Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Arch Toxicol ; 94(9): 2965-2979, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32577786

RESUMEN

Road traffic is one of the main sources of particulate emissions into the environment and has an increasing, negative impact on the release of potentially dangerous materials. Vehicle brakes release a significant amount of wear particles, and knowledge regarding their possible adverse effects is limited. One of the most dangerous elements contained in brake pads is copper (Cu), known to be toxic for human health. Therefore, our aim was to study the cell toxicity of particulate matter (PM) produced by different combinations of braking discs and pads containing different amounts of Cu. We investigated whether brake-derived microparticles have toxic effects on lung cells proportionally to their Cu content. Analyte content was measured in friction materials by XRFS and in PM2.5 captured during braking tests using SEM/EDX. The biological impact of brake-derived PM2.5 was investigated on a human epithelial alveolar cell line (A549). Cell viability, oxidative stress, mitochondrial membrane potential, apoptosis, and the pro-inflammatory response of the cells, as well as gene expression, were assessed following exposure to increasing PM2.5 concentrations (1, 10, 100, 200, and 500 µg/ml). The brake debris with the lowest Cu content did not induce significant changes in biological effects on A549 cells compared to normal controls, except for ROS production and IL6 gene expression. PM2.5 containing higher Cu quantities induced cell toxicity that correlated with Cu concentration. Our data suggest that the toxicity of PM2.5 from the brake system is mainly related to Cu content, thus confirming that eliminating Cu from brake pads will be beneficial for human health in urbanized environments.


Asunto(s)
Contaminantes Atmosféricos/toxicidad , Cobre/toxicidad , Material Particulado/toxicidad , Células Epiteliales Alveolares/efectos de los fármacos , Humanos , Estrés Oxidativo , Emisiones de Vehículos
2.
Blood ; 129(19): 2636-2644, 2017 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-28288980

RESUMEN

Polymorphonuclear neutrophils (PMNs) have previously been reported to mediate phagocytosis of anti-CD20-opsonized B cells from patients with chronic lymphocytic leukemia (CLL). However, recent data have suggested that PMNs, like macrophages, can also mediate trogocytosis. We have performed experiments to more precisely investigate this point and to discriminate between trogocytosis and phagocytosis. In live-cell time-lapse microscopy experiments, we could not detect any significant phagocytosis by purified PMNs of anti-CD20-opsonized CLL B cells, but could detect only the repeated close contact between effectors and targets, which suggested trogocytosis. Similarly, in flow cytometry assays using CLL B-cell targets labeled with the membrane dye PKH67 and opsonized with rituximab or obinutuzumab, we observed that a mean of 50% and 75% of PMNs had taken a fraction of the dye from CLL B cells at 3 and 20 hours, respectively, with no significant decrease in absolute live or total CLL B-cell numbers, confirming that trogocytosis occurs, rather than phagocytosis. Trogocytosis was accompanied by loss of membrane CD20 from CLL B cells, which was evident with rituximab but not obinutuzumab. We conclude that PMNs mediate mostly trogocytosis rather than phagocytosis of anti-CD20-opsonized CLL B cells, and we discuss the implications of this finding in patients with CLL treated with rituximab or obinutuzumab in vivo.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Antineoplásicos/farmacología , Leucemia Linfocítica Crónica de Células B/inmunología , Neutrófilos/efectos de los fármacos , Fagocitosis/efectos de los fármacos , Rituximab/farmacología , Antígenos CD20/inmunología , Supervivencia Celular/efectos de los fármacos , Humanos , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Leucemia Linfocítica Crónica de Células B/patología , Neutrófilos/inmunología , Neutrófilos/patología
3.
J Appl Toxicol ; 39(11): 1484-1491, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31025406

RESUMEN

Exposure to atmospheric particulate matter (PM) can affect human health, causing asthma, atherosclerosis, renal disease and cancer. In the last few years, outdoor air pollution has increased globally, leading to a public health emergency. Epidemiological studies have reported a correlation between the development of severe respiratory and systemic diseases and exposure to PM. To evaluate the toxic effect of PM of different origins, conventional experimental toxicological investigations have been conducted in animals; however, animal experimentation poses major ethical issues and usually differs from human conditions. As an alternative, human cell cultures are increasingly being used to investigate cellular and molecular mechanisms of PM toxicity. Although 2D cell cultures have been proven helpful, they are far from being a valid alternative to animal tests. Recently, 3D cell culture and organ-on-chip technology have provided systems that are more complex and that can be more informative for toxicity studies. In this review, the results of the 2D systems that are most frequently used for PM toxicity evaluations are summarized with a special focus on their limitations. We also examined to which extent 3D cell culture and particularly the organ-on-chip technology may overcome these limitations and represent effective tools to improve airborne PM toxicity evaluations.


Asunto(s)
Contaminantes Atmosféricos/toxicidad , Alternativas a las Pruebas en Animales/métodos , Material Particulado/toxicidad , Pruebas de Toxicidad/métodos , Técnicas de Cultivo de Célula/métodos , Células Cultivadas , Células Epiteliales/efectos de los fármacos , Células Epiteliales/ultraestructura , Humanos , Sistema Respiratorio/citología , Sistema Respiratorio/efectos de los fármacos
4.
Am J Physiol Heart Circ Physiol ; 310(1): H49-59, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26497959

RESUMEN

Intimal hyperplasia (IH) is the first cause of failure of an arteriovenous fistula (AVF). The aim of the present study was to investigate the effects on endothelial cells (ECs) of shear stress waveforms derived from AVF areas prone to develop IH. We used a cone-and-plate device to obtain real-time control of shear stress acting on EC cultures. We exposed human umbilical vein ECs for 48 h to different shear stimulations calculated in a side-to-end AVF model. Pulsatile unidirectional flow, representative of low-risk stenosis areas, induced alignment of ECs and actin fiber orientation with flow. Shear stress patterns of reciprocating flow, derived from high-risk stenosis areas, did not affect EC shape or cytoskeleton organization, which remained similar to static cultures. We also evaluated flow-induced EC expression of genes known to be involved in cytoskeletal remodeling and expression of cell adhesion molecules. Unidirectional flow induced a significant increase in Kruppel-like factor 2 mRNA expression, whereas it significantly reduced phospholipase D1, α4-integrin, and Ras p21 protein activator 1 mRNA expression. Reciprocating flow did not increase Kruppel-like factor 2 mRNA expression compared with static controls but significantly increased mRNA expression of phospholipase D1, α4-integrin, and Ras p21 protein activator 1. Reciprocating flow selectively increased monocyte chemoattractant protein-1 and IL-8 production. Furthermore, culture medium conditioned by ECs exposed to reciprocating flows selectively increased smooth muscle cell proliferation compared with unidirectional flow. Our results indicate that protective vascular effects induced in ECs by unidirectional pulsatile flow are not induced by reciprocating shear forces, suggesting a mechanism by which oscillating flow conditions may induce the development of IH in AVF and vascular access dysfunction.


Asunto(s)
Derivación Arteriovenosa Quirúrgica/efectos adversos , Hemodinámica , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Mecanotransducción Celular , Diálisis Renal , Citoesqueleto de Actina/metabolismo , Proliferación Celular , Forma de la Célula , Células Cultivadas , Medios de Cultivo Condicionados/metabolismo , Citocinas/genética , Citocinas/metabolismo , Regulación de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Hiperplasia , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Comunicación Paracrina , Flujo Pulsátil , ARN Mensajero/metabolismo , Transducción de Señal , Estrés Mecánico , Factores de Tiempo
5.
Am J Pathol ; 183(5): 1527-38, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24160324

RESUMEN

Islet transplantation is a poorly investigated long-term strategy for insulin replacement and for treatment of complications in patients with diabetes. We investigated whether islet transplantation and insulin treatment can relieve diabetic neuropathy and rescue the residual endogenous pancreatic ß cells. We used a multimodal approach, with five groups of Sprague-Dawley rats studied for 8 months: control rats, diabetic rats, insulin-treated diabetic rats with moderate or mild hyperglycemia, and diabetic rats transplanted with microencapsulated islets. Islet transplantation normalized glycemia and increased body and muscle weight; it was also effective in reducing proteinuria and altered liver function. Transplantation significantly improved tail nerve conduction velocity, Na(+)-K(+)-ATPase activity, and morphological alterations in the sciatic nerve as evidenced by decrease in g-ratio; it also restored thermal and ameliorated mechanical nociceptive thresholds. Morphometric analysis of pancreas indicated a significant ß-cell volume increase in transplanted rats, compared with mildly and moderately hyperglycemic rats. Thus, allogeneic islet transplantation had a positive systemic effect in diabetic rats and induced regression of the established neuropathy and restitution of the typical characteristics of the islets. These findings strongly reinforce the need for improving glycemic control, not only to reverse established diabetic complications but also to improve ß-cell status in diabetic pancreas.


Asunto(s)
Complicaciones de la Diabetes/patología , Complicaciones de la Diabetes/terapia , Células Secretoras de Insulina/patología , Insulina/administración & dosificación , Trasplante de Islotes Pancreáticos , Animales , Glucemia/metabolismo , Peso Corporal/efectos de los fármacos , Complicaciones de la Diabetes/sangre , Complicaciones de la Diabetes/fisiopatología , Glucagón/metabolismo , Prueba de Tolerancia a la Glucosa , Hiperglucemia/complicaciones , Hiperglucemia/patología , Insulina/farmacología , Insulina/uso terapéutico , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/enzimología , Masculino , Conducción Nerviosa/efectos de los fármacos , Nocicepción/efectos de los fármacos , Umbral del Dolor/efectos de los fármacos , Proteinuria/complicaciones , Proteinuria/patología , Ratas , Ratas Endogámicas Lew , Ratas Sprague-Dawley , Nervio Ciático/efectos de los fármacos , Nervio Ciático/patología , Nervio Ciático/fisiopatología , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo
6.
Nephron Exp Nephrol ; 126(2): 113, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24854652

RESUMEN

BACKGROUND: Chronic kidney disease (CKD) is a global public health issue with an estimated prevalence of 8-16% worldwide. End-stage renal disease eventually develops every year in 0.15-0.2% of patients with overt CKD, and renal replacement therapy (RRT) with dialysis or transplantation is required. Although approximately 2 million people worldwide are currently on RRT to sustain life, this likely represents less than 10% of those who need it. The kidney transplant approach is also seriously impaired by limited graft survival and by the scarce availability of donors. Innovative tissue-engineering strategies have been recently proposed to overcome these challenges. It is anticipated that these novel approaches will also be cost-effective in the long term. Although the initial setup of these innovative technologies could be quite expensive, there would be a single application for each patient, with no additional costs thereafter, compared to the lifelong costs of dialysis or immunosuppressive medications required for transplantation. One of the most innovative tools currently being investigated in experimental models is based on the idea of using decellularized kidneys to engineer a new functional organ as a potential future treatment option for end-stage renal disease. SUMMARY: In the last 5 years, several interesting observations have been reported regarding the possibility of using an acellular matrix from the whole kidney and the attempt to recellularize this scaffold using stem or differentiated cells. This review provides an overview of the decellularization methods tested so far and their effects on the resulting extracellular matrix structure and composition. In addition, we also discuss methods recently described by us and others for the perfusion of kidney scaffolds for recellularization. KEY MESSAGES: Despite difficulties in achieving the import goal of kidney engineering in the laboratory, we discuss the problems with and limits of the experimental results obtained so far and point out the strategies that need to be adopted in order for this line of research to advance.


Asunto(s)
Enfermedades Renales/terapia , Riñón/citología , Ingeniería de Tejidos/métodos , Andamios del Tejido , Animales , Ratas , Porcinos
7.
Electrophoresis ; 34(7): 1068-75, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23161152

RESUMEN

The purification of Langerhans islets from fragments of pancreatic exocrine tissue is a critical stage for the further transplantation of insulin secreting islets in patients affected by type I diabetes. Aim of our work was the evaluation of dielectrophoresis as a promising method for pancreatic islets isolation without physical contact in miniaturized lab-on-chip devices. DEP exploits the dielectric properties of particles suspended in a fluid, in a region where the amplitude of the electric field is characterized by a high gradient. Langerhans islets are aggregates of cells and have a minimum diameter of 50 microns. Dielectric models of pancreatic islets as cell aggregates were derived from single pancreatic beta cells model. Numerical simulations were performed to optimize the exact shape and size of the quadrupole microelectrode configuration and to determine the DEP forces acting on islets. A custom electronic setup was developed for the generation of sinusoidal signals with proper voltage and frequency and used to perform DEP experiments with samples of Langerhans islets. Dielectric models were found sufficiently accurate and negative DEP, showing repulsion from the electrodes, was observed for pancreatic islets. The results of this work demonstrate that Langerhans islet can be manipulated without physical contact by dielectrophoresis, a technique that can be applied on cell aggregates in miniaturized lab-on-chip devices.


Asunto(s)
Electroforesis/instrumentación , Electroforesis/métodos , Islotes Pancreáticos/citología , Técnicas Analíticas Microfluídicas/instrumentación , Técnicas Analíticas Microfluídicas/métodos , Animales , Separación Celular/instrumentación , Separación Celular/métodos , Simulación por Computador , Conductividad Eléctrica , Islotes Pancreáticos/química , Modelos Biológicos , Ratas
8.
Ann Surg ; 255(5): 867-80, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22330032

RESUMEN

The present review illustrates the state of the art of regenerative medicine (RM) as applied to surgical diseases and demonstrates that this field has the potential to address some of the unmet needs in surgery. RM is a multidisciplinary field whose purpose is to regenerate in vivo or ex vivo human cells, tissues, or organs to restore or establish normal function through exploitation of the potential to regenerate, which is intrinsic to human cells, tissues, and organs. RM uses cells and/or specially designed biomaterials to reach its goals and RM-based therapies are already in use in several clinical trials in most fields of surgery. The main challenges for investigators are threefold: Creation of an appropriate microenvironment ex vivo that is able to sustain cell physiology and function in order to generate the desired cells or body parts; identification and appropriate manipulation of cells that have the potential to generate parenchymal, stromal and vascular components on demand, both in vivo and ex vivo; and production of smart materials that are able to drive cell fate.


Asunto(s)
Cirugía General/tendencias , Medicina Regenerativa , Animales , Materiales Biocompatibles/uso terapéutico , Prótesis Vascular , Trasplante de Células , Sulfatos de Condroitina/uso terapéutico , Colágeno/uso terapéutico , Procedimientos Quirúrgicos Dermatologicos , Tracto Gastrointestinal/cirugía , Insuficiencia Cardíaca/terapia , Humanos , Fallo Renal Crónico/cirugía , Laringe/cirugía , Trasplante de Hígado , Enfermedades Respiratorias/cirugía , Piel Artificial , Andamios del Tejido , Cicatrización de Heridas/fisiología , Heridas y Lesiones/cirugía
9.
Biomed Mater ; 17(5)2022 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-35944550

RESUMEN

To address the need of alternatives to autologous vessels for small-calibre vascular applications (e.g. cardiac surgery), a bio-hybrid semi-degradable material composed of silk fibroin (SF) and polyurethane (Silkothane®) was herein used to fabricate very small-calibre grafts (Øin= 1.5 mm) via electrospinning. Bio-hybrid grafts werein vitrocharacterized in terms of morphology and mechanical behaviour, and compared to similar grafts of pure SF. Similarly, two native vessels from a rodent model (abdominal aorta and vena cava) were harvested and characterized. Preliminary implants were performed on Lewis rats to confirm the suitability of Silkothane® grafts for small-calibre applications, specifically as aortic insertion and femoral shunt. The manufacturing process generated pliable grafts consisting of a randomized fibrous mesh and exhibiting similar geometrical features to rat aortas. Both Silkothane® and pure SF grafts showed radial compliances in the range from 1.37 ± 0.86 to 1.88 ± 1.01% 10-2mmHg-1, lower than that of native vessels. The Silkothane® small-calibre devices were also implanted in rats demonstrating to be adequate for vascular applications; all the treated rats survived the surgery for three months after implantation, and 16 rats out of 17 (94%) still showed blood flow inside the graft at sacrifice. The obtained results lay the basis for a deeper investigation of the interaction between the Silkothane® graft and the implant site, which may deal with further analysis on the potentialities in terms of degradability and tissue formation, on longer time-points.


Asunto(s)
Fibroínas , Injerto Vascular , Animales , Prótesis Vascular , Poliuretanos , Ratas , Ratas Endogámicas Lew
10.
Biomolecules ; 11(5)2021 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-33946440

RESUMEN

Cell's microenvironment has been shown to exert influence on cell behavior. In particular, matrix-cell interactions strongly impact cell morphology and function. The purpose of this study was to analyze the influence of different culture substrate materials on phenotype and functional properties of lung epithelial adenocarcinoma (A549) cells. A549 cells were seeded onto two different biocompatible, commercially available substrates: a polyester coverslip (Thermanox™ Coverslips), that was used as cell culture plate control, and a polydimethylsiloxane membrane (PDMS, Elastosil® Film) investigated in this study as alternative material for A549 cells culture. The two substrates influenced cell morphology and the actin cytoskeleton organization. Further, the Yes-associated protein (YAP) and its transcriptional coactivator PDZ-binding motif (TAZ) were translocated to the nucleus in A549 cells cultured on polyester substrate, yet it remained mostly cytosolic in cells on PDMS substrate. By SEM analysis, we observed that cells grown on Elastosil® Film maintained an alveolar Type II cell morphology. Immunofluorescence staining for surfactant-C revealing a high expression of surfactant-C in cells cultured on Elastosil® Film, but not in cells cultured on Thermanox™ Coverslips. A549 cells grown onto Elastosil® Film exhibited morphology and functionality that suggest retainment of alveolar epithelial Type II phenotype, while A549 cells grown onto conventional plastic substrates acquired an alveolar Type I phenotype.


Asunto(s)
Células Epiteliales Alveolares/citología , Células Epiteliales Alveolares/efectos de los fármacos , Dimetilpolisiloxanos/farmacología , Poliésteres/farmacología , Alveolos Pulmonares/citología , Alveolos Pulmonares/efectos de los fármacos , Células A549 , Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Técnicas de Cultivo de Célula/métodos , Línea Celular Tumoral , Medios de Cultivo , Humanos , Lipopéptidos/biosíntesis , Pulmón/citología , Pulmón/efectos de los fármacos , Microscopía Electroquímica de Rastreo , Péptidos Cíclicos/biosíntesis , Factores de Transcripción/biosíntesis , Proteínas Señalizadoras YAP
11.
Cells ; 9(8)2020 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-32796521

RESUMEN

Stem cell fate and behavior are affected by the bidirectional communication of cells and their local microenvironment (the stem cell niche), which includes biochemical cues, as well as physical and mechanical factors. Stem cells are normally cultured in conventional two-dimensional monolayer, with a mechanical environment very different from the physiological one. Here, we compare culture of rat mesenchymal stem cells on flat culture supports and in the "Nichoid", an innovative three-dimensional substrate micro-engineered to recapitulate the architecture of the physiological niche in vitro. Two versions of the culture substrates Nichoid (single-layered or "2D Nichoid" and multi-layered or "3D Nichoid") were fabricated via two-photon laser polymerization in a biocompatible hybrid organic-inorganic photoresist (SZ2080). Mesenchymal stem cells, isolated from rat bone marrow, were seeded on flat substrates and on 2D and 3D Nichoid substrates and maintained in culture up to 2 weeks. During cell culture, we evaluated cell morphology, proliferation, cell motility and the expression of a panel of 89 mesenchymal stem cells' specific genes, as well as intracellular structures organization. Our results show that mesenchymal stem cells adhered and grew in the 3D Nichoid with a comparable proliferation rate as compared to flat substrates. After seeding on flat substrates, cells displayed large and spread nucleus and cytoplasm, while cells cultured in the 3D Nichoid were spatially organized in three dimensions, with smaller and spherical nuclei. Gene expression analysis revealed the upregulation of genes related to stemness and to mesenchymal stem cells' features in Nichoid-cultured cells, as compared to flat substrates. The observed changes in cytoskeletal organization of cells cultured on 3D Nichoids were also responsible for a different localization of the mechanotransducer transcription factor YAP, with an increase of the cytoplasmic retention in cells cultured in the 3D Nichoid. This difference could be explained by alterations in the import of transcription factors inside the nucleus due to the observed decrease of mean nuclear pore diameter, by transmission electron microscopy. Our data show that 3D distribution of cell volume has a profound effect on mesenchymal stem cells structure and on their mechanobiological response, and highlight the potential use of the 3D Nichoid substrate to strengthen the potential effects of MSC in vitro and in vivo.


Asunto(s)
Células Madre Mesenquimatosas/citología , Animales , Western Blotting , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Células Cultivadas , Técnica del Anticuerpo Fluorescente , Adhesiones Focales/fisiología , Masculino , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/ultraestructura , Microscopía Electrónica de Rastreo , Microscopía Electrónica de Transmisión , Reacción en Cadena de la Polimerasa , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa
12.
Int J Stem Cells ; 13(1): 116-126, 2020 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-31887847

RESUMEN

BACKGROUND AND OBJECTIVES: Transplantation of pancreatic islets is an intriguing new therapeutic option to face the worldwide spread problem of Type-I diabetes. Currently, its clinical use is limited by several problems, mainly based on the high number of islets required to restore normoglycaemia and by the low survival of the transplanted tissue. A promising attempt to overcome the limits to such an approach was represented by the use of Mesenchymal Stem Cells (MSC). Despite the encouraging results obtained with murine-derived MSC, little is still known about their protective mechanisms. The aim of the present study was to verify the effectiveness, (besides murine MSC), of clinically relevant human-derived MSC (hMSC) on protecting pancreatic islets, thus also shedding light on the putative differences between MSC of different origin. METHODS AND RESULTS: Threefold kinds of co-cultures were therefore in vitro set up (direct, indirect and mixed), to analyze the hMSC effect on pancreatic islet survival and function and to study the putative mechanisms involved. Although in a different way with respect to murine MSC, also human derived cells demonstrated to be effective on protecting pancreatic islet survival. This effect could be due to the release of some trophic factors, such as VEGF and Il-6, and by the reduction of inflammatory cytokine TNF-α. CONCLUSIONS: Therefore, hMSC confirmed their great clinical potential to improve the feasibility of pancreatic islet transplantation therapy against diabetes.

13.
Cell Transplant ; 18(2): 195-201, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19499707

RESUMEN

Immunoisolation of pancreatic islets is extensively investigated for glycemic control in diabetic experimental animals. We previously reported that subcutaneous xenotransplantation of bovine islets protected by a selective polysulfone membrane successfully controlled glycemia in diabetic rats for up to 20 days. We then wondered whether immunoisolated islets have adequate oxygen supply in this device, where only diffusive transport allows cell function and survival. Here we set up an experimental technique to measure oxygen consumption rate (OCR) using a Clark's electrode inserted in a glass thermostated chamber connected to a data recorder and acquisition system. Bovine islets were isolated from 6-month-old calves, encapsulated in sodium alginate microcapsules or inserted in polysulfone hollow fibers. After 1 and 2 days in culture a series of measurements was performed using free islets (at normal or high-glucose concentration), islets encapsulated in microcapsules, or in hollow fibers. In free islets OCR averaged from 2.0 +/- 0.8 pmol/IEQ/min at low-glucose concentration and from 2.5 +/- 1.0 pmol/IEQ/min at high-glucose concentration (p < 0.01). OCR in islets encapsulated in microcapsules and in hollow fibers was comparable, and not significantly different from that measured in free islets. Two days after isolation OCR averaged 2.3 +/- 0.6 in free islets, 2.3 +/- 0.9 in alginate microcapsules, and 2.2 +/- 0.7 pmol/IEQ/min in hollow fibers. These results show that OCR by bovine islets is comparable to that previously reported for other species. OCR increases in islets stimulated with high glucose and may be considered as a functional index. Moreover, islet encapsulation in alginate microcapsule, as well as in hollow fiber membranes, did not significantly affect in vitro OCR, suggesting adequate islet oxygenation in these conditions.


Asunto(s)
Islotes Pancreáticos/metabolismo , Consumo de Oxígeno , Alginatos , Animales , Cápsulas , Bovinos , Células Cultivadas , Ácido Glucurónico , Ácidos Hexurónicos , Islotes Pancreáticos/citología , Polímeros , Sulfonas
14.
Biomed Mater ; 14(2): 025007, 2019 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-30620939

RESUMEN

Clinically available alternatives of vascular access for long-term haemodialysis-currently limited to native arteriovenous fistulae and synthetic grafts-suffer from several drawbacks and are associated to high failure rates. Bioprosthetic grafts and tissue-engineered blood vessels are costly alternatives without clearly demonstrated increased performance. In situ tissue engineering could be the ideal approach to provide a vascular access that profits from the advantages of vascular grafts in the short-term (e.g. early cannulation) and of fistulae in the long-term (e.g. high success rates driven by biointegration). Hence, in this study a three-layered silk fibroin/polyurethane vascular graft was developed by electrospinning to be applied as long-term haemodialysis vascular access pursuing a 'hybrid' in situ engineering approach (i.e. based on a semi-degradable scaffold). This Silkothane® graft was characterized concerning morphology, mechanics, physical properties, blood contact and vascular cell adhesion/viability. The full three-layered graft structure, influenced by the polyurethane presence, ensured mechanical properties that are a determinant factor for the success of a vascular access (e.g. vein-graft compliance matching). The Silkothane® graft demonstrated early cannulation potential in line with self-sealing commercial synthetic arteriovenous grafts, and a degradability driven by enzymatic activity. Moreover, the fibroin-only layers and extracellular matrix-like morphology, presented by the graft, revealed to be crucial in providing a non-haemolytic character, long clotting time, and favourable adhesion of human umbilical vein endothelial cells with increasing viability after 3 and 7 d. Accordingly, the proposed approach may represent a step forward towards an in situ engineered hybrid vascular access with potentialities for vein-graft anastomosis stability, early cannulation, and biointegration.


Asunto(s)
Prótesis Vascular , Fibroínas/química , Poliuretanos/química , Diálisis Renal/instrumentación , Ingeniería de Tejidos/métodos , Dispositivos de Acceso Vascular , Animales , Materiales Biocompatibles/química , Pruebas de Coagulación Sanguínea , Bombyx , Adhesión Celular , Supervivencia Celular , Electroquímica , Hemólisis , Células Endoteliales de la Vena Umbilical Humana , Humanos , Inflamación , Permeabilidad , Diálisis Renal/métodos , Estrés Mecánico , Suturas , Resistencia a la Tracción
15.
Sci Rep ; 9(1): 8001, 2019 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-31142801

RESUMEN

Generating new kidneys using tissue engineering technologies is an innovative strategy for overcoming the shortage of donor organs for transplantation. Here we report how to efficiently engineer the kidney vasculature of decellularized rat kidney scaffolds by using human induced pluripotent stem cell (hiPSCs)-derived endothelial cells (hiPSC-ECs). In vitro, hiPSC-ECs responded to flow stress by acquiring an alignment orientation, and attached to and proliferated on the acellular kidney sections, maintaining their phenotype. The hiPSC-ECs were able to self-organize into chimeric kidney organoids to form vessel-like structures. Ex vivo infusion of hiPSC-ECs through the renal artery and vein of acellular kidneys resulted in the uniform distribution of the cells in all the vasculature compartments, from glomerular capillaries to peritubular capillaries and small vessels. Ultrastructural analysis of repopulated scaffolds through transmission and scanning electron microscopy demonstrated the presence of continuously distributed cells along the vessel wall, which was also confirmed by 3D reconstruction of z-stack images showing the continuity of endothelial cell coverage inside the vessels. Notably, the detection of fenestrae in the endothelium of glomerular capillaries but not in the vascular capillaries was clear evidence of site-specific endothelial cell specialisation.


Asunto(s)
Riñón/química , Neovascularización Fisiológica/genética , Organoides/crecimiento & desarrollo , Ingeniería de Tejidos , Andamios del Tejido/química , Animales , Vasos Sanguíneos/química , Vasos Sanguíneos/crecimiento & desarrollo , Diferenciación Celular/genética , Células Endoteliales/citología , Células Endoteliales/metabolismo , Endotelio/química , Endotelio/crecimiento & desarrollo , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Riñón/crecimiento & desarrollo , Organoides/química , Ratas
16.
JOP ; 9(5): 601-11, 2008 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-18762691

RESUMEN

CONTEXT: Expansion and culture of beta cell progenitors in vitro may represent an alternative to the use of differentiated beta cells from donor pancreata. OBJECTIVE: The aim of our study was to investigate to what extent exocrine or endocrine pancreatic cells can be differentiated in insulin-producing cells in vitro. SETTING: Bovine exocrine tissue (n=4) and islets (n=4) were cultured in DMEM with serum. INTERVENTIONS: After 7 days, the cells were trypsinized and cultured in the same medium for cell proliferation, or in DMEM/F-12 containing growth factors to induce cell differentiation. MAIN OUTCOME MEASURE: Proliferating capacity after 4 weeks in culture. In addition, insulin expression was evaluated by RT-PCR and by immunohistochemical staining. RESULTS: After 4 weeks of culture, cells from exocrine tissue showed a 69.5+/-10.0 fold increase, while cells from islets showed a 31.2+/-11.4 fold increase (P=0.059). In differentiating medium, monolayers from exocrine and islet tissue were organized into islet-like structures containing cells which stained positively for insulin. Morphometrical analysis and RT-PCR confirmed the presence of insulin in the cells at the protein and the mRNA level. CONCLUSIONS: In our experimental conditions, cells from pancreatic tissue proliferated and differentiated in insulin-containing cells. However, the level of insulin as well as mRNA expression is only a small fraction of that shown by fresh islets. Only selective identification of cell precursors may allow efficient generation of insulin-producing cells in vitro.


Asunto(s)
Diferenciación Celular , Células Secretoras de Insulina/fisiología , Páncreas/fisiología , Animales , Bovinos , Recuento de Células , Proliferación Celular , Células Cultivadas , Eficiencia , Insulina/metabolismo , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Páncreas/citología , Esferoides Celulares/citología , Esferoides Celulares/metabolismo , Técnicas de Cultivo de Tejidos
17.
Adv Healthc Mater ; 7(21): e1800430, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30230709

RESUMEN

For patients with severe kidney or liver failure the best solution is currently organ transplantation. However, not all patients are eligible for transplantation and due to limited organ availability, most patients are currently treated with therapies using artificial kidney and artificial liver devices. These therapies, despite their relative success in preserving the patients' life, have important limitations since they can only replace part of the natural kidney or liver functions. As blood detoxification (and other functions) in these highly perfused organs is achieved by specialized cells, it seems relevant to review the approaches leading to bioengineered organs fulfilling most of the native organ functions. There, the culture of cells of specific phenotypes on adapted scaffolds that can be perfused takes place. In this review paper, first the functions of kidney and liver organs are briefly described. Then artificial kidney/liver devices, bioartificial kidney devices, and bioartificial liver devices are focused on, as well as biohybrid constructs obtained by decellularization and recellularization of animal organs. For all organs, a thorough overview of the literature is given and the perspectives for their application in the clinic are discussed.


Asunto(s)
Órganos Bioartificiales , Bioingeniería/métodos , Animales , Humanos , Riñón/citología , Hígado/citología , Hígado Artificial , Ingeniería de Tejidos/métodos
18.
J Appl Biomater Funct Mater ; 15(4): e326-e333, 2017 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-29131298

RESUMEN

Renal transplantation is currently the most effective treatment for end-stage renal disease, which represents one of the major current public health problems. However, the number of available donor kidneys is drastically insufficient to meet the demand, causing prolonged waiting lists. For this reason, tissue engineering offers great potential to increase the pool of donated organs for kidney transplantation, by way of seeding cells on supporting scaffolding material. Biological scaffolds are prepared by removing cellular components from the donor organs using a decellularization process with detergents, enzymes or other cell lysing solutions. Extracellular matrix which makes up the scaffold is critical to directing the cell attachment and to creating a suitable environment for cell survival, proliferation and differentiation. Researchers are now studying whole intact scaffolds produced from the kidneys of animals or humans without adversely affecting extracellular matrix, biological activity and mechanical integrity. The process of recellularization includes cell seeding strategies and the choice of the cell source to repopulate the scaffold. This is the most difficult phase, due to the complexity of the kidney. Indeed, no studies have provided sufficient results of complete renal scaffold repopulation and differentiation. This review summarizes the research that has been conducted to obtain decellularized kidney scaffolds and to repopulate the scaffolds, evaluating the best cell sources, the cell seeding methods and the cell differentiation in kidney scaffolds.


Asunto(s)
Matriz Extracelular/fisiología , Riñón , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Animales , Diferenciación Celular , Matriz Extracelular/química , Humanos , Riñón/citología , Riñón/patología , Técnicas de Cultivo de Órganos , Células Madre/citología , Células Madre/fisiología , Fracciones Subcelulares/química , Fracciones Subcelulares/fisiología
19.
Sci Rep ; 7: 43502, 2017 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-28266553

RESUMEN

The rising number of patients needing renal replacement therapy, alongside the significant clinical and economic limitations of current therapies, creates an imperative need for new strategies to treat kidney diseases. Kidney bioengineering through the production of acellular scaffolds and recellularization with stem cells is one potential strategy. While protocols for obtaining organ scaffolds have been developed successfully, scaffold recellularization is more challenging. We evaluated the potential of in vivo and in vitro kidney scaffold recellularization procedures. Our results show that acellular scaffolds implanted in rats cannot be repopulated with host cells, and in vitro recellularization is necessary. However, we obtained very limited and inconsistent cell seeding when using different infusion protocols, regardless of injection site. We also obtained experimental and theoretical data indicating that uniform cell delivery into the kidney scaffolds cannot be obtained using these infusion protocols, due to the permeability of the extracellular matrix of the scaffold. Our results highlight the major physical barriers that limit in vitro recellularization of acellular kidney scaffolds and the obstacles that must be investigated to effectively advance this strategy for regenerative medicine.


Asunto(s)
Riñón , Regeneración , Ingeniería de Tejidos , Andamios del Tejido , Animales , Matriz Extracelular , Masculino , Ratas , Medicina Regenerativa
20.
Exp Neurol ; 288: 75-84, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27851902

RESUMEN

Type-1 Diabetes is generally treated with exogenous insulin administration. Despite treatment, a very common long term consequence of diabetes is the development of a disabling and painful peripheral neuropathy. The transplantation of pancreatic islets is an advanced alternative therapeutic approach, but its clinical application is still very limited, mainly because of the great number of islets required to complete the procedure and of their short-term survival. An intriguing method to improve the performance of pancreatic islets transplantation is the co-transplantation of Mesenchymal Stem Cells (MSCs), adult stem cells already known to support the survival of different cellular populations. In this proof-of-concept study, we demonstrated using an in vivo model of diabetes, the ability of allogenic MSCs to reduce the number of pancreatic islets necessary to achieve glycemic control in diabetic rats, and overall their positive effect on diabetic neuropathy, with the reduction of all the neuropathic signs showed after disease induction. The cutback of the pancreatic islet number required to control glycemia and the regression of the painful neuropathy make MSC co-transplantation a very promising tool to improve the clinical feasibility of pancreatic islet transplantation for diabetes treatment.


Asunto(s)
Neuropatías Diabéticas/cirugía , Neuropatías Diabéticas/terapia , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/fisiología , Análisis de Varianza , Animales , Antibióticos Antineoplásicos/farmacología , Glucemia/metabolismo , Peso Corporal/efectos de los fármacos , Neuropatías Diabéticas/sangre , Neuropatías Diabéticas/fisiopatología , Modelos Animales de Enfermedad , Masculino , Fibras Nerviosas Mielínicas/patología , Conducción Nerviosa/efectos de los fármacos , Umbral del Dolor/efectos de los fármacos , Páncreas/patología , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Estreptozocina/farmacología , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA