ciliary localization
Recently Published Documents


TOTAL DOCUMENTS

42
(FIVE YEARS 16)

H-INDEX

14
(FIVE YEARS 2)

Author(s):  
Yang Yue ◽  
Martin F. Engelke ◽  
T. Lynne Blasius ◽  
Kristen J. Verhey

The kinesin-4 motor KIF7 is a conserved regulator of the Hedgehog signaling pathway. In vertebrates, Hedgehog signaling requires the primary cilium, and KIF7 and Gli transcription factors accumulate at the cilium tip in response to Hedgehog activation. Unlike conventional kinesins, KIF7 is an immotile kinesin and its mechanism of ciliary accumulation is unknown. We generated KIF7 variants with altered microtubule binding or motility. We demonstrate that microtubule binding of KIF7 is not required for the increase in KIF7 or Gli localization at the cilium tip in response to Hedgehog signaling. In addition, we show that the immotile behavior of KIF7 is required to prevent ciliary localization of Gli transcription factors in the absence of Hedgehog signaling. Using an engineered kinesin-2 motor that enables acute inhibition of intraflagellar transport (IFT), we demonstrate that kinesin-2 KIF3A/KIF3B/KAP mediates the translocation of KIF7 to the cilium tip in response to Hedgehog pathway activation. Together, these results suggest that KIF7’s role at the tip of the cilium is unrelated to its ability to bind to microtubules.


Biology Open ◽  
2021 ◽  
Vol 10 (9) ◽  
Author(s):  
Sayaka Fujisawa ◽  
Hantian Qiu ◽  
Shohei Nozaki ◽  
Shuhei Chiba ◽  
Yohei Katoh ◽  
...  

ABSTRACT INPP5E, a phosphoinositide 5-phosphatase, localizes on the ciliary membrane via its C-terminal prenyl moiety, and maintains the distinct ciliary phosphoinositide composition. The ARL3 GTPase contributes to the ciliary membrane localization of INPP5E by stimulating the release of PDE6D bound to prenylated INPP5E. Another GTPase, ARL13B, which is localized on the ciliary membrane, contributes to the ciliary membrane retention of INPP5E by directly binding to its ciliary targeting sequence. However, as ARL13B was shown to act as a guanine nucleotide exchange factor (GEF) for ARL3, it is also possible that ARL13B indirectly mediates the ciliary INPP5E localization via activating ARL3. We here show that INPP5E is delocalized from cilia in both ARL3-knockout (KO) and ARL13B-KO cells. However, some of the abnormal phenotypes were different between these KO cells, while others were found to be common, indicating the parallel roles of ARL3 and ARL13B, at least concerning some cellular functions. For several variants of ARL13B, their ability to interact with INPP5E, rather than their ability as an ARL3-GEF, was associated with whether they could rescue the ciliary localization of INPP5E in ARL13B-KO cells. These observations together indicate that ARL13B determines the ciliary localization of INPP5E, mainly by its direct binding to INPP5E.


Hypertension ◽  
2021 ◽  
Vol 78 (Suppl_1) ◽  
Author(s):  
Deng-Fu Guo ◽  
Donald A Morgan ◽  
Paul Williams ◽  
Yuying Zhao ◽  
Mohamed Shaban ◽  
...  

Bardet-Biedl syndrome (BBS) proteins have emerged as critical regulators of various physiological functions including energy and glucose homeostasis, autonomic function and blood pressure. BBS3 protein is a cilia related protein that mediates the ciliary localization of the BBSome, a protein complex composed of eight BBS protein. Consistent with this, we found that BBS3 gene knockout in human RPE1 cells disrupt the ciliary localization of BBS2 and BBS9 proteins, indicating a loss of BBSome-mediated cargo trafficking to cilia. We previously demonstrated that disruption of the BBSome through Bbs1 gene deletion in the anorexigenic pro-opiomelanocortin (POMC) neurons altered energy homeostasis and sympathetic nerve traffic. However, the significance of BBS3-mediated ciliary localization of the BBSome in POMC neurons is not clear. To address this, we investigated the consequence of Bbs3 gene deletion in POMC neurons . We generated mice lacking the Bbs3 gene in POMC neurons by crossing Bbs3 fl/fl mice with mice expressing inducible Cre in POMC neurons (POMC ERCre ). To visualize Cre recombinase we further crossed POMC ERCre /Bbs3 fl/fl mice with tdTomato reporter mice. To induce Cre expression tamoxifen (75 mg/kg for 5 days) was injected at 6 weeks of age. Interestingly, both male and female POMC ERCre /Bbs3 fl/fl mice did not develop obesity as indicated by the lack of difference in body weight (male 28.1 + 0.5 vs 28.3 + 1.2g, female 21.9 + 0.7 vs 21.4 + 0.5g), fat mass (male 2.9 + .0.4 vs 3.4 + 0.5g, female 2.7 + .3 vs 2.3 + 0.2g) and weight of white and brown fat pads. Insulin and glucose tolerance tests revealed that POMC ERCre /Bbs3 fl/fl mice have glucose intolerance, whereas insulin sensitivity was normal. Blood pressure and renal SNA were comparable between POMC ERCre /Bbs3 fl/fl and control mice measured in the conscious state. In contrast, measurement of renal SNA response to changes in arterial pressure evoked by infusion of sodium nitroprusside and phenylephrine showed reduced baroreflex sensitivity in POMC ERCre /Bbs3 fl/fl mice. These findings demonstrate that POMC neuron BBS3, that mediate cargo transport to cilia, is not necessary for the regulation of energy homeostasis, but is involved in the regulation of glucose homeostasis and baroreflex sensitivity.


eLife ◽  
2021 ◽  
Vol 10 ◽  
Author(s):  
Sun-Hee Hwang ◽  
Bandarigoda N Somatilaka ◽  
Kevin White ◽  
Saikat Mukhopadhyay

The role of compartmentalized signaling in primary cilia during tissue morphogenesis is not well understood. The cilia-localized G-protein-coupled receptor—Gpr161 represses hedgehog pathway via cAMP signaling. We engineered a knock-in at Gpr161 locus in mice to generate a variant (Gpr161mut1), which was ciliary localization defective but cAMP signaling competent. Tissue phenotypes from hedgehog signaling depend on downstream bifunctional Gli transcriptional factors functioning as activators/repressors. Compared to knockout (ko), Gpr161mut1/ko had delayed embryonic lethality, moderately increased hedgehog targets and partially down-regulated Gli3-repressor. Unlike ko, the Gpr161mut1/ko neural tube did not show Gli2-activator-dependent expansion of ventral-most progenitors. Instead, the intermediate neural tube showed progenitor expansion that depends on loss of Gli3-repressor. Increased extraciliary receptor (Gpr161mut1/mut1) prevented ventralization. Morphogenesis in limb buds and midface requires Gli-repressor; these tissues in Gpr161mut1/mut1 manifested hedgehog hyperactivation phenotypes—polydactyly and midfacial widening. Thus, ciliary and extraciliary Gpr161 pools likely establish tissue-specific Gli-repressor thresholds in determining morpho-phenotypic outcomes.


Cells ◽  
2021 ◽  
Vol 10 (8) ◽  
pp. 1885
Author(s):  
Dinu Antony ◽  
Han G. Brunner ◽  
Miriam Schmidts

Although ubiquitously present, the relevance of cilia for vertebrate development and health has long been underrated. However, the aberration or dysfunction of ciliary structures or components results in a large heterogeneous group of disorders in mammals, termed ciliopathies. The majority of human ciliopathy cases are caused by malfunction of the ciliary dynein motor activity, powering retrograde intraflagellar transport (enabled by the cytoplasmic dynein-2 complex) or axonemal movement (axonemal dynein complexes). Despite a partially shared evolutionary developmental path and shared ciliary localization, the cytoplasmic dynein-2 and axonemal dynein functions are markedly different: while cytoplasmic dynein-2 complex dysfunction results in an ultra-rare syndromal skeleto-renal phenotype with a high lethality, axonemal dynein dysfunction is associated with a motile cilia dysfunction disorder, primary ciliary dyskinesia (PCD) or Kartagener syndrome, causing recurrent airway infection, degenerative lung disease, laterality defects, and infertility. In this review, we provide an overview of ciliary dynein complex compositions, their functions, clinical disease hallmarks of ciliary dynein disorders, presumed underlying pathomechanisms, and novel developments in the field.


2021 ◽  
Vol 5 (Supplement_1) ◽  
pp. A548-A549
Author(s):  
Kathryn M Brewer ◽  
Ruchi Bansal ◽  
Staci E Engle ◽  
Patrick J Antonellis ◽  
Theodore R Cummins ◽  
...  

Abstract Puberty and reproduction are initiated and controlled through the hypothalamic-pituitary-gonadal (HPG) axis. A critical surge of luteinizing hormone (LH) and follicle stimulating hormone (FSH) are released from the anterior pituitary upon release of gonadotrophins from gonadotrophin releasing hormone (GnRH) neurons. Thus, GnRH neurons are key regulators of the HPG axis. GnRH neurons become active when kisspeptin (Kiss1) neuropeptides are released from neurons in the arcuate nucleus. Kiss1 binds to the Kiss1 receptor (Kiss1R), a G-protein coupled receptor (GPCR) which localizes to the primary cilia of GnRH neurons. Loss-of-function mutations of Kiss1R cause hypogonadism in mouse and human models while gain-of-function mutations are associated with precocious puberty. Interestingly, the subset of GnRH neurons that express Kiss1R are observed to be polyciliated, possessing more than one primary cilia, an uncommon property as most neurons only possess a single, primary cilium. The mechanism and conditions leading to GnRH neuron polyciliation are unknown. It is also unclear if multiple cilia impact Kiss1R or other GPCR signaling in these neurons. Here, we utilize cultured mouse primary hypothalamic neurons to begin addressing some of these questions. We have confirmed with qPCR that the ligands GnRH and Kiss1, as well as Kiss1R, are all expressed in these cultures. Surprisingly, when treated with Kiss1 and GnRH ligands we observed a small subset of polyciliated neurons compared to vehicle treated neurons. These observations mirror what is seen during sexual maturation in vivo and suggest that our model system may help elucidate fundamental questions about how ciliary localization of Kiss1r and other GPCRs participate in initiation of puberty and regulation of reproduction. Future studies will focus on the mechanisms of polyciliation and the conditions needed to induce the formation of new cilia in GnRH neurons. Investigating neuronal polyciliation could provide insights into new signaling paradigm in hypogonadism and HPG signaling.


2021 ◽  
Vol 22 (9) ◽  
pp. 4313
Author(s):  
Clare L. Thompson ◽  
Megan McFie ◽  
J. Paul Chapple ◽  
Philip Beales ◽  
Martin M. Knight

Primary cilia and associated intraflagellar transport are essential for skeletal development, joint homeostasis, and the response to mechanical stimuli, although the mechanisms remain unclear. Polycystin-2 (PC2) is a member of the transient receptor potential polycystic (TRPP) family of cation channels, and together with Polycystin-1 (PC1), it has been implicated in cilia-mediated mechanotransduction in epithelial cells. The current study investigates the effect of mechanical stimulation on the localization of ciliary polycystins in chondrocytes and tests the hypothesis that they are required in chondrocyte mechanosignaling. Isolated chondrocytes were subjected to mechanical stimulation in the form of uniaxial cyclic tensile strain (CTS) in order to examine the effects on PC2 ciliary localization and matrix gene expression. In the absence of strain, PC2 localizes to the chondrocyte ciliary membrane and neither PC1 nor PC2 are required for ciliogenesis. Cartilage matrix gene expression (Acan, Col2a) is increased in response to 10% CTS. This response is inhibited by siRNA-mediated loss of PC1 or PC2 expression. PC2 ciliary localization requires PC1 and is increased in response to CTS. Increased PC2 cilia trafficking is dependent on the activation of transient receptor potential cation channel subfamily V member 4 (TRPV4) activation. Together, these findings demonstrate for the first time that polycystins are required for chondrocyte mechanotransduction and highlight the mechanosensitive cilia trafficking of PC2 as an important component of cilia-mediated mechanotransduction.


2021 ◽  
pp. jcs.258364
Author(s):  
Kirill Ukhanov ◽  
Cedric Uytingco ◽  
Warren Green ◽  
Lian Zhang ◽  
Stephane Schurmans ◽  
...  

The lipid composition of the primary cilia membrane is emerging as a critical regulator of cilia formation, maintenance, and function. Here, we show that conditional deletion of the phosphoinositide 5’-phosphatase gene, Inpp5e, causative of Joubert syndrome in terminally developed mouse olfactory sensory neurons (OSNs) led to a dramatic remodeling of ciliary phospholipids that was accompanied by marked elongation of cilia. PI(4,5)P2 normally restricted to the proximal segment redistributed to the entire length of cilia in Inpp5e knockout mice with a reduction in PI(3,4)P2 and elevation of PI(3,4,5)P3 in the dendritic knob. The redistribution of phosphoinositides impaired odor adaptation, resulting in less efficient recovery and altered inactivation kinetics of the odor-evoked electrical response and the odor-induced elevation of cytoplasmic Ca2+. Gene replacement by adenoviral expression of Inpp5e restored the ciliary localization of PI(4,5)P2 and odor response kinetics in OSNs. Our findings support the role of phosphoinositides as a modulator of the odor response and in ciliary biology of native multi-ciliated OSNs.


Author(s):  
Pablo Barbeito ◽  
Francesc R. Garcia-Gonzalo

Primary cilia are hair-like projections of the cell membrane supported by an inner microtubule scaffold, the axoneme, which polymerizes out of a membrane-docked centriole at the ciliary base. By working as specialized signaling compartments, primary cilia provide an optimal environment for many G protein-coupled receptors (GPCRs) and their effectors to efficiently transmit their signals to the rest of the cell. For this to occur, however, all necessary receptors and signal transducers must first accumulate at the ciliary membrane. Serotonin receptor 6 (HTR6) and Somatostatin receptor 3 (SSTR3) are two GPCRs whose signaling in brain neuronal cilia affects cognition and is implicated in psychiatric, neurodegenerative, and oncologic diseases. Over a decade ago, the third intracellular loops (IC3s) of HTR6 and SSTR3 were shown to contain ciliary localization sequences (CLSs) that, when grafted onto non-ciliary GPCRs, could drive their ciliary accumulation. Nevertheless, these CLSs were dispensable for ciliary targeting of HTR6 and SSTR3, suggesting the presence of additional CLSs, which we have recently identified in their C-terminal tails. Herein, we review the discovery and mapping of these CLSs, as well as the state of the art regarding how these CLSs may orchestrate ciliary accumulation of these GPCRs by controlling when and where they interact with the ciliary entry and exit machinery via adaptors such as TULP3, RABL2 and the BBSome.


Sign in / Sign up

Export Citation Format

Share Document