing on recent evidences that demonstrate the capability of this enzyme to modulate the signaling of pro-fibrotic factors such as FGF-2 and TGF-ß and consequently to control the epithelial-mesenchymal transition in renal tubular cells. We also emphasize the need of the research community to undertake studies and clinical trials to assess the potential clinical employment of this enzyme as diagnostic and prognostic tool and/or its role as therapeutic target for new pharmacological interventions.


Asunto(s)
Glucuronidasa/metabolismo , Riñón/enzimología , Riñón/patología , Biomarcadores/metabolismo , Transición Epitelial-Mesenquimal , Fibrosis , Humanos , Terapia Molecular Dirigida
15.
BMC Gastroenterol ; 15: 118, 2015 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-26369804

RESUMEN

BACKGROUND: Everolimus (EVE), a mammalian target of rapamycin inhibitor, has been proposed as liver transplant immunosuppressive drug, gaining wide interest also for the treatment of cancer. Although an appropriate tolerance, it may induce several adverse effects, such as fibro-interstitial pneumonitis due to the acquisition of activated myofibroblasts. The exact molecular mechanism associated with epithelial to mesenchymal transition (EMT) may be crucial also in the liver context. This work examines the role and the molecular mediators of EMT in hepatic stellate cell (HSC) and human liver cancer cells (HepG2) and the potential role of EVE to maintain the epithelial phenotype rather than to act as a potential initiators of EMT. METHODS: Real time-PCR and western blot have been used to assess the capability of EVE at low-therapeutic (10 nM) and high (100 nM) dose to induce an in vitro EMT in HSC and HepG2. RESULTS: Biomolecular experiments demonstrated that low concentration of EVE (10 nM) did not modify the gene expression of alpha-smooth muscle actin (α-SMA), Vimentin (VIM), Fibronectin (FN) in both HSC and HepG2 cells, whereas EVE at 100 nM induced a significant over-expression of all the three above-mentioned genes and an increment of α-SMA and FN protein levels. Additionally, 100 nM of EVE induced a significant phosphorylation of AKT and an up-regulation of TGF-ß expression in HSC and HepG2 cells. DISCUSSION: Our data, although obtained in an in vitro model, revealed, for the first time, that high concentration of EVE may induce EMT in liver cells confirming previous published evidences obtained in renal cells. Additionally, they suggested that mTOR-I should be administered at the lowest dose able to maximize their important and specific therapeutic properties minimizing or avoiding fibrosis-related adverse effects. CONCLUSIONS: In summary, if confirmed by additional studies, our results could be useful for researchers to standardize new therapeutic immunosuppressive and anticancer drugs protocols.


Asunto(s)
Transición Epitelial-Mesenquimal/efectos de los fármacos , Everolimus/farmacología , Expresión Génica/efectos de los fármacos , Células Estrelladas Hepáticas/efectos de los fármacos , Inmunosupresores/farmacología , Actinas/genética , Actinas/metabolismo , Animales , Everolimus/administración & dosificación , Fibronectinas/genética , Fibronectinas/metabolismo , Células Hep G2 , Células Estrelladas Hepáticas/fisiología , Humanos , Inmunosupresores/administración & dosificación , Fenotipo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Wistar , Factor de Crecimiento Transformador beta/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Vimentina/genética
16.
Nephrol Dial Transplant ; 29 Suppl 1: i74-i79, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24493873

RESUMEN

Experimental data in cell cultures and animal models suggest that sulodexide and glycosaminoglycans are potentially effective drugs to treat chronic kidney diseases and prevent progression to renal failure. However, no conclusive evidence support the use of them in human renal disease. In acute and chronic glomerulonephritis, only few studies have been performed. Sulodexide has been more intensely investigated in diabetic nephropathy (DN) where the body of data supports its effectiveness as an antialbuminuric agent in early stages. Unfortunately, there is no study in DN patients on the effect of sulodexide on clinical end points.


Asunto(s)
Anticoagulantes/uso terapéutico , Glicosaminoglicanos/uso terapéutico , Enfermedades Renales/tratamiento farmacológico , Animales , Progresión de la Enfermedad , Humanos
17.
J Biol Chem ; 287(2): 1478-88, 2012 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-22102278

RESUMEN

The epithelial-mesenchymal transition (EMT) of proximal tubular epithelial cells (PTECs) into myofibroblasts contributes to the establishment of fibrosis that leads to end stage renal disease. FGF-2 induces EMT in PTECs. Because the interaction between FGF-2 and its receptor is mediated by heparan sulfate (HS) and syndecans, we speculated that a deranged HS/syndecans regulation impairs FGF-2 activity. Heparanase is crucial for the correct turnover of HS/syndecans. The aim of the present study was to assess the role of heparanase on epithelial-mesenchymal transition induced by FGF-2 in renal tubular cells. In human kidney 2 (HK2) PTEC cultures, although FGF-2 induces EMT in the wild-type clone, it is ineffective in heparanase-silenced cells. The FGF-2 induced EMT is through a stable activation of PI3K/AKT which is only transient in heparanase-silenced cells. In PTECs, FGF-2 induces an autocrine loop which sustains its signal through multiple mechanisms (reduction in syndecan-1, increase in heparanase, and matrix metalloproteinase 9). Thus, heparanase is necessary for FGF-2 to produce EMT in PTECs and to sustain FGF-2 intracellular signaling. Heparanase contributes to a synergistic loop for handling syndecan-1, facilitating FGF-2 induced-EMT. In conclusion, heparanase plays a role in the tubular-interstitial compartment favoring the FGF-2-dependent EMT of tubular cells. Hence, heparanase is an interesting pharmacological target for the prevention of renal fibrosis.


Asunto(s)
Transición Epitelial-Mesenquimal , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Glucuronidasa/metabolismo , Fallo Renal Crónico/metabolismo , Túbulos Renales/metabolismo , Sindecano-1/metabolismo , Comunicación Autocrina/genética , Línea Celular , Activación Enzimática/genética , Factor 2 de Crecimiento de Fibroblastos/genética , Fibrosis , Glucuronidasa/genética , Humanos , Fallo Renal Crónico/patología , Túbulos Renales/patología , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal/genética , Sindecano-1/genética
18.
J Transl Med ; 11: 292, 2013 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-24256696

RESUMEN

BACKGROUND: Everolimus (EVE) is a drug widely used in several renal transplant protocols. Although characterized by a relatively low nephrotoxicity, it may induce several adverse effects including severe fibro-interstitial pneumonitis. The exact molecular/biological mechanism associated to these pro-fibrotic effects is unknown, but epithelial to mesenchymal transition (EMT) may have a central role. Additionally, heparanase, an enzyme recently associated with the progression of chronic allograft nephropathy, could contribute to activate this machinery in renal cells. METHODS: Several biomolecular strategies (RT-PCR, immunofluorescence, zymography and migration assay) have been used to assess the capability of EVE (10, 100, 200 and 500 nM) to induce an in vitro heparanase-mediated EMT in wild-type (WT) and Heparanase (HPSE)-silenced immortalized human renal epithelial proximal tubular cells (HK-2). Additionally, microarray technology was used to find additional biological elements involved in EVE-induced EMT. RESULTS: Biomolecular experiments demonstrated a significant up-regulation (more than 1.5 fold increase) of several genes encoding for well known EMT markers [(alpha-smooth muscle actin (α-SMA), Vimentin (VIM), Fibronectin (FN) and matrix metalloproteinase-9 (MMP9)], enhancement of MMP9 protein level and increment of cells motility in WT HK2 cells treated with high concentrations of EVE (higher than 100 nM). Similarly, immunofluorescence analysis showed that 100 nM of EVE increased α-SMA, VIM and FN protein expression in WT HK2 cells. All these effects were absent in both HPSE- and AKT-silenced cell lines. AKT is a protein having a central role in EMT. Additionally, microarray analysis identified other 2 genes significantly up-regulated in 100 nM EVE-treated cells (p < 0.005 and FDR < 5%): transforming growth factor beta-2 (TGFß2) and epidermal growth factor receptor (EGFR). Real-time PCR analysis validated microarray. CONCLUSIONS: Our in vitro study reveals new biological/cellular aspects of the pro-fibrotic activity of EVE and it demonstrates, for the first time, that an heparanase-mediated EMT of renal tubular cells may be activated by high doses of this drug. Additionally, our results suggest that clinicians should administer the adequate dosage of EVE in order to increase efficacy and reduce adverse effects. Finally heparanase could be a new potential therapeutic target useful to prevent/minimize drug-related systemic fibrotic adverse effects.


Asunto(s)
Transición Epitelial-Mesenquimal/efectos de los fármacos , Glucuronidasa/metabolismo , Inmunosupresores/farmacología , Túbulos Renales Proximales/efectos de los fármacos , Sirolimus/análogos & derivados , Actinas/genética , Línea Celular Transformada , Everolimus , Fibronectinas/genética , Expresión Génica/efectos de los fármacos , Humanos , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/enzimología , Metaloproteinasa 9 de la Matriz/genética , Sirolimus/farmacología , Vimentina/genética
19.
Methods Mol Biol ; 2619: 99-106, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36662465

RESUMEN

Glycosaminoglycans, the building blocks of proteoglycans, play a central role in the extracellular matrix and regulate a number of cellular processes. Therefore, any imbalance in their levels can lead to significant changes in cell behavior and phenotype. Additionally, glycosaminoglycans and their derivatives can be deployed as therapeutic agents in pathological conditions. Since cell morphology is a critical indicator of specialized cellular functions, its study can provide valuable insight. Scanning electron microscopy is a high-resolution imaging technique that makes for an ideal tool to observe the cellular appearance in 2D and 3D cultures under different conditions and/or substrates. In this chapter we provide a step-by-step protocol to study the influence of exogenously added glycosaminoglycans in the morphology of cells using scanning electron microscopy.


Asunto(s)
Glicosaminoglicanos , Proteoglicanos , Microscopía Electrónica de Rastreo , Matriz Extracelular/fisiología
20.
Biomedicines ; 11(7)2023 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-37509428

RESUMEN

We investigated how the extracellular matrix (ECM) affects LoVo colorectal cancer cells behavior during a spatiotemporal invasion. Epithelial-to-mesenchymal transition (EMT) markers, matrix-degrading enzymes, and morphological phenotypes expressed by LoVo-S (doxorubicin-sensitive) and higher aggressive LoVo-R (doxorubicin-resistant) were evaluated in cells cultured for 3 and 24 h on Millipore filters covered by Matrigel, mimicking the basement membrane, or type I Collagen reproducing a desmoplastic lamina propria. EMT and invasiveness were investigated with RT-qPCR, Western blot, and scanning electron microscopy. As time went by, most gene expressions decreased, but in type I Collagen samples, a strong reduction and high increase in MMP-2 expression in LoVo-S and -R cells occurred, respectively. These data were confirmed by the development of an epithelial morphological phenotype in LoVo-S and invading phenotypes with invadopodia in LoVo-R cells as well as by protein-level analysis. We suggest that the duration of culturing and type of substrate influence the morphological phenotype and aggressiveness of both these cell types differently. In particular, the type I collagen meshwork, consisting of large fibrils confining inter fibrillar micropores, affects the two cell types differently. It attenuates drug-sensitive LoVo-S cell aggressiveness but improves a proteolytic invasion in drug-resistant LoVo-R cells as time goes by. Experimental studies on CRC cells should examine the peri-tumoral ECM components, as well as the dynamic physical conditions of TME, which affect the behavior and aggressiveness of both drug-sensitive and drug-resistant LoVo cells differently.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA