Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-38464230

RESUMEN

Nephronophthisis (NPHP) and autosomal dominant Polycystic Kidney Disease (ADPKD) are two genetically distinct forms of Polycystic Kidney Disease (PKD), yet both diseases present with kidney cysts and a gradual decline in renal function. Prevailing dogma in PKD is that changes in kidney architecture account for the decline in kidney function, but the molecular/cellular basis of such coupling is unknown. To address this question, we induced a form of proteome reprogramming by deleting Fbxw7 encoding FBW7, the recognition receptor of the SCF FBW7 E3 ubiquitin ligase in different segments of the kidney tubular system. Deletion of Fbxw7 in the medulla led to a juvenile-adult NPHP-like phenotype, where the decline in renal function was due to SOX9-mediated interstitial fibrosis rather than cystogenesis. In contrast, the decline of renal function in ADPKD is coupled to cystic expansion via the abnormal accumulation of FBW7 in the proximal tubules and other cell types in the renal cortex. We propose that FBW7 functions at the apex of a protein network that determines renal function in ADPKD by sensing architectural changes induced by cystic expansion.

3.
Blood Cells Mol Dis ; 102: 102760, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37267696

RESUMEN

Hematopoiesis and lineage commitment are regulated by several conserved cell-intrinsic signaling pathways, including MAPKs and ß-catenin/TCF/LEF. The Inhibitor of MyoD Family A (I-MFA), a transcriptional repressor and tumor suppressor gene, interacts with these pathways and is dysregulated in chronic and acute myeloid leukemias, suggesting it may play a role in development and differentiation during hematopoiesis. To study this, immune cell populations in the bone marrow (BM) and periphery were analyzed in mice lacking Mdfi, encoding I-MFA (I-MFA-/-), and wild type (WT) controls. I-MFA-/- mice had reduced spleen and BM cellularity, with significant hyposplenism, compared to WT mice. In blood, total red blood cells and platelet counts were significantly reduced in I-MFA-/- mice, accompanied by a reduction in megakaryocyte (MK)/erythrocyte progenitor cells and an increase in myeloid progenitors in BM compared to WT mice. The K562 cell line exhibits PMA-induced MK differentiation, and shRNA knockdown of I-MFA resulted in reduced differentiation compared to control, with an increase and prolongation in phospho-JNK and phospho-ERK signaling. Overexpression of I-MFA promoted MK differentiation. These results suggest I-MFA plays a cell-intrinsic role in the response to differentiation signals, an effect that can be explored in the context of hematological cancers or other blood proliferative disorders.


Asunto(s)
Médula Ósea , Megacariocitos , Ratones , Animales , Médula Ósea/metabolismo , Diferenciación Celular , Hematopoyesis , Células de la Médula Ósea/patología , Linaje de la Célula
4.
ACS Nano ; 17(10): 9326-9337, 2023 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-37129853

RESUMEN

The RAS-transformed cells utilize macropinocytosis to acquire amino acids to support their uncontrolled growth. However, targeting RAS to inhibit macropinocytosis remains a challenge. Here, we report that gold nanoparticles (GNP) inhibit macropinocytosis by decreasing KRAS activation. Using surface-modified and unmodified GNP, we showed that unmodified GNP specifically sequestered both wild-type and mutant KRAS and inhibited its activation, irrespective of growth factor stimulation, while surface-passivated GNP had no effect. Alteration of KRAS activation is reflected on downstream signaling cascades, macropinocytosis and tumor cell growth in vitro, and two independent preclinical human xenograft models of pancreatic cancer in vivo. The current study demonstrates NP-mediated inhibition of macropinocytosis and KRAS activation and provides translational opportunities to inhibit tumor growth in a number of cancers where activation of KRAS plays a major role.


Asunto(s)
Nanopartículas del Metal , Neoplasias Pancreáticas , Humanos , Oro/farmacología , Proteínas Proto-Oncogénicas p21(ras)/genética , Pinocitosis , Neoplasias Pancreáticas/patología , Proliferación Celular , Línea Celular Tumoral , Mutación
5.
Cells ; 10(11)2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34831200

RESUMEN

The primary cilium, an antenna-like structure that protrudes out from the cell surface, is present in most cell types. It is a microtubule-based organelle that serves as a mega-signaling center and is important for sensing biochemical and mechanical signals to carry out various cellular processes such as proliferation, migration, differentiation, and many others. At any given time, cilia length is determined by a dynamic balance of cilia assembly and disassembly processes. Abnormally short or long cilia can cause a plethora of human diseases commonly referred to as ciliopathies, including, but not limited to, skeletal malformations, obesity, autosomal dominant polycystic kidney disease, retinal degeneration, and bardet-biedl syndrome. While the process of cilia assembly is studied extensively, the process of cilia disassembly and its biological role(s) are less well understood. This review discusses current knowledge on ciliary disassembly and how different cellular processes and molecular signals converge to carry out this process. This information will help us understand how the process of ciliary disassembly is regulated, identify the key steps that need further investigation, and possibly design therapeutic targets for a subset of ciliopathies that are causally linked to defective ciliary disassembly.


Asunto(s)
Cilios/metabolismo , Animales , Humanos , Microtúbulos/metabolismo , Modelos Biológicos , Polimerizacion , Transducción de Señal
6.
Commun Biol ; 4(1): 1066, 2021 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-34518642

RESUMEN

Structural defects in primary cilia have robust effects in diverse tissues and systems. However, how disorders of ciliary length lead to functional outcomes are unknown. We examined the functional role of a ciliary length control mechanism of FBW7-mediated destruction of NDE1, in mesenchymal stem cell (MSC) differentiation. We show that FBW7 functions as a master regulator of both negative (NDE1) and positive (TALPID3) regulators of ciliogenesis, with an overall positive net effect on primary cilia formation, MSC differentiation to osteoblasts, and bone architecture. Deletion of Fbxw7 suppresses ciliation, Hedgehog activity, and differentiation, which are partially rescued in Fbxw7/Nde1-null cells. We also show that NDE1, despite suppressing ciliogenesis, promotes MSC differentiation by increasing the activity of the Hedgehog pathway by direct binding and enhancing GLI2 activity in a cilia-independent manner. We propose that FBW7 controls a protein-protein interaction network coupling ciliary structure and function, which is essential for stem cell differentiation.


Asunto(s)
Cilios/metabolismo , Proteína 7 que Contiene Repeticiones F-Box-WD/genética , Proteínas Asociadas a Microtúbulos/genética , Animales , Diferenciación Celular , Proteína 7 que Contiene Repeticiones F-Box-WD/metabolismo , Masculino , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Transducción de Señal
7.
Life Sci Alliance ; 3(9)2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32651191

RESUMEN

The primary cilium is a microtubule-based, antenna-like organelle housing several signaling pathways. It follows a cyclic pattern of assembly and deciliation (disassembly and/or shedding), as cells exit and re-enter the cell cycle, respectively. In general, primary cilia loss leads to kidney cystogenesis. However, in animal models of autosomal dominant polycystic kidney disease, a major disease caused by mutations in the polycystin genes (Pkd1 or Pkd2), primary cilia ablation or acceleration of deciliation suppresses cystic growth, whereas deceleration of deciliation enhances cystogenesis. Here, we show that deciliation is delayed in the cystic epithelium of a mouse model of postnatal deletion of Pkd1 and in Pkd1- or Pkd2-null cells in culture. Mechanistic experiments show that PKD1 depletion activates the centrosomal integrity/mitotic surveillance pathway involving 53BP1, USP28, and p53 leading to a delay in deciliation. Reduced deciliation rate causes prolonged activation of cilia-based signaling pathways that could promote cystic growth. Our study links polycystins to cilia dynamics, identifies cellular deciliation downstream of the centrosomal integrity pathway, and helps explain pro-cystic effects of primary cilia in autosomal dominant polycystic kidney disease.


Asunto(s)
Cilios/metabolismo , Canales Catiónicos TRPP/metabolismo , Animales , Centrosoma/metabolismo , Cilios/fisiología , Modelos Animales de Enfermedad , Femenino , Riñón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Mitosis/fisiología , Mutación , Células 3T3 NIH , Enfermedades Renales Poliquísticas/genética , Riñón Poliquístico Autosómico Dominante/genética , Transducción de Señal , Canales Catiónicos TRPP/genética , Proteína p53 Supresora de Tumor , Proteína 1 de Unión al Supresor Tumoral P53 , Ubiquitina Tiolesterasa
8.
Cell Signal ; 72: 109640, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32305669

RESUMEN

Naturally occurring mutations in two separate genes, PKD1 and PKD2, are responsible for the vast majority of all cases of autosomal dominant polycystic kidney disease (ADPKD), one of the most common genetic diseases affecting 1 in 1000 Americans. The hallmark of ADPKD is the development of epithelial cysts in the kidney, liver, and pancreas. PKD1 encodes a large plasma membrane protein (PKD1, PC1, or Polycystin-1) with a long extracellular domain and has been speculated to function as an atypical G protein coupled receptor. PKD2 encodes an ion channel of the Transient Receptor Potential superfamily (TRPP2, PKD2, PC2, or Polycystin-2). Despite the identification of these genes more than 20 years ago, the molecular function of their encoded proteins and the mechanism(s) by which mutations in PKD1 and PKD2 cause ADPKD remain elusive. Genetic, biochemical, and functional evidence suggests they form a multiprotein complex present in multiple locations in the cell, including the plasma membrane, endoplasmic reticulum, and the primary cilium. Over the years, numerous interacting proteins have been identified using directed and unbiased approaches, and shown to modulate function, cellular localization, and protein stability and turnover of Polycystins. Delineation of the molecular composition of the Polycystin complex can have a significant impact on understanding their cellular function in health and disease states and on the identification of more specific and effective therapeutic targets.


Asunto(s)
Complejos Multiproteicos/metabolismo , Canales Catiónicos TRPP/metabolismo , Citoesqueleto de Actina/metabolismo , Animales , Proteínas de Unión al GTP/metabolismo , Humanos , Complejos Multiproteicos/química , Unión Proteica , Transducción de Señal , Canales Catiónicos TRPP/química
9.
JCI Insight ; 5(8)2020 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-32213715

RESUMEN

Familial hypocalciuric hypercalcemia (FHH) is a genetic condition associated with hypocalciuria, hypercalcemia, and, in some cases, inappropriately high levels of circulating parathyroid hormone (PTH). FHH is associated with inactivating mutations in the gene encoding the Ca2+-sensing receptor (CaSR), a GPCR, and GNA11 encoding G protein subunit α 11 (Gα11), implicating defective GPCR signaling as the root pathophysiology for FHH. However, the downstream mechanism by which CaSR activation inhibits PTH production/secretion is incompletely understood. Here, we show that mice lacking the transient receptor potential canonical channel 1 (TRPC1) develop chronic hypercalcemia, hypocalciuria, and elevated PTH levels, mimicking human FHH. Ex vivo and in vitro studies revealed that TRPC1 serves a necessary and sufficient mediator to suppress PTH secretion from parathyroid glands (PTGs) downstream of CaSR in response to high extracellular Ca2+ concentration. Gα11 physically interacted with both the N- and C-termini of TRPC1 and enhanced CaSR-induced TRPC1 activity in transfected cells. These data identify TRPC1-mediated Ca2+ signaling as an essential component of the cellular apparatus controlling PTH secretion in the PTG downstream of CaSR.


Asunto(s)
Hormona Paratiroidea/metabolismo , Canales Catiónicos TRPC/metabolismo , Animales , Señalización del Calcio/fisiología , Femenino , Humanos , Hipercalcemia/congénito , Hipercalcemia/metabolismo , Masculino , Ratones , Ratones Noqueados , Glándulas Paratiroides/metabolismo , Ratas
10.
Am J Physiol Renal Physiol ; 318(3): F673-F682, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31984795

RESUMEN

Overproduction of extracellular matrix proteins, including fibronectin by mesangial cells (MCs), contributes to diabetic nephropathy. Inhibitor of myogenic differentiation family isoform a (I-mfa) is a multifunctional cytosolic protein functioning as a transcriptional modulator or plasma channel protein regulator. However, its renal effects are unknown. The present study was conducted to determine whether I-mfa regulated fibronectin production by glomerular MCs. In human MCs, overexpression of I-mfa significantly increased fibronectin abundance. Silencing I-mfa significantly reduced the level of fibronectin mRNA and blunted transforming growth factor-ß1-stimulated production of fibronectin. We further found that high glucose increased I-mfa protein content in a time course (≥48 h) and concentration (≥25 mM)-dependent manner. Although high glucose exposure increased I-mfa at the protein level, it did not significantly alter transcripts of I-mfa in MCs. Furthermore, the abundance of I-mfa protein was significantly increased in the renal cortex of rats with diabetic nephropathy. The I-mfa protein level was also elevated in the glomerulus of mice with diabetic kidney disease. However, there was no significant difference in glomerular I-mfa mRNA levels between mice with and without diabetic nephropathy. Moreover, H2O2 significantly increased I-mfa protein abundance in a dose-dependent manner in cultured human MCs. The antioxidants polyethylene glycol-catalase, ammonium pyrrolidithiocarbamate, and N-acetylcysteine significantly blocked the high glucose-induced increase of I-mfa protein. Taken together, our results suggest that I-mfa, increased by high glucose/diabetes through the production of reactive oxygen species, stimulates fibronectin production by MCs.


Asunto(s)
Fibronectinas/metabolismo , Factores Reguladores Miogénicos/metabolismo , Animales , Antioxidantes , Glucemia , Diabetes Mellitus Experimental , Dieta Alta en Grasa , Glomérulos Renales , Masculino , Células Mesangiales , Ratones , Factores Reguladores Miogénicos/genética , ARN Mensajero , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno
11.
Cell Calcium ; 85: 102110, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31785581

RESUMEN

Calcium signaling plays a central role in bone development and homeostasis. Store operated calcium entry (SOCE) is an important calcium influx pathway mediated by calcium release activated calcium (CRAC) channels in the plasma membrane. Stromal interaction molecule 1 (STIM1) is an endoplasmic reticulum calcium sensing protein important for SOCE. We generated a mouse model expressing the STIM1 R304W mutation, causing Stormorken syndrome in humans. Stim1R304W/R304W mice showed perinatal lethality, and the only three animals that survived into adulthood presented with reduced growth, low body weight, and thoracic kyphosis. Radiographs revealed a reduced number of ribs in the Stim1R304W/R304W mice. Microcomputed tomography data revealed decreased cortical bone thickness and increased trabecular bone volume fraction in Stim1R304W/R304W mice, which had thinner and more compact bone compared to wild type mice. The Stim1R304W/+ mice showed an intermediate phenotype. Histological analyses showed that the Stim1R304W/R304W mice had abnormal bone architecture, with markedly increased number of trabeculae and reduced bone marrow cavity. Homozygous mice showed STIM1 positive osteocytes and osteoblasts. These findings highlight the critical role of the gain-of-function (GoF) STIM1 R304W protein in skeletal development and homeostasis in mice. Furthermore, the novel feature of bilateral subgingival hair growth on the lower incisors in the Stim1R304W/R304W mice and 25 % of the heterozygous mice indicate that the GoF STIM1 R304W protein also induces an abnormal epithelial cell fate.


Asunto(s)
Hueso Esponjoso/patología , Encía/crecimiento & desarrollo , Cabello/crecimiento & desarrollo , Molécula de Interacción Estromal 1/metabolismo , Animales , Huesos/anomalías , Huesos/patología , Hueso Cortical/diagnóstico por imagen , Hueso Cortical/patología , Cabello/ultraestructura , Homocigoto , Incisivo/patología , Cifosis/genética , Cifosis/patología , Megacariocitos/metabolismo , Megacariocitos/patología , Ratones , Mutación , Osteoblastos/metabolismo , Osteoblastos/patología , Osteocitos/metabolismo , Osteocitos/patología , Costillas/diagnóstico por imagen , Costillas/patología , Esplenomegalia/patología , Tórax/patología , Microtomografía por Rayos X
12.
Cell Rep ; 22(6): 1560-1573, 2018 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-29425510

RESUMEN

Transient receptor potential (TRP) channels are regulated by diverse stimuli comprising thermal, chemical, and mechanical modalities. They are also commonly regulated by phosphatidylinositol-4,5-bisphosphate (PIP2), with underlying mechanisms largely unknown. We here revealed an intramolecular interaction of the TRPP3 N and C termini (N-C) that is functionally essential. The interaction was mediated by aromatic Trp81 in pre-S1 domain and cationic Lys568 in TRP-like domain. Structure-function analyses revealed similar N-C interaction in TRPP2 as well as TRPM8/-V1/-C4 via highly conserved tryptophan and lysine/arginine residues. PIP2 bound to cationic residues in TRPP3, including K568, thereby disrupting the N-C interaction and negatively regulating TRPP3. PIP2 had similar negative effects on TRPP2. Interestingly, we found that PIP2 facilitates the N-C interaction in TRPM8/-V1, resulting in channel potentiation. The intramolecular N-C interaction might represent a shared mechanism underlying the gating and PIP2 regulation of TRP channels.


Asunto(s)
Activación del Canal Iónico/fisiología , Fosfatidilinositol 4,5-Difosfato/química , Fosfatidilinositol 4,5-Difosfato/metabolismo , Canales de Potencial de Receptor Transitorio/química , Canales de Potencial de Receptor Transitorio/metabolismo , Animales , Humanos , Unión Proteica , Dominios Proteicos , Relación Estructura-Actividad , Xenopus laevis
13.
J Am Soc Nephrol ; 28(10): 2973-2984, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28620080

RESUMEN

Autosomal dominant polycystic kidney disease (ADPKD) is caused by inactivating mutations in PKD1 (85%) or PKD2 (15%). The ADPKD proteins encoded by these genes, polycystin-1 (PC1) and polycystin-2 (PC2), form a plasma membrane receptor-ion channel complex. However, the mechanisms controlling the subcellular localization of PC1 and PC2 are poorly understood. Here, we investigated the involvement of the retromer complex, an ancient protein module initially discovered in yeast that regulates the retrieval, sorting, and retrograde transport of membrane receptors. Using yeast two-hybrid, biochemical, and cellular assays, we determined that PC2 binds two isoforms of the retromer-associated protein sorting nexin 3 (SNX3), including a novel isoform that binds PC2 in a direct manner. Knockdown of SNX3 or the core retromer protein VPS35 increased the surface expression of endogenous PC1 and PC2 in vitro and in vivo and increased Wnt-activated PC2-dependent whole-cell currents. These findings indicate that an SNX3-retromer complex regulates the surface expression and function of PC1 and PC2. Molecular targeting of proteins involved in the endosomal sorting of PC1 and PC2 could lead to new therapeutic approaches in ADPKD.


Asunto(s)
Endocitosis , Nexinas de Clasificación/metabolismo , Canales Catiónicos TRPP/metabolismo , Animales , Células HEK293 , Células HeLa , Humanos , Túbulos Renales/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Xenopus
14.
Nat Commun ; 8: 14634, 2017 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-28530221

RESUMEN

Cancer cells actively promote aerobic glycolysis to sustain their metabolic requirements through mechanisms not always clear. Here, we demonstrate that the gatekeeper of mitochondrial Ca2+ uptake, Mitochondrial Calcium Uptake 1 (MICU1/CBARA1) drives aerobic glycolysis in ovarian cancer. We show that MICU1 is overexpressed in a panel of ovarian cancer cell lines and that MICU1 overexpression correlates with poor overall survival (OS). Silencing MICU1 in vitro increases oxygen consumption, decreases lactate production, inhibits clonal growth, migration and invasion of ovarian cancer cells, whereas silencing in vivo inhibits tumour growth, increases cisplatin efficacy and OS. Mechanistically, silencing MICU1 activates pyruvate dehydrogenase (PDH) by stimulating the PDPhosphatase-phosphoPDH-PDH axis. Forced-expression of MICU1 in normal cells phenocopies the metabolic aberrations of malignant cells. Consistent with the in vitro and in vivo findings we observe a significant correlation between MICU1 and pPDH (inactive form of PDH) expression with poor prognosis. Thus, MICU1 could serve as an important therapeutic target to normalize metabolic aberrations responsible for poor prognosis in ovarian cancer.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Proteínas de Transporte de Catión/metabolismo , Resistencia a Antineoplásicos , Glucólisis , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Neoplasias Ováricas/metabolismo , Animales , Antineoplásicos/uso terapéutico , Apoptosis , Calcio/metabolismo , Línea Celular Tumoral , Cisplatino/uso terapéutico , Femenino , Humanos , Ratones Desnudos , Análisis por Micromatrices , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/mortalidad , Fosforilación Oxidativa , Fenotipo , Complejo Piruvato Deshidrogenasa/metabolismo
15.
Nat Cell Biol ; 18(7): 752-764, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27214281

RESUMEN

WNT ligands induce Ca(2+) signalling on target cells. PKD1 (polycystin 1) is considered an orphan, atypical G-protein-coupled receptor complexed with TRPP2 (polycystin 2 or PKD2), a Ca(2+)-permeable ion channel. Inactivating mutations in their genes cause autosomal dominant polycystic kidney disease (ADPKD), one of the most common genetic diseases. Here, we show that WNTs bind to the extracellular domain of PKD1 and induce whole-cell currents and Ca(2+) influx dependent on TRPP2. Pathogenic PKD1 or PKD2 mutations that abrogate complex formation, compromise cell surface expression of PKD1, or reduce TRPP2 channel activity suppress activation by WNTs. Pkd2(-/-) fibroblasts lack WNT-induced Ca(2+) currents and are unable to polarize during directed cell migration. In Xenopus embryos, pkd1, Dishevelled 2 (dvl2) and wnt9a act within the same pathway to preserve normal tubulogenesis. These data define PKD1 as a WNT (co)receptor and implicate defective WNT/Ca(2+) signalling as one of the causes of ADPKD.


Asunto(s)
Calcio/metabolismo , Vía de Señalización Wnt , Animales , Membrana Celular/metabolismo , Proteínas Dishevelled/metabolismo , Fibroblastos/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Unión Proteica , Canales Catiónicos TRPP/metabolismo , Xenopus
16.
Cells ; 5(1)2016 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-26840332

RESUMEN

Cilia and flagella are evolutionarily conserved, membrane-bound, microtubule-based organelles on the surface of most eukaryotic cells. They play important roles in coordinating a variety of signaling pathways during growth, development, cell mobility, and tissue homeostasis. Defects in ciliary structure or function are associated with multiple human disorders called ciliopathies. These diseases affect diverse tissues, including, but not limited to the eyes, kidneys, brain, and lungs. Many processes must be coordinated simultaneously in order to initiate ciliogenesis. These include cell cycle, vesicular trafficking, and axonemal extension. Centrioles play a central role in both cell cycle progression and ciliogenesis, making the transition between basal bodies and mitotic spindle organizers integral to both processes. The maturation of centrioles involves a functional shift from cell division toward cilium nucleation which takes place concurrently with its migration and fusion to the plasma membrane. Several proteinaceous structures of the distal appendages in mother centrioles are required for this docking process. Ciliary assembly and maintenance requires a precise balance between two indispensable processes; so called assembly and disassembly. The interplay between them determines the length of the resulting cilia. These processes require a highly conserved transport system to provide the necessary substances at the tips of the cilia and to recycle ciliary turnover products to the base using a based microtubule intraflagellar transport (IFT) system. In this review; we discuss the stages of ciliogenesis as well as mechanisms controlling the lengths of assembled cilia.

17.
EMBO J ; 34(19): 2424-40, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26206584

RESUMEN

Primary cilia start forming within the G1 phase of the cell cycle and continue to grow as cells exit the cell cycle (G0). They start resorbing when cells re-enter the cell cycle (S phase) and are practically invisible in mitosis. The mechanisms by which cilium biogenesis and disassembly are coupled to the cell cycle are complex and not well understood. We previously identified the centrosomal phosphoprotein NDE1 as a negative regulator of ciliary length and showed that its levels inversely correlate with ciliogenesis. Here, we identify the tumor suppressor FBW7 (also known as FBXW7, CDC4, AGO, or SEL-10) as the E3 ligase that mediates the destruction of NDE1 upon entry into G1. CDK5, a kinase active in G1/G0, primes NDE1 for FBW7-mediated recognition. Cells depleted of FBW7 or CDK5 show enhanced levels of NDE1 and a reduction in ciliary length, which is corrected in cells depleted of both FBW7 or CDK5 and NDE1. These data show that cell cycle-dependent mechanisms can control ciliary length through a CDK5-FBW7-NDE1 pathway.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Ciclo Celular/fisiología , Quinasa 5 Dependiente de la Ciclina/metabolismo , Proteínas F-Box/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteolisis , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación/fisiología , Animales , Células 3T3 BALB , Proteínas de Ciclo Celular/genética , Cilios/genética , Cilios/metabolismo , Quinasa 5 Dependiente de la Ciclina/genética , Proteínas F-Box/genética , Proteína 7 que Contiene Repeticiones F-Box-WD , Células HEK293 , Humanos , Ratones , Proteínas Asociadas a Microtúbulos/genética , Transducción de Señal/fisiología , Ubiquitina-Proteína Ligasas/genética
18.
Handb Exp Pharmacol ; 222: 15-51, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24756701

RESUMEN

The TRPC1 ion channel was the first mammalian TRP channel to be cloned. In humans, it is encoded by the TRPC1 gene located in chromosome 3. The protein is predicted to consist of six transmembrane segments with the N- and C-termini located in the cytoplasm. The extracellular loop connecting transmembrane segments 5 and 6 participates in the formation of the ionic pore region. Inside the cell, TRPC1 is present in the endoplasmic reticulum, plasma membrane, intracellular vesicles, and primary cilium, an antenna-like sensory organelle functioning as a signaling platform. In human and rodent tissues, it shows an almost ubiquitous expression. TRPC1 interacts with a diverse group of proteins including ion channel subunits, receptors, and cytosolic proteins to mediate its effect on Ca(2+) signaling. It primarily functions as a cation nonselective channel within pathways controlling Ca(2+) entry in response to cell surface receptor activation. Through these pathways, it affects basic cell functions, such as proliferation and survival, differentiation, secretion, and cell migration, as well as cell type-specific functions such as chemotropic turning of neuronal growth cones and myoblast fusion. The biological role of TRPC1 has been studied in genetically engineered mice where the Trpc1 gene has been experimentally ablated. Although these mice live to adulthood, they show defects in several organs and tissues, such as the cardiovascular, central nervous, skeletal and muscular, and immune systems. Genetic and functional studies have implicated TRPC1 in diabetic nephropathy, Parkinson's disease, Huntington's disease, Duchenne muscular dystrophy, cancer, seizures, and Darier-White skin disease.


Asunto(s)
Canales Catiónicos TRPC/metabolismo , Animales , Permeabilidad de la Membrana Celular , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Humanos , Activación del Canal Iónico , Potenciales de la Membrana , Ratones , Ratones Noqueados , Fenotipo , Conformación Proteica , Relación Estructura-Actividad , Canales Catiónicos TRPC/química , Canales Catiónicos TRPC/deficiencia , Canales Catiónicos TRPC/genética
19.
Proc Natl Acad Sci U S A ; 111(11): 4197-202, 2014 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-24591628

RESUMEN

Signaling through the store-operated Ca(2+) release-activated Ca(2+) (CRAC) channel regulates critical cellular functions, including gene expression, cell growth and differentiation, and Ca(2+) homeostasis. Loss-of-function mutations in the CRAC channel pore-forming protein ORAI1 or the Ca(2+) sensing protein stromal interaction molecule 1 (STIM1) result in severe immune dysfunction and nonprogressive myopathy. Here, we identify gain-of-function mutations in the cytoplasmic domain of STIM1 (p.R304W) associated with thrombocytopenia, bleeding diathesis, miosis, and tubular myopathy in patients with Stormorken syndrome, and in ORAI1 (p.P245L), associated with a Stormorken-like syndrome of congenital miosis and tubular aggregate myopathy but without hematological abnormalities. Heterologous expression of STIM1 p.R304W results in constitutive activation of the CRAC channel in vitro, and spontaneous bleeding accompanied by reduced numbers of thrombocytes in zebrafish embryos, recapitulating key aspects of Stormorken syndrome. p.P245L in ORAI1 does not make a constitutively active CRAC channel, but suppresses the slow Ca(2+)-dependent inactivation of the CRAC channel, thus also functioning as a gain-of-function mutation. These data expand our understanding of the phenotypic spectrum of dysregulated CRAC channel signaling, advance our knowledge of the molecular function of the CRAC channel, and suggest new therapies aiming at attenuating store-operated Ca(2+) entry in the treatment of patients with Stormorken syndrome and related pathologic conditions.


Asunto(s)
Trastornos de las Plaquetas Sanguíneas/genética , Canales de Calcio/genética , Señalización del Calcio/genética , Dislexia/genética , Ictiosis/genética , Proteínas de la Membrana/genética , Trastornos Migrañosos/genética , Miosis/genética , Miopatías Estructurales Congénitas/genética , Proteínas de Neoplasias/genética , Bazo/anomalías , Animales , Secuencia de Bases , Niño , Cartilla de ADN/genética , Eritrocitos Anormales , Femenino , Humanos , Datos de Secuencia Molecular , Fatiga Muscular/genética , Mutagénesis Sitio-Dirigida , Mutación/genética , Proteína ORAI1 , Técnicas de Placa-Clamp , Linaje , Análisis de Secuencia de ADN , Molécula de Interacción Estromal 1 , Pez Cebra
20.
J Biol Chem ; 288(31): 22219-32, 2013 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-23770672

RESUMEN

Ca(2+) signaling is essential for bone homeostasis and skeletal development. Here, we show that the transient receptor potential canonical 1 (TRPC1) channel and the inhibitor of MyoD family, I-mfa, function antagonistically in the regulation of osteoclastogenesis. I-mfa null mice have an osteopenic phenotype characterized by increased osteoclast numbers and surface, which are normalized in mice lacking both Trpc1 and I-mfa. In vitro differentiation of pre-osteoclasts derived from I-mfa-deficient mice leads to an increased number of mature osteoclasts and higher bone resorption per osteoclast. These parameters return to normal levels in osteoclasts derived from double mutant mice. Consistently, whole cell currents activated in response to the depletion of intracellular Ca(2+) stores are larger in pre-osteoclasts derived from I-mfa knock-out mice compared with currents in wild type mice and normalized in cells derived from double mutant mice, suggesting a cell-autonomous effect of I-mfa on TRPC1 in these cells. A new splice variant of TRPC1 (TRPC1ε) was identified in early pre-osteoclasts. Heterologous expression of TRPC1ε in HEK293 cells revealed that it is unique among all known TRPC1 isoforms in its ability to amplify the activity of the Ca(2+) release-activated Ca(2+) (CRAC) channel, mediating store-operated currents. TRPC1ε physically interacts with Orai1, the pore-forming subunit of the CRAC channel, and I-mfa is recruited to the TRPC1ε-Orai1 complex through TRPC1ε suppressing CRAC channel activity. We propose that the positive and negative modulation of the CRAC channel by TRPC1ε and I-mfa, respectively, fine-tunes the dynamic range of the CRAC channel regulating osteoclastogenesis.


Asunto(s)
Osteoclastos/citología , Canales Catiónicos TRPC/fisiología , Animales , Secuencia de Bases , División Celular , Línea Celular , Codón , Cartilla de ADN , Humanos , Ratones , Ratones Noqueados , Biosíntesis de Proteínas , ARN Mensajero/genética , Canales Catiónicos TRPC/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...