Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Cell ; 150(3): 533-48, 2012 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-22863007

RESUMEN

Nephronophthisis-related ciliopathies (NPHP-RC) are degenerative recessive diseases that affect kidney, retina, and brain. Genetic defects in NPHP gene products that localize to cilia and centrosomes defined them as "ciliopathies." However, disease mechanisms remain poorly understood. Here, we identify by whole-exome resequencing, mutations of MRE11, ZNF423, and CEP164 as causing NPHP-RC. All three genes function within the DNA damage response (DDR) pathway. We demonstrate that, upon induced DNA damage, the NPHP-RC proteins ZNF423, CEP164, and NPHP10 colocalize to nuclear foci positive for TIP60, known to activate ATM at sites of DNA damage. We show that knockdown of CEP164 or ZNF423 causes sensitivity to DNA damaging agents and that cep164 knockdown in zebrafish results in dysregulated DDR and an NPHP-RC phenotype. Our findings link degenerative diseases of the kidney and retina, disorders of increasing prevalence, to mechanisms of DDR.


Asunto(s)
Daño del ADN , Proteínas de Unión al ADN/metabolismo , Exoma , Enfermedades Renales Quísticas/genética , Proteínas de Microtúbulos/metabolismo , Animales , Cilios/metabolismo , Técnicas de Silenciamiento del Gen , Genes Recesivos , Humanos , Proteína Homóloga de MRE11 , Ratones , Proteínas , Transducción de Señal , Pez Cebra/embriología , Pez Cebra/metabolismo
2.
Hum Mol Genet ; 29(15): 2508-2522, 2020 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-32620959

RESUMEN

Bardet-Biedl syndrome (BBS) is a pleiotropic autosomal recessive ciliopathy affecting multiple organs. The development of potential disease-modifying therapy for BBS will require concurrent targeting of multi-systemic manifestations. Here, we show for the first time that monosialodihexosylganglioside accumulates in Bbs2-/- cilia, indicating impairment of glycosphingolipid (GSL) metabolism in BBS. Consequently, we tested whether BBS pathology in Bbs2-/- mice can be reversed by targeting the underlying ciliary defect via reduction of GSL metabolism. Inhibition of GSL synthesis with the glucosylceramide synthase inhibitor Genz-667161 decreases the obesity, liver disease, retinal degeneration and olfaction defect in Bbs2-/- mice. These effects are secondary to preservation of ciliary structure and signaling, and stimulation of cellular differentiation. In conclusion, reduction of GSL metabolism resolves the multi-organ pathology of Bbs2-/- mice by directly preserving ciliary structure and function towards a normal phenotype. Since this approach does not rely on the correction of the underlying genetic mutation, it might translate successfully as a treatment for other ciliopathies.


Asunto(s)
Síndrome de Bardet-Biedl/genética , Cilios/genética , Ciliopatías/genética , Proteínas/genética , Animales , Síndrome de Bardet-Biedl/tratamiento farmacológico , Síndrome de Bardet-Biedl/patología , Diferenciación Celular/efectos de los fármacos , Cilios/patología , Ciliopatías/tratamiento farmacológico , Ciliopatías/patología , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Gangliósidos/biosíntesis , Gangliósidos/genética , Glucosiltransferasas/antagonistas & inhibidores , Glucosiltransferasas/genética , Glicoesfingolípidos/biosíntesis , Glicoesfingolípidos/genética , Ratones Noqueados
3.
Hum Mol Genet ; 25(11): 2245-2255, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27053712

RESUMEN

Polycystic kidney diseases (PKDs) comprise a subgroup of ciliopathies characterized by the formation of fluid-filled kidney cysts and progression to end-stage renal disease. A mechanistic understanding of cystogenesis is crucial for the development of viable therapeutic options. Here, we identify CDK5, a kinase active in post mitotic cells, as a new and important mediator of PKD progression. We show that long-lasting attenuation of PKD in the juvenile cystic kidneys (jck) mouse model of nephronophthisis by pharmacological inhibition of CDK5 using either R-roscovitine or S-CR8 is accompanied by sustained shortening of cilia and a more normal epithelial phenotype, suggesting this treatment results in a reprogramming of cellular differentiation. Also, a knock down of Cdk5 in jck cells using small interfering RNA results in significant shortening of ciliary length, similar to what we observed with R-roscovitine. Finally, conditional inactivation of Cdk5 in the jck mice significantly attenuates cystic disease progression and is associated with shortening of ciliary length as well as restoration of cellular differentiation. Our results suggest that CDK5 may regulate ciliary length by affecting tubulin dynamics via its substrate collapsin response mediator protein 2. Taken together, our data support therapeutic approaches aimed at restoration of ciliogenesis and cellular differentiation as a promising strategy for the treatment of renal cystic diseases.


Asunto(s)
Cilios/efectos de los fármacos , Quinasa 5 Dependiente de la Ciclina/genética , Fallo Renal Crónico/tratamiento farmacológico , Enfermedades Renales Poliquísticas/tratamiento farmacológico , Animales , Diferenciación Celular/efectos de los fármacos , Cilios/patología , Quinasa 5 Dependiente de la Ciclina/antagonistas & inhibidores , Modelos Animales de Enfermedad , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Riñón/efectos de los fármacos , Riñón/patología , Fallo Renal Crónico/genética , Fallo Renal Crónico/patología , Ratones , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Enfermedades Renales Poliquísticas/genética , Enfermedades Renales Poliquísticas/patología , Purinas/administración & dosificación , Roscovitina , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo
4.
Am J Physiol Renal Physiol ; 310(11): F1414-22, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27076647

RESUMEN

Polycystic kidney diseases (PKDs) are genetic diseases characterized by renal cyst formation with increased cell proliferation, apoptosis, and transition to a secretory phenotype at the expense of terminal differentiation. Despite recent progress in understanding PKD pathogenesis and the emergence of potential therapies, the key molecular mechanisms promoting cystogenesis are not well understood. Here, we demonstrate that mechanisms including endoplasmic reticulum stress, oxidative damage, and compromised mitochondrial function all contribute to nephronophthisis-associated PKD. Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) is emerging as a critical mediator of these cellular processes. Therefore, we reasoned that pharmacological targeting of CaMKII may translate into effective inhibition of PKD in jck mice. Our data demonstrate that CaMKII is activated within cystic kidney epithelia in jck mice. Blockade of CaMKII with a selective inhibitor results in effective inhibition of PKD in jck mice. Mechanistic experiments in vitro and in vivo demonstrated that CaMKII inhibition relieves endoplasmic reticulum stress and oxidative damage and improves mitochondrial integrity and membrane potential. Taken together, our data support CaMKII inhibition as a new and effective therapeutic avenue for the treatment of cystic diseases.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Estrés del Retículo Endoplásmico/fisiología , Riñón/metabolismo , Mitocondrias/metabolismo , Estrés Oxidativo/fisiología , Enfermedades Renales Poliquísticas/metabolismo , Animales , Ratones
5.
Hum Mol Genet ; 21(15): 3397-407, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22563011

RESUMEN

Genetic forms of polycystic kidney diseases (PKDs), including nephronophthisis, are characterized by formation of fluid-filled cysts in the kidneys and progression to end-stage renal disease. No therapies are currently available to treat cystic diseases, making it imperative to dissect molecular mechanisms in search of therapeutic targets. Accumulating evidence suggests a pathogenic role for glucosylceramide (GlcCer) in multiple forms of PKD. It is not known, however, whether other structural glycosphingolipids (GSLs) or bioactive signaling sphingolipids (SLs) modulate cystogenesis. Therefore, we set out to address the role of a specific GSL (ganglioside GM3) and signaling SL (sphingosine-1-phosphate, S1P) in PKD progression, using the jck mouse model of nephronopthisis. To define the role of GM3 accumulation in cystogenesis, we crossed jck mice with mice carrying a targeted mutation in the GM3 synthase (St3gal5) gene. GM3-deficient jck mice displayed milder PKD, revealing a pivotal role for ganglioside GM3. Mechanistic changes in regulation of the cell-cycle machinery and Akt-mTOR signaling were consistent with reduced cystogenesis. Dramatic overexpression of sphingosine kinase 1 (Sphk1) mRNA in jck kidneys suggested a pathogenic role for S1P. Surprisingly, genetic loss of Sphk1 exacerbated cystogenesis and was associated with increased levels of GlcCer and GM3. On the other hand, increasing S1P accumulation through pharmacologic inhibition of S1P lyase had no effect on the progression of cystogenesis or kidney GSL levels. Together, these data suggest that genes involved in the SL metabolism may be modifiers of cystogenesis, and suggest GM3 synthase as a new anti-cystic therapeutic target.


Asunto(s)
Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Enfermedades Renales Poliquísticas/genética , Sialiltransferasas/genética , Animales , Modelos Animales de Enfermedad , Glucosilceramidas/metabolismo , Glicoesfingolípidos/metabolismo , Ratones , Proteína Oncogénica v-akt/genética , Proteína Oncogénica v-akt/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Enfermedades Renales Poliquísticas/enzimología , Sialiltransferasas/metabolismo , Esfingosina/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
6.
Commun Biol ; 7(1): 932, 2024 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-39095617

RESUMEN

While significant advances have been made in understanding renal pathophysiology, less is known about the role of glycosphingolipid (GSL) metabolism in driving organ dysfunction. Here, we used a small molecule inhibitor of glucosylceramide synthase to modulate GSL levels in three mouse models of distinct renal pathologies: Alport syndrome (Col4a3 KO), polycystic kidney disease (Nek8jck), and steroid-resistant nephrotic syndrome (Nphs2 cKO). At the tissue level, we identified a core immune-enriched transcriptional signature that was shared across models and enriched in human polycystic kidney disease. Single nuclei analysis identified robust transcriptional changes across multiple kidney cell types, including epithelial and immune lineages. To further explore the role of GSL modulation in macrophage biology, we performed in vitro studies with homeostatic and inflammatory bone marrow-derived macrophages. Cumulatively, this study provides a comprehensive overview of renal dysfunction and the effect of GSL modulation on kidney-derived cells in the setting of renal dysfunction.


Asunto(s)
Glucosiltransferasas , Macrófagos , Animales , Macrófagos/metabolismo , Macrófagos/efectos de los fármacos , Ratones , Glucosiltransferasas/metabolismo , Glucosiltransferasas/genética , Glucosiltransferasas/antagonistas & inhibidores , Ratones Noqueados , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Riñón/patología , Riñón/metabolismo , Riñón/efectos de los fármacos , Masculino
7.
Sci Rep ; 12(1): 9340, 2022 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-35660779

RESUMEN

A significant population of patients with chronic kidney disease (CKD) develops cardiac hypertrophy, which can lead to heart failure and sudden cardiac death. Soluble klotho (sKL), the shed ectodomain of the transmembrane protein klotho, protects the heart against hypertrophic growth. We have shown that sKL protects the heart by regulating the formation and function of lipid rafts by targeting the sialic acid moiety of gangliosides, GM1/GM3. Reduction in circulating sKL contributes to an increased risk of cardiac hypertrophy in mice. sKL replacement therapy has been considered but its use is limited by the inability to mass produce the protein. Therefore, alternative methods to protect the heart are proposed. Glucosylation of ceramide catalyzed by glucosylceramide synthase is the entry step for the formation of gangliosides. Here we show that oral administration of a glucosylceramide synthase inhibitor (GCSi) reduces plasma and heart tissue glycosphingolipids, including gangliosides. Administration of GCSi is protective in two mouse models of cardiac stress-induction, one with isoproterenol overstimulation and the other with 5/6 nephrectomy-induced CKD. Treatment with GCSi does not alter the severity of renal dysfunction and hypertension in CKD. These results provide proof of principle for targeting glucosylceramide synthase to decrease gangliosides as a treatment for cardiac hypertrophy. They also support the hypothesis that sKL protects the heart by targeting gangliosides.


Asunto(s)
Cardiomegalia , Insuficiencia Renal Crónica , Animales , Cardiomegalia/tratamiento farmacológico , Cardiomegalia/prevención & control , Gangliósidos/metabolismo , Glucosiltransferasas , Humanos , Ratones , Insuficiencia Renal Crónica/complicaciones , Insuficiencia Renal Crónica/tratamiento farmacológico
8.
Cells ; 11(4)2022 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-35203245

RESUMEN

Col4a3-/- Alport mice serve as an animal model for renal fibrosis. MicroRNA-21 (miR-21) expression has been shown to be increased in the kidneys of Alport syndrome patients. Here, we investigated the nephroprotective effects of Lademirsen anti-miR-21 therapy. We used a fast-progressing Col4a3-/- mouse model with a 129/SvJ background and an intermediate-progressing F1 hybrid mouse model with a mixed genetic background, with angiotensin-converting enzyme inhibitor (ACEi) monotherapy in combination with anti-miR-21 therapy. In the fast-progressing model, the anti miR-21 and ACEi therapies showed an additive effect in the reduction in fibrosis, the decline of proteinuria, the preservation of kidney function and increased survival. In the intermediate-progressing F1 model, the anti-miR-21 and ACEi therapies individually improved kidney pathology. Both also improved kidney function and survival; however, the combination showed a significant additive effect, particularly for survival. RNA sequencing (RNA-seq) gene expression profiling revealed that the anti-miR-21 and ACEi therapies modulate several common pathways. However, anti-miR-21 was particularly effective at normalizing the expression profiles of the genes involved in renal tubulointerstitial injury pathways. In conclusion, significant additive effects were detected for the combination of anti-miR-21 and ACEi therapies on kidney function, pathology and survival in Alport mouse models, as well as a strong differential effect of anti-miR-21 on the renal expression of fibrotic factors. These results support the addition of anti-miR-21 to the current standard of care (ACEi) in ongoing clinical trials in patients with Alport syndrome.


Asunto(s)
Inhibidores de la Enzima Convertidora de Angiotensina , MicroARNs , Nefritis Hereditaria , Insuficiencia Renal , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Animales , Antagomirs , Colágeno Tipo IV/genética , Colágeno Tipo IV/metabolismo , Modelos Animales de Enfermedad , Fibrosis , Humanos , Ratones , Ratones Noqueados , MicroARNs/antagonistas & inhibidores , Nefritis Hereditaria/tratamiento farmacológico , Nefritis Hereditaria/genética , Insuficiencia Renal/tratamiento farmacológico
10.
J Clin Invest ; 114(10): 1433-43, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15545994

RESUMEN

Polycystin-1, which is encoded by a gene that is mutated in autosomal dominant polycystic kidney disease (ADPKD), is involved in cell-matrix interactions as well as in ciliary signaling. The precise mechanisms by which it functions, however, remain unclear. Here we find that polycystin-1 undergoes a proteolytic cleavage that releases its C-terminal tail (CTT), which enters the nucleus and initiates signaling processes. The cleavage occurs in vivo in association with alterations in mechanical stimuli. Polycystin-2, the product of the second gene mutated in ADPKD, modulates the signaling properties of the polycystin-1 CTT. These data reveal a novel pathway by which polycystin-1 transmits messages directly to the nucleus.


Asunto(s)
Núcleo Celular/metabolismo , Proteínas/química , Proteínas/metabolismo , Transducción de Señal , Secuencia de Aminoácidos , Animales , Células CHO , Células COS , Línea Celular , Chlorocebus aethiops , Cricetinae , Cricetulus , Perros , Embrión de Mamíferos , Células Epiteliales/citología , Túbulos Renales/citología , Túbulos Renales/embriología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Riñón Poliquístico Autosómico Dominante/genética , Riñón Poliquístico Autosómico Dominante/patología , Proteínas/genética , Eliminación de Secuencia , Estrés Mecánico , Canales Catiónicos TRPP , Factor de Transcripción AP-1/metabolismo
11.
Physiol Rep ; 4(12)2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27356569

RESUMEN

Development of a disease-modifying therapy to treat autosomal dominant polycystic kidney disease (ADPKD) requires well-characterized preclinical models that accurately reflect the pathology and biochemical changes associated with the disease. Using a Pkd1 conditional knockout mouse, we demonstrate that subtly altering the timing and extent of Pkd1 deletion can have a significant impact on the origin and severity of kidney cyst formation. Pkd1 deletion on postnatal day 1 or 2 results in cysts arising from both the cortical and medullary regions, whereas deletion on postnatal days 3-8 results in primarily medullary cyst formation. Altering the extent of Pkd1 deletion by modulating the tamoxifen dose produces dose-dependent changes in the severity, but not origin, of cystogenesis. Limited Pkd1 deletion produces progressive kidney cystogenesis, accompanied by interstitial fibrosis and loss of kidney function. Cyst growth occurs in two phases: an early, rapid growth phase, followed by a later, slow growth period. Analysis of biochemical pathway changes in cystic kidneys reveals dysregulation of the cell cycle, increased proliferation and apoptosis, activation of Mek-Erk, Akt-mTOR, and Wnt-ß-catenin signaling pathways, and altered glycosphingolipid metabolism that resemble the biochemical changes occurring in human ADPKD kidneys. These pathways are normally active in neonatal mouse kidneys until repressed around 3 weeks of age; however, they remain active following Pkd1 deletion. Together, this work describes the key parameters to accurately model the pathological and biochemical changes associated with ADPKD in a conditional mouse model.


Asunto(s)
Eliminación de Gen , Enfermedades Renales Poliquísticas/genética , Canales Catiónicos TRPP/metabolismo , Animales , Modelos Animales de Enfermedad , Fibrosis , Riñón/metabolismo , Riñón/patología , Sistema de Señalización de MAP Quinasas , Ratones , Enfermedades Renales Poliquísticas/metabolismo , Enfermedades Renales Poliquísticas/patología , Canales Catiónicos TRPP/genética , Vía de Señalización Wnt
12.
Oncogene ; 21(9): 1423-33, 2002 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-11857085

RESUMEN

CBL and the related CBL-B protein are two members of a family of RING finger type ubiquitin E3 ligases that are believed to function as negative regulators of signal transduction in hematopoietic and immune cells. In mice, expression of v-Cbl causes lymphomas, and targeted disruption of either the CBL gene or the CBL-B gene can result in a lymphoproliferative disorder or hypersensitivity of lymphocytes. CBL is one of the most prominent targets of the BCR/ABL tyrosine kinase oncogene. We compared the role of CBL and CBL-B in signal transduction of BCR/ABL using pairs of cell lines before and after expression of BCR/ABL. In contrast to CBL, BCR/ABL was found to rapidly downregulate the expression of CBL-B protein. The decrease in CBL-B protein induced by BCR/ABL was associated with downregulation of CBL-B mRNA. Downregulation and tyrosine phosphorylation of CBL-B required BCR/ABL kinase activity. However, despite their known similarities in structure and function, we found CBL and CBL-B proteins to be involved in distinct signaling complexes. CBL was predominantly in a complex with phosphatidylinositol 3'-kinase and CRKL, while CBL-B was not associated with any significant phosphatidylinositol 3'-kinase activity. A major CBL-B associated protein was identified as mono-ubiquitinated Vav, a nucleotide exchange factor for Rac1. These results demonstrate that BCR/ABL signals differentially through CBL and CBL-B, with downregulation of the CBL-B protein potentially contributing to BCR/ABL-mediated transformation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Proteínas Portadoras/metabolismo , Transformación Celular Neoplásica/metabolismo , Fosfoproteínas/metabolismo , Proteínas Oncogénicas de Retroviridae/metabolismo , Transducción de Señal , Ubiquitina-Proteína Ligasas , Animales , Proteínas Portadoras/genética , Línea Celular Transformada , Movimiento Celular , Transformación Celular Neoplásica/genética , Regulación hacia Abajo , Proteínas de Fusión bcr-abl/genética , Proteínas de Fusión bcr-abl/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genes abl/genética , Humanos , Peróxido de Hidrógeno/farmacología , Ratones , Proteínas Nucleares/metabolismo , Proteína Oncogénica v-cbl , Proteínas Oncogénicas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfoproteínas/genética , Fosforilación , Fosfotirosina/metabolismo , Unión Proteica , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-cbl , Proteínas Proto-Oncogénicas c-pim-1 , Proteínas Proto-Oncogénicas c-vav , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Oncogénicas de Retroviridae/genética , Transducción de Señal/efectos de los fármacos , Transfección
13.
Nat Med ; 16(7): 788-92, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20562878

RESUMEN

Polycystic kidney disease (PKD) represents a family of genetic disorders characterized by renal cystic growth and progression to kidney failure. No treatment is currently available for people with PKD, although possible therapeutic interventions are emerging. Despite genetic and clinical heterogeneity, PKDs have in common defects of cystic epithelia, including increased proliferation, apoptosis and activation of growth regulatory pathways. Sphingolipids and glycosphingolipids are emerging as major regulators of these cellular processes. We sought to evaluate the therapeutic potential for glycosphingolipid modulation as a new approach to treat PKD. Here we demonstrate that kidney glucosylceramide (GlcCer) and ganglioside GM3 levels are higher in human and mouse PKD tissue as compared to normal tissue, regardless of the causative mutation. Blockade of GlcCer accumulation with the GlcCer synthase inhibitor Genz-123346 effectively inhibits cystogenesis in mouse models orthologous to human autosomal dominant PKD (Pkd1 conditional knockout mice) and nephronophthisis (jck and pcy mice). Molecular analysis in vitro and in vivo indicates that Genz-123346 acts through inhibition of the two key pathways dysregulated in PKD: Akt protein kinase-mammalian target of rapamycin signaling and cell cycle machinery. Taken together, our data suggest that inhibition of GlcCer synthesis represents a new and effective treatment option for PKD.


Asunto(s)
Dioxanos/farmacología , Glucosilceramidas/biosíntesis , Enfermedades Renales Poliquísticas/metabolismo , Pirrolidinas/farmacología , Animales , Ciclo Celular , Modelos Animales de Enfermedad , Gangliósido G(M3)/metabolismo , Glucosiltransferasas/antagonistas & inhibidores , Glicoesfingolípidos/metabolismo , Humanos , Ratones , Ratones Noqueados , Enfermedades Renales Poliquísticas/tratamiento farmacológico , Ratas
14.
Am J Physiol Renal Physiol ; 294(1): F73-83, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17928412

RESUMEN

Development of novel therapies for polycystic kidney disease (PKD) requires assays that adequately reflect disease biology and are adaptable to high-throughput screening. Here we describe an embryonic cystic kidney organ culture model and demonstrate that a new mutant allele of the Pkd1 gene (Pkd1(tm1Bdgz)) modulates cystogenesis in this model. Cyst formation induced by cAMP is influenced by the dosage of the mutant allele: Pkd1(tm1Bdgz) -/- cultures develop a larger cystic area compared with +/+ counterparts, while Pkd1(tm1Bdgz) +/- cultures show an intermediate phenotype. A similar relationship between the degree of cystogenesis and mutant gene dosage is seen in cystic kidney organ cultures derived from mice with a mutated Nek8 gene (Nek8(jck)). Both Pkd1- and Nek8- cultures display altered cell-cell junctions, with reduced E-cadherin expression and altered desmosomal protein expression, similar to ADPKD epithelia. Additionally, characteristic ciliary abnormalities are identified in cystic kidney cultures, with elevated ciliary polycystin 1 expression in Nek8 homozygous cultures and elevated ciliary Nek8 protein expression in Pkd1 homozygotes. These data suggest that the Nek8 and Pkd1 genes function in a common pathway to regulate cystogenesis. Moreover, compound Pkd1 and Nek8 heterozygous adult mice develop a more aggressive cystic disease than animals with a mutation in either gene alone. Finally, we validate the kidney organ culture cystogenesis assay as a therapeutic testing platform using the CDK inhibitor roscovitine. Therefore, embryonic kidney organ culture represents a relevant model for studying molecular cystogenesis and a rapid tool for the screening for therapies that block cystic growth.


Asunto(s)
Adhesión Celular/fisiología , Cilios/metabolismo , Mutación/genética , Enfermedades Renales Poliquísticas/metabolismo , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Canales Catiónicos TRPP/metabolismo , Alelos , Animales , Cadherinas/metabolismo , Adhesión Celular/genética , Cilios/efectos de los fármacos , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Quistes/metabolismo , Quistes/fisiopatología , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Noqueados , Quinasas Relacionadas con NIMA , Técnicas de Cultivo de Órganos , Enfermedades Renales Poliquísticas/fisiopatología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas , Purinas/farmacología , Roscovitina
15.
J Am Soc Nephrol ; 17(10): 2821-31, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16928806

RESUMEN

Significant progress in understanding the molecular mechanisms of polycystic kidney disease (PKD) has been made in recent years. Translating this understanding into effective therapeutics will require testing in animal models that closely resemble human PKD by multiple parameters. Similar to autosomal dominant PKD, juvenile cystic kidney (jck) mice develop cysts in multiple nephron segments, including cortical collecting ducts, distal tubules, and loop of Henle. The jck mice display gender dimorphism in kidney disease progression with more aggressive disease in male mice. Gonadectomy experiments show that testosterone aggravates the severity of the disease in jck male mice, while female gonadal hormones have protective effects. EGF receptor is overexpressed and mislocalized in jck cystic epithelia, a hallmark of human disease. Increased cAMP levels in jck kidneys and activation of the B-Raf/extracellular signal-regulated kinase pathway are demonstrated. The effect of jck mutation on the expression of Nek8, a NIMA-related (never in mitosis A) kinase, and polycystins in jck cilia is shown for the first time. Nek8 overexpression and loss of ciliary localization in jck epithelia are accompanied by enhanced expression of polycystins along the cilia. The primary cilia in jck kidneys are significantly more lengthened than the cilia in wild-type mice, suggesting a role for Nek8 in controlling ciliary length. Collectively, these data demonstrate that the jck mice should be useful for testing potential therapies and for studying the molecular mechanisms that link ciliary structure/function and cystogenesis.


Asunto(s)
Cilios/metabolismo , Trastornos de la Motilidad Ciliar/fisiopatología , Enfermedades Renales Quísticas/patología , Riñón Poliquístico Autosómico Dominante/etiología , Animales , Cilios/genética , AMP Cíclico/metabolismo , Progresión de la Enfermedad , Femenino , Hormonas Esteroides Gonadales/farmacología , Humanos , Immunoblotting , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Quinasas Relacionadas con NIMA , Riñón Poliquístico Autosómico Dominante/fisiopatología , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas , Caracteres Sexuales , Transducción de Señal , Tasa de Supervivencia , Canales Catiónicos TRPP/metabolismo
16.
Histochem Cell Biol ; 124(6): 487-97, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16187067

RESUMEN

Mutations in polycystin-1 (PC-1) are responsible for autosomal dominant polycystic kidney disease (ADPKD), characterized by formation of fluid-filled tubular cysts. The PC-1 is a multifunctional protein essential for tubular differentiation and maturation found in desmosomal junctions of epithelial cells where its primary function is to mediate cell-cell adhesion. To address the impact of mutated PC-1 on intercellular adhesion, we have analyzed the structure/function of desmosomal junctions in primary cells derived from ADPKD cysts. Primary epithelial cells from normal kidney showed co-localization of PC-1 and desmosomal proteins at cell-cell contacts. A striking difference was seen in ADPKD cells, where PC-1 and desmosomal proteins were lost from the intercellular junction membrane, despite unchanged protein expression levels. Instead, punctate intracellular expression for PC-1 and desmosomal proteins was detected. The N-cadherin, but not E-cadherin was expressed in adherens junctions of ADPKD cells. These data together with co-sedimentation analysis demonstrate that, in the absence of functional PC-1, desmosomal junctions cannot be properly assembled and remain sequestered in cytoplasmic compartments. Taken together, our results demonstrate that PC-1 is crucial for formation of intercellular contacts. We propose that abnormal expression of PC-1 causes disregulation of cellular adhesion complexes leading to increased proliferation, loss of polarity and, ultimately, cystogenesis.


Asunto(s)
Desmosomas/patología , Células Epiteliales/patología , Uniones Intercelulares/patología , Riñón Poliquístico Autosómico Dominante/metabolismo , Riñón Poliquístico Autosómico Dominante/patología , Cadherinas/metabolismo , Cadherinas Desmosómicas/genética , Cadherinas Desmosómicas/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/ultraestructura , Regulación de la Expresión Génica , Humanos , Inmunohistoquímica , Uniones Intercelulares/metabolismo , Modelos Biológicos , Mutación , Riñón Poliquístico Autosómico Dominante/genética , Proteínas/genética , Proteínas/metabolismo , Sensibilidad y Especificidad , Canales Catiónicos TRPP
17.
Hum Mol Genet ; 11(8): 923-36, 2002 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-11971874

RESUMEN

Polycystin-1 is a protein mutated in the majority of cases of autosomal dominant polycystic kidney disease (ADPKD), but its role in the molecular pathway of tubulogenesis and cystogenesis is not understood. To define the role of polycystin-1 during dynamic changes in formation of intercellular contacts and cell polarity accompanying epithelial morphogenesis, we have utilized a 3D MDCK in vitro model of tubulogenesis and cystogenesis. Here we demonstrate that polycystin-1 is a novel component of desmosomal junctions of epithelial cells. A striking downregulation of polycystin-1 mRNA was detected in cysts as compared to tubules, leading to altered protein expression and localization. While polycystin-1 is localized to basolateral membranes of MDCK tubules, it is only detected in cytoplasmic pools in cystic cells. Furthermore, the expression of polycystin-1 is modulated during distinct stages of HGF-induced tubulogenesis from MDCK cysts. Thus, polycystin-1 is not detected in intercellular contacts at early steps of tubulogenesis, but assumes its basolateral localization at the time of cell polarization and lumen formation. An important role of polycystin-1 is further demonstrated using the pancreatic ductal epithelial cell line SU.86.86 which undergoes in vitro differentiation resulting in the formation of domes. Dome formation is thought to parallel tubular differentiation and morphogenesis in vivo. Our data reveal significant upregulation of polycystin-1 mRNA and protein levels in domes. Collectively, our results demonstrate a critical importance of controlled level of polycystin-1 expression for proper tubular differentiation and maturation. We suggest that the loss of polycystin-1 from its basolateral location in tubular epithelium may alter critical pathways controlling normal tubulogenesis leading to cystic transformation.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Proteínas/genética , Uniones Adherentes/metabolismo , Animales , Membrana Celular/metabolismo , Polaridad Celular , Perros , Regulación hacia Abajo , Células Epiteliales , Epitelio/embriología , Técnica del Anticuerpo Fluorescente , Humanos , Técnicas In Vitro , Túbulos Renales/embriología , Túbulos Renales/metabolismo , Ratones , Microscopía Confocal , Morfogénesis , Páncreas/citología , Páncreas/embriología , Páncreas/metabolismo , Riñón Poliquístico Autosómico Dominante/genética , Riñón Poliquístico Autosómico Dominante/metabolismo , Transporte de Proteínas/fisiología , Canales Catiónicos TRPP , Células Tumorales Cultivadas , Regulación hacia Arriba/fisiología
18.
Blood ; 99(1): 282-9, 2002 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-11756183

RESUMEN

Follicular lymphomas (FLs) are neoplastic counterparts of normal germinal center (GC) B cells. FLs are characterized by t(14;18) with deregulation of the Bcl-2 (BCL2) gene. The presence of t(14;18) and overexpression of Bcl-2 is necessary, but not sufficient, to cause this disease. An array containing 588 complementary DNAs (cDNAs) was used to compare the gene expression between GC B cells and FL cells. To specifically monitor genes expressed in normal GC B and FL cells and not the entire tissue compartment, normal and malignant B cells were purified from tissues. Using the array, 37 genes were up-regulated and 28 were down-regulated in FL cells as compared to normal GC B cells. The expression level of each differentially expressed gene was verified by quantitative polymerase chain reaction. Following these studies 24 genes were up-regulated and 8 genes down-regulated with a P value less than.1. Included among the genes that were up-regulated in FLs were cell cycle regulator proteins CDK10, p120, p21CIP1, and p16INK4A; transcription factors/regulators Pax-5 and Id-2, which are involved in normal B-cell development; and genes involved in cell-cell interactions, tumor necrosis factor, interleukin-2R gamma (IL-2R gamma), and IL-4R alpha. Among the genes that were down-regulated in FLs were MRP8 and MRP14, which are involved in adhesion. Interestingly, several of these genes are localized within chromosomal regions already described to be altered in FLs. These findings provide a basis for future studies into the pathogenesis and pathophysiology of FL and may lead to the identification of potential therapeutic targets as well as antigens for immunotherapeutic strategies.


Asunto(s)
Linfocitos B/química , Perfilación de la Expresión Génica , Linfoma Folicular/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Comunicación Celular/genética , Ciclo Celular/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Quinasas Ciclina-Dependientes/genética , Ciclinas/genética , ADN Complementario/genética , Proteínas de Unión al ADN/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas Nucleares/genética , Factor de Transcripción PAX5 , ARN Mensajero/análisis , Receptores CXCR4/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción/genética , Factor de Necrosis Tumoral alfa/genética , ARNt Metiltransferasas
19.
Br J Haematol ; 119(2): 492-5, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12406091

RESUMEN

Follicular lymphomas (FLs) localize in lymphoid tissues and recapitulate the structure of normal secondary follicles. The chemokine/chemokine receptor pair CXCL13/CXCR5 is required for the architectural organization of B cells within lymphoid follicles. In this study, we showed that CXCL13 was secreted by FL cells. FL cells expressed CXCR5 and migrated in response to CXCL13. Furthermore, we observed a synergistic effect between CXCL13 and CXCL12 (SDF-1), a chemokine produced by stromal cells in lymphoid tissues. The production of CXCL13 by FL cells and CXCL12 by stromal cells probably directs and participates in the accumulation of FL cells within specific anatomic sites.


Asunto(s)
Quimiocinas CXC/metabolismo , Quimiotaxis de Leucocito , Linfoma Folicular/metabolismo , Apoptosis , Linfocitos B/metabolismo , Linfocitos B/patología , Ligando de CD40/farmacología , Células Cultivadas , Quimiocina CXCL13 , Citometría de Flujo , Humanos , Interleucina-4/farmacología , Linfoma Folicular/patología , Receptores CXCR5 , Receptores de Quimiocina , Receptores de Citocinas/metabolismo , Estimulación Química , Células Tumorales Cultivadas
20.
Br J Haematol ; 122(5): 745-59, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12930384

RESUMEN

In order fully to identify secondary chromosomal alterations, such as duplications, additions and marker chromosomes that remained unresolved by G banding, 60 cases of t(14;18)-positive follicular lymphoma (FL) were analysed by multicolour karyotyping techniques [multicolour fluorescence in situ hybridization (MFISH)/multicolour banding for chromosome 1 (MBAND1)]. A total of 165 additional structural chromosomal aberrations were delineated. An increased frequency of chromosomal gains involving X, 1q, 2, 3q27-q29, 5, 6p11-p21, 7, 8, 11, 12, 14q32, 17q, 18 and 21 and deletions of 1p36, 3q28-q29, 6q, 10q22-q24 and 17p11-p13 was revealed by the MFISH/MBAND1 analysis. Balanced translocations other than t(14;18) were uncommon, whereas unbalanced translocations were numerous. Deletion of 1p36 and duplication of 1p33-p35, 1p12-p21 and 1q21-q41 were regularly involved in chromosome 1 alterations, seen in 53% of the cases. A strong correlation was demonstrated between gains of individual chromosomal bands and increased gene expression, including 1q22/MNDA, 6p21/CDKN1A, 12q13-q14/SAS, 17q23/ZNF161, 18q21/BCL2 and Xq13/IL2RG. Unfavourable overall survival was associated with del(1)(p36) and dup(18q). These data support the notion that translocation events are primarily responsible for FL disease initiation, whereas the unbalanced chromosomal gains and losses that mirror the gene expression patterns characterize clonal evolution and disease progression, and thus provide further insights into the biology of FL.


Asunto(s)
Cromosomas Humanos Par 14 , Cromosomas Humanos Par 18 , Linfoma Folicular/genética , Translocación Genética , Adulto , Anciano , Anciano de 80 o más Años , Aberraciones Cromosómicas , Progresión de la Enfermedad , Femenino , Eliminación de Gen , Humanos , Hibridación Fluorescente in Situ , Masculino , Persona de Mediana Edad , Pronóstico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA