Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Biomaterials ; 291: 121910, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36403325

RESUMEN

Renal tubular cells frequently lose differentiation markers and physiological properties when propagated in conventional cell culture conditions. Embedding cells in 3D microenvironments or controlling their 3D assembly by bioprinting can enhance their physiological properties, which is beneficial for modeling diseases in vitro. A potential cellular source for modeling renal tubular physiology and kidney diseases in vitro are directly reprogrammed induced renal tubular epithelial cells (iRECs). iRECs were cultured in various biomaterials and as bioprinted tubular structures. They showed high compatibility with the embedding substrates and dispensing methods. The morphology of multicellular aggregates was substantially influenced by the 3D microenvironment. Transcriptomic analyses revealed signatures of differentially expressed genes specific to each of the selected biomaterials. Using a new cellular model for autosomal-dominant polycystic kidney disease, Pkd1-/- iRECs showed disrupted morphology in bioprinted tubules and a marked upregulation of the Aldehyde dehydrogenase 1a1 (Aldh1a1). In conclusion, 3D microenvironments strongly influence the morphology and expression profiles of iRECs, help to unmask disease phenotypes, and can be adapted to experimental demands. Combining a direct reprogramming approach with appropriate biomaterials will facilitate construction of biomimetic kidney tubules and disease models at the microscale.


Asunto(s)
Biomimética , Enfermedades Renales Poliquísticas , Humanos , Riñón , Células Epiteliales , Materiales Biocompatibles
2.
Biochem Biophys Res Commun ; 584: 19-25, 2021 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-34753064

RESUMEN

The primary cilium is a sensory organelle at the cell surface with integral functions in cell signaling. It contains a microtubular axoneme that is rooted in the basal body (BB) and serves as a scaffold for the movement of intraflagellar transport (IFT) particles by Kinesin-2 along the cilium. Ift88, a member of the anterograde moving IFT-B1 complex, as well as the Kinesin-2 subunit Kif3a are required for cilia formation. To facilitate signaling, the cilium restricts the access of molecules to its membrane ("ciliary gate"). This is thought to be mediated by cytoskeletal barriers ("subciliary domains") originating from the BB subdistal/distal appendages, the periciliary membrane compartment (PCMC) as well as the transition fibers and zone (TF/TZ). The PCMC is a poorly characterized membrane domain surrounding the ciliary base with exclusion of certain apical membrane proteins. Here we describe that Ift88, but not Kinesin-2, is required for the establishment of the PCMC in MDCK cells. Likewise, in C. elegans mutants of the Ift88 ortholog osm-5 fail to establish the PCMC, while Kinesin-2 deficient osm-3 mutants form PCMCs normally. Furthermore, disruption of IFT-B1 into two subcomplexes, while disrupting ciliogenesis, does not interfere with PCMC formation. Our findings suggest that cilia are not a prerequisite for the formation of the PCMC, and that separate machineries with partially overlapping functions are required for the establishment of each.


Asunto(s)
Membrana Celular/metabolismo , Cilios/metabolismo , Células Epiteliales/metabolismo , Cinesinas/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Animales , Cuerpos Basales/metabolismo , Caenorhabditis elegans/citología , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Citoesqueleto/metabolismo , Perros , Células de Riñón Canino Madin Darby , Microscopía Fluorescente , Proteínas del Tejido Nervioso/metabolismo , Transducción de Señal
3.
Sci Rep ; 11(1): 15139, 2021 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-34301992

RESUMEN

Autosomal dominant polycystic kidney disease (ADPKD) affects more than 12 million people worldwide. Mutations in PKD1 and PKD2 cause cyst formation through unknown mechanisms. To unravel the pathogenic mechanisms in ADPKD, multiple studies have investigated transcriptional mis-regulation in cystic kidneys from patients and mouse models, and numerous dysregulated genes and pathways have been described. Yet, the concordance between studies has been rather limited. Furthermore, the cellular and genetic diversity in cystic kidneys has hampered the identification of mis-expressed genes in kidney epithelial cells with homozygous PKD mutations, which are critical to identify polycystin-dependent pathways. Here we performed transcriptomic analyses of Pkd1- and Pkd2-deficient mIMCD3 kidney epithelial cells followed by a meta-analysis to integrate all published ADPKD transcriptomic data sets. Based on the hypothesis that Pkd1 and Pkd2 operate in a common pathway, we first determined transcripts that are differentially regulated by both genes. RNA sequencing of genome-edited ADPKD kidney epithelial cells identified 178 genes that are concordantly regulated by Pkd1 and Pkd2. Subsequent integration of existing transcriptomic studies confirmed 31 previously described genes and identified 61 novel genes regulated by Pkd1 and Pkd2. Cluster analyses then linked Pkd1 and Pkd2 to mRNA splicing, specific factors of epithelial mesenchymal transition, post-translational protein modification and epithelial cell differentiation, including CD34, CDH2, CSF2RA, DLX5, HOXC9, PIK3R1, PLCB1 and TLR6. Taken together, this model-based integrative analysis of transcriptomic alterations in ADPKD annotated a conserved core transcriptomic profile and identified novel candidate genes for further experimental studies.


Asunto(s)
Células Epiteliales/patología , Epitelio/patología , Riñón Poliquístico Autosómico Dominante/genética , Transcripción Genética/genética , Animales , Células Cultivadas , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica/genética , Ratones , Ratones Endogámicos C57BL , Mutación/genética , Enfermedades Renales Poliquísticas/genética , Enfermedades Renales Poliquísticas/patología , Riñón Poliquístico Autosómico Dominante/patología , Transducción de Señal/genética , Canales Catiónicos TRPP/genética
4.
J Am Soc Nephrol ; 31(5): 1035-1049, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32238474

RESUMEN

BACKGROUND: The inactivation of the ciliary proteins polycystin 1 or polycystin 2 leads to autosomal dominant polycystic kidney disease (ADPKD). Although signaling by primary cilia and interstitial inflammation both play a critical role in the disease, the reciprocal interactions between immune and tubular cells are not well characterized. The transcription factor STAT3, a component of the cilia proteome that is involved in crosstalk between immune and nonimmune cells in various tissues, has been suggested as a factor fueling ADPKD progression. METHOD: To explore how STAT3 intersects with cilia signaling, renal inflammation, and cyst growth, we used conditional murine models involving postdevelopmental ablation of Pkd1, Stat3, and cilia, as well as cultures of cilia-deficient or STAT3-deficient tubular cell lines. RESULTS: Our findings indicate that, although primary cilia directly modulate STAT3 activation in vitro, the bulk of STAT3 activation in polycystic kidneys occurs through an indirect mechanism in which primary cilia trigger macrophage recruitment to the kidney, which in turn promotes Stat3 activation. Surprisingly, although inactivating Stat3 in Pkd1-deficient tubules slightly reduced cyst burden, it resulted in a massive infiltration of the cystic kidneys by macrophages and T cells, precluding any improvement of kidney function. We also found that Stat3 inactivation led to increased expression of the inflammatory chemokines CCL5 and CXCL10 in polycystic kidneys and cultured tubular cells. CONCLUSIONS: STAT3 appears to repress the expression of proinflammatory cytokines and restrict immune cell infiltration in ADPKD. Our findings suggest that STAT3 is not a critical driver of cyst growth in ADPKD but rather plays a major role in the crosstalk between immune and tubular cells that shapes disease expression.


Asunto(s)
Túbulos Renales/metabolismo , Riñón Poliquístico Autosómico Dominante/patología , Factor de Transcripción STAT3/fisiología , Anciano de 80 o más Años , Animales , Células Cultivadas , Quimiocina CCL5/metabolismo , Quimiocina CXCL10/metabolismo , Cilios/metabolismo , Perros , Humanos , Inflamación , Túbulos Renales/patología , Macrófagos/fisiología , Células de Riñón Canino Madin Darby , Ratones , Ratones Endogámicos C57BL , Riñón Poliquístico Autosómico Dominante/genética , Riñón Poliquístico Autosómico Dominante/inmunología , Riñón Poliquístico Autosómico Dominante/metabolismo , Organismos Libres de Patógenos Específicos , Linfocitos T/fisiología , Canales Catiónicos TRPP/deficiencia , Canales Catiónicos TRPP/metabolismo
5.
EMBO J ; 37(15)2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29925518

RESUMEN

Polycystic kidney disease (PKD) and other renal ciliopathies are characterized by cysts, inflammation, and fibrosis. Cilia function as signaling centers, but a molecular link to inflammation in the kidney has not been established. Here, we show that cilia in renal epithelia activate chemokine signaling to recruit inflammatory cells. We identify a complex of the ciliary kinase LKB1 and several ciliopathy-related proteins including NPHP1 and PKD1. At homeostasis, this ciliary module suppresses expression of the chemokine CCL2 in tubular epithelial cells. Deletion of LKB1 or PKD1 in mouse renal tubules elevates CCL2 expression in a cell-autonomous manner and results in peritubular accumulation of CCR2+ mononuclear phagocytes, promoting a ciliopathy phenotype. Our findings establish an epithelial organelle, the cilium, as a gatekeeper of tissue immune cell numbers. This represents an unexpected disease mechanism for renal ciliopathies and establishes a new model for how epithelial cells regulate immune cells to affect tissue homeostasis.


Asunto(s)
Quimiocina CCL2/metabolismo , Cilios/patología , Enfermedades Renales Quísticas/congénito , Riñón Poliquístico Autosómico Dominante/patología , Proteína Quinasa C/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Quinasas Activadas por AMP , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas Portadoras/metabolismo , Línea Celular , Proteínas del Citoesqueleto , Perros , Células Epiteliales/metabolismo , Femenino , Células HEK293 , Humanos , Enfermedades Renales Quísticas/patología , Túbulos Renales/citología , Túbulos Renales/patología , Macrófagos/metabolismo , Células de Riñón Canino Madin Darby , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fagocitosis/fisiología , Riñón Poliquístico Autosómico Dominante/genética , Proteína Quinasa C/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Pez Cebra
6.
Pflugers Arch ; 469(2): 303-311, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27987038

RESUMEN

Recent advances in genome editing technologies have enabled the rapid and precise manipulation of genomes, including the targeted introduction, alteration, and removal of genomic sequences. However, respective methods have been described mainly in non-differentiated or haploid cell types. Genome editing of well-differentiated renal epithelial cells has been hampered by a range of technological issues, including optimal design, efficient expression of multiple genome editing constructs, attainable mutation rates, and best screening strategies. Here, we present an easily implementable workflow for the rapid generation of targeted heterozygous and homozygous genomic sequence alterations in renal cells using transcription activator-like effector nucleases (TALENs) and the clustered regularly interspaced short palindromic repeat (CRISPR) system. We demonstrate the versatility of established protocols by generating novel cellular models for studying autosomal dominant polycystic kidney disease (ADPKD). Furthermore, we show that cell culture-validated genetic modifications can be readily applied to mouse embryonic stem cells (mESCs) for the generation of corresponding mouse models. The described procedure for efficient genome editing can be applied to any cell type to study physiological and pathophysiological functions in the context of precisely engineered genotypes.


Asunto(s)
Diferenciación Celular/genética , Células Epiteliales/metabolismo , Genoma/genética , Riñón/metabolismo , Animales , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , ADN Complementario/genética , Células Madre Embrionarias/metabolismo , Edición Génica/métodos , Genotipo , Humanos , Ratones , Enfermedades Renales Poliquísticas/genética , Nucleasas de los Efectores Tipo Activadores de la Transcripción/genética
7.
Nat Cell Biol ; 18(6): 657-67, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27214279

RESUMEN

Autophagy is an adaptation mechanism that is vital for cellular homeostasis in response to various stress conditions. Previous reports indicate that there is a functional interaction between the primary cilium (PC) and autophagy. The PC, a microtubule-based structure present at the surface of numerous cell types, is a mechanical sensor. Here we show that autophagy induced by fluid flow regulates kidney epithelial cell volume in vitro and in vivo. PC ablation blocked autophagy induction and cell-volume regulation. In addition, inhibition of autophagy in ciliated cells impaired the flow-dependent regulation of cell volume. PC-dependent autophagy can be triggered either by mTOR inhibition or a mechanism dependent on the polycystin 2 channel. Only the LKB1-AMPK-mTOR signalling pathway was required for the flow-dependent regulation of cell volume by autophagy. These findings suggest that therapies regulating autophagy should be considered in developing treatments for PC-related diseases.


Asunto(s)
Autofagia , Fenómenos Fisiológicos Celulares , Cilios/fisiología , Células Epiteliales/citología , Células Epiteliales/fisiología , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/fisiología , Proteínas Quinasas Activadas por AMP/fisiología , Animales , Tamaño de la Célula , Perros , Immunoblotting , Células de Riñón Canino Madin Darby , Ratones , Ratones Noqueados , Microscopía Fluorescente , Proteínas Serina-Treonina Quinasas/fisiología , Transducción de Señal , Serina-Treonina Quinasas TOR/fisiología
8.
Clin Kidney J ; 8(6): 690-4, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26613025

RESUMEN

COL4A5 mutations are a known cause of Alport syndrome, which typically manifests with haematuria, hearing loss and ocular symptoms. Here we report on a 16-year-old male patient with a negative family history who presented with proteinuria, progressive renal failure and haemolysis, but without overt haematuria or hearing loss. A renal biopsy revealed features of atypical IgA nephropathy, while a second biopsy a year later showed features of focal segmental glomerulosclerosis, but was finally diagnosed as chronic thrombotic microangiopathy. Targeted sequencing of candidate genes for steroid-resistant nephrotic syndrome and congenital thrombotic microangiopathy was negative. Despite all therapeutic efforts, including angiotensin-converting enzyme inhibition, immunosuppressive therapy, plasma exchanges and rituximab, the patient progressed to end-stage renal disease. When a male cousin presented with nephrotic syndrome years later, whole-exome sequencing identified a shared disruptive COL4A5 mutation (p.F222C) that showed X-linked segregation. Thus, mutations in COL4A5 give rise to a broader spectrum of clinical presentation than commonly suspected, highlighting the benefits of comprehensive rather than candidate genetic testing in young patients with otherwise unexplained glomerular disease. Our results are in line with an increasing number of atypical presentations of single-gene disorders identified through genome-wide sequencing.

9.
Cell Logist ; 4(1): e28928, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24843830

RESUMEN

Apical lumen formation is a key step during epithelial morphogenesis of tubular organs. Appropriate transport and targeting of apical proteins to the apical membrane initiation site (AMIS) plays a crucial role in establishing a solitary, central lumen. FIP5, a Rab11-interacting protein, is an important regulator that directs apical endosome trafficking along microtubules toward the AMIS during cytokinesis. However, it is unknown which molecular motor(s) transports FIP5-positive apical endosomes during lumen initiation, and how this process is regulated. In this study, we demonstrate that the interaction of FIP5 with the microtubule motor, Kinesin-2, is required for the movement of FIP5-endosomes and delivery of these endosomes from centrosomes to the cleavage furrow during apical lumen initiation. Loss of Kinesin-2 disrupts targeting of apical proteins to the AMIS and results in multiple lumen formation in MDCK cysts. Our data provide more details to the molecular mechanism of FIP5-dependent apical trafficking during apical lumen formation.

10.
Cell ; 154(4): 859-74, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23953116

RESUMEN

Mammalian target of rapamycin complex 1 (mTORC1) controls growth and survival in response to metabolic cues. Oxidative stress affects mTORC1 via inhibitory and stimulatory inputs. Whereas downregulation of TSC1-TSC2 activates mTORC1 upon oxidative stress, the molecular mechanism of mTORC1 inhibition remains unknown. Here, we identify astrin as an essential negative mTORC1 regulator in the cellular stress response. Upon stress, astrin inhibits mTORC1 association and recruits the mTORC1 component raptor to stress granules (SGs), thereby preventing mTORC1-hyperactivation-induced apoptosis. In turn, balanced mTORC1 activity enables expression of stress factors. By identifying astrin as a direct molecular link between mTORC1, SG assembly, and the stress response, we establish a unifying model of mTORC1 inhibition and activation upon stress. Importantly, we show that in cancer cells, apoptosis suppression during stress depends on astrin. Being frequently upregulated in tumors, astrin is a potential clinically relevant target to sensitize tumors to apoptosis.


Asunto(s)
Apoptosis , Neoplasias de la Mama/metabolismo , Proteínas de Ciclo Celular/metabolismo , Complejos Multiproteicos/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Gránulos Citoplasmáticos/metabolismo , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina , Estrés Oxidativo , Proteína Reguladora Asociada a mTOR
11.
Nat Genet ; 45(8): 951-6, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23793029

RESUMEN

Nephronophthisis is an autosomal recessive cystic kidney disease that leads to renal failure in childhood or adolescence. Most NPHP gene products form molecular networks. Here we identify ANKS6 as a new NPHP family member that connects NEK8 (NPHP9) to INVS (NPHP2) and NPHP3. We show that ANKS6 localizes to the proximal cilium and confirm its role in renal development through knockdown experiments in zebrafish and Xenopus laevis. We also identify six families with ANKS6 mutations affected by nephronophthisis, including severe cardiovascular abnormalities, liver fibrosis and situs inversus. The oxygen sensor HIF1AN hydroxylates ANKS6 and INVS and alters the composition of the ANKS6-INVS-NPHP3 module. Knockdown of Hif1an in Xenopus results in a phenotype that resembles loss of other NPHP proteins. Network analyses uncovered additional putative NPHP proteins and placed ANKS6 at the center of this NPHP module, explaining the overlapping disease manifestation caused by mutation in ANKS6, NEK8, INVS or NPHP3.


Asunto(s)
Enfermedades Renales Quísticas/genética , Cinesinas/genética , Proteínas Nucleares/genética , Proteínas Quinasas/genética , Factores de Transcripción/genética , Animales , Cilios/metabolismo , Consanguinidad , Exones , Técnicas de Silenciamiento del Gen , Humanos , Intrones , Enfermedades Renales Quísticas/metabolismo , Cinesinas/metabolismo , Ratones , Mutación , Quinasas Relacionadas con NIMA , Proteínas Nucleares/metabolismo , Fenotipo , Enfermedades Renales Poliquísticas/genética , Unión Proteica , Mapas de Interacción de Proteínas , Proteínas Quinasas/metabolismo , Transporte de Proteínas , Factores de Transcripción/metabolismo , Xenopus/embriología , Xenopus/metabolismo , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
12.
Nephrol Dial Transplant ; 28(3): 518-26, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23314319

RESUMEN

Since the discovery that proteins mutated in different forms of polycystic kidney disease (PKD) are tightly associated with primary cilia, strong efforts have been made to define the role of this organelle in the pathogenesis of cyst formation. Cilia are filiform microtubular structures, anchored in the basal body and extending from the apical membrane into the tubular lumen. Early work established that cilia act as flow sensors, eliciting calcium transients in response to bending, which involve the two proteins mutated in autosomal dominant PKD (ADPKD), polycystin-1 and -2. Loss of cilia alone is insufficient to cause cyst formation. Nevertheless, a large body of evidence links flow sensing by cilia to aspects relevant for cyst formation such as cell polarity, Stat6- and mammalian target of rapamycin signalling. This review summarizes the current literature on cilia and flow sensing with respect to PKD and discusses how these findings intercalate with different aspects of cyst formation.


Asunto(s)
Cilios/patología , Mecanotransducción Celular/fisiología , Enfermedades Renales Poliquísticas/patología , Animales , Humanos
13.
Kidney Int ; 79(5): 502-11, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21085109

RESUMEN

The immunosuppressive drug rapamycin has helped to identify a large signaling network around the target of rapamycin (TOR) protein that integrates information on nutrient availability and growth factors to control protein synthesis and cell size. Studies using rapamycin in animal models of kidney disease indicate that mTOR deregulation has a role in glomerular disease, polycystic kidney disease, and renal cancer. The role of mTOR activation in podocytes is context dependent, and indirect evidence suggests that mTOR may have a role in chronic podocyte loss. Several lines of evidence show that cyst formation in polycystic kidney disease (PKD) involves mTOR activation and its upstream regulator TSC. Polycystin 1 regulates mTOR activity through different pathways, and TSC intersects with the primary cilium, a crucial cell organelle in the pathogenesis of PKD. Data from hamartoma syndromes provide clear evidence that mutation of members of the mTOR network results in renal cancers. The detailed analysis of renal cell carcinomas has revealed a positive feedback loop involving VHL and mTOR. Rapamycin and its derivatives have been approved for the treatment of advanced renal cancer and are being investigated for the treatment of PKD. Discrepancies exist between the effects of rapamycin in animal models and the clinical experience with patients, precluding the widespread use of mTOR inhibitors in kidney disease. The details of mTOR signaling in the kidney need to be clarified to hopefully develop targeted treatments for renal disease in the future.


Asunto(s)
Inmunosupresores/uso terapéutico , Enfermedades Renales/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Sirolimus/uso terapéutico , Serina-Treonina Quinasas TOR/fisiología , Animales , Autofagia , Cilios/fisiología , Ensayos Clínicos como Asunto , Nefropatías Diabéticas/etiología , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina , Complejos Multiproteicos , Nefronas/patología , Podocitos/patología , Proteínas/fisiología , Canales Catiónicos TRPP/fisiología
14.
Proc Natl Acad Sci U S A ; 107(47): 20388-93, 2010 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-21059920

RESUMEN

Mutations of inversin cause type II nephronophthisis, an infantile autosomal recessive disease characterized by cystic kidney disease and developmental defects. Inversin regulates Wnt signaling and is required for convergent extension movements during early embryogenesis. We now show that Inversin is essential for Xenopus pronephros formation, involving two distinct and opposing forms of cell movements. Knockdown of Inversin abrogated both proximal pronephros extension and distal tubule differentiation, phenotypes similar to that of Xenopus deficient in Frizzled-8. Exogenous Inversin rescued the pronephric defects caused by lack of Frizzled-8, indicating that Inversin acts downstream of Frizzled-8 in pronephros morphogenesis. Depletion of Inversin prevents the recruitment of Dishevelled in response to Frizzled-8 and impeded the accumulation of Dishevelled at the apical membrane of tubular epithelial cells in vivo. Thus, defective tubule morphogenesis seems to contribute to the renal pathology observed in patients with nephronophthisis type II.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Riñón/embriología , Receptores de Superficie Celular/metabolismo , Transducción de Señal/fisiología , Factores de Transcripción/metabolismo , Proteínas de Xenopus/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Proteínas Dishevelled , Fluorescencia , Hibridación in Situ , Riñón/metabolismo , Ratones , Microscopía Confocal , Oligonucleótidos/genética , Fosfoproteínas/metabolismo , Proteínas Wnt/metabolismo , Xenopus
15.
Nat Cell Biol ; 12(11): 1115-22, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20972424

RESUMEN

The mTOR pathway is the central regulator of cell size. External signals from growth factors and nutrients converge on the mTORC1 multi-protein complex to modulate downstream targets, but how the different inputs are integrated and translated into specific cellular responses is incompletely understood. Deregulation of the mTOR pathway occurs in polycystic kidney disease (PKD), where cilia (filiform sensory organelles) fail to sense urine flow because of inherited mutations in ciliary proteins. We therefore investigated if cilia have a role in mTOR regulation. Here, we show that ablation of cilia in transgenic mice results in enlarged cells when compared with control animals. In vitro analysis demonstrated that bending of the cilia by flow is required for mTOR downregulation and cell-size control. Surprisingly, regulation of cell size by cilia is independent of flow-induced calcium transients, or Akt. However, the tumour-suppressor protein Lkb1 localises in the cilium, and flow results in increased AMPK phosphorylation at the basal body. Conversely, knockdown of Lkb1 prevents normal cell-size regulation under flow conditions. Our results demonstrate that the cilium regulates mTOR signalling and cell size, and identify the cilium-basal body compartment as a spatially restricted activation site for Lkb1 signalling.


Asunto(s)
Tamaño de la Célula , Cilios/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas/metabolismo , Proteínas Quinasas Activadas por AMP , Animales , Calcio/metabolismo , Línea Celular , Cilios/química , Perros , Cinesinas/deficiencia , Cinesinas/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Transgénicos , Complejos Multiproteicos , Fosforilación , Transducción de Señal , Serina-Treonina Quinasas TOR
17.
Hum Mol Genet ; 19(1): 16-24, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19801576

RESUMEN

Autosomal dominant polycystic liver disease (PCLD) is caused by mutations of either PRKCSH or Sec63, two proteins associated with the endoplasmic reticulum (ER). Both proteins are involved in carbohydrate processing, folding and translocation of newly synthesized glycoproteins. It is postulated that defective quality control of proteins initiates endoplasmic reticulum-associated degradation (ERAD), which disrupts hepatic homeostasis in patients with PRKCSH or Sec63 mutations. However, the precise molecular mechanisms are not known. Here, we show that over-expression or depletion of PRKCSH in zebrafish embryos leads to pronephric cysts, abnormal body curvature and situs inversus. Identical phenotypic changes are induced by depletion or over-expression of TRPP2. Increased PRKCSH levels ameliorate developmental abnormalities caused by over-expressed TRPP2, whereas excess TRPP2 can compensate the loss PRKCSH, indicating that the proteins share a common signaling pathway. PRKCSH binds the C-terminal domain of TRPP2, and both proteins co-localize within the ER. Furthermore, PRKCSH interacts with Herp, and inhibits Herp-mediated ubiquitination of TRPP2. Our findings suggest that PRKCSH functions as a chaperone-like molecule, which prevents ERAD of TRPP2. Dysequilibrium between TRPP2 and PRKCSH may lead to cyst formation in PCLD patients with PRKCSH mutations, and thereby account for the overlapping manifestations observed in PCLD and autosomal dominant polycystic kidney disease.


Asunto(s)
Proteínas Portadoras/metabolismo , Chaperonas Moleculares/metabolismo , Mutación/genética , Riñón Poliquístico Autosómico Dominante/metabolismo , Procesamiento Proteico-Postraduccional , Canales Catiónicos TRPP/metabolismo , Ubiquitinas/metabolismo , Proteínas de Pez Cebra/metabolismo , Animales , Proteínas Portadoras/genética , Perros , Embrión no Mamífero/anomalías , Embrión no Mamífero/efectos de los fármacos , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Chaperonas Moleculares/genética , Nefronas/efectos de los fármacos , Nefronas/metabolismo , Nefronas/patología , Oligonucleótidos Antisentido/farmacología , Unión Proteica/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Canales Catiónicos TRPP/genética , Ubiquitinación/efectos de los fármacos , Ubiquitinas/genética , Pez Cebra/embriología , Proteínas de Pez Cebra/genética
18.
Proc Natl Acad Sci U S A ; 106(21): 8579-84, 2009 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-19439659

RESUMEN

Spatial organization of cells and their appendages is controlled by the planar cell polarity pathway, a signaling cascade initiated by the protocadherin Fat in Drosophila. Vertebrates express 4 Fat molecules, Fat1-4. We found that depletion of Fat1 caused cyst formation in the zebrafish pronephros. Knockdown of the PDZ domain containing the adaptor protein Scribble intensified the cyst-promoting phenotype of Fat1 depletion, suggesting that Fat1 and Scribble act in overlapping signaling cascades during zebrafish pronephros development. Supporting the genetic interaction with Fat1, Scribble recognized the PDZ-binding site of Fat1. Depletion of Yes-associated protein 1 (YAP1), a transcriptional co-activator inhibited by Hippo signaling, ameliorated the cyst formation in Fat1-deficient zebrafish, whereas Scribble inhibited the YAP1-induced cyst formation. Thus, reduced Hippo signaling and subsequent YAP1 disinhibition seem to play a role in the development of pronephric cysts after depletion of Fat1 or Scribble. We hypothesize that Hippo signaling is required for normal pronephros development in zebrafish and that Scribble is a candidate link between Fat and the Hippo signaling cascade in vertebrates.


Asunto(s)
Riñón/embriología , Riñón/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente , Cadherinas/genética , Cadherinas/metabolismo , Línea Celular , Embrión no Mamífero/embriología , Embrión no Mamífero/metabolismo , Humanos , Proteínas de la Membrana/genética , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Regiones Promotoras Genéticas/genética , Unión Proteica , Proteínas Serina-Treonina Quinasas/genética , Serina-Treonina Quinasa 3 , Pez Cebra/genética , Proteínas de Pez Cebra/genética
19.
J Cell Biol ; 182(3): 437-47, 2008 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-18695040

RESUMEN

The primary cilium has evolved as a multifunctional cellular compartment that decorates most vertebrate cells. Cilia sense mechanical stimuli in various organs, but the molecular mechanisms that convert the deflection of cilia into intracellular calcium transients have remained elusive. Polycystin-2 (TRPP2), an ion channel mutated in polycystic kidney disease, is required for cilia-mediated calcium transients but lacks mechanosensitive properties. We find here that TRPP2 utilizes TRPV4 to form a mechano- and thermosensitive molecular sensor in the cilium. Depletion of TRPV4 in renal epithelial cells abolishes flow-induced calcium transients, demonstrating that TRPV4, like TRPP2, is an essential component of the ciliary mechanosensor. Because TRPV4-deficient zebrafish and mice lack renal cysts, our findings challenge the concept that defective ciliary flow sensing constitutes the fundamental mechanism of cystogenesis.


Asunto(s)
Canales Catiónicos TRPP/metabolismo , Canales Catiónicos TRPV/metabolismo , Animales , Señalización del Calcio , Línea Celular , Cilios/metabolismo , Quistes/metabolismo , Células Epiteliales/metabolismo , Humanos , Oocitos/metabolismo , Unión Proteica , Transporte de Proteínas , Temperatura
20.
Pflugers Arch ; 456(6): 1025-35, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18425533

RESUMEN

Kidney cysts are characterized by an abnormal tubular geometry that may result from loss of orientation and random cell divisions during renal development. Since cystic kidney disease is caused by mutations of ciliary proteins and cilia act as flow sensors in the kidney, we examined three polarized events in Madin Darby Canine Kidney cells under flow: cell division, cell migration, and centriole movement. We found that the mitotic orientation of dividing cells was not affected by flow and was randomly distributed in relation to the direction of the flow. Flow did not alter the direction and speed of cell migration in a wound-healing assay. However, flow resulted in increased motility of centrioles and biased centrioles to move along the axis of the flow. This effect was lost after flow-induced calcium signaling was abolished by a mutant polycystin 2. Our findings suggest that the cilium may translate fluid flow into altered centriole movements to provide tubular epithelial cells with the spatial orientation required to establish and/or maintain a normal tubular geometry.


Asunto(s)
Centriolos/fisiología , Corriente Citoplasmática/fisiología , Células Epiteliales/fisiología , Túbulos Renales/fisiología , Animales , División Celular/fisiología , Línea Celular , Movimiento Celular/fisiología , Centriolos/ultraestructura , Centrosoma/fisiología , Cilios/fisiología , Cilios/ultraestructura , Cámaras de Difusión de Cultivos , Perros , Células Epiteliales/ultraestructura , Colorantes Fluorescentes , Fura-2 , Indicadores y Reactivos , Túbulos Renales/citología , Túbulos Renales/metabolismo , Nefronas/citología , Nefronas/fisiología , Plásmidos/genética , Plásmidos/fisiología , Proteínas/metabolismo , Cicatrización de Heridas/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA