Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Cell Biol ; 36(1): 223-35, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26503786

ABSTRACT

Primary cilia are essential sensory and signaling organelles present on nearly every mammalian cell type. Defects in primary cilia underlie a class of human diseases collectively termed ciliopathies. Primary cilia are restricted subcellular compartments, and specialized mechanisms coordinate the localization of proteins to cilia. Moreover, trafficking of proteins into and out of cilia is required for proper ciliary function, and this process is disrupted in ciliopathies. The somatostatin receptor subtype 3 (Sstr3) is selectively targeted to primary cilia on neurons in the mammalian brain and is implicated in learning and memory. Here, we show that Sstr3 localization to cilia is dynamic and decreases in response to somatostatin treatment. We further show that somatostatin treatment stimulates ß-arrestin recruitment into Sstr3-positive cilia and this recruitment can be blocked by mutations in Sstr3 that impact agonist binding or phosphorylation. Importantly, somatostatin treatment fails to decrease Sstr3 ciliary localization in neurons lacking ß-arrestin 2. Together, our results implicate ß-arrestin in the modulation of Sstr3 ciliary localization and further suggest a role for ß-arrestin in the mediation of Sstr3 ciliary signaling.


Subject(s)
Arrestins/metabolism , Cilia/metabolism , Memory/physiology , Neurons/metabolism , Receptors, Somatostatin/metabolism , Animals , Brain/metabolism , Cells, Cultured , Learning/physiology , Mice , Signal Transduction/physiology , beta-Arrestin 2 , beta-Arrestins
2.
Oncotarget ; 6(31): 31792-804, 2015 Oct 13.
Article in English | MEDLINE | ID: mdl-26397139

ABSTRACT

Targeted radioiodine therapy for thyroid cancer is based on selective stimulation of Na+/I- Symporter (NIS)-mediated radioactive iodide uptake (RAIU) in thyroid cells by thyrotropin. Patients with advanced thyroid cancer do not benefit from radioiodine therapy due to reduced or absent NIS expression. To identify inhibitors that can be readily translated into clinical care, we examined oncological pipeline inhibitors targeting Akt, MEK, PI3K, Hsp90 or BRAF in their ability to increase RAIU in thyroid cells expressing BRAFV600E or RET/PTC3 oncogene. Our data showed that (1) PI3K inhibitor GDC-0941 outperformed other inhibitors in RAIU increase mainly by decreasing iodide efflux rate to a great extent; (2) RAIU increase by all inhibitors was extensively reduced by TGF-ß, a cytokine secreted in the invasive fronts of thyroid cancers; (3) RAIU reduction by TGF-ß was mainly mediated by NIS reduction and could be reversed by Apigenin, a plant-derived flavonoid; and (4) In the presence of TGF-ß, GDC-0941 with Apigenin co-treatment had the highest RAIU level in both BRAFV600E expressing cells and RET/PTC3 expressing cells. Taken together, Apigenin may serve as a dietary supplement along with small molecule inhibitors to improve radioiodine therapeutic efficacy on invasive tumor margins thereby minimizing future metastatic events.


Subject(s)
Apigenin/pharmacology , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/metabolism , Iodine Radioisotopes/administration & dosage , Small Molecule Libraries/pharmacology , Thyroid Gland/drug effects , Thyroid Gland/metabolism , Animals , Blotting, Western , Cells, Cultured , Immunoenzyme Techniques , Iodine Radioisotopes/pharmacokinetics , Proto-Oncogene Proteins c-ret/genetics , Proto-Oncogene Proteins c-ret/metabolism , RNA, Messenger/genetics , Radionuclide Imaging , Rats , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Thyroid Gland/diagnostic imaging , Tissue Distribution , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
3.
Proc Natl Acad Sci U S A ; 111(28): 10335-40, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24982149

ABSTRACT

Most central neurons in the mammalian brain possess an appendage called a primary cilium that projects from the soma into the extracellular space. The importance of these organelles is highlighted by the fact that primary cilia dysfunction is associated with numerous neuropathologies, including hyperphagia-induced obesity, hypogonadism, and learning and memory deficits. Neuronal cilia are enriched for signaling molecules, including certain G protein-coupled receptors (GPCRs), suggesting that neuronal cilia sense and respond to neuromodulators in the extracellular space. However, the impact of cilia on signaling to central neurons has never been demonstrated. Here, we show that the kisspeptin receptor (Kiss1r), a GPCR that is activated by kisspeptin to regulate the onset of puberty and adult reproductive function, is enriched in cilia projecting from mouse gonadotropin-releasing hormone (GnRH) neurons. Interestingly, GnRH neurons in adult animals are multiciliated and the percentage of GnRH neurons possessing multiple Kiss1r-positive cilia increases during postnatal development in a progression that correlates with sexual maturation. Remarkably, disruption of cilia selectively on GnRH neurons leads to a significant reduction in kisspeptin-mediated GnRH neuronal activity. To our knowledge, this result is the first demonstration of cilia disruption affecting central neuronal activity and highlights the importance of cilia for proper GPCR signaling.


Subject(s)
Gonadotropin-Releasing Hormone/metabolism , Kisspeptins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Receptors, G-Protein-Coupled/metabolism , Reproduction/physiology , Signal Transduction/physiology , Animals , Cell Line , Cilia/genetics , Cilia/metabolism , Female , Gonadotropin-Releasing Hormone/genetics , Kisspeptins/genetics , Male , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Neurons/cytology , Receptors, G-Protein-Coupled/genetics , Receptors, Kisspeptin-1 , Sexual Maturation/physiology
4.
Cell Mol Life Sci ; 71(11): 2165-78, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24104765

ABSTRACT

Primary cilia with a diameter of ~200 nm have been implicated in development and disease. Calcium signaling within a primary cilium has never been directly visualized and has therefore remained a speculation. Fluid-shear stress and dopamine receptor type-5 (DR5) agonist are among the few stimuli that require cilia for intracellular calcium signal transduction. However, it is not known if these stimuli initiate calcium signaling within the cilium or if the calcium signal originates in the cytoplasm. Using an integrated single-cell imaging technique, we demonstrate for the first time that calcium signaling triggered by fluid-shear stress initiates in the primary cilium and can be distinguished from the subsequent cytosolic calcium response through the ryanodine receptor. Importantly, this flow-induced calcium signaling depends on the ciliary polycystin-2 calcium channel. While DR5-specific agonist induces calcium signaling mainly in the cilioplasm via ciliary CaV1.2, thrombin specifically induces cytosolic calcium signaling through the IP3 receptor. Furthermore, a non-specific calcium ionophore triggers both ciliary and cytosolic calcium responses. We suggest that cilia not only act as sensory organelles but also function as calcium signaling compartments. Cilium-dependent signaling can spread to the cytoplasm or be contained within the cilioplasm. Our study thus provides the first model to understand signaling within the cilioplasm of a living cell.


Subject(s)
Calcium Signaling , Cilia/metabolism , Epithelial Cells/metabolism , Mechanotransduction, Cellular , TRPP Cation Channels/metabolism , Animals , Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/metabolism , Calcium Ionophores/pharmacology , Cilia/drug effects , Cytoplasm/drug effects , Cytoplasm/metabolism , Epithelial Cells/cytology , Epithelial Cells/drug effects , Gene Expression Regulation , Inositol 1,4,5-Trisphosphate Receptors/genetics , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Kidney/cytology , Kidney/drug effects , Kidney/metabolism , Molecular Imaging , Primary Cell Culture , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Rheology , Ryanodine Receptor Calcium Release Channel/genetics , Ryanodine Receptor Calcium Release Channel/metabolism , Single-Cell Analysis , Stress, Mechanical , Swine , TRPP Cation Channels/genetics , Thrombin/pharmacology
6.
PLoS One ; 7(9): e46304, 2012.
Article in English | MEDLINE | ID: mdl-23029470

ABSTRACT

Nearly every cell type in the mammalian body projects from its cell surface a primary cilium that provides important sensory and signaling functions. Defects in the formation or function of primary cilia have been implicated in the pathogenesis of many human developmental disorders and diseases, collectively termed ciliopathies. Most neurons in the brain possess cilia that are enriched for signaling proteins such as G protein-coupled receptors and adenylyl cyclase type 3, suggesting neuronal cilia sense neuromodulators in the brain and contribute to non-synaptic signaling. Indeed, disruption of neuronal cilia or loss of neuronal ciliary signaling proteins is associated with obesity and learning and memory deficits. As the functions of primary cilia are defined by the signaling proteins that localize to the ciliary compartment, identifying the complement of signaling proteins in cilia can provide important insights into their physiological roles. Here we report for the first time that different GPCRs can colocalize within the same cilium. Specifically, we found the ciliary GPCRs, melanin-concentrating hormone receptor 1 (Mchr1) and somatostatin receptor 3 (Sstr3) colocalizing within cilia in multiple mouse brain regions. In addition, we have evidence suggesting Mchr1 and Sstr3 form heteromers. As GPCR heteromerization can affect ligand binding properties as well as downstream signaling, our findings add an additional layer of complexity to neuronal ciliary signaling.


Subject(s)
Brain/physiology , Cilia/metabolism , Neurons/metabolism , Receptors, Somatostatin/metabolism , Animals , Brain/cytology , Cells, Cultured , Cilia/genetics , Gene Expression , HEK293 Cells , Humans , Mice , Neurons/cytology , Protein Multimerization , Receptors, Somatostatin/genetics , Signal Transduction/physiology
7.
Cell Mol Life Sci ; 68(17): 2951-60, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21152952

ABSTRACT

Primary cilia are nearly ubiquitous cellular appendages that provide important sensory and signaling functions. Ciliary dysfunction underlies numerous human diseases, collectively termed ciliopathies. Primary cilia have distinct functions on different cell types and these functions are defined by the signaling proteins that localize to the ciliary membrane. Neurons throughout the mammalian brain possess primary cilia upon which certain G protein-coupled receptors localize. Yet, the precise signaling proteins present on the vast majority of neuronal cilia are unknown. Here, we report that dopamine receptor 1 (D1) localizes to cilia on mouse central neurons, thereby implicating neuronal cilia in dopamine signaling. Interestingly, ciliary localization of D1 is dynamic, and the receptor rapidly translocates to and from cilia in response to environmental cues. Notably, the translocation of D1 from cilia requires proteins mutated in the ciliopathy Bardet-Biedl syndrome (BBS), and we find that one of the BBS proteins, Bbs5, specifically interacts with D1.


Subject(s)
Carrier Proteins/metabolism , Cilia/metabolism , Receptors, Dopamine D1/metabolism , Animals , Bardet-Biedl Syndrome/metabolism , Cells, Cultured , Cyclic AMP/metabolism , Cytoskeletal Proteins , Humans , Mice , Mice, Knockout , Neurons/cytology , Phosphate-Binding Proteins , Proteins/metabolism , Receptors, Dopamine D1/analysis , Receptors, G-Protein-Coupled/metabolism , Signal Transduction
8.
Cell Mol Life Sci ; 67(19): 3287-97, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20544253

ABSTRACT

Primary cilia are a class of cilia that are typically solitary, immotile appendages present on nearly every mammalian cell type. Primary cilia are believed to perform specialized sensory and signaling functions that are important for normal development and cellular homeostasis. Indeed, primary cilia dysfunction is now linked to numerous human diseases and genetic disorders. Collectively, primary cilia disorders are termed as ciliopathies and present with a wide range of clinical features, including cystic kidney disease, retinal degeneration, obesity, polydactyly, anosmia, intellectual disability, and brain malformations. Although significant progress has been made in elucidating the functions of primary cilia on some cell types, the precise functions of most primary cilia remain unknown. This is particularly true for primary cilia on neurons throughout the mammalian brain. This review will introduce primary cilia and ciliary signaling pathways with a focus on neuronal cilia and their putative functions and roles in human diseases.


Subject(s)
Neurons/physiology , Signal Transduction , Cilia/metabolism , Cilia/physiology , Genetic Diseases, Inborn/metabolism , Homeostasis , Humans , Kidney Diseases, Cystic/metabolism , Neurons/cytology , Polycystic Kidney Diseases/metabolism , Polydactyly/metabolism , Retinal Degeneration/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...