Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 91
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Kidney Int ; 96(4): 971-982, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31285081

RESUMEN

The clinical heterogeneity of idiopathic nephrotic syndrome in childhood may reflect different mechanisms of disease that are as yet unclear. Here, we evaluated the association between an atypical presence of IgM on the surface of T cells (T-cell IgM) and the response to steroid therapy in a total of 153 pediatric patients with idiopathic nephrotic syndrome in different phases of disease. At disease onset, T-cell IgM median levels were significantly elevated and predictive of risk of relapse in 47 patients. They were also significantly increased comparing 58 steroid-dependent to 8 infrequently relapsing and 14 frequently relapsing patients, especially during relapse, whereas they were within the normal range in 7 genetic steroid-resistant patients. T-cell IgM in vivo was not affected by the amount of total circulating IgM, nor by concomitant acute infections or oral immunosuppression. However, it was affected by rituximab treatment in 21 steroid-dependent patients. By in vitro experiments, elevated T-cell IgM was not influenced by total circulating IgM levels or by the presence of other circulating factors, and there was no distinctive antigen-specificity or atypical IgM polymerization. Rather, we found that increased T-cell IgM correlates with reduced IgM sialylation, which influences T-cell response to steroid inhibition and T-cell production of podocyte-damaging factors. Thus, the atypical presence of IgM on the surface of T cells may predispose a subset of steroid-sensitive pediatric patients with idiopathic nephrotic syndrome to a poor response to steroid therapy since disease onset.


Asunto(s)
Glucocorticoides/farmacología , Inmunoglobulina M/metabolismo , Síndrome Nefrótico/tratamiento farmacológico , Linfocitos T/inmunología , Adolescente , Niño , Preescolar , Resistencia a Medicamentos/genética , Quimioterapia Combinada/métodos , Femenino , Estudios de Seguimiento , Glucocorticoides/uso terapéutico , Humanos , Inmunoglobulina M/análisis , Inmunoglobulina M/inmunología , Lactante , Masculino , Síndrome Nefrótico/sangre , Síndrome Nefrótico/genética , Síndrome Nefrótico/inmunología , Podocitos , Estudios Prospectivos , Recurrencia , Rituximab/farmacología , Rituximab/uso terapéutico , Ácidos Siálicos/metabolismo , Linfocitos T/metabolismo , Resultado del Tratamiento
2.
J Am Soc Nephrol ; 28(7): 2144-2157, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28270414

RESUMEN

Mammalian target of rapamycin (mTOR) signaling is involved in a variety of kidney diseases. Clinical trials administering mTOR inhibitors to patients with FSGS, a prototypic podocyte disease, led to conflicting results, ranging from remission to deterioration of kidney function. Here, we combined complex genetic titration of mTOR complex 1 (mTORC1) levels in murine glomerular disease models, pharmacologic studies, and human studies to precisely delineate the role of mTOR in FSGS. mTORC1 target genes were significantly induced in microdissected glomeruli from both patients with FSGS and a murine FSGS model. Furthermore, a mouse model with constitutive mTORC1 activation closely recapitulated human FSGS. Notably, the complete knockout of mTORC1 by induced deletion of both Raptor alleles accelerated the progression of murine FSGS models. However, lowering mTORC1 signaling by deleting just one Raptor allele ameliorated the progression of glomerulosclerosis. Similarly, low-dose treatment with the mTORC1 inhibitor rapamycin efficiently diminished disease progression. Mechanistically, complete pharmacologic inhibition of mTOR in immortalized podocytes shifted the cellular energy metabolism toward reduced rates of oxidative phosphorylation and anaerobic glycolysis, which correlated with increased production of reactive oxygen species. Together, these data suggest that podocyte injury and loss is commonly followed by adaptive mTOR activation. Prolonged mTOR activation, however, results in a metabolic podocyte reprogramming leading to increased cellular stress and dedifferentiation, thus offering a treatment rationale for incomplete mTOR inhibition.


Asunto(s)
Glomeruloesclerosis Focal y Segmentaria/etiología , Glomeruloesclerosis Focal y Segmentaria/prevención & control , Inmunosupresores/farmacología , Inmunosupresores/uso terapéutico , Complejos Multiproteicos/antagonistas & inhibidores , Complejos Multiproteicos/fisiología , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología , Sirolimus/uso terapéutico , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/fisiología , Animales , Progresión de la Enfermedad , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones
3.
Genome Res ; 23(11): 1862-73, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23950145

RESUMEN

Cell-lineage-specific transcripts are essential for differentiated tissue function, implicated in hereditary organ failure, and mediate acquired chronic diseases. However, experimental identification of cell-lineage-specific genes in a genome-scale manner is infeasible for most solid human tissues. We developed the first genome-scale method to identify genes with cell-lineage-specific expression, even in lineages not separable by experimental microdissection. Our machine-learning-based approach leverages high-throughput data from tissue homogenates in a novel iterative statistical framework. We applied this method to chronic kidney disease and identified transcripts specific to podocytes, key cells in the glomerular filter responsible for hereditary and most acquired glomerular kidney disease. In a systematic evaluation of our predictions by immunohistochemistry, our in silico approach was significantly more accurate (65% accuracy in human) than predictions based on direct measurement of in vivo fluorescence-tagged murine podocytes (23%). Our method identified genes implicated as causal in hereditary glomerular disease and involved in molecular pathways of acquired and chronic renal diseases. Furthermore, based on expression analysis of human kidney disease biopsies, we demonstrated that expression of the podocyte genes identified by our approach is significantly related to the degree of renal impairment in patients. Our approach is broadly applicable to define lineage specificity in both cell physiology and human disease contexts. We provide a user-friendly website that enables researchers to apply this method to any cell-lineage or tissue of interest. Identified cell-lineage-specific transcripts are expected to play essential tissue-specific roles in organogenesis and disease and can provide starting points for the development of organ-specific diagnostics and therapies.


Asunto(s)
Linaje de la Célula , Biología Computacional/métodos , Enfermedades Renales/etiología , Podocitos/metabolismo , Insuficiencia Renal Crónica/genética , Animales , Inteligencia Artificial , Biopsia , Diferenciación Celular/genética , Simulación por Computador , Bases de Datos Genéticas , Perfilación de la Expresión Génica , Genoma Humano , Humanos , Enfermedades Renales/genética , Enfermedades Renales/patología , Ratones , Nanotecnología , Especificidad de Órganos/genética , Organogénesis/genética , Podocitos/citología , Podocitos/patología , Insuficiencia Renal Crónica/patología
4.
J Pathol ; 235(5): 731-44, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25408545

RESUMEN

Idiopathic focal segmental glomerulosclerosis (FSGS) is a progressive and proteinuric kidney disease that starts with podocyte injury. Podocytes cover the external side of the glomerular capillary by a complex web of primary and secondary ramifications. Similar to dendritic spines of neuronal cells, podocyte processes rely on a dynamic actin-based cytoskeletal architecture to maintain shape and function. Brain-derived neurotrophic factor (BDNF) is a pleiotropic neurotrophin that binds to the tropomyosin-related kinase B receptor (TrkB) and has crucial roles in neuron maturation, survival, and activity. In neuronal cultures, exogenously added BDNF increases the number and size of dendritic spines. In animal models, BDNF administration is beneficial in both central and peripheral nervous system disorders. Here we show that BDNF has a TrkB-dependent trophic activity on podocyte cell processes; by affecting microRNA-134 and microRNA-132 signalling, BDNF up-regulates Limk1 translation and phosphorylation, and increases cofilin phosphorylation, which results in actin polymerization. Importantly, BDNF effectively repairs podocyte damage in vitro, and contrasts proteinuria and glomerular lesions in in vivo models of FSGS, opening a potential new perspective to the treatment of podocyte disorders.


Asunto(s)
Citoesqueleto de Actina/efectos de los fármacos , Actinas/metabolismo , Factor Neurotrófico Derivado del Encéfalo/farmacología , Glomeruloesclerosis Focal y Segmentaria/prevención & control , MicroARNs/metabolismo , Podocitos/efectos de los fármacos , Células 3T3 , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/patología , Factores Despolimerizantes de la Actina/metabolismo , Animales , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Glomeruloesclerosis Focal y Segmentaria/inducido químicamente , Glomeruloesclerosis Focal y Segmentaria/genética , Glomeruloesclerosis Focal y Segmentaria/metabolismo , Glomeruloesclerosis Focal y Segmentaria/patología , Larva/efectos de los fármacos , Larva/metabolismo , Quinasas Lim/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , MicroARNs/genética , Fosforilación , Podocitos/metabolismo , Podocitos/patología , Polimerizacion , Proteinuria/metabolismo , Proteinuria/prevención & control , Interferencia de ARN , Receptor trkB/metabolismo , Proteínas Recombinantes/farmacología , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Transfección , Pez Cebra
5.
Int J Mol Sci ; 17(10)2016 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-27754425

RESUMEN

Warm renal ischemia performed during partial nephrectomy has been found to be associated with kidney disease. Since endogenous ouabain (EO) is a neuro-endocrine hormone involved in renal damage, we evaluated the role of EO in renal ischemia-reperfusion injury (IRI). We measured plasma and renal EO variations and markers of glomerular and tubular damage (nephrin, KIM-1, Kidney-Injury-Molecule-1, α1 Na-K ATPase) and the protective effect of the ouabain inhibitor, rostafuroxin. We studied five groups of rats: (1) normal; (2) infused for eight weeks with ouabain (30 µg/kg/day, OHR) or (3) saline; (4) ouabain; or (5) saline-infused rats orally treated with 100 µg/kg/day rostafuroxin for four weeks. In group 1, 2-3 h after IRI, EO increased in ischemic kidneys while decreased in plasma. Nephrin progressively decreased and KIM-1 mRNA increased starting from 24 h. Ouabain infusion (group 2) increased blood pressure (from 111.7 to 153.4 mmHg) and ouabain levels in plasma and kidneys. In OHR ischemic kidneys at 120 h from IRI, nephrin, and KIM-1 changes were greater than those detected in the controls infused with saline (group 3). All these changes were blunted by rostafuroxin treatment (groups 4 and 5). These findings support the role of EO in IRI and suggest that rostafuroxin pre-treatment of patients before partial nephrectomy with warm ischemia may reduce IRI, particularly in those with high EO.


Asunto(s)
Androstanoles/uso terapéutico , Enfermedades Renales/tratamiento farmacológico , Enfermedades Renales/metabolismo , Riñón/efectos de los fármacos , Ouabaína/metabolismo , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/metabolismo , Animales , Riñón/metabolismo , Riñón/patología , Enfermedades Renales/sangre , Enfermedades Renales/patología , Nefrectomía , Ouabaína/sangre , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/sangre , Daño por Reperfusión/patología
6.
Curr Opin Nephrol Hypertens ; 24(3): 239-44, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-26066473

RESUMEN

PURPOSE OF REVIEW: Podocytes are the main gatekeeper of protein filtration in the glomerulus. When podocytes work less efficiently, this translates to the appearance of proteins in the urine, a condition that, if not promptly treated, leads to progression of glomerular damage and renal failure. RECENT FINDINGS: Novel gene mutations have been uncovered in patients with nephrotic syndrome combined with a better definition of the role of podocin mutations. Although the importance of the inflammasome pathway and of the mechanisms of autophagy in podocyte health and disease have been increasingly recognized, a precise relationship between these processes still needs to be assessed. Numerous potential therapeutic targets have been identified and numerous data support the possibility of boosting podocyte regeneration. However, translation of experimental results into the clinic could largely depend on the avoidance of undesired side-effects; nanomedicine could provide the means to target old and novel drugs specifically to the podocytes. SUMMARY: Podocytes are key cells in the glomerulus, and their damage inevitably leads to proteinuria and glomerular dysfunction. The more is known about the causes and mechanisms of podocyte damage, the more it will be possible to find new cures for glomerular diseases of the kidney.


Asunto(s)
Enfermedades Renales/terapia , Glomérulos Renales/lesiones , Podocitos/metabolismo , Proteinuria/metabolismo , Regeneración/fisiología , Animales , Autofagia/fisiología , Humanos , Enfermedades Renales/metabolismo , Glomérulos Renales/metabolismo
7.
Nephrol Dial Transplant ; 30(12): 1965-71, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25829327

RESUMEN

The incidence and prevalence of chronic kidney disease represents an important problem for public health. In renal diseases, the main histologic alterations derive from the development of renal fibrosis which results from the loss of the balance between pro- and anti-fibrotic factors. Tyrosine kinase receptors (RTKs) and matricellular proteins (MPs) are nowadays studied as potential modulators of renal injury. RTKs regulate cell cycle, migration, metabolism and cellular differentiation. Discoidin domain receptor-1 (DDR-1) is an RTK that has been extensively studied in cancer, and lung and renal diseases. It modulates inflammatory recruitment, extracellular matrix deposition and fibrosis; in renal diseases, it appears to act independently of the underlying disease. MPs regulate cell-matrix interactions and matrix accumulation, cellular adhesion and migration, and expression of inflammatory cells. Periostin is an MP, mainly studied in bone, heart, lung and cancer. Several studies demonstrated that it mediates cell-matrix interactions, migration of inflammatory cells and development of fibrosis. Recently, it has been reported in several nephropathies. In this review, we discuss the potential pathological roles of DDR-1 and periostin focussing on the kidney in both experimental models and human diseases.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Insuficiencia Renal Crónica/metabolismo , Receptor con Dominio Discoidina 1 , Humanos
8.
J Am Soc Nephrol ; 25(7): 1415-29, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24676639

RESUMEN

Podocyte injury and resulting albuminuria are hallmarks of diabetic nephropathy, but targeted therapies to halt or prevent these complications are currently not available. Here, we show that the immune-related molecule B7-1/CD80 is a critical mediator of podocyte injury in type 2 diabetic nephropathy. We report the induction of podocyte B7-1 in kidney biopsy specimens from patients with type 2 diabetes. Genetic and epidemiologic studies revealed the association of two single nucleotide polymorphisms at the B7-1 gene with diabetic nephropathy. Furthermore, increased levels of the soluble isoform of the B7-1 ligand CD28 correlated with the progression to ESRD in individuals with type 2 diabetes. In vitro, high glucose conditions prompted the phosphatidylinositol 3 kinase-dependent upregulation of B7-1 in podocytes, and the ectopic expression of B7-1 in podocytes increased apoptosis and induced disruption of the cytoskeleton that were reversed by the B7-1 inhibitor CTLA4-Ig. Podocyte expression of B7-1 was also induced in vivo in two murine models of diabetic nephropathy, and treatment with CTLA4-Ig prevented increased urinary albumin excretion and improved kidney pathology in these animals. Taken together, these results identify B7-1 inhibition as a potential therapeutic strategy for the prevention or treatment of diabetic nephropathy.


Asunto(s)
Antígeno B7-1/fisiología , Diabetes Mellitus Tipo 1/complicaciones , Nefropatías Diabéticas/etiología , Podocitos , Adulto , Anciano , Animales , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Regulación hacia Arriba
9.
Kidney Int ; 86(5): 979-90, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24827776

RESUMEN

A functionally active endocannabinoid system is present within the kidney. The cannabinoid receptor type 2 (CB2) is expressed by both inflammatory cells and podocytes, and its activation has beneficial effects in experimental diabetic nephropathy. To further explore the role of CB2 in diabetic nephropathy, we studied renal functional and structural abnormalities in streptozotocin-induced diabetic CB2 knockout mice. In diabetic mice, deletion of the CB2 receptor albuminuria, the downregulation of podocin and nephrin, mesangial expansion, overexpression of extracellular matrix components, monocyte infiltration, and reduced renal function were all exacerbated. To investigate the relative contributions of podocytes and monocytes to the phenotype of diabetic knockout mice, bone marrow transplantation experiments were performed. The lack of CB2 on bone marrow-derived cells was shown to be important in driving the enhanced glomerular monocyte accrual found in diabetic knockout mice. Absence of CB2 on resident glomerular cells had a major role in worsening diabetic nephropathy, both functional and structural abnormalities, likely by enhanced MCP-1 and CB1 signaling. Studies in cultured podocytes demonstrated that CB2 expression is not altered by a high glucose milieu but is downregulated by mechanical stretch, mimicking glomerular capillary hypertension. Thus, CB2 deletion worsens diabetic nephropathy, independent of bone marrow-derived cells.


Asunto(s)
Diabetes Mellitus Experimental/complicaciones , Nefropatías Diabéticas/etiología , Glomérulos Renales/metabolismo , Receptor Cannabinoide CB2/deficiencia , Estreptozocina , Acetilglucosamina/orina , Albuminuria/etiología , Albuminuria/metabolismo , Animales , Trasplante de Médula Ósea , Línea Celular , Proliferación Celular , Quimiocina CCL2/metabolismo , Quimiotaxis de Leucocito , Creatinina/sangre , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Experimental/fisiopatología , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/patología , Nefropatías Diabéticas/fisiopatología , Matriz Extracelular/metabolismo , Femenino , Glomérulos Renales/patología , Glomérulos Renales/fisiopatología , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/metabolismo , Podocitos/metabolismo , Receptor Cannabinoide CB2/genética , Receptores CCR2/metabolismo , Factores de Tiempo
10.
J Pharmacol Exp Ther ; 351(2): 278-87, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25187430

RESUMEN

Glomerulopathies are important causes of morbidity and mortality. Selective therapies that address the underlying mechanisms are still lacking. Recently, two mechanisms, mutant ß-adducin and ouabain, have been found to be involved in glomerular podocytopathies and proteinuria through nephrin downregulation. The main purpose of the present study was to investigate whether rostafuroxin, a novel antihypertensive agent developed as a selective inhibitor of Src-SH2 interaction with mutant adducin- and ouabain-activated Na,K-ATPase, may protect podocytes from adducin- and ouabain-induced effects, thus representing a novel pharmacologic approach for the therapy of podocytopathies and proteinuria caused by the aforementioned mechanisms. To study the effect of rostafuroxin on podocyte protein changes and proteinuria, mice carrying mutant ß-adducin and ouabain hypertensive rats were orally treated with 100 µg/kg per day rostafuroxin. Primary podocytes from congenic rats carrying mutant α-adducin or ß-adducin (NB) from Milan hypertensive rats and normal rat podocytes incubated with 10(-9) M ouabain were cultured with 10(-9) M rostafuroxin. The results indicated that mutant ß-adducin and ouabain caused podocyte nephrin loss and proteinuria in animal models. These alterations were reproduced in primary podocytes from NB rats and normal rats incubated with ouabain. Treatment of animals, or incubation of cultured podocytes with rostafuroxin, reverted mutant ß-adducin- and ouabain-induced effects on nephrin protein expression and proteinuria. We conclude that rostafuroxin prevented podocyte lesions and proteinuria due to mutant ß-adducin and ouabain in animal models. This suggests a potential therapeutic effect of rostafuroxin in patients with glomerular disease progression associated with these two mechanisms.


Asunto(s)
Androstanoles/farmacología , Proteínas de Unión a Calmodulina/metabolismo , Variación Genética/genética , Ouabaína/efectos adversos , Podocitos/efectos de los fármacos , Proteinuria/tratamiento farmacológico , Animales , Antihipertensivos/farmacología , Proteínas de Unión a Calmodulina/genética , Modelos Animales de Enfermedad , Femenino , Hipertensión/inducido químicamente , Hipertensión/tratamiento farmacológico , Hipertensión/genética , Hipertensión/metabolismo , Riñón/efectos de los fármacos , Riñón/metabolismo , Masculino , Ratones , Ratones Noqueados , Podocitos/metabolismo , Proteinuria/inducido químicamente , Proteinuria/genética , Proteinuria/metabolismo , Ratas , Ratas Sprague-Dawley , ATPasa Intercambiadora de Sodio-Potasio/metabolismo
11.
Curr Opin Nephrol Hypertens ; 23(3): 239-44, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24709948

RESUMEN

PURPOSE OF REVIEW: Segmental glomerulosclerosis is the end-point of a series of processes with have podocyte damage as a common denominator. This review summarizes the important advances that have been made in the past 2 years leading to the comprehension of several molecular mechanisms of regulation of podocyte physiology and pathology. RECENT FINDINGS: From recent studies it has become clear that the dynamic cytoskeleton of podocyte foot processes has to be highly regulated to maintain cell shape and function. The importance of intracellular calcium in this process has started to be revealed, together with the channels and the organelles appointed to calcium entry and buffering.Novel data highlight the centrality and the complexity of the mammalian target of rapamycin pathways, which are implicated in the regulation of autophagy. Similarities between podocytes and neuronal cells have been extended to the process of dynamin-regulated endocytosis, and further data in mice and humans provide support to the idea that podocytes can be directly targeted by old and new drugs. SUMMARY: Research is bringing numerous advances regarding the role of podocytes in the development of glomerulosclerosis, which can lead to novel and specific therapeutic approaches, as well as to a more rational use of drugs already in use. Consequently, renal biopsy becomes the indispensable instrument not only for diagnosis but also to precisely detect molecular therapeutic targets and guide personalized therapy.


Asunto(s)
Glomeruloesclerosis Focal y Segmentaria/patología , Podocitos/patología , Animales , Biopsia , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Glomeruloesclerosis Focal y Segmentaria/tratamiento farmacológico , Glomeruloesclerosis Focal y Segmentaria/genética , Glomeruloesclerosis Focal y Segmentaria/metabolismo , Glomeruloesclerosis Focal y Segmentaria/fisiopatología , Humanos , Ratones , Terapia Molecular Dirigida , Podocitos/efectos de los fármacos , Podocitos/metabolismo , Pronóstico , Factores de Riesgo , Transducción de Señal
12.
Cereb Cortex ; 23(9): 2179-89, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22791805

RESUMEN

The metabotropic glutamate type 1 (mGlu1) and type 5 (mGlu5) receptors, the only members of group I mGlu receptors, are implicated in synaptic plasticity and mechanisms of feedback control of glutamate release. They exhibit nearly complementary distributions throughout the central nervous system, well evident in the cerebellum, where mGlu1 receptor is most intensely expressed while mGlu5 receptor is not. Despite their different distribution, they show a similar subcellular localization and use common transducing pathways. We recently described the Grm1(crv4) mouse with motor coordination deficits and renal anomalies caused by a spontaneous mutation inactivating the mGlu1 receptor. To define the neuropathological mechanisms in these mice, we evaluated expression and function of the mGlu5 receptor in cerebral and cerebellar cortices. Western blot and immunofluorescence analyses showed mGlu5 receptor overexpression. Quantitative reverse transcriptase-polymerase chain reaction results indicated that the up-regulation is already evident at RNA level. Functional studies confirmed an enhanced glutamate release from cortical cerebral and cerebellar synaptosomes when compared with wild-type that is abolished by the mGlu5 receptor-specific inhibitor, 2-methyl-6-(phenylethynyl) pyridine hydrochloride (MPEP). Finally, acute MPEP treatment of Grm1(crv4/crv4) mice induced an evident although incomplete improvement of motor coordination, suggesting that mGlu5 receptors enhanced activity worsens, instead of improving, the motor-coordination defects in the Grm1(crv4/crv4) mice.


Asunto(s)
Encéfalo/fisiopatología , Trastornos del Movimiento/fisiopatología , Receptor del Glutamato Metabotropico 5/fisiología , Receptores de Glutamato Metabotrópico/deficiencia , Animales , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Antagonistas de Aminoácidos Excitadores/farmacología , Femenino , Ácido Glutámico/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Mutación , Piridinas/farmacología , Receptor del Glutamato Metabotropico 5/antagonistas & inhibidores , Receptor del Glutamato Metabotropico 5/metabolismo , Sinaptosomas/fisiología
13.
J Immunol ; 187(3): 1304-13, 2011 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-21724994

RESUMEN

FcγRIIB-deficient mice generated in 129 background (FcγRIIB(129)(-/-)) if back-crossed into C57BL/6 background exhibit a hyperactive phenotype and develop lethal lupus. Both in mice and humans, the Fcγr2b gene is located within a genomic interval on chromosome 1 associated with lupus susceptibility. In mice, the 129-derived haplotype of this interval, named Sle16, causes loss of self-tolerance in the context of the B6 genome, hampering the analysis of the specific contribution of FcγRIIB deficiency to the development of lupus in FcγRIIB(129)(-/-) mice. Moreover, in humans genetic linkage studies revealed contradictory results regarding the association of "loss of function" mutations in the Fcγr2b gene and susceptibility to systemic lupus erythematosis. In this study, we demonstrate that FcγRIIB(-/-) mice generated by gene targeting in B6-derived ES cells (FcγRIIB(B6)(-/-)), lacking the 129-derived flanking Sle16 region, exhibit a hyperactive phenotype but fail to develop lupus indicating that in FcγRIIB(129)(-/-) mice, not FcγRIIB deficiency but epistatic interactions between the C57BL/6 genome and the 129-derived Fcγr2b flanking region cause loss of tolerance. The contribution to the development of autoimmune disease by the resulting autoreactive B cells is amplified by the absence of FcγRIIB, culminating in lethal lupus. In the presence of the Yaa lupus-susceptibility locus, FcγRIIB(B6)(-/-) mice do develop lethal lupus, confirming that FcγRIIB deficiency only amplifies spontaneous autoimmunity determined by other loci.


Asunto(s)
Predisposición Genética a la Enfermedad/prevención & control , Inmunoglobulina G/metabolismo , Nefritis Lúpica/genética , Nefritis Lúpica/inmunología , Receptores de IgG/fisiología , Animales , Células Cultivadas , Cruzamientos Genéticos , Modelos Animales de Enfermedad , Células Madre Embrionarias/inmunología , Células Madre Embrionarias/metabolismo , Femenino , Marcación de Gen , Humanos , Inmunofenotipificación , Nefritis Lúpica/prevención & control , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Receptores de IgG/deficiencia , Receptores de IgG/genética
14.
Nat Med ; 12(9): 1081-7, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16906157

RESUMEN

Rapidly progressive glomerulonephritis (RPGN) is a clinical syndrome characterized by loss of renal function within days to weeks and by glomerular crescents on biopsy. The pathogenesis of this disease is unclear, but circulating factors are believed to have a major role. Here, we show that deletion of the Von Hippel-Lindau gene (Vhlh) from intrinsic glomerular cells of mice is sufficient to initiate a necrotizing crescentic glomerulonephritis and the clinical features that accompany RPGN. Loss of Vhlh leads to stabilization of hypoxia-inducible factor alpha subunits (HIFs). Using gene expression profiling, we identified de novo expression of the HIF target gene Cxcr4 (ref. 3) in glomeruli from both mice and humans with RPGN. The course of RPGN is markedly improved in mice treated with a blocking antibody to Cxcr4, whereas overexpression of Cxcr4 alone in podocytes of transgenic mice is sufficient to cause glomerular disease. Collectively, these results indicate an alternative mechanism for the pathogenesis of RPGN and glomerular disease in an animal model and suggest novel molecular pathways for intervention in this disease.


Asunto(s)
Glomerulonefritis/fisiopatología , Glomérulos Renales/metabolismo , Receptores CXCR4/biosíntesis , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proliferación Celular , Perfilación de la Expresión Génica , Glomerulonefritis/etiología , Glomerulonefritis/patología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Podocitos/citología , Podocitos/metabolismo , Regulación hacia Arriba
15.
Biologicals ; 41(6): 439-45, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24135082

RESUMEN

Focal segmental glomerulosclerosis (FSGS) is the most frequent acquired renal condition resulting in end stage kidney disease in children. We describe a cell therapy treatment with human allogeneic bone marrow mesenchymal stem cells (MSC) in a 13-year-old patient developing recurrent FSGS after renal transplantation, which was not responding to conventional therapy. This treatment relied on the following measurements:clinical and laboratory evaluation of renal function, proteome array, biopsy, short tandem repeat assay. Before MSC treatment, the patient needed weekly plasmapheresis to achieve proteinuria-to-creatininuria ratio below 5. After three MSC infusions without adverse events, the patient has a stable renal function and the proteinuria target was reached without plasmapheresis. In addition, some circulating inflammatory factors decreased and their levels were still low after one year. This is the first report of an MSC treatment in an FSGS patient. Even though different factors may have contributed to the clinical results, after MSC infusion a stable reduction in the serum level of several inflammatory factors has been registered and the patient does not need anymore plasmapheresis to keep proteinuria under control. In addition, this encouraging single case let us identify some putative efficacy biomarkers that could be of clinical interest in chronic kidney diseases.


Asunto(s)
Glomeruloesclerosis Focal y Segmentaria/cirugía , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Adolescente , Supervivencia Celular , Células Cultivadas , Citometría de Flujo , Glomeruloesclerosis Focal y Segmentaria/etiología , Glomeruloesclerosis Focal y Segmentaria/fisiopatología , Humanos , Inmunofenotipificación , Trasplante de Riñón/efectos adversos , Masculino , Células Madre Mesenquimatosas/metabolismo , Trasplante Homólogo , Resultado del Tratamiento
16.
J Biol Chem ; 286(35): 30795-30805, 2011 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-21680739

RESUMEN

Genetic alterations of α-actinin-4 can cause podocyte injury through multiple mechanisms. Although a mechanism involving gain-of-α-actinin-4 function was well described and is responsible for a dominantly inherited form of human focal segmental glomerulosclerosis (FSGS), evidence supporting mechanisms involving loss-of-α-actinin-4 function in human glomerular diseases remains elusive. Here we show that α-actinin-4 deficiency occurs in multiple human primary glomerulopathies including sporadic FSGS, minimal change disease, and IgA nephropathy. Furthermore, we identify a close correlation between the levels of α-actinin-4 and CLP36, which form a complex in normal podocytes, in human glomerular diseases. siRNA-mediated depletion of α-actinin-4 in human podocytes resulted in a marked reduction of the CLP36 level. Additionally, two FSGS-associated α-actinin-4 mutations (R310Q and Q348R) inhibited the complex formation between α-actinin-4 and CLP36. Inhibition of the α-actinin-4-CLP36 complex, like loss of α-actinin-4, markedly reduced the level of CLP36 in podocytes. Finally, reduction of the CLP36 level or disruption of the α-actinin-4-CLP36 complex significantly inhibited RhoA activity and generation of traction force in podocytes. Our studies reveal a critical role of the α-actinin-4-CLP36 complex in podocytes and provide an explanation as to how α-actinin-4 deficiency or mutations found in human patients could contribute to podocyte defects and glomerular failure through a loss-of-function mechanism.


Asunto(s)
Actinina/genética , Glomérulos Renales/metabolismo , Proteínas de Microfilamentos/genética , Podocitos/metabolismo , Actinina/deficiencia , Animales , Biopsia , Detergentes/farmacología , Glomerulonefritis por IGA/metabolismo , Glomeruloesclerosis Focal y Segmentaria/metabolismo , Humanos , Inmunohistoquímica/métodos , Riñón/metabolismo , Proteínas con Dominio LIM , Ratones , Proteínas de Microfilamentos/deficiencia , Mutación , Mapeo de Interacción de Proteínas/métodos , Proteinuria/metabolismo , Factores de Transcripción
17.
Hum Mol Genet ; 19(15): 2998-3010, 2010 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-20472742

RESUMEN

Uromodulin-associated kidney diseases (UAKD) are autosomal-dominant disorders characterized by alteration of urinary concentrating ability, tubulo-interstitial fibrosis, hyperuricaemia and renal cysts at the cortico-medullary junction. UAKD are caused by mutations in UMOD, the gene encoding uromodulin. Although uromodulin is the most abundant protein secreted in urine, its physiological role remains elusive. Several in vitro studies demonstrated that mutations in uromodulin lead to endoplasmic reticulum (ER) retention of mutant protein, but their relevance in vivo has not been studied. We here report on the generation and characterization of the first transgenic mouse model for UAKD. Transgenic mice that express the C147W mutant uromodulin (Tg(Umod)(C147W)), corresponding to the well-established patient mutation C148W, were compared with expression-matched transgenic mice expressing the wild-type protein (Tg(Umod)(wt)). Tg(Umod)(C147W) mice recapitulate most of the UAKD features, with urinary concentrating defect of renal origin and progressive renal injury, i.e. tubulo-interstitial fibrosis with inflammatory cell infiltration, tubule dilation and specific damage of the thick ascending limb of Henle's loop, leading to mild renal failure. As observed in patients, Tg(Umod)(C147W) mice show a marked reduction of urinary uromodulin excretion. Mutant uromodulin trafficking to the plasma membrane is indeed impaired as it is retained in the ER of expressing cells leading to ER hyperplasia. The Tg(Umod)(C147W) mice represent a unique model that recapitulates most of the features associated with UAKD. Our data clearly demonstrate a gain-of-toxic function of uromodulin mutations providing insights into the pathogenetic mechanism of the disease. These findings may also be relevant for other tubulo-interstitial or ER-storage disorders.


Asunto(s)
Túbulos Renales/patología , Mucoproteínas/metabolismo , Insuficiencia Renal/complicaciones , Insuficiencia Renal/orina , Secuencia de Aminoácidos , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/ultraestructura , Inflamación/patología , Espacio Intracelular/metabolismo , Túbulos Renales/ultraestructura , Ratones , Ratones Transgénicos , Modelos Biológicos , Datos de Secuencia Molecular , Mucoproteínas/química , Proteínas Mutantes/metabolismo , Transporte de Proteínas , Insuficiencia Renal/patología , Uromodulina , Privación de Agua
18.
Am J Pathol ; 178(3): 1257-69, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21356376

RESUMEN

The metabotropic glutamate (mGlu) receptor 1 (GRM1) has been shown to play an important role in neuronal cells by triggering, through calcium release from intracellular stores, various signaling pathways that finally modulate neuron excitability, synaptic plasticity, and mechanisms of feedback regulation of neurotransmitter release. Herein, we show that Grm1 is expressed in glomerular podocytes and that a glomerular phenotype is exhibited by Grm1(crv4) mice carrying a spontaneous recessive inactivating mutation of the gene. Homozygous Grm1(crv4/crv4) and, to a lesser extent, heterozygous mice show albuminuria, podocyte foot process effacement, and reduced levels of nephrin and other proteins known to contribute to the maintenance of podocyte cell structure. Overall, the present data extend the role of mGlu1 receptor to the glomerular filtration barrier. The regulatory action of mGlu1 receptor in dendritic spine morphology and in the control of glutamate release is well acknowledged in neuronal cells. Analogously, we speculate that mGlu1 receptor may regulate foot process morphology and intercellular signaling in the podocyte.


Asunto(s)
Albuminuria/patología , Glomérulos Renales/patología , Receptores de Glutamato Metabotrópico/deficiencia , Albuminuria/complicaciones , Albuminuria/metabolismo , Animales , Western Blotting , Células Cultivadas , Doxorrubicina/farmacología , Técnica del Anticuerpo Fluorescente , Regulación de la Expresión Génica/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Humanos , Corteza Renal/efectos de los fármacos , Corteza Renal/metabolismo , Corteza Renal/patología , Enfermedades Renales/complicaciones , Enfermedades Renales/metabolismo , Enfermedades Renales/patología , Glomérulos Renales/metabolismo , Glomérulos Renales/ultraestructura , Ratones , Fenotipo , Podocitos/efectos de los fármacos , Podocitos/metabolismo , Podocitos/patología , Receptores de Glutamato Metabotrópico/genética , Receptores de Glutamato Metabotrópico/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
19.
J Pathol ; 225(1): 118-28, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21630272

RESUMEN

Nephrin is an immunoglobulin-like adhesion molecule first discovered as a major component of the podocyte slit diaphragm, where its integrity is essential to the function of the glomerular filtration barrier. Outside the kidney, nephrin has been shown in other restricted locations, most notably in the central nervous system (CNS) of embryonic and newborn rodents. With the aim of better characterizing nephrin expression and its role in the CNS of adult rodents, we studied its expression pattern and possible binding partners in CNS tissues and cultured neuronal cells and compared these data to those obtained in control renal tissues and podocyte cell cultures. Our results show that, besides a number of locations already found in embryos and newborns, endogenous nephrin in adult rodent CNS extends to the pons and corpus callosum and is expressed by granule cells and Purkinje cells of the cerebellum, with a characteristic alternating expression pattern. In primary neuronal cells we find nephrin expression close to synaptic proteins and demonstrate that nephrin co-immunoprecipitates with Fyn kinase, glutamate receptors and the scaffolding molecule PSD95, an assembly that is reminiscent of those made by synaptic adhesion molecules. This role seems to be confirmed by our findings of impaired maturation and reduced glutamate exocytosis occurring in Neuro2A cells upon nephrin silencing. Of note, we disclose that the very same nephrin interactions occur in renal glomeruli and cultured podocytes, supporting our hypothesis that podocytes organize and use similar molecular intercellular signalling modules to those used by neuronal cells.


Asunto(s)
Encéfalo/metabolismo , Proteínas de la Membrana/biosíntesis , Receptores de Glutamato/metabolismo , Animales , Células Cultivadas , Corteza Cerebral/metabolismo , Glomérulos Renales/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas de Neoplasias , Neuronas/metabolismo , Podocitos/metabolismo , Proteínas Tirosina Quinasas , Células de Purkinje/metabolismo , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Familia-src Quinasas/metabolismo
20.
J Immunol ; 185(7): 4457-69, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20805419

RESUMEN

Systemic lupus erythematosus (SLE) is characterized by increased vascular risk due to premature atherosclerosis independent of traditional risk factors. We previously proposed that IFN-α plays a crucial role in premature vascular damage in SLE. IFN-α alters the balance between endothelial cell apoptosis and vascular repair mediated by endothelial progenitor cells (EPCs) and myeloid circulating angiogenic cells (CACs). In this study, we demonstrate that IFN-α promotes an antiangiogenic signature in SLE and control EPCs/CACs, characterized by transcriptional repression of IL-1α and ß, IL-1R1, and vascular endothelial growth factor A, and upregulation of IL-1R antagonist and the decoy receptor IL-1R2. IL-1ß promotes significant improvement in the functional capacity of lupus EPCs/CACs, therefore abrogating the deleterious effects of IFN-α. The beneficial effects from IL-1 are mediated, at least in part, by increases in EPC/CAC proliferation, by decreases in EPC/CAC apoptosis, and by preventing the skewing of CACs toward nonangiogenic pathways. IFN-α induces STAT2 and 6 phosphorylation in EPCs/CACs, and JAK inhibition abrogates the transcriptional antiangiogenic changes induced by IFN-α in these cells. Immunohistochemistry of renal biopsies from patients with lupus nephritis, but not anti-neutrophil cytoplasmic Ab-positive vasculitis, showed this pathway to be operational in vivo, with increased IL-1R antagonist, downregulation of vascular endothelial growth factor A, and glomerular and blood vessel decreased capillary density, compared with controls. Our study introduces a novel putative pathway by which type I IFNs may interfere with vascular repair in SLE through repression of IL-1-dependent pathways. This could promote atherosclerosis and loss of renal function in this disease.


Asunto(s)
Aterosclerosis/metabolismo , Interferón-alfa/metabolismo , Interleucina-1/metabolismo , Lupus Eritematoso Sistémico/metabolismo , Neovascularización Fisiológica , Adulto , Aterosclerosis/inmunología , Diferenciación Celular/fisiología , Células Endoteliales/metabolismo , Femenino , Expresión Génica , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Inmunohistoquímica , Proteína Antagonista del Receptor de Interleucina 1/biosíntesis , Interleucina-1/inmunología , Riñón/irrigación sanguínea , Riñón/metabolismo , Riñón/patología , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/patología , Nefritis Lúpica/metabolismo , Nefritis Lúpica/patología , Masculino , Microscopía Fluorescente , Análisis de Secuencia por Matrices de Oligonucleótidos , Receptores de Interleucina-1/biosíntesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/fisiología , Células Madre , Factor A de Crecimiento Endotelial Vascular/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA