Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Neurosci ; 2022 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-35882560

RESUMO

A rod-shaped appendage called a primary cilium projects from the soma of most central neurons in the mammalian brain. The importance of cilia within the nervous system is highlighted by the fact that human syndromes linked to primary cilia dysfunction, collectively termed ciliopathies, are associated with numerous neuropathologies, including hyperphagia-induced obesity, neuropsychiatric disorders, and learning and memory deficits. Neuronal cilia are enriched with signaling molecules, including specific G protein-coupled receptors (GPCRs) and their downstream effectors, suggesting they act as sensory organelles that respond to neuromodulators in the extracellular space. We previously showed that GPCR ciliary localization is disrupted in neurons from mouse models of the ciliopathy Bardet-Biedl syndrome (BBS). Based on this finding we hypothesized that mislocalization of ciliary GPCRs may impact receptor signaling and contribute to the BBS phenotypes. Here, we show that disrupting localization of the ciliary GPCR dopamine receptor 1 (D1) in male and female mice, either by loss of a BBS protein or loss of the cilium itself, specifically in D1-expressing neurons, results in obesity. Interestingly, the weight gain is associated with reduced locomotor activity, rather than increased food intake. Moreover, loss of a BBS protein or cilia on D1-expressing neurons leads to a reduction in D1-mediated signaling. Together, these results indicate that cilia impact D1 activity in the nervous system and underscore the importance of neuronal cilia for proper GPCR signaling.SIGNIFICANCE STATEMENT:Most mammalian neurons possess solitary appendages called primary cilia. These rod-shaped structures are enriched with signaling proteins, such as G protein-coupled receptors (GPCRs), suggesting they respond to neuromodulators. This study examines the consequences of disrupting ciliary localization of the GPCR dopamine receptor 1 (D1) in D1-expressing neurons. Remarkably, mice that have either abnormal accumulation of D1 in cilia or loss of D1 ciliary localization become obese. In both cases the obesity is associated with lower locomotor activity rather than overeating. As D1 activation increases locomotor activity, these results are consistent with a reduction in D1 signaling. Indeed, we found that D1-mediated signaling is reduced in brain slices from both mouse models. Thus, cilia impact D1 signaling in the brain.

2.
Front Cell Dev Biol ; 10: 1092161, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36699005

RESUMO

In the brain, primary cilia are found on most, if not all, central neurons. The importance of neuronal cilia is underscored by the fact that human diseases caused by primary cilia dysfunction, which are known as ciliopathies, are associated with neuropathologies, including neuropsychiatric disorders and learning and memory deficits. Neuronal cilia are enriched for certain G protein-coupled receptors and their downstream effectors, suggesting they sense and respond to neuromodulators in the extracellular milieu. GPCR ciliary localization is disrupted in neurons from mouse models of the ciliopathy Bardet-Biedl syndrome, with GPCRs failing to localize to cilia, indicating the Bardet-Biedl syndrome proteins are required for trafficking of G protein-coupled receptors into neuronal cilia. Yet, dopamine receptor 1 accumulates in cilia in the absence of Bardet-Biedl syndrome proteins, suggesting Bardet-Biedl syndrome proteins are required for normal ciliary import and export. To further explore the roles of the Bardet-Biedl syndrome proteins in neuronal cilia, we examined localization of ciliary signaling proteins in a new constitutive Bbs1 knockout mouse model. Interestingly, we find that two additional ciliary G protein-coupled receptors (Gpr161 and Gpr19) abnormally accumulate in cilia on Bardet-Biedl syndrome neurons. In addition, we find that the GPCR signaling protein ß-arrestin accumulates in a subset of cilia in the brain, suggesting the presence of additional unidentified ciliary G protein-coupled receptors. These results confirm the importance of the Bardet-Biedl syndrome proteins in establishing ciliary GPCR pathways and indicate that loss of Bbs1 leads to complex changes in the localization of signaling proteins in the brain.

3.
Life Sci Alliance ; 4(3)2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33372037

RESUMO

G protein-coupled receptors (GPCRs) are the most common pharmacological target in human clinical practice. To perform their functions, many GPCRs must accumulate inside primary cilia, microtubule-based plasma membrane protrusions working as cellular antennae. Nevertheless, the molecular mechanisms underlying GPCR ciliary targeting remain poorly understood. Serotonin receptor 6 (HTR6) and somatostatin receptor 3 (SSTR3) are two brain-enriched ciliary GPCRs involved in cognition and pathologies such as Alzheimer's disease and cancer. Although the third intracellular loops (IC3) of HTR6 and SSTR3 suffice to target non-ciliary GPCRs to cilia, these IC3s are dispensable for ciliary targeting of HTR6 and SSTR3 themselves, suggesting these GPCRs contain additional ciliary targeting sequences (CTSs). Herein, we discover and characterize novel CTSs in HTR6 and SSTR3 C-terminal tails (CT). These CT-CTSs (CTS2) act redundantly with IC3-CTSs (CTS1), each being sufficient for ciliary targeting. In HTR6, RKQ and LPG motifs are critical for CTS1 and CTS2 function, respectively, whereas in SSTR3 these roles are mostly fulfilled by AP[AS]CQ motifs in IC3 and juxtamembrane residues in CT. Furthermore, we shed light on how these CTSs promote ciliary targeting by modulating binding to ciliary trafficking adapters TULP3 and RABL2.


Assuntos
Membrana Celular/metabolismo , Cílios/metabolismo , Receptores de Serotonina/química , Receptores de Serotonina/metabolismo , Receptores de Somatostatina/química , Receptores de Somatostatina/metabolismo , Transdução de Sinais/genética , Sequência de Aminoácidos , Animais , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos , Domínios e Motivos de Interação entre Proteínas , Transporte Proteico/genética , Transfecção
4.
Microsc Microanal ; 26(1): 157-165, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31931893

RESUMO

The voltage-gated sodium channel [pore-forming subunit of the neuronal voltage-gated sodium channel (NaV1.6)] has recently been found in cardiac myocytes. Emerging studies indicate a role for NaV1.6 in ionic homeostasis as well as arrhythmogenesis. Little is known about the spatial organization of these channels in cardiac muscle, mainly due to the lack of high-fidelity antibodies. Therefore, we developed and rigorously validated a novel rabbit polyclonal NaV1.6 antibody and undertook super-resolution microscopy studies of NaV1.6 localization in cardiac muscle. We developed and validated a novel rabbit polyclonal antibody against a C-terminal epitope on the neuronal sodium channel 1.6 (NaV1.6). Raw sera showed high affinity in immuno-fluorescence studies, which was improved with affinity purification. The antibody was rigorously validated for specificity via multiple approaches. Lastly, we used this antibody in proximity ligation assay (PLA) and super-resolution STochastic Optical Reconstruction Microscopy (STORM) studies, which revealed enrichment of NaV1.6 in close proximity to ryanodine receptor (RyR2), a key calcium (Ca2+) cycling protein, in cardiac myocytes. In summary, our novel NaV1.6 antibody demonstrates high degrees of specificity and fidelity in multiple preparations. It enabled multimodal microscopic studies and revealed that over half of the NaV1.6 channels in cardiac myocytes are located within 100 nm of ryanodine receptor Ca2+ release channels.


Assuntos
Miocárdio/citologia , Canal de Sódio Disparado por Voltagem NAV1.6/análise , Canal de Liberação de Cálcio do Receptor de Rianodina/análise , Animais , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia de Fluorescência , Imagem Óptica
5.
Methods Mol Biol ; 1957: 271-289, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30919360

RESUMO

Primary cilia (PC) are microtubule-based organelles that behave like a cellular antenna controlling key signaling pathways during development and tissue homeostasis. The ciliary membrane is highly enriched for G protein-coupled receptors (GPCRs), and PC are a crucial signaling compartment for this large receptor family. Downstream effectors of GPCR signaling are also present in cilia, and evidence obtained by our labs and others demonstrated that ß-arrestin (ßarr) family members are differentially recruited to PC and have investigated the role of GPCR activation in this process. In this chapter, we provide methods based on fluorescence microscopy on fixed or live cells suitable for investigating targeting and recruitment of ßarrs at PC.


Assuntos
Corpos Basais/metabolismo , Centrossomo/metabolismo , Cílios/metabolismo , Microscopia de Fluorescência/métodos , beta-Arrestina 2/metabolismo , Animais , Corpos Basais/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Centrossomo/efeitos dos fármacos , Cílios/efeitos dos fármacos , DNA/metabolismo , Recuperação de Fluorescência Após Fotodegradação , Proteínas de Fluorescência Verde/metabolismo , Hipocampo/citologia , Humanos , Camundongos , Microtúbulos/efeitos dos fármacos , Microtúbulos/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Plasmídeos/metabolismo , Somatostatina/farmacologia
6.
Nat Rev Nephrol ; 15(4): 199-219, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30733609

RESUMO

Primary cilia project in a single copy from the surface of most vertebrate cell types; they detect and transmit extracellular cues to regulate diverse cellular processes during development and to maintain tissue homeostasis. The sensory capacity of primary cilia relies on the coordinated trafficking and temporal localization of specific receptors and associated signal transduction modules in the cilium. The canonical Hedgehog (HH) pathway, for example, is a bona fide ciliary signalling system that regulates cell fate and self-renewal in development and tissue homeostasis. Specific receptors and associated signal transduction proteins can also localize to primary cilia in a cell type-dependent manner; available evidence suggests that the ciliary constellation of these proteins can temporally change to allow the cell to adapt to specific developmental and homeostatic cues. Consistent with important roles for primary cilia in signalling, mutations that lead to their dysfunction underlie a pleiotropic group of diseases and syndromic disorders termed ciliopathies, which affect many different tissues and organs of the body. In this Review, we highlight central mechanisms by which primary cilia coordinate HH, G protein-coupled receptor, WNT, receptor tyrosine kinase and transforming growth factor-ß (TGFß)/bone morphogenetic protein (BMP) signalling and illustrate how defects in the balanced output of ciliary signalling events are coupled to developmental disorders and disease progression.


Assuntos
Cílios/metabolismo , Transtornos da Motilidade Ciliar/metabolismo , Proteínas Hedgehog/metabolismo , Organogênese , Diferenciação Celular , Movimento Celular , Cílios/patologia , Transtornos da Motilidade Ciliar/patologia , Homeostase , Humanos , Transdução de Sinais
7.
Genesis ; 56(8): e23217, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29806135

RESUMO

The neuropeptide, melanin concentrating hormone (MCH), and its G protein-coupled receptor, melanin concentrating hormone receptor 1 (Mchr1), are expressed centrally in adult rodents. MCH signaling has been implicated in diverse behaviors such as feeding, sleep, anxiety, as well as addiction and reward. While a model utilizing the Mchr1 promoter to drive constitutive expression of Cre recombinase (Mchr1-Cre) exists, there is a need for an inducible Mchr1-Cre to determine the roles for this signaling pathway in neural development and adult neuronal function. Here, we generated a BAC transgenic mouse where the Mchr1 promotor drives expression of tamoxifen inducible CreER recombinase. Many aspects of the Mchr1-Cre expression pattern are recapitulated by the Mchr1-CreER model, though there are also notable differences. Most strikingly, compared to the constitutive model, the new Mchr1-CreER model shows strong expression in adult animals in hypothalamic brain regions involved in feeding behavior but diminished expression in regions involved in reward, such as the nucleus accumbens. The inducible Mchr1-CreER allele will help reveal the potential for Mchr1 signaling to impact neural development and subsequent behavioral phenotypes, as well as contribute to the understanding of the MCH signaling pathway in terminally differentiated adult neurons and the diverse behaviors that it influences.


Assuntos
Hormônios Hipotalâmicos/fisiologia , Melaninas/fisiologia , Hormônios Hipofisários/fisiologia , Receptores de Somatostatina/fisiologia , Animais , Encéfalo/metabolismo , Encéfalo/fisiologia , Hormônios Hipotalâmicos/metabolismo , Hipotálamo/metabolismo , Integrases , Melaninas/metabolismo , Camundongos , Camundongos Transgênicos , Modelos Animais , Neurônios/metabolismo , Neuropeptídeos/metabolismo , Hormônios Hipofisários/metabolismo , Receptores de Somatostatina/metabolismo , Transdução de Sinais , Tamoxifeno
8.
Dev Cell ; 42(3): 286-300.e4, 2017 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-28787594

RESUMO

Appropriate growth and synaptic integration of GABAergic inhibitory interneurons are essential for functional neural circuits in the brain. Here, we demonstrate that disruption of primary cilia function following the selective loss of ciliary GTPase Arl13b in interneurons impairs interneuronal morphology and synaptic connectivity, leading to altered excitatory/inhibitory activity balance. The altered morphology and connectivity of cilia mutant interneurons and the functional deficits are rescued by either chemogenetic activation of ciliary G-protein-coupled receptor (GPCR) signaling or the selective induction of Sstr3, a ciliary GPCR, in Arl13b-deficient cilia. Our results thus define a specific requirement for primary cilia-mediated GPCR signaling in interneuronal connectivity and inhibitory circuit formation.


Assuntos
Interneurônios/metabolismo , Transdução de Sinais , Sinapses/metabolismo , Potenciais Sinápticos , Fatores de Ribosilação do ADP/genética , Fatores de Ribosilação do ADP/metabolismo , Animais , Células Cultivadas , Cílios/metabolismo , Interneurônios/citologia , Interneurônios/fisiologia , Camundongos , Neurogênese , Receptores de Somatostatina/genética , Receptores de Somatostatina/metabolismo , Sinapses/fisiologia
9.
Artigo em Inglês | MEDLINE | ID: mdl-28159877

RESUMO

G-protein-coupled receptors (GPCRs) are the largest and most versatile family of signaling receptors in humans. They respond to diverse external signals, such as photons, proteins, peptides, chemicals, hormones, lipids, and sugars, and mediate a myriad of functions in the human body. Signaling through GPCRs can be optimized by enriching receptors and downstream effectors in discrete cellular domains. Many GPCRs have been found to be selectively targeted to cilia on numerous mammalian cell types. Moreover, investigations into the pathophysiology of human ciliopathies have implicated GPCR ciliary signaling in a number of developmental and cellular pathways. Thus, cilia are now appreciated as an increasingly important nexus for GPCR signaling. Yet, we are just beginning to understand the precise signaling pathways mediated by most ciliary GPCRs and how they impact cellular function and mammalian physiology.


Assuntos
Cílios/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Humanos , Modelos Biológicos , Receptores Odorantes/metabolismo , Receptores Odorantes/fisiologia , Transdução de Sinais
10.
Mol Cell Biol ; 36(1): 223-35, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26503786

RESUMO

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.


Assuntos
Arrestinas/metabolismo , Cílios/metabolismo , Memória/fisiologia , Neurônios/metabolismo , Receptores de Somatostatina/metabolismo , Animais , Encéfalo/metabolismo , Células Cultivadas , Aprendizagem/fisiologia , Camundongos , Transdução de Sinais/fisiologia , beta-Arrestina 2 , beta-Arrestinas
11.
Proc Natl Acad Sci U S A ; 111(28): 10335-40, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24982149

RESUMO

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.


Assuntos
Hormônio Liberador de Gonadotropina/metabolismo , Kisspeptinas/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Reprodução/fisiologia , Transdução de Sinais/fisiologia , Animais , Linhagem Celular , Cílios/genética , Cílios/metabolismo , Feminino , Hormônio Liberador de Gonadotropina/genética , Kisspeptinas/genética , Masculino , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Neurônios/citologia , Receptores Acoplados a Proteínas G/genética , Receptores de Kisspeptina-1 , Maturidade Sexual/fisiologia
13.
Cell Mol Life Sci ; 71(11): 2165-78, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24104765

RESUMO

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.


Assuntos
Sinalização do Cálcio , Cílios/metabolismo , Células Epiteliais/metabolismo , Mecanotransdução Celular , Canais de Cátion TRPP/metabolismo , Animais , Canais de Cálcio Tipo L/genética , Canais de Cálcio Tipo L/metabolismo , Ionóforos de Cálcio/farmacologia , Cílios/efeitos dos fármacos , Citoplasma/efeitos dos fármacos , Citoplasma/metabolismo , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Regulação da Expressão Gênica , Receptores de Inositol 1,4,5-Trifosfato/genética , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Rim/citologia , Rim/efeitos dos fármacos , Rim/metabolismo , Imagem Molecular , Cultura Primária de Células , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/genética , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Reologia , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Análise de Célula Única , Estresse Mecânico , Suínos , Canais de Cátion TRPP/genética , Trombina/farmacologia
14.
J Neurosci ; 33(6): 2626-38, 2013 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-23392690

RESUMO

The formation of primary cilia is a highly choreographed process that can be disrupted in developing neurons by overexpressing neuromodulatory G-protein-coupled receptors GPCRs or by blocking intraflagellar transport. Here, we examined the effects of overexpressing the ciliary GPCRs, 5HT6 and SSTR3, on cilia structure and the differentiation of neocortical neurons. Neuronal overexpression of 5HT6 and SSTR3 was achieved by electroporating mouse embryo cortex in utero with vectors encoding these receptors. We found that overexpression of ciliary GPCRs in cortical neurons, especially 5HT6, induced the formation of long (>30 µm) and often forked cilia. These changes were associated with increased levels of intraflagellar transport proteins and accelerated ciliogenesis in neonatal neocortex, the induction of which required Kif3a, an anterograde motor critical for cilia protein trafficking and growth. GPCR overexpression also altered the complement of signaling molecules within the cilia. We found that SSTR3 and type III adenylyl cyclase (ACIII), proteins normally enriched in neuronal cilia, were rarely detected in 5HT6-elongated cilia. Intriguingly, the changes in cilia structure were accompanied by changes in neuronal morphology. Specifically, disruption of normal ciliogenesis in developing neocortical neurons, either by overexpressing cilia GPCRs or a dominant-negative form of Kif3a, significantly impaired dendrite outgrowth. Remarkably, coexpression of ACIII with 5HT6 restored ACIII to cilia, normalized cilia structure, and restored dendrite outgrowth, effects that were not observed in neurons coexpressing ACIII and dominant-negative form of Kif3a. Collectively, our data suggest the formation of neuronal dendrites in developing neocortex requires structurally normal cilia enriched with ACIII.


Assuntos
Adenilil Ciclases/fisiologia , Cílios/enzimologia , Dendritos/enzimologia , Neocórtex/enzimologia , Neurônios/enzimologia , Receptores de Serotonina/biossíntese , Animais , Células Cultivadas , Cílios/fisiologia , Feminino , Cinesinas/biossíntese , Masculino , Camundongos , Células NIH 3T3 , Neocórtex/embriologia , Neurogênese/fisiologia , Neurônios/citologia , Gravidez
15.
PLoS One ; 7(9): e46304, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23029470

RESUMO

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.


Assuntos
Encéfalo/fisiologia , Cílios/metabolismo , Neurônios/metabolismo , Receptores de Somatostatina/metabolismo , Animais , Encéfalo/citologia , Células Cultivadas , Cílios/genética , Expressão Gênica , Células HEK293 , Humanos , Camundongos , Neurônios/citologia , Multimerização Proteica , Receptores de Somatostatina/genética , Transdução de Sinais/fisiologia
16.
Cell Mol Life Sci ; 68(17): 2951-60, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21152952

RESUMO

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.


Assuntos
Proteínas de Transporte/metabolismo , Cílios/metabolismo , Receptores de Dopamina D1/metabolismo , Animais , Síndrome de Bardet-Biedl/metabolismo , Células Cultivadas , AMP Cíclico/metabolismo , Proteínas do Citoesqueleto , Humanos , Camundongos , Camundongos Knockout , Neurônios/citologia , Proteínas de Ligação a Fosfato , Proteínas/metabolismo , Receptores de Dopamina D1/análise , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais
17.
Cell Mol Life Sci ; 67(19): 3287-97, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20544253

RESUMO

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.


Assuntos
Neurônios/fisiologia , Transdução de Sinais , Cílios/metabolismo , Cílios/fisiologia , Doenças Genéticas Inatas/metabolismo , Homeostase , Humanos , Doenças Renais Císticas/metabolismo , Neurônios/citologia , Doenças Renais Policísticas/metabolismo , Polidactilia/metabolismo , Degeneração Retiniana/metabolismo
18.
Methods Cell Biol ; 91: 111-21, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-20409783

RESUMO

Primary cilia were first detected on neurons in the mammalian brain over 40 years ago using electron microscopy. However, this approach is very labor intensive and has inherent limitations that restrict its utility for studying neuronal cilia. While the study of cilia in other tissues was greatly facilitated by the identification of specific ciliary markers, historically there have been no markers for neuronal cilia. Fortunately, recent developments make the study of neuronal cilia more practical. First, specific proteins have been shown to selectively localize to neuronal cilia and can serve as markers by immunolabeling. Second, neurons have been shown to possess cilia in culture, which allows for the use of additional approaches, such as live-cell imaging of neuronal cilia. This chapter provides an overview of the current techniques for visualizing neuronal cilia in tissue as well as fixed and living cells. These approaches allow for the identification of additional neuronal ciliary proteins and provide a basis for future functional studies.


Assuntos
Biomarcadores/metabolismo , Cílios , Neurônios , Animais , Encéfalo/citologia , Encéfalo/metabolismo , Cílios/metabolismo , Cílios/ultraestrutura , Imuno-Histoquímica/métodos , Camundongos , Neurônios/citologia , Neurônios/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Coloração e Rotulagem/métodos
19.
Proc Natl Acad Sci U S A ; 105(11): 4242-6, 2008 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-18334641

RESUMO

Primary cilia are ubiquitous cellular appendages that provide important yet not well understood sensory and signaling functions. Ciliary dysfunction underlies numerous human genetic disorders. However, the precise defects in cilia function and the basis of disease pathophysiology remain unclear. Here, we report that the proteins disrupted in the human ciliary disorder Bardet-Biedl syndrome (BBS) are required for the localization of G protein-coupled receptors to primary cilia on central neurons. We demonstrate a lack of ciliary localization of somatostatin receptor type 3 (Sstr3) and melanin-concentrating hormone receptor 1 (Mchr1) in neurons from mice lacking the Bbs2 or Bbs4 gene. Because Mchr1 is involved in the regulation of feeding behavior and BBS is associated with hyperphagia-induced obesity, our results suggest that altered signaling caused by mislocalization of ciliary signaling proteins underlies the BBS phenotypes. Our results also provide a potential molecular mechanism to link cilia defects with obesity.


Assuntos
Síndrome de Bardet-Biedl/metabolismo , Cílios/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas/metabolismo , Receptores de Somatostatina/metabolismo , Animais , Síndrome de Bardet-Biedl/genética , Células Cultivadas , Camundongos , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/deficiência , Proteínas Associadas aos Microtúbulos/genética , Mutação/genética , Proteínas/genética
20.
Mol Biol Cell ; 19(4): 1540-7, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18256283

RESUMO

Primary cilia are sensory organelles present on most mammalian cells. The functions of cilia are defined by the signaling proteins localized to the ciliary membrane. Certain G protein-coupled receptors (GPCRs), including somatostatin receptor 3 (Sstr3) and serotonin receptor 6 (Htr6), localize to cilia. As Sstr3 and Htr6 are the only somatostatin and serotonin receptor subtypes that localize to cilia, we hypothesized they contain ciliary localization sequences. To test this hypothesis we expressed chimeric receptors containing fragments of Sstr3 and Htr6 in the nonciliary receptors Sstr5 and Htr7, respectively, in ciliated cells. We found the third intracellular loop of Sstr3 or Htr6 is sufficient for ciliary localization. Comparison of these loops revealed a loose consensus sequence. To determine whether this consensus sequence predicts ciliary localization of other GPCRs, we compared it with the third intracellular loop of all human GPCRs. We identified the consensus sequence in melanin-concentrating hormone receptor 1 (Mchr1) and confirmed Mchr1 localizes to primary cilia in vitro and in vivo. Thus, we have identified a putative GPCR ciliary localization sequence and used this sequence to identify a novel ciliary GPCR. As Mchr1 mediates feeding behavior and metabolism, our results implicate ciliary signaling in the regulation of body weight.


Assuntos
Cílios/metabolismo , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação/genética , Linhagem Celular , Sequência Consenso , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Túbulos Renais Coletores/citologia , Túbulos Renais Coletores/metabolismo , Camundongos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Receptores Acoplados a Proteínas G/genética , Receptores de Serotonina/química , Receptores de Serotonina/genética , Receptores de Serotonina/metabolismo , Receptores de Somatostatina/química , Receptores de Somatostatina/genética , Receptores de Somatostatina/metabolismo , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Homologia de Sequência de Aminoácidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...