Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Int J Mol Sci ; 24(14)2023 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-37511568

RESUMEN

Induced pluripotent stem cells (iPSC) have huge potential as cell therapy for various diseases, given their potential for unlimited self-renewal and capability to differentiate into a wide range of cell types. Although autologous iPSCs represents the ideal source for patient-tailored regenerative medicine, the high costs of the extensive and time-consuming production process and the impracticability for treating acute conditions hinder their use for broad applications. An allogeneic iPSC-based strategy may overcome these issues, but it carries the risk of triggering an immune response. So far, several approaches based on genome-editing techniques to silence human leukocyte antigen class I (HLA-I) or II (HLA-II) expression have been explored to overcome the immune rejection of allogeneic iPSCs. In this study, we employed the CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats/CRISPR associated protein 9) system to delete the ß2-Microglobulin (B2M) and the Class II Major Histocompatibility Complex Transactivator (CIITA) genes, essential for the correct surface expression of HLA-I and HLA-II proteins. The resulting hypoimmunogenic iPSC line has a normal karyotype, expresses the pluripotency stem cell markers, and is capable of differentiating into the three embryonic germ layers. Furthermore, we showed that it specifically retains the ability to differentiate towards different liver cells, such as endothelial-like cells, hepatocyte-like cells, and hepatic stellate-like cells. Our results indicate that hypoimmunogenic iPSCs could give a new cost-effective and off-the-shelf opportunity for cell therapy in liver diseases.


Asunto(s)
Células Madre Pluripotentes Inducidas , Células Madre Pluripotentes , Humanos , Medicina Regenerativa , Edición Génica/métodos , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/metabolismo , Hígado
2.
Biomedicines ; 9(12)2021 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-34944624

RESUMEN

No effective treatments are available for familial steroid-resistant Focal Segmental Glomerulosclerosis (FSGS), characterized by proteinuria due to ultrastructural abnormalities in glomerular podocytes. Here, we studied a private PAX2 mutation identified in a patient who developed FSGS in adulthood. By generating adult podocytes using patient-specific induced pluripotent stem cells (iPSC), we developed an in vitro model to dissect the role of this mutation in the onset of FSGS. Despite the PAX2 mutation, patient iPSC properly differentiated into podocytes that exhibited a normal structure and function when compared to control podocytes. However, when exposed to an environmental trigger, patient podocytes were less viable and more susceptible to cell injury. Fixing the mutation improved their phenotype and functionality. Using a branching morphogenesis assay, we documented developmental defects in patient-derived ureteric bud-like tubules that were totally rescued by fixing the mutation. These data strongly support the hypothesis that the PAX2 mutation has a dual effect, first in renal organogenesis, which could account for a suboptimal nephron number at birth, and second in adult podocytes, which are more susceptible to cell death caused by environmental triggers. These abnormalities might translate into the development of proteinuria in vivo, with a progressive decline in renal function, leading to FSGS.

3.
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
4.
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
5.
CRISPR J ; 2: 108-120, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30998089

RESUMEN

Focal segmental glomerulosclerosis (FSGS) is defined by focal (involving few glomeruli) and segmental sclerosis of the glomerular tuft that manifests with nephrotic syndrome. Mutations in genes involved in the maintenance of structure and function of podocytes have been found in a minority of these patients. A family with adult-onset autosomal dominant FSGS was recently found to carry a new germline missense heterozygous mutation (p.G189R) in the octapeptide domain of the transcription factor PAX2. Here, we efficiently corrected this point mutation in patient-derived induced pluripotent stem cells (iPSCs) by means of CRISPR-Cas9-based homology-directed repair. The iPSC lines were differentiated into podocytes, which were tested for their motility. Editing the PAX2 p.G189R mutation restored podocyte motility, which was altered in podocytes derived from patient iPSCs.


Asunto(s)
Glomeruloesclerosis Focal y Segmentaria/genética , Glomeruloesclerosis Focal y Segmentaria/terapia , Factor de Transcripción PAX2/genética , Adulto , Proteína 9 Asociada a CRISPR/genética , Sistemas CRISPR-Cas/genética , Diferenciación Celular , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Ingeniería Genética/métodos , Mutación de Línea Germinal/genética , Glomeruloesclerosis Focal y Segmentaria/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/fisiología , Glomérulos Renales/metabolismo , Mutación/genética , Factor de Transcripción PAX2/análisis , Podocitos/química , Podocitos/metabolismo , Podocitos/fisiología , Polimorfismo de Nucleótido Simple/genética
6.
Nat Commun ; 8(1): 983, 2017 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-29042548

RESUMEN

Mesenchymal stromal cells (MSCs) are renoprotective and drive regeneration following injury, although cellular targets of such an effect are still ill-defined. Here, we show that human umbilical cord (UC)-MSCs transplanted into mice stimulate tubular cells to regain mitochondrial mass and function, associated with enhanced microtubule-rich projections that appear to mediate mitochondrial trafficking to create a reparative dialogue among adjacent tubular cells. Treatment with UC-MSCs in mice with cisplatin-induced acute kidney injury (AKI) regulates mitochondrial biogenesis in proximal tubuli by enhancing PGC1α expression, NAD+ biosynthesis and Sirtuin 3 (SIRT3) activity, thus fostering antioxidant defenses and ATP production. The functional role of SIRT3 in tubular recovery is highlighted by data that in SIRT3-deficient mice with AKI, UC-MSC treatment fails to induce renoprotection. These data document a previously unrecognized mechanism through which UC-MSCs facilitate renal repair, so as to induce global metabolic reprogramming of damaged tubular cells to sustain energy supply.Mesenchymal stromal cells drive renal regeneration following injury. Here, the authors show that human mesenchymal stromal cells, when transplanted into mice with acute kidney injury, stimulate renal tubular cell growth and enhance mitochondrial function via SIRT3.


Asunto(s)
Lesión Renal Aguda/terapia , Túbulos Renales/metabolismo , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Mitocondrias/metabolismo , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/fisiopatología , Adenosina Trifosfato/metabolismo , Animales , Proliferación Celular , Cisplatino/efectos adversos , Femenino , Humanos , Ratones , Ratones SCID , Mitocondrias/genética , NAD/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Sirtuina 3/genética , Sirtuina 3/metabolismo
7.
Sci Rep ; 7(1): 11518, 2017 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-28912528

RESUMEN

We have previously shown that rat allogeneic DC, made immature by adenoviral gene transfer of the dominant negative form of IKK2, gave rise in-vitro to a unique population of CD4+CD25- regulatory T cells (dnIKK2-Treg). These cells inhibited Tcell response in-vitro, without needing cell-to-cell contact, and induced kidney allograft survival prolongation in-vivo. Deep insight into the mechanisms behind dnIKK2-Treg-induced suppression of Tcell proliferation remained elusive. Here we document that dnIKK2-Treg release extracellular vesicles (EV) riched in exosomes, fully accounting for the cell-contact independent immunosuppressive activity of parent cells. DnIKK2-Treg-EV contain a unique molecular cargo of specific miRNAs and iNOS, which, once delivered into target cells, blocked cell cycle progression and induced apoptosis. DnIKK2-Treg-EV-exposed T cells were in turn converted into regulatory cells. Notably, when administered in-vivo, dnIKK2-Treg-EV prolonged kidney allograft survival. DnIKK2-Treg-derived EV could be a tool for manipulating the immune system and for discovering novel potential immunosuppressive molecules in the context of allotransplantation.


Asunto(s)
Aloinjertos/fisiología , Vesículas Extracelulares/metabolismo , Tolerancia Inmunológica , Inmunosupresores/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Animales , Ratas
8.
Am J Kidney Dis ; 70(1): 145-150, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28242136

RESUMEN

Dabrafenib and trametinib, BRAF and MEK inhibitors, respectively, are effective targeted metastatic melanoma therapies, but little is known about their nephrotoxicity. Although tubulointerstitial injury has been the most widely reported renal side effect of targeted melanoma therapy, nephrotic syndrome has not been reported before. We report on a patient with metastatic melanoma who developed nephrotic syndrome during dabrafenib and trametinib treatment. Kidney biopsy showed diffuse loss of podocyte cytoarchitecture, extensive foot-process effacement, and glomerular endothelial injury. Kidney function and glomerular ultrastructural changes recovered fully after drug withdrawal. In vitro, BRAF inhibition decreased PLCε1 expression in podocytes, accompanied by a reduction in nephrin expression and an increase in permeability to albumin. Additionally, these drugs inhibited the podocyte-vascular endothelial growth factor (VEGF) system. In addition to implications for nephrotic syndrome pathophysiology, we suggest that patients given dabrafenib and trametinib be monitored closely for potential glomerular damage.


Asunto(s)
Antineoplásicos/efectos adversos , Imidazoles/efectos adversos , Melanoma/tratamiento farmacológico , Síndrome Nefrótico/inducido químicamente , Oximas/efectos adversos , Podocitos/efectos de los fármacos , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Piridonas/efectos adversos , Pirimidinonas/efectos adversos , Transducción de Señal/efectos de los fármacos , Neoplasias Cutáneas/tratamiento farmacológico , Anciano , Femenino , Humanos , Proteínas Proto-Oncogénicas B-raf/fisiología
9.
Stem Cell Res ; 17(1): 130-9, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27299470

RESUMEN

Generating human podocytes in vitro could offer a unique opportunity to study human diseases. Here, we describe a simple and efficient protocol for obtaining functional podocytes in vitro from human induced pluripotent stem cells. Cells were exposed to a three-step protocol, which induced their differentiation into intermediate mesoderm, then into nephron progenitors and, finally, into mature podocytes. After differentiation, cells expressed the main podocyte markers, such as synaptopodin, WT1, α-Actinin-4, P-cadherin and nephrin at the protein and mRNA level, and showed the low proliferation rate typical of mature podocytes. Exposure to Angiotensin II significantly decreased the expression of podocyte genes and cells underwent cytoskeleton rearrangement. Cells were able to internalize albumin and self-assembled into chimeric 3D structures in combination with dissociated embryonic mouse kidney cells. Overall, these findings demonstrate the establishment of a robust protocol that, mimicking developmental stages, makes it possible to derive functional podocytes in vitro.


Asunto(s)
Células Madre Pluripotentes Inducidas/metabolismo , Podocitos/citología , Actinina/genética , Actinina/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Diferenciación Celular , Células Cultivadas , Cuerpos Embrioides/metabolismo , Cuerpos Embrioides/fisiología , Humanos , Inmunohistoquímica , Células Madre Pluripotentes Inducidas/citología , Cariotipo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Microscopía Electrónica de Rastreo , Microscopía Fluorescente , Podocitos/metabolismo , Sinaptofisina/genética , Sinaptofisina/metabolismo
10.
Sci Rep ; 6: 28445, 2016 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-27345360

RESUMEN

Podocyte loss is the initial event in the development of glomerulosclerosis, the structural hallmark of progressive proteinuric nephropathies. Understanding mechanisms underlying glomerular injury is the key challenge for identifying novel therapeutic targets. In mice with protein-overload induced by bovine serum albumin (BSA), we evaluated whether the alternative pathway (AP) of complement mediated podocyte depletion and podocyte-dependent parietal epithelial cell (PEC) activation causing glomerulosclerosis. Factor H (Cfh(-/-)) or factor B-deficient mice were studied in comparison with wild-type (WT) littermates. WT+BSA mice showed podocyte depletion accompanied by glomerular complement C3 and C3a deposits, PEC migration to capillary tuft, proliferation, and glomerulosclerosis. These changes were more prominent in Cfh(-/-) +BSA mice. The pathogenic role of AP was documented by data that factor B deficiency preserved glomerular integrity. In protein-overload mice, PEC dysregulation was associated with upregulation of CXCR4 and GDNF/c-Ret axis. In vitro studies provided additional evidence of a direct action of C3a on proliferation and CXCR4-related migration of PECs. These effects were enhanced by podocyte-derived GDNF. In patients with proteinuric nephropathy, glomerular C3/C3a paralleled PEC activation, CXCR4 and GDNF upregulation. These results indicate that mechanistically uncontrolled AP complement activation is not dispensable for podocyte-dependent PEC activation resulting in glomerulosclerosis.


Asunto(s)
Complemento C3a/metabolismo , Glomeruloesclerosis Focal y Segmentaria/patología , Adulto , Animales , Bovinos , Proliferación Celular , Células Cultivadas , Factor B del Complemento/deficiencia , Factor B del Complemento/genética , Factor H de Complemento/deficiencia , Factor H de Complemento/genética , Modelos Animales de Enfermedad , Células Epiteliales/citología , Células Epiteliales/metabolismo , Femenino , Glomeruloesclerosis Focal y Segmentaria/metabolismo , Humanos , Riñón/metabolismo , Riñón/patología , Riñón/ultraestructura , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Podocitos/citología , Podocitos/metabolismo , Proteinuria/etiología , Albúmina Sérica Bovina/administración & dosificación , Regulación hacia Arriba , Adulto Joven
11.
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
12.
J Am Soc Nephrol ; 27(3): 699-705, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26116358

RESUMEN

Chronic renal insufficiency inexorably progresses in patients, such as it does after partial renal ablation in rats. However, the progression of renal diseases can be delayed by angiotensin II blockers that stabilize renal function or increase GFR, even in advanced phases of the disease. Regression of glomerulosclerosis can be induced by angiotensin II antagonism, but the effect of these treatments on the entire vascular tree is unclear. Here, using microcomputed tomography and scanning electron microscopy, we compared the size and extension of kidney blood vessels in untreated Wistar rats with those in untreated and angiotensin II antagonist-treated Munich Wistar Frömter (MWF) rats that spontaneously develop kidney disease with age. The kidney vasculature underwent progressive rarefaction in untreated MWF rats, substantially affecting intermediate and small vessels. Microarray analysis showed increased Tgf-ß and endothelin-1 gene expression with age. Notably, 10-week inhibition of the renin-angiotensin system regenerated kidney vasculature and normalized Tgf-ß and endothelin-1 gene expression in aged MWF rats. These changes were associated with reduced apoptosis, increased endothelial cell proliferation, and restoration of Nrf2 expression, suggesting mechanisms by which angiotensin II antagonism mediates regeneration of capillary segments. These results have important implications in the clinical setting of chronic renal insufficiency.


Asunto(s)
Antagonistas de Receptores de Angiotensina/farmacología , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Capilares/fisiología , Glomérulos Renales/irrigación sanguínea , Neovascularización Fisiológica/efectos de los fármacos , Insuficiencia Renal Crónica/tratamiento farmacológico , Actinas/metabolismo , Animales , Apoptosis , Capilares/metabolismo , Capilares/ultraestructura , Proliferación Celular , Células Endoteliales/fisiología , Endotelina-1/genética , Expresión Génica , Microscopía Electrónica de Rastreo , Factor 2 Relacionado con NF-E2/metabolismo , Ratas , Ratas Wistar , Sistema Renina-Angiotensina/efectos de los fármacos , Factor de Crecimiento Transformador beta/genética , Microtomografía por Rayos X
13.
PLoS One ; 10(5): e0127172, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25993470

RESUMEN

BACKGROUND: Angiotensin II promotes insulin resistance. The mechanism underlying this abnormality, however, is still poorly defined. In a different setting, skeletal muscle metabolism and insulin signaling are regulated by Sirtuin3. OBJECTIVE: Here, we investigate whether angiotensin II-induced insulin resistance in skeletal muscle is associated with Sirtuin3 dysregulation and whether pharmacological manipulation of Sirtuin3 confers protection. STUDY DESIGN: Parental and GLUT4-myc L6 rat skeletal muscle cells exposed to angiotensin II are used as in vitro models of insulin resistance. GLUT4 translocation, glucose uptake, intracellular molecular signals such as mitochondrial reactive oxygen species, Sirtuin3 protein expression and activity, along with its downstream targets and upstream regulators, are analyzed both in the absence and presence of acetyl-L-carnitine. The role of Sirtuin3 in GLUT4 translocation and intracellular molecular signaling is also studied in Sirtuin3-silenced as well as over-expressing cells. RESULTS: Angiotensin II promotes insulin resistance in skeletal muscle cells via mitochondrial oxidative stress, resulting in a two-fold increase in superoxide generation. In this context, reactive oxygen species open the mitochondrial permeability transition pore and significantly lower Sirtuin3 levels and activity impairing the cell antioxidant defense. Angiotensin II-induced Sirtuin3 dysfunction leads to the impairment of AMP-activated protein kinase/nicotinamide phosphoribosyltransferase signaling. Acetyl-L-carnitine, by lowering angiotensin II-induced mitochondrial superoxide formation, prevents Sirtuin3 dysfunction. This phenomenon implies the restoration of manganese superoxide dismutase antioxidant activity and AMP-activated protein kinase activation. Acetyl-L-carnitine protection is abrogated by specific Sirtuin3 siRNA. CONCLUSIONS: Our data demonstrate that angiotensin II-induced insulin resistance fosters mitochondrial superoxide generation, in turn leading to Sirtuin3 dysfunction. The present results also highlight Sirtuin3 as a therapeutic target for the insulin-sensitizing effects of acetyl-L-carnitine.


Asunto(s)
Angiotensina II/farmacología , Resistencia a la Insulina , Músculo Esquelético/fisiología , Sirtuina 3/fisiología , Acetilcarnitina/farmacología , Animales , Línea Celular , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Músculo Esquelético/efectos de los fármacos , Ratas , Especies Reactivas de Oxígeno/metabolismo
14.
PLoS One ; 10(3): e0120235, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25811887

RESUMEN

New intervention tools for severely damaged kidneys are in great demand to provide patients with a valid alternative to whole organ replacement. For repairing or replacing injured tissues, emerging approaches focus on using stem and progenitor cells. Embryonic kidneys represent an interesting option because, when transplanted to sites such as the renal capsule of healthy animals, they originate new renal structures. Here, we studied whether metanephroi possess developmental capacity when transplanted under the kidney capsule of MWF male rats, a model of spontaneous nephropathy. We found that six weeks post-transplantation, renal primordia developed glomeruli and tubuli able to filter blood and to produce urine in cyst-like structures. Newly developed metanephroi were able to initiate a regenerative-like process in host renal tissues adjacent to the graft in MWF male rats as indicated by an increase in cell proliferation and vascular density, accompanied by mRNA and protein upregulation of VEGF, FGF2, HGF, IGF-1 and Pax-2. The expression of SMP30 and NCAM was induced in tubular cells. Oxidative stress and apoptosis markedly decreased. Our study shows that embryonic kidneys generate functional nephrons when transplanted into animals with severe renal disease and at the same time activate events at least partly mimicking those observed in kidney tissues during renal regeneration.


Asunto(s)
Enfermedades Renales/terapia , Trasplante de Riñón , Regeneración , Animales , Apoptosis , Biomarcadores , Proliferación Celular , Femenino , Fibroblastos/metabolismo , Fibroblastos/trasplante , Expresión Génica , Supervivencia de Injerto , Enfermedades Renales/patología , Enfermedades Renales/fisiopatología , Trasplante de Riñón/métodos , Masculino , Neovascularización Fisiológica , Estrés Oxidativo , Ratas , Regeneración/genética
15.
Stem Cell Reports ; 4(4): 685-98, 2015 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-25754206

RESUMEN

The application of cell-based therapies in regenerative medicine is gaining recognition. Here, we show that human bone marrow stromal cells (BMSCs), also known as bone-marrow-derived mesenchymal cells, can be reprogrammed into renal proximal tubular-like epithelial cells using cell-free extracts. Streptolysin-O-permeabilized BMSCs exposed to HK2-cell extracts underwent morphological changes-formation of "domes" and tubule-like structures-and acquired epithelial functional properties such as transepithelial-resistance, albumin-binding, and uptake and specific markers E-cadherin and aquaporin-1. Transmission electron microscopy revealed the presence of brush border microvilli and tight intercellular contacts. RNA sequencing showed tubular epithelial transcript abundance and revealed the upregulation of components of the EGFR pathway. Reprogrammed BMSCs integrated into self-forming kidney tissue and formed tubular structures. Reprogrammed BMSCs infused in immunodeficient mice with cisplatin-induced acute kidney injury engrafted into proximal tubuli, reduced renal injury and improved function. Thus, reprogrammed BMSCs are a promising cell resource for future cell therapy.


Asunto(s)
Diferenciación Celular , Reprogramación Celular , Riñón/citología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Animales , Línea Celular Transformada , Sistema Libre de Células , Femenino , Perfilación de la Expresión Génica , Humanos , Técnicas In Vitro , Túbulos Renales Proximales/citología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/ultraestructura , Ratones , Ratones Endogámicos NOD , Ratones SCID , Transcriptoma
16.
J Clin Invest ; 125(2): 715-26, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25607838

RESUMEN

Acute kidney injury (AKI) is a public health concern with an annual mortality rate that exceeds those of breast and prostate cancer, heart failure, and diabetes combined. Oxidative stress and mitochondrial damage are drivers of AKI-associated pathology; however, the pathways that mediate these events are poorly defined. Here, using a murine cisplatin-induced AKI model, we determined that both oxidative stress and mitochondrial damage are associated with reduced levels of renal sirtuin 3 (SIRT3). Treatment with the AMPK agonist AICAR or the antioxidant agent acetyl-l-carnitine (ALCAR) restored SIRT3 expression and activity, improved renal function, and decreased tubular injury in WT animals, but had no effect in Sirt3-/- mice. Moreover, Sirt3-deficient mice given cisplatin experienced more severe AKI than WT animals and died, and neither AICAR nor ALCAR treatment prevented death in Sirt3-/- AKI mice. In cultured human tubular cells, cisplatin reduced SIRT3, resulting in mitochondrial fragmentation, while restoration of SIRT3 with AICAR and ALCAR improved cisplatin-induced mitochondrial dysfunction. Together, our results indicate that SIRT3 is protective against AKI and suggest that enhancing SIRT3 to improve mitochondrial dynamics has potential as a strategy for improving outcomes of renal injury.


Asunto(s)
Lesión Renal Aguda/enzimología , Túbulos Renales Proximales/enzimología , Mitocondrias/metabolismo , Dinámicas Mitocondriales , Sirtuina 3/metabolismo , Acetilcarnitina/farmacología , Lesión Renal Aguda/genética , Lesión Renal Aguda/patología , Animales , Antineoplásicos/farmacología , Antioxidantes/farmacología , Línea Celular , Cisplatino/farmacocinética , Humanos , Túbulos Renales Proximales/patología , Ratones , Ratones Noqueados , Mitocondrias/genética , Mitocondrias/patología , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética , Sirtuina 3/genética , Complejo Vitamínico B/farmacología
17.
J Am Soc Nephrol ; 25(3): 523-33, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24371298

RESUMEN

Activation of endothelin-A receptor (ET(A)R) by endothelin-1 (ET-1) drives epithelial-to-mesenchymal transition in ovarian tumor cells through ß-arrestin signaling. Here, we investigated whether this pathogenetic pathway could affect podocyte phenotype in proliferative glomerular disorders. In cultured mouse podocytes, ET-1 caused loss of the podocyte differentiation marker synaptopodin and acquisition of the mesenchymal marker α-smooth muscle actin. ET-1 promoted podocyte migration via ET(A)R activation and increased ß-arrestin-1 expression. Activated ET(A)R recruited ß-arrestin-1 to form a trimeric complex with Src leading to epithelial growth factor receptor (EGFR) transactivation and ß-catenin phosphorylation, which promoted gene transcription of Snail. Increased Snail expression fostered ET-1-induced migration as confirmed by Snail knockdown experiments. Silencing of ß-arrestin-1 prevented podocyte phenotypic changes and motility and inhibited ET(A)R-driven signaling. In vitro findings were confirmed in doxorubicin (Adriamycin)-induced nephropathy. Mice receiving Adriamycin developed renal injury with loss of podocytes and hyperplastic lesion formation; ß-arrestin-1 expression increased in visceral podocytes and in podocytes entrapped in pseudo-crescents. Administration of the selective ET(A)R antagonist sitaxsentan prevented podocyte loss, formation of the hyperplastic lesions, and normalized expression of glomerular ß-arrestin-1 and Snail. Increased ß-arrestin-1 levels in podocytes retrieved from crescents of patients with proliferative glomerulopathies confirmed the translational relevance of these findings and suggest the therapeutic potential of ET(A)R antagonism for a group of diseases still needing a specific treatment.


Asunto(s)
Arrestinas/fisiología , Endotelina-1/metabolismo , Glomerulonefritis/inducido químicamente , Podocitos/fisiología , Receptor de Endotelina A/metabolismo , Animales , Movimiento Celular , Modelos Animales de Enfermedad , Doxorrubicina , Receptores ErbB/metabolismo , Femenino , Glomerulonefritis/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Persona de Mediana Edad , Factores de Transcripción de la Familia Snail , Factores de Transcripción/metabolismo , Activación Transcripcional , beta Catenina/metabolismo , beta-Arrestina 1 , beta-Arrestinas , Familia-src Quinasas/metabolismo
18.
Am J Pathol ; 183(1): 119-30, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23707238

RESUMEN

In nondiabetic rat models of renal disease, angiotensin II (Ang II) perpetuates podocyte injury and promotes progression to end-stage kidney disease. Herein, we wanted to explore the role of Ang II in diabetic nephropathy by a translational approach spanning from in vitro to in vivo rat and human studies, and to dissect the intracellular pathways involved. In isolated perfused rat kidneys and in cultured human podocytes, Ang II down-regulated nephrin expression via Notch1 activation and nuclear translocation of Snail. Hairy enhancer of split-1 was a Notch1-downstream gene effector that activated Snail in cultured podocytes. In vitro changes of the Snail/nephrin axis were similar to those in renal biopsy specimens of Zucker diabetic fatty rats and patients with advanced diabetic nephropathy, and were normalized by pharmacological inhibition of the renin-angiotensin system. Collectively, the present studies provide evidence that Ang II plays a relevant role in perpetuating glomerular injury in experimental and human diabetic nephropathy via persistent activation of Notch1 and Snail signaling in podocytes, eventually resulting in down-regulation of nephrin expression, the integrity of which is crucial for the glomerular filtration barrier.


Asunto(s)
Angiotensina II/metabolismo , Nefropatías Diabéticas/metabolismo , Proteínas de la Membrana/metabolismo , Receptor Notch1/metabolismo , Factores de Transcripción/metabolismo , Anciano , Animales , Estudios de Casos y Controles , Células Cultivadas , Diabetes Mellitus Tipo 2/complicaciones , Regulación hacia Abajo , Femenino , Humanos , Inmunohistoquímica , Riñón/metabolismo , Riñón/patología , Modelos Lineales , Masculino , Microscopía Electrónica de Transmisión , Persona de Mediana Edad , Ratas , Ratas Sprague-Dawley , Ratas Zucker , Reacción en Cadena en Tiempo Real de la Polimerasa , Factores de Transcripción de la Familia Snail
19.
Stem Cells Dev ; 22(5): 772-80, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23082760

RESUMEN

Bone marrow-mesenchymal stem cells (BM-MSC) ameliorate renal dysfunction and repair tubular damage of acute kidney injury by locally releasing growth factors, including the insulin-like growth factor-1 (IGF-1). The restricted homing of BM-MSC at the site of injury led us to investigate a possible gene-based communication mechanism between BM-MSC and tubular cells. Human BM-MSC (hBM-MSC) released microparticles and exosomes (Exo) enriched in mRNAs. A selected pattern of transcripts was detected in Exo versus parental cells. Exo expressed the IGF-1 receptor (IGF-1R), but not IGF-1 mRNA, while hBM-MSC contained both mRNAs. R- cells lacking IGF-1R exposed to hBM-MSC-derived Exo acquired the human IGF-1R transcript that was translated in the corresponding protein. Transfer of IGF-1R mRNA from Exo to cisplatin-damaged proximal tubular cells (proximal tubular epithelial cell [PTEC]) increased PTEC proliferation. Coincubation of damaged PTEC with Exo and soluble IGF-1 further enhanced cell proliferation. These findings suggest that horizontal transfer of the mRNA for IGF-1R to tubular cells through Exo potentiates tubular cell sensitivity to locally produced IGF-1 providing a new mechanism underlying the powerful renoprotection of few BM-MSC observed in vivo.


Asunto(s)
Exosomas , Transferencia de Gen Horizontal , Células Madre Mesenquimatosas/fisiología , ARN Mensajero/genética , Receptores de Factores de Crecimiento/genética , Lesión Renal Aguda/genética , Lesión Renal Aguda/metabolismo , Adulto , Animales , Células de la Médula Ósea/metabolismo , Comunicación Celular , Proliferación Celular , Células Cultivadas , Humanos , Factor I del Crecimiento Similar a la Insulina/genética , Túbulos Renales Proximales/metabolismo , Ratones , Ratones Endogámicos C57BL , Interferencia de ARN , ARN Interferente Pequeño
20.
Stem Cells ; 28(3): 513-22, 2010 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-20049901

RESUMEN

In search for new sources of mesenchymal stem cells (MSCs) for renal repair in acute kidney injury (AKI), we investigated the potential of human cord blood (CB)-MSCs to cure mice with AKI. Infusion of CB-MSCs in immunodeficient mice with cisplatin-induced AKI ameliorated both renal function and tubular cell injury, and prolonged survival. Transplanted CB-MSCs localized in peritubular areas, limited capillary alterations and neutrophil infiltration. Apoptosis reduced and tubular cell proliferation increased by virtue of stem cell capacity to produce growth factors. The reno-protective effect of CB-MSCs was further confirmed by their ability to inhibit oxidative damage and to induce the prosurvival factor Akt in tubular cells. The evidence that CB-MSCs in vitro increased the production of growth factors and inhibited IL-1 beta and TNFalpha synthesis when cocultured with damaged proximal tubular cells indicates a regenerative and anti-inflammatory action of stem cell treatment. Altogether these results highlight the potential of human CB-MSCs as future cell therapy for testing in human AKI.


Asunto(s)
Diferenciación Celular/fisiología , Trasplante de Células Madre de Sangre del Cordón Umbilical/métodos , Supervivencia de Injerto/fisiología , Enfermedades Renales/cirugía , Riñón/cirugía , Trasplante de Células Madre Mesenquimatosas/métodos , Enfermedad Aguda , Animales , Antiinflamatorios/metabolismo , Apoptosis/fisiología , Técnicas de Cultivo de Célula , Proliferación Celular , Técnicas de Cocultivo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Riñón/patología , Riñón/fisiopatología , Enfermedades Renales/fisiopatología , Túbulos Renales/citología , Túbulos Renales/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones SCID , Estrés Oxidativo/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...