1
|
Fakhari S, Campolina-Silva G, Asayesh F, Girardet L, Scott-Boyer MP, Droit A, Soulet D, Greener J, Belleannée C. Shear stress effects on epididymal epithelial cell via primary cilia mechanosensory signaling. J Cell Physiol 2024:e31475. [PMID: 39508588 DOI: 10.1002/jcp.31475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/24/2024] [Accepted: 10/17/2024] [Indexed: 11/15/2024]
Abstract
Shear stress, resulting from fluid flow, is a fundamental mechanical stimulus affecting various cellular functions. The epididymis, essential for sperm maturation, offers a compelling model to study the effects of shear stress on cellular behavior. This organ undergoes extensive proliferation and differentiation until puberty, achieving full functionality as spermatozoa commence their post-testicular maturation. Although the mechanical tension exerted by testicular fluid is hypothesized to drive epithelial proliferation and differentiation, the precise mechanisms remain unclear. Here we assessed whether the responsiveness of the epididymal cells to shear stress depends on functional primary cilia by combining microfluidic strategies on immortalized epididymal cells, calcium signaling assays, and high-throughput gene expression analysis. We identified 97 genes overexpressed in response to shear stress, including early growth response (Egr) 2/3, cellular communication network factor (Ccn) 1/2, and Fos proto-oncogene (Fos). While shear stress triggered a rapid increase of intracellular Ca2+, this response was abrogated following the impairment of primary ciliogenesis through pharmacological and siRNA approaches. Overall, our findings provide valuable insights into how mechanical forces influence the development of the male reproductive system, a requisite to sperm maturation.
Collapse
Affiliation(s)
- Sepideh Fakhari
- Department of Obstetrics, Gynecology, and Reproduction, Centre de recherche en Reproduction, Développement et Santé Intergénérationnelle, Faculty of Medicine, Québec City, Quebec, Canada
- Centre de recherche du centre hospitalier universitaire de Québec - Université Laval, Québec City, Quebec, Canada
- Department of Chemistry, Faculty of Science and Engineering, Québec City, Quebec, Canada
| | - Gabriel Campolina-Silva
- Department of Obstetrics, Gynecology, and Reproduction, Centre de recherche en Reproduction, Développement et Santé Intergénérationnelle, Faculty of Medicine, Québec City, Quebec, Canada
- Centre de recherche du centre hospitalier universitaire de Québec - Université Laval, Québec City, Quebec, Canada
| | - Farnaz Asayesh
- Department of Obstetrics, Gynecology, and Reproduction, Centre de recherche en Reproduction, Développement et Santé Intergénérationnelle, Faculty of Medicine, Québec City, Quebec, Canada
| | - Laura Girardet
- Department of Obstetrics, Gynecology, and Reproduction, Centre de recherche en Reproduction, Développement et Santé Intergénérationnelle, Faculty of Medicine, Québec City, Quebec, Canada
- Centre de recherche du centre hospitalier universitaire de Québec - Université Laval, Québec City, Quebec, Canada
| | - Marie-Pier Scott-Boyer
- Proteomics Platform, Québec Genomic Center, Université Laval, CHU de Québec Research Center (CHUL), Québec City, Quebec, Canada
| | - Arnaud Droit
- Proteomics Platform, Québec Genomic Center, Université Laval, CHU de Québec Research Center (CHUL), Québec City, Quebec, Canada
| | - Denis Soulet
- Centre de recherche du centre hospitalier universitaire de Québec - Université Laval, Québec City, Quebec, Canada
- Faculté de pharmacie, Université Laval, Québec City, Quebec, Canada
| | - Jesse Greener
- Centre de recherche du centre hospitalier universitaire de Québec - Université Laval, Québec City, Quebec, Canada
- Department of Chemistry, Faculty of Science and Engineering, Québec City, Quebec, Canada
| | - Clémence Belleannée
- Department of Obstetrics, Gynecology, and Reproduction, Centre de recherche en Reproduction, Développement et Santé Intergénérationnelle, Faculty of Medicine, Québec City, Quebec, Canada
- Centre de recherche du centre hospitalier universitaire de Québec - Université Laval, Québec City, Quebec, Canada
| |
Collapse
|
2
|
Limerick A, McCabe EA, Turner JS, Kuang KW, Brautigan DL, Hao Y, Chu CY, Fu SH, Ahmadi S, Xu W, Fu Z. An Epilepsy-Associated CILK1 Variant Compromises KATNIP Regulation and Impairs Primary Cilia and Hedgehog Signaling. Cells 2024; 13:1258. [PMID: 39120290 PMCID: PMC11311665 DOI: 10.3390/cells13151258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/17/2024] [Accepted: 07/23/2024] [Indexed: 08/10/2024] Open
Abstract
Mutations in human CILK1 (ciliogenesis associated kinase 1) are linked to ciliopathies and epilepsy. Homozygous point and nonsense mutations that extinguish kinase activity impair primary cilia function, whereas mutations outside the kinase domain are not well understood. Here, we produced a knock-in mouse equivalent to the human CILK1 A615T variant identified in juvenile myoclonic epilepsy (JME). This residue is in the intrinsically disordered C-terminal region of CILK1 separate from the kinase domain. Mouse embryo fibroblasts (MEFs) with either heterozygous or homozygous A612T mutant alleles exhibited a higher ciliation rate, shorter individual cilia, and upregulation of ciliary Hedgehog signaling. Thus, a single A612T mutant allele was sufficient to impair primary cilia and ciliary signaling in MEFs. Gene expression profiles of wild-type versus mutant MEFs revealed profound changes in cilia-related molecular functions and biological processes. The CILK1 A615T mutant protein was not increased to the same level as the wild-type protein when co-expressed with scaffold protein KATNIP (katanin-interacting protein). Our data show that KATNIP regulation of a JME-associated single-residue variant of CILK1 is compromised, and this impairs the maintenance of primary cilia and Hedgehog signaling.
Collapse
Affiliation(s)
- Ana Limerick
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA; (A.L.); (E.A.M.); (J.S.T.); (K.W.K.); (C.Y.C.); (S.H.F.); (S.A.)
| | - Ellie A. McCabe
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA; (A.L.); (E.A.M.); (J.S.T.); (K.W.K.); (C.Y.C.); (S.H.F.); (S.A.)
| | - Jacob S. Turner
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA; (A.L.); (E.A.M.); (J.S.T.); (K.W.K.); (C.Y.C.); (S.H.F.); (S.A.)
| | - Kevin W. Kuang
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA; (A.L.); (E.A.M.); (J.S.T.); (K.W.K.); (C.Y.C.); (S.H.F.); (S.A.)
| | - David L. Brautigan
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA; (D.L.B.); (W.X.)
| | - Yi Hao
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22908, USA;
| | - Cheuk Ying Chu
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA; (A.L.); (E.A.M.); (J.S.T.); (K.W.K.); (C.Y.C.); (S.H.F.); (S.A.)
| | - Sean H. Fu
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA; (A.L.); (E.A.M.); (J.S.T.); (K.W.K.); (C.Y.C.); (S.H.F.); (S.A.)
| | - Sean Ahmadi
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA; (A.L.); (E.A.M.); (J.S.T.); (K.W.K.); (C.Y.C.); (S.H.F.); (S.A.)
| | - Wenhao Xu
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA; (D.L.B.); (W.X.)
| | - Zheng Fu
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA; (A.L.); (E.A.M.); (J.S.T.); (K.W.K.); (C.Y.C.); (S.H.F.); (S.A.)
| |
Collapse
|
3
|
Riaz A, Mehmood K, Alhamzi G, Alharbi KAM. Electroosmotic flow of cobalt-ferrite nanoparticles in water and ethylene glycol through a ciliary annulus: A biomedical application. Electrophoresis 2024; 45:1198-1211. [PMID: 37592853 DOI: 10.1002/elps.202300137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 08/02/2023] [Accepted: 08/08/2023] [Indexed: 08/19/2023]
Abstract
Unique magnetic characteristics of cobalt-ferrite nanoparticles make them suitable for biological imaging and therapeutic applications. Understanding their activity in nanofluids via the ciliary annulus could lead to better contrast agents for magnetic resonance imaging and improved cancer therapy and other medical therapies. This article provides a comprehensive analysis of the theoretical conclusions regarding the transport of a nanofluid by electroosmosis across a ciliary annulus. The nanofluid consists of cobalt-ferrite nanoparticles (CoFe2O4), water (H2O), and ethylene glycol (C2H6O2). As part of the investigation into constructing a physical model, mathematical analysis is performed based on the conservation of mass, momentum, and energy. Dimension-free analysis and mathematical constraints are utilized to learn more about the system. By generating differential equations and including suitable boundary conditions, one can obtain exact solutions, which can then be visually inspected. Recent studies have demonstrated an inverse relationship between flow velocity and cilia length, zeta potential, and Helmholtz-Smoluchowski velocity. The streamlines show that the growth of the trapping boluses is affected by several factors, including the nanoparticles' volume fraction, the cilia's length, the amplitude ratio, the eccentricity, and the zeta potential. These results not only shed light on how nanofluids move, but they also have potential applications in microfluidics, heat transfer, and biomedical engineering.
Collapse
Affiliation(s)
- Arshad Riaz
- Department of Mathematics, Division of Science and Technology, University of Education, Lahore, Pakistan
| | - Kinza Mehmood
- Department of Mathematics, Division of Science and Technology, University of Education, Lahore, Pakistan
| | - Ghaliah Alhamzi
- Department of Mathematics and Statistics, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Kingdom of Saudi Arabia
| | - Khalid Abdulkhaliq M Alharbi
- Mechanical Engineering Department, College of Engineering, Umm Al-Qura University, Makkah, Kingdom of Saudi Arabia
| |
Collapse
|
4
|
Limerick A, McCabe EA, Turner JS, Kuang KW, Brautigan DL, Hao Y, Chu C, Fu SH, Ahmadi S, Xu W, Fu Z. An epilepsy-associated CILK1 variant compromises KATNIP regulation and impairs primary cilia and Hedgehog signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.14.594243. [PMID: 38798407 PMCID: PMC11118389 DOI: 10.1101/2024.05.14.594243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Mutations in human CILK1 (ciliogenesis associated kinase 1) are linked to ciliopathies and epilepsy. Homozygous point and nonsense mutations that extinguish kinase activity impair primary cilia function, whereas mutations outside the kinase domain are not well understood. Here, we produced a knock-in mouse equivalent of the human CILK1 A615T variant identified in juvenile myoclonic epilepsy (JME). This residue is in the C-terminal region of CILK1 separate from the kinase domain. Mouse embryo fibroblasts (MEF) with either heterozygous or homozygous A612T mutant alleles exhibited a higher ciliation rate, shorter individual cilia and up-regulation of ciliary Hedgehog signaling. Thus, a single A612T mutant allele was sufficient to impair primary cilia and ciliary signaling in MEFs. Gene expression profiles of wild type versus mutant MEFs revealed profound changes in cilia-related molecular functions and biological processes. CILK1 A615T mutant protein was not increased to the same level as the wild type protein when co-expressed with scaffold protein KATNIP (katanin-interacting protein). Our data show that KATNIP regulation of a JME-associated single residue variant of CILK1 is compromised and this impairs the maintenance of primary cilia and Hedgehog signaling.
Collapse
Affiliation(s)
- Ana Limerick
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Ellie A. McCabe
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Jacob S. Turner
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Kevin W. Kuang
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - David L. Brautigan
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Yi Hao
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22908, USA
| | - Cherry Chu
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Sean H. Fu
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Sean Ahmadi
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Wenhao Xu
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Zheng Fu
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
5
|
Parashar A, Jha D, Mehta V, Chauhan B, Ghosh P, Deb PK, Jaiswal M, Prajapati SK. Sonic hedgehog signalling pathway contributes in age-related disorders and Alzheimer's disease. Ageing Res Rev 2024; 96:102271. [PMID: 38492808 DOI: 10.1016/j.arr.2024.102271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 03/10/2024] [Accepted: 03/11/2024] [Indexed: 03/18/2024]
Abstract
Alzheimer's disease (AD) is caused by the aging process and manifested by cognitive deficits and progressive memory loss. During aging, several conditions, including hypertension, diabetes, and cholesterol, have been identified as potential causes of AD by affecting Sonic hedgehog (Shh) signalling. In addition to being essential for cell differentiation and proliferation, Shh signalling is involved in tissue repair and the prevention of neurodegeneration. Neurogenesis is dependent on Shh signalling; inhibition of this pathway results in neurodegeneration. Several protein-protein interactions that are involved in Shh signalling are implicated in the pathophysiology of AD like overexpression of the protein nexin-1 inhibits the Shh pathway in AD. A protein called Growth Arrest Specific-1 works with another protein called cysteine dioxygenase (CDO) to boost Shh signalling. CDO is involved in the development of the central nervous system (CNS). Shh signalling strengthened the blood brain barrier and therefore prevent the entry of amyloid beta and other toxins to the brain from periphery. Further, several traditional remedies used for AD and dementia, including Epigallocatechin gallate, yokukansan, Lycium barbarum polysaccharides, salvianolic acid, and baicalin, are known to stimulate the Shh pathway. In this review, we elaborated that the Shh signalling exerts a substantial influence on the pathogenesis of AD. In this article, we have tried to explore the various possible connections between the Shh signalling and various known pathologies of AD.
Collapse
Affiliation(s)
- Arun Parashar
- School of Pharmaceutical Sciences, Shoolini University of Biotechnology & Management Sciences, Solan 173 212, India.
| | - Dhruv Jha
- Birla Institute of Technology, India
| | - Vineet Mehta
- Department of Pharmacology, Government College of Pharmacy, Rohru, District Shimla, Himachal Pradesh 171207, India
| | - Bonney Chauhan
- School of Pharmaceutical Sciences, Shoolini University of Biotechnology & Management Sciences, Solan 173 212, India
| | - Pappu Ghosh
- School of Pharmaceutical Sciences, Shoolini University of Biotechnology & Management Sciences, Solan 173 212, India
| | - Prashanta Kumar Deb
- School of Pharmaceutical Sciences, Shoolini University of Biotechnology & Management Sciences, Solan 173 212, India
| | | | | |
Collapse
|
6
|
Everett T, Ten Eyck TW, Wu CH, Shelowitz AL, Stansbury SM, Firek A, Setlow B, McIntyre JC. Cilia loss on distinct neuron populations differentially alters cocaine-induced locomotion and reward. J Psychopharmacol 2024; 38:200-212. [PMID: 38151883 PMCID: PMC11078551 DOI: 10.1177/02698811231219058] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
BACKGROUND Neuronal primary cilia are being recognized for their role in mediating signaling associated with a variety of neurobehaviors, including responses to drugs of abuse. They function as signaling hubs, enriched with a diverse array of G-protein coupled receptors (GPCRs), including several associated with motivation and drug-related behaviors. However, our understanding of how cilia regulate neuronal function and behavior is still limited. AIMS The objective of the current study was to investigate the contributions of primary cilia on specific neuronal populations to behavioral responses to cocaine. METHODS To test the consequences of cilia loss on cocaine-induced locomotion and reward-related behavior, we selectively ablated cilia from dopaminergic or GAD2-GABAergic neurons in mice. RESULTS Cilia ablation on either population of neurons failed to significantly alter acute locomotor responses to cocaine at a range of doses. With repeated administration, mice lacking cilia on GAD2-GABAergic neurons showed no difference in locomotor sensitization to cocaine compared to wild-type (WT) littermates, whereas mice lacking cilia on dopaminergic neurons exhibited reduced locomotor sensitization to cocaine at 10 and 30 mg/kg. Mice lacking cilia on GAD2-GABAergic neurons showed no difference in cocaine conditioned place preference (CPP), whereas mice lacking cilia on dopaminergic neurons exhibited reduced CPP compared to WT littermates. CONCLUSIONS Combined with previous findings using amphetamine, our results show that behavioral effects of cilia ablation are cell- and drug type-specific, and that neuronal cilia contribute to modulation of both the locomotor-inducing and rewarding properties of cocaine.
Collapse
Affiliation(s)
- Thomas Everett
- Department of Neuroscience, University of Florida, Gainesville, FL 32610
| | - Tyler W. Ten Eyck
- Department of Neuroscience, University of Florida, Gainesville, FL 32610
| | - Chang-Hung Wu
- Department of Neuroscience, University of Florida, Gainesville, FL 32610
| | | | - Sofia M. Stansbury
- Department of Neuroscience, University of Florida, Gainesville, FL 32610
| | - Alexandra Firek
- Department of Neuroscience, University of Florida, Gainesville, FL 32610
| | - Barry Setlow
- Department of Psychiatry, University of Florida, Gainesville, FL 32610
- Center for Addiction Research and Education, University of Florida, Gainesville, FL 32610
| | - Jeremy C. McIntyre
- Department of Neuroscience, University of Florida, Gainesville, FL 32610
- Center for Addiction Research and Education, University of Florida, Gainesville, FL 32610
| |
Collapse
|
7
|
Winsky-Sommerer R, King HA, Iadevaia V, Möller-Levet C, Gerber AP. A post-transcriptional regulatory landscape of aging in the female mouse hippocampus. Front Aging Neurosci 2023; 15:1119873. [PMID: 37122377 PMCID: PMC10135431 DOI: 10.3389/fnagi.2023.1119873] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 02/27/2023] [Indexed: 05/02/2023] Open
Abstract
Aging is associated with substantial physiological changes and constitutes a major risk factor for neurological disorders including dementia. Alterations in gene expression upon aging have been extensively studied; however, an in-depth characterization of post-transcriptional regulatory events remains elusive. Here, we profiled the age-related changes of the transcriptome and translatome in the female mouse hippocampus by RNA sequencing of total RNA and polysome preparations at four ages (3-, 6-, 12-, 20-month-old); and we implemented a variety of bioinformatics approaches to unravel alterations in transcript abundance, alternative splicing, and polyadenylation site selection. We observed mostly well-coordinated transcriptome and translatome expression signatures across age including upregulation of transcripts related to immune system processes and neuroinflammation, though transcripts encoding ribonucleoproteins or associated with mitochondrial functions, calcium signaling and the cell-cycle displayed substantial discordant profiles, suggesting translational control associated with age-related deficits in hippocampal-dependent behavior. By contrast, alternative splicing was less preserved, increased with age and was associated with distinct functionally-related transcripts encoding proteins acting at synapses/dendrites, RNA-binding proteins; thereby predicting regulatory roles for RBM3 and CIRBP. Only minor changes in polyadenylation site selection were identified, indicating pivotal 3'-end selection in young adults compared to older groups. Overall, our study provides a comprehensive resource of age-associated post-transcriptional regulatory events in the mouse hippocampus, enabling further examination of the molecular features underlying age-associated neurological diseases.
Collapse
Affiliation(s)
| | | | | | | | - André P. Gerber
- Faculty of Health and Medical Sciences, School of Biosciences and Medicine, University of Surrey, Guildford, Surrey, United Kingdom
| |
Collapse
|
8
|
Kowal TJ, Dhande OS, Wang B, Wang Q, Ning K, Liu W, Berbari NF, Hu Y, Sun Y. Distribution of prototypical primary cilia markers in subtypes of retinal ganglion cells. J Comp Neurol 2022; 530:2176-2187. [PMID: 35434813 PMCID: PMC9219574 DOI: 10.1002/cne.25326] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 01/27/2022] [Accepted: 03/21/2022] [Indexed: 11/07/2022]
Abstract
Loss of retinal ganglion cells (RGCs) underlies several forms of retinal disease including glaucomatous optic neuropathy, a leading cause of irreversible blindness. Several rare genetic disorders associated with cilia dysfunction have retinal degeneration as a clinical hallmark. Much of the focus of ciliopathy associated blindness is on the connecting cilium of photoreceptors; however, RGCs also possess primary cilia. It is unclear what roles RGC cilia play, what proteins and signaling machinery localize to RGC cilia, or how RGC cilia are differentiated across the subtypes of RGCs. To better understand these questions, we assessed the presence or absence of a prototypical cilia marker Arl13b and a widely distributed neuronal cilia marker AC3 in different subtypes of mouse RGCs. Interestingly, not all RGC subtype cilia are the same and there are significant differences even among these standard cilia markers. Alpha-RGCs positive for osteopontin, calretinin, and SMI32 primarily possess AC3-positive cilia. Directionally selective RGCs that are CART positive or Trhr positive localize either Arl13b or AC3, respectively, in cilia. Intrinsically photosensitive RGCs differentially localize Arl13b and AC3 based on melanopsin expression. Taken together, we characterized the localization of gold standard cilia markers in different subtypes of RGCs and conclude that cilia within RGC subtypes may be differentially organized. Future studies aimed at understanding RGC cilia function will require a fundamental ability to observe the cilia across subtypes as their signaling protein composition is elucidated. A comprehensive understanding of RGC cilia may reveal opportunities to understanding how their dysfunction leads to retinal degeneration.
Collapse
Affiliation(s)
- Tia J. Kowal
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, 94304, USA
| | - Onkar S. Dhande
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, 94304, USA
| | - Biao Wang
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, 94304, USA
| | - Qing Wang
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, 94304, USA
| | - Ke Ning
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, 94304, USA
| | - Wendy Liu
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, 94304, USA
| | - Nicolas F. Berbari
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis IN 46202 USA
| | - Yang Hu
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, 94304, USA
| | - Yang Sun
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, 94304, USA
- Palo Alto Veterans Administration, Palo Alto, CA 94304
| |
Collapse
|
9
|
Jalalvand E, Alvelid J, Coceano G, Edwards S, Robertson B, Grillner S, Testa I. ExSTED microscopy reveals contrasting functions of dopamine and somatostatin CSF-c neurons along the lamprey central canal. eLife 2022; 11:73114. [PMID: 35103591 PMCID: PMC8809891 DOI: 10.7554/elife.73114] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 01/19/2022] [Indexed: 01/04/2023] Open
Abstract
Cerebrospinal fluid-contacting (CSF-c) neurons line the central canal of the spinal cord and a subtype of CSF-c neurons expressing somatostatin, forms a homeostatic pH regulating system. Despite their importance, their intricate spatial organization is poorly understood. The function of another subtype of CSF-c neurons expressing dopamine is also investigated. Imaging methods with a high spatial resolution (5-10 nm) are used to resolve the synaptic and ciliary compartments of each individual cell in the spinal cord of the lamprey to elucidate their signalling pathways and to dissect the cellular organization. Here, light-sheet and expansion microscopy resolved the persistent ventral and lateral organization of dopamine- and somatostatin-expressing CSF-c neuronal subtypes. The density of somatostatin-containing dense-core vesicles, resolved by stimulated emission depletion microscopy, was shown to be markedly reduced upon each exposure to either alkaline or acidic pH and being part of a homeostatic response inhibiting movements. Their cilia symmetry was unravelled by stimulated emission depletion microscopy in expanded tissues as sensory with 9 + 0 microtubule duplets. The dopaminergic CSF-c neurons on the other hand have a motile cilium with the characteristic 9 + 2 duplets and are insensitive to pH changes. This novel experimental workflow elucidates the functional role of CSF-c neuron subtypes in situ paving the way for further spatial and functional cell-type classification.
Collapse
Affiliation(s)
- Elham Jalalvand
- Department of Applied Physics and Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Jonatan Alvelid
- Department of Applied Physics and Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Giovanna Coceano
- Department of Applied Physics and Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Steven Edwards
- Department of Applied Physics and Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Brita Robertson
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Sten Grillner
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Ilaria Testa
- Department of Applied Physics and Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| |
Collapse
|
10
|
Duittoz A, Cayla X, Fleurot R, Lehnert J, Khadra A. Gonadotrophin-releasing hormone and kisspeptin: It takes two to tango. J Neuroendocrinol 2021; 33:e13037. [PMID: 34533248 DOI: 10.1111/jne.13037] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 08/25/2021] [Accepted: 08/26/2021] [Indexed: 01/06/2023]
Abstract
Kisspeptin (Kp), a family of peptides comprising products of the Kiss1 gene, was discovered 20 years ago; it is recognised as the major factor controlling the activity of the gonadotrophin-releasing hormone (GnRH) neurones and thus the activation of the reproductive axis in mammals. It has been widely documented that the effects of Kp on reproduction through its action on GnRH neurones are mediated by the GPR54 receptor. Kp controls the activation of the reproductive axis at puberty, maintains reproductive axis activity in adults and is involved in triggering ovulation in some species. Although there is ample evidence coming from both conditional knockout models and conditional-induced Kp neurone death implicating the Kp/GPR54 pathway in the control of reproduction, the mechanism(s) underlying this process may be more complex than a sole direct control of GnRH neuronal activity by Kp. In this review, we provide an overview of the recent advances made in elucidating the interplay between Kp- and GnRH- neuronal networks with respect to regulating the reproductive axis. We highlight the existence of a possible mutual regulation between GnRH and Kp neurones, as well as the implication of Kp-dependent volume transmission in this process. We also discuss the capacity of heterodimerisation between GPR54 and GnRH receptor (GnRH-R) and its consequences on signalling. Finally, we illustrate the role of mathematical modelling that accounts for the synergy between GnRH-R and GPR54 in explaining the role of these two receptors when defining GnRH neuronal activity and GnRH pulsatile release.
Collapse
Affiliation(s)
- Anne Duittoz
- Physiologie de la Reproduction et des Comportements (PRC) UMR7247 INRA, CNRS, Centre INRAe Val de Loire, Université de Tours, IFCE, Nouzilly, France
| | - Xavier Cayla
- Physiologie de la Reproduction et des Comportements (PRC) UMR7247 INRA, CNRS, Centre INRAe Val de Loire, Université de Tours, IFCE, Nouzilly, France
| | - Renaud Fleurot
- Physiologie de la Reproduction et des Comportements (PRC) UMR7247 INRA, CNRS, Centre INRAe Val de Loire, Université de Tours, IFCE, Nouzilly, France
| | - Jonas Lehnert
- Department of Quantitative Life Sciences, McGill University, Montreal, QC, Canada
| | - Anmar Khadra
- Department of Quantitative Life Sciences, McGill University, Montreal, QC, Canada
- Department of Physiology, McGill University, Montréal, QC, Canada
| |
Collapse
|
11
|
Ki SM, Jeong HS, Lee JE. Primary Cilia in Glial Cells: An Oasis in the Journey to Overcoming Neurodegenerative Diseases. Front Neurosci 2021; 15:736888. [PMID: 34658775 PMCID: PMC8514955 DOI: 10.3389/fnins.2021.736888] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 08/31/2021] [Indexed: 12/29/2022] Open
Abstract
Many neurodegenerative diseases have been associated with defects in primary cilia, which are cellular organelles involved in diverse cellular processes and homeostasis. Several types of glial cells in both the central and peripheral nervous systems not only support the development and function of neurons but also play significant roles in the mechanisms of neurological disease. Nevertheless, most studies have focused on investigating the role of primary cilia in neurons. Accordingly, the interest of recent studies has expanded to elucidate the role of primary cilia in glial cells. Correspondingly, several reports have added to the growing evidence that most glial cells have primary cilia and that impairment of cilia leads to neurodegenerative diseases. In this review, we aimed to understand the regulatory mechanisms of cilia formation and the disease-related functions of cilia, which are common or specific to each glial cell. Moreover, we have paid close attention to the signal transduction and pathological mechanisms mediated by glia cilia in representative neurodegenerative diseases. Finally, we expect that this field of research will clarify the mechanisms involved in the formation and function of glial cilia to provide novel insights and ideas for the treatment of neurodegenerative diseases in the future.
Collapse
Affiliation(s)
- Soo Mi Ki
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, South Korea
| | - Hui Su Jeong
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, South Korea
| | - Ji Eun Lee
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, South Korea
- Samsung Medical Center, Samsung Biomedical Research Institute, Seoul, South Korea
| |
Collapse
|
12
|
Di Nardo A, Lenoël I, Winden KD, Rühmkorf A, Modi ME, Barrett L, Ercan-Herbst E, Venugopal P, Behne R, Lopes CAM, Kleiman RJ, Bettencourt-Dias M, Sahin M. Phenotypic Screen with TSC-Deficient Neurons Reveals Heat-Shock Machinery as a Druggable Pathway for mTORC1 and Reduced Cilia. Cell Rep 2021; 31:107780. [PMID: 32579942 PMCID: PMC7381997 DOI: 10.1016/j.celrep.2020.107780] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 04/20/2020] [Accepted: 05/27/2020] [Indexed: 02/08/2023] Open
Abstract
Tuberous sclerosis complex (TSC) is a neurogenetic disorder that leads to elevated mechanistic targeting of rapamycin complex 1 (mTORC1) activity. Cilia can be affected by mTORC1 signaling, and ciliary deficits are associated with neurodevelopmental disorders. Here, we examine whether neuronal cilia are affected in TSC. We show that cortical tubers from TSC patients and mutant mouse brains have fewer cilia. Using high-content image-based assays, we demonstrate that mTORC1 activity inversely correlates with ciliation in TSC1/2-deficientneurons.To investigate the mechanistic relationship between mTORC1 and cilia, we perform a phenotypic screen for mTORC1 inhibitors with TSC1/2-deficient neurons. We identify inhibitors ofthe heat shock protein 90 (Hsp90) that suppress mTORC1 through regulation of phosphatidylinositol 3-kinase (PI3K)/Akt signaling. Pharmacological inhibition of Hsp90 rescues ciliation through downregulation of Hsp27. Our study uncovers the heat-shock machinery as a druggable signaling node to restore mTORC1 activity and cilia due to loss of TSC1/2, and it provides broadly applicable platforms for studying TSC-related neuronal dysfunction. Di Nardo et al. find that cortical tubers from TSC patients and mutant mouse brains have fewer cilia. An image-based screening of mTORC1 activity in TSC1/2-deficient neurons leads to the identification of the heat-shock machinery as a druggable signaling node to restore mTORC1 activity and cilia.
Collapse
Affiliation(s)
- Alessia Di Nardo
- F.M. Kirby Neurobiology Center, Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Isadora Lenoël
- F.M. Kirby Neurobiology Center, Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Kellen D Winden
- F.M. Kirby Neurobiology Center, Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Alina Rühmkorf
- F.M. Kirby Neurobiology Center, Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Meera E Modi
- F.M. Kirby Neurobiology Center, Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Lee Barrett
- F.M. Kirby Neurobiology Center, Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ebru Ercan-Herbst
- F.M. Kirby Neurobiology Center, Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Pooja Venugopal
- F.M. Kirby Neurobiology Center, Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Robert Behne
- F.M. Kirby Neurobiology Center, Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Carla A M Lopes
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande 6, Oeiras 2780-156, Portugal
| | - Robin J Kleiman
- F.M. Kirby Neurobiology Center, Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | - Mustafa Sahin
- F.M. Kirby Neurobiology Center, Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
13
|
Zhang Y, Zhang X, Dai Y, Song M, Zhou Y, Zhou J, Yan X, Shen Y. The decrease of intraflagellar transport impairs sensory perception and metabolism in ageing. Nat Commun 2021; 12:1789. [PMID: 33741976 PMCID: PMC7979750 DOI: 10.1038/s41467-021-22065-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 02/26/2021] [Indexed: 12/22/2022] Open
Abstract
Sensory perception and metabolic homeostasis are known to deteriorate with ageing, impairing the health of aged animals, while mechanisms underlying their deterioration remain poorly understood. The potential interplay between the declining sensory perception and the impaired metabolism during ageing is also barely explored. Here, we report that the intraflagellar transport (IFT) in the cilia of sensory neurons is impaired in the aged nematode Caenorhabditis elegans due to a daf-19/RFX-modulated decrease of IFT components. We find that the reduced IFT in sensory cilia thus impairs sensory perception with ageing. Moreover, we demonstrate that whereas the IFT-dependent decrease of sensory perception in aged worms has a mild impact on the insulin/IGF-1 signalling, it remarkably suppresses AMP-activated protein kinase (AMPK) signalling across tissues. We show that upregulating daf-19/RFX effectively enhances IFT, sensory perception, AMPK activity and autophagy, promoting metabolic homeostasis and longevity. Our study determines an ageing pathway causing IFT decay and sensory perception deterioration, which in turn disrupts metabolism and healthy ageing.
Collapse
Affiliation(s)
- Yincong Zhang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiaona Zhang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yumin Dai
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Mengjiao Song
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yifei Zhou
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jun Zhou
- Institute of Biomedical Sciences, College of Life Sciences, Key Laboratory of Animal Resistance Biology of Shandong Province, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Shandong Normal University, Jinan, Shandong, China
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Xiumin Yan
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yidong Shen
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
14
|
Briuglia S, Calabrò M, Capra AP, Briguori S, La Rosa MA, Crisafulli C. Molecular Pathways within Autism Spectrum Disorder Endophenotypes. J Mol Neurosci 2021; 71:1357-1367. [PMID: 33492615 DOI: 10.1007/s12031-020-01782-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 12/16/2020] [Indexed: 12/19/2022]
Abstract
Autism spectrum disorder (ASD) is a condition that includes a number of neurodevelopmental mental disorders. Recent genetic/genomic investigations have reported an increased prevalence of copy number variations (CNVs) in individuals with autism. Despite the extensive evidence of a genetic component, the genes involved are not known and the background is heterogeneous among subjects. As such, it is highly likely that multiple events (molecular cascades) are implicated in the development of autism. The aim of this work was to shed some light on the biological background behind this condition. We hypothesized that the heterogeneous alterations found within different individuals may converge into one or more specific biological functions (pathways) linked to the heterogeneous phenotypes commonly observed in subjects with ASD. We analyzed a sample of 107 individuals for CNV alterations and checked the genes located within the altered loci (1366). Then, we characterized the subjects for distinct phenotypes. After creating subsamples based on symptoms, the CNVs related to each specific symptom were used to create distinct networks associated with each phenotype (18 in total in the sample under analysis). These networks were independently clustered and enriched to identify potential common pathways involved in autism and variably combined with the clinical phenotype. The first 10 pathways of the analysis are discussed.
Collapse
Affiliation(s)
- Silvana Briuglia
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Torre Biologica Via C. Valeria-Gazzi, Messina, 98125, Italy
| | - Marco Calabrò
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Torre Biologica Via C. Valeria-Gazzi, Messina, 98125, Italy
| | - Anna Paola Capra
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Torre Biologica Via C. Valeria-Gazzi, Messina, 98125, Italy
| | - Sara Briguori
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Torre Biologica Via C. Valeria-Gazzi, Messina, 98125, Italy
| | - Maria Angela La Rosa
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Torre Biologica Via C. Valeria-Gazzi, Messina, 98125, Italy
| | - Concetta Crisafulli
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Torre Biologica Via C. Valeria-Gazzi, Messina, 98125, Italy.
| |
Collapse
|
15
|
Ramos C, Roberts JB, Jasso KR, Ten Eyck TW, Everett T, Pozo P, Setlow B, McIntyre JC. Neuron-specific cilia loss differentially alters locomotor responses to amphetamine in mice. J Neurosci Res 2020; 99:827-842. [PMID: 33175436 DOI: 10.1002/jnr.24755] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 10/07/2020] [Accepted: 10/25/2020] [Indexed: 12/20/2022]
Abstract
The neural mechanisms that underlie responses to drugs of abuse are complex, and impacted by a number of neuromodulatory peptides. Within the past 10 years it has been discovered that several of the receptors for neuromodulators are enriched in the primary cilia of neurons. Primary cilia are microtubule-based organelles that project from the surface of nearly all mammalian cells, including neurons. Despite what we know about cilia, our understanding of how cilia regulate neuronal function and behavior is still limited. The primary objective of this study was to investigate the contributions of primary cilia on specific neuronal populations to behavioral responses to amphetamine. To test the consequences of cilia loss on amphetamine-induced locomotor activity we selectively ablated cilia from dopaminergic or GAD2-GABAergic neurons in mice. Cilia loss had no effect on baseline locomotion in either mouse strain. In mice lacking cilia on dopaminergic neurons, locomotor activity compared to wild- type mice was reduced in both sexes in response to acute administration of 3.0 mg/kg amphetamine. In contrast, changes in the locomotor response to amphetamine in mice lacking cilia on GAD2-GABAergic neurons were primarily driven by reductions in locomotor activity in males. Following repeated amphetamine administration (1.0 mg kg-1 day-1 over 5 days), mice lacking cilia on GAD2-GABAergic neurons exhibited enhanced sensitization of the locomotor stimulant response to the drug, whereas mice lacking cilia on dopaminergic neurons did not differ from wild-type controls. These results indicate that cilia play neuron-specific roles in both acute and neuroplastic responses to psychostimulant drugs of abuse.
Collapse
Affiliation(s)
- Carlos Ramos
- Department of Neuroscience, University of Florida, Gainesville, FL, USA
| | - Jonté B Roberts
- Department of Neuroscience, University of Florida, Gainesville, FL, USA
| | - Kalene R Jasso
- Department of Neuroscience, University of Florida, Gainesville, FL, USA
| | - Tyler W Ten Eyck
- Department of Neuroscience, University of Florida, Gainesville, FL, USA
| | - Thomas Everett
- Department of Neuroscience, University of Florida, Gainesville, FL, USA
| | - Patricia Pozo
- Department of Neuroscience, University of Florida, Gainesville, FL, USA
| | - Barry Setlow
- Department of Psychiatry, University of Florida, Gainesville, FL, USA.,Center for Addiction Research and Education, University of Florida, Gainesville, FL, USA
| | - Jeremy C McIntyre
- Department of Neuroscience, University of Florida, Gainesville, FL, USA.,Center for Addiction Research and Education, University of Florida, Gainesville, FL, USA
| |
Collapse
|
16
|
Saternos H, Ley S, AbouAlaiwi W. Primary Cilia and Calcium Signaling Interactions. Int J Mol Sci 2020; 21:E7109. [PMID: 32993148 PMCID: PMC7583801 DOI: 10.3390/ijms21197109] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/23/2020] [Accepted: 09/24/2020] [Indexed: 02/06/2023] Open
Abstract
The calcium ion (Ca2+) is a diverse secondary messenger with a near-ubiquitous role in a vast array of cellular processes. Cilia are present on nearly every cell type in either a motile or non-motile form; motile cilia generate fluid flow needed for a variety of biological processes, such as left-right body patterning during development, while non-motile cilia serve as the signaling powerhouses of the cell, with vital singling receptors localized to their ciliary membranes. Much of the research currently available on Ca2+-dependent cellular actions and primary cilia are tissue-specific processes. However, basic stimuli-sensing pathways, such as mechanosensation, chemosensation, and electrical sensation (electrosensation), are complex processes entangled in many intersecting pathways; an overview of proposed functions involving cilia and Ca2+ interplay will be briefly summarized here. Next, we will focus on summarizing the evidence for their interactions in basic cellular activities, including the cell cycle, cell polarity and migration, neuronal pattering, glucose-mediated insulin secretion, biliary regulation, and bone formation. Literature investigating the role of cilia and Ca2+-dependent processes at a single-cellular level appears to be scarce, though overlapping signaling pathways imply that cilia and Ca2+ interact with each other on this level in widespread and varied ways on a perpetual basis. Vastly different cellular functions across many different cell types depend on context-specific Ca2+ and cilia interactions to trigger the correct physiological responses, and abnormalities in these interactions, whether at the tissue or the single-cell level, can result in diseases known as ciliopathies; due to their clinical relevance, pathological alterations of cilia function and Ca2+ signaling will also be briefly touched upon throughout this review.
Collapse
Affiliation(s)
| | | | - Wissam AbouAlaiwi
- Department of Pharmacology and Experimental Therapeutics, University of Toledo Health Science Campus, Toledo, OH 43614, USA; (H.S.); (S.L.)
| |
Collapse
|
17
|
Wei Z, Sakamuru S, Zhang L, Zhao J, Huang R, Kleinstreuer NC, Chen Y, Shu Y, Knudsen TB, Xia M. Identification and Profiling of Environmental Chemicals That Inhibit the TGFβ/SMAD Signaling Pathway. Chem Res Toxicol 2019; 32:2433-2444. [PMID: 31652400 PMCID: PMC7341485 DOI: 10.1021/acs.chemrestox.9b00228] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The transforming growth factor beta (TGFβ) superfamily of secreted signaling molecules and their cognate receptors regulate cell fate and behaviors relevant to many developmental and disease processes. Disruption of TGFβ signaling during embryonic development can, for example, affect morphogenesis and differentiation through complex pathways that may be SMAD (Small Mothers Against Decapentaplegic) dependent or SMAD independent. In the present study, the SMAD Binding Element (SBE)-beta lactamase (bla) HEK 293T cell line, which responds to the activation of the SMAD2/3/4 complex, was used in a quantitative high-throughput screening (qHTS) assay to identify potential TGFβ disruptors in the Tox21 10K compound library. From the primary screening we identified several kinase inhibitors, organometallic compounds, and dithiocarbamates (DTCs) that inhibited TGFβ1-induced SMAD signaling of reporter gene activation independent of cytotoxicity. Counterscreen of SBE antagonists on human embryonic neural stem cells demonstrated cytotoxicity, providing additional evidence to support evaluation of these compounds for developmental toxicity. We profiled the inhibitory patterns of putative SBE antagonists toward other developmental signaling pathways, including wingless-related integration site (WNT), retinoic acid α receptor (RAR), and sonic hedgehog (SHH). The profiling results from SBE-bla assay identify chemicals that disrupt TGFβ/SMAD signaling as part of an integrated qHTS approach for prioritizing putative developmental toxicants.
Collapse
Affiliation(s)
- Zhengxi Wei
- National Center for Advancing Translational Sciences, National Institutes of Health, MD, USA
| | - Srilatha Sakamuru
- National Center for Advancing Translational Sciences, National Institutes of Health, MD, USA
| | - Li Zhang
- National Center for Advancing Translational Sciences, National Institutes of Health, MD, USA
| | - Jinghua Zhao
- National Center for Advancing Translational Sciences, National Institutes of Health, MD, USA
| | - Ruili Huang
- National Center for Advancing Translational Sciences, National Institutes of Health, MD, USA
| | - Nicole C. Kleinstreuer
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, USA
| | - Yanling Chen
- Division of Molecular Biology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Laurel, MD, USA
| | - Yan Shu
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201, USA
| | - Thomas B. Knudsen
- National Center for Computational Toxicology, Office of Research and Development, United States Environmental Protection Agency, Research Triangle Park, NC, USA
| | - Menghang Xia
- National Center for Advancing Translational Sciences, National Institutes of Health, MD, USA
| |
Collapse
|
18
|
Fu Z, Gailey CD, Wang EJ, Brautigan DL. Ciliogenesis associated kinase 1: targets and functions in various organ systems. FEBS Lett 2019; 593:2990-3002. [PMID: 31506943 DOI: 10.1002/1873-3468.13600] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 08/22/2019] [Accepted: 09/04/2019] [Indexed: 12/12/2022]
Abstract
Ciliogenesis associated kinase 1 (CILK1) was previously known as intestinal cell kinase because it was cloned from that origin. However, CILK1 is now recognized as a widely expressed and highly conserved serine/threonine protein kinase. Mutations in the human CILK1 gene have been associated with ciliopathies, a group of human genetic disorders with defects in the primary cilium. In mice, both Cilk1 knock-out and Cilk1 knock-in mutations have recapitulated human ciliopathies. Thus, CILK1 has a fundamental role in the function of the cilium. Several candidate substrates have been proposed for CILK1 and the challenge is to relate these to the mutant phenotypes. In this review, we summarize what is known about CILK1 functions and targets, and discuss gaps in current knowledge that motivate further experimentation to fully understand the role of CILK1 in organ development in humans.
Collapse
Affiliation(s)
- Zheng Fu
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Casey D Gailey
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Eric J Wang
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - David L Brautigan
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
19
|
Gomez-Acevedo H, Patterson JD, Sardar S, Gokden M, Das BC, Ussery DW, Rodriguez A. SMARC-B1 deficient sinonasal carcinoma metastasis to the brain with next generation sequencing data: a case report of perineural invasion progressing to leptomeningeal invasion. BMC Cancer 2019; 19:827. [PMID: 31438887 PMCID: PMC6704572 DOI: 10.1186/s12885-019-6043-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 08/15/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND SMARCB1-deficient sinonasal carcinoma (SDSC) is an aggressive subtype of head and neck cancers that has a poor prognosis despite multimodal therapy. We present a unique case with next generation sequencing data of a patient who had SDSC with perineural invasion to the trigeminal nerve that progressed to a brain metastasis and eventually leptomeningeal spread. CASE PRESENTATION A 42 year old female presented with facial pain and had resection of a tumor along the V2 division of the trigeminal nerve on the right. She underwent adjuvant stereotactic radiation. She developed further neurological symptoms and imaging demonstrated the tumor had infiltrated into the cavernous sinus as well as intradurally. She had surgical resection for removal of her brain metastasis and decompression of the cavernous sinus. Following her second surgery, she had adjuvant radiation and chemotherapy. Several months later she had quadriparesis and imaging was consistent with leptomeningeal spread. She underwent palliative radiation and ultimately transitioned quickly to comfort care and expired. Overall survival from time of diagnosis was 13 months. Next generation sequencing was carried out on her primary tumor and brain metastasis. The brain metastatic tissue had an increased tumor mutational burden in comparison to the primary. CONCLUSIONS This is the first report of SDSC with perineural invasion progressing to leptomeningeal carcinomatosis. Continued next generation sequencing of the primary and metastatic tissue by clinicians is encouraged toprovide further insights into metastatic progression of rare solid tumors.
Collapse
Affiliation(s)
- Horacio Gomez-Acevedo
- Department of Biomedical Informatics, University of Arkansas for Medical Science, Little Rock, AR 72205 USA
| | - John D. Patterson
- Department of Neurosurgery, University of Arkansas for Medical Sciences, Little Rock, AR 72205 USA
| | - Sehrish Sardar
- Department of Neurosurgery, University of Arkansas for Medical Sciences, Little Rock, AR 72205 USA
| | - Murat Gokden
- Division of Neuropathology, Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR 72205 USA
| | - Bhaskar C. Das
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - David W. Ussery
- Department of Biomedical Informatics, University of Arkansas for Medical Science, Little Rock, AR 72205 USA
| | - Analiz Rodriguez
- Department of Neurosurgery, University of Arkansas for Medical Sciences, Little Rock, AR 72205 USA
| |
Collapse
|
20
|
Kumar S, Reynolds K, Ji Y, Gu R, Rai S, Zhou CJ. Impaired neurodevelopmental pathways in autism spectrum disorder: a review of signaling mechanisms and crosstalk. J Neurodev Disord 2019; 11:10. [PMID: 31202261 PMCID: PMC6571119 DOI: 10.1186/s11689-019-9268-y] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 05/02/2019] [Indexed: 12/11/2022] Open
Abstract
Background The development of an autistic brain is a highly complex process as evident from the involvement of various genetic and non-genetic factors in the etiology of the autism spectrum disorder (ASD). Despite being a multifactorial neurodevelopmental disorder, autistic patients display a few key characteristics, such as the impaired social interactions and elevated repetitive behaviors, suggesting the perturbation of specific neuronal circuits resulted from abnormal signaling pathways during brain development in ASD. A comprehensive review for autistic signaling mechanisms and interactions may provide a better understanding of ASD etiology and treatment. Main body Recent studies on genetic models and ASD patients with several different mutated genes revealed the dysregulation of several key signaling pathways, such as WNT, BMP, SHH, and retinoic acid (RA) signaling. Although no direct evidence of dysfunctional FGF or TGF-β signaling in ASD has been reported so far, a few examples of indirect evidence can be found. This review article summarizes how various genetic and non-genetic factors which have been reported contributing to ASD interact with WNT, BMP/TGF-β, SHH, FGF, and RA signaling pathways. The autism-associated gene ubiquitin-protein ligase E3A (UBE3A) has been reported to influence WNT, BMP, and RA signaling pathways, suggesting crosstalk between various signaling pathways during autistic brain development. Finally, the article comments on what further studies could be performed to gain deeper insights into the understanding of perturbed signaling pathways in the etiology of ASD. Conclusion The understanding of mechanisms behind various signaling pathways in the etiology of ASD may help to facilitate the identification of potential therapeutic targets and design of new treatment methods.
Collapse
Affiliation(s)
- Santosh Kumar
- Department of Biochemistry and Molecular Medicine, Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, University of California at Davis School of Medicine, 2425 Stockton Blvd, Sacramento, CA, 95817, USA.
| | - Kurt Reynolds
- Department of Biochemistry and Molecular Medicine, Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, University of California at Davis School of Medicine, 2425 Stockton Blvd, Sacramento, CA, 95817, USA
| | - Yu Ji
- Department of Biochemistry and Molecular Medicine, Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, University of California at Davis School of Medicine, 2425 Stockton Blvd, Sacramento, CA, 95817, USA
| | - Ran Gu
- Department of Biochemistry and Molecular Medicine, Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, University of California at Davis School of Medicine, 2425 Stockton Blvd, Sacramento, CA, 95817, USA
| | - Sunil Rai
- Department of Biochemistry and Molecular Medicine, Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, University of California at Davis School of Medicine, 2425 Stockton Blvd, Sacramento, CA, 95817, USA
| | - Chengji J Zhou
- Department of Biochemistry and Molecular Medicine, Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, University of California at Davis School of Medicine, 2425 Stockton Blvd, Sacramento, CA, 95817, USA.
| |
Collapse
|
21
|
Shi W, Ma Z, Zhang G, Wang C, Jiao Z. Novel functions of the primary cilium in bone disease and cancer. Cytoskeleton (Hoboken) 2019; 76:233-242. [PMID: 31108028 DOI: 10.1002/cm.21529] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 05/15/2019] [Accepted: 05/16/2019] [Indexed: 11/09/2022]
Abstract
The primary cilium, a sensory organelle that emanates from the cell surface of most mammalian cell types during growth arrest, has attracted the attention of many researchers over the past decade. Recently, a large number of new findings have assigned novel functions and roles to the primary cilium in signal transduction and related diseases, which has greatly augmented the importance of the cilium in human health and development. Here, we review emerging evidence supporting the primary cilium as a sensory organelle in signal transduction in microgravity, electromagnetic field sensing, chemosensation and tumorigenesis. We also present an overview of signal transduction crosstalk associated with the primary cilium in bone disease and cancer, including primary cilium-related Ca2+ signaling, parathyroid hormone signaling, cAMP signaling, BMP/Smad1/5/8 signaling and Wnt signaling. We anticipate that emerging discoveries about the function of the primary cilium will provide novel insight into the molecular mechanisms of stimulus sensation, signal transduction and pathogenesis.
Collapse
Affiliation(s)
- Wengui Shi
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, People's Republic of China
| | - Zhijian Ma
- The First Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, People's Republic of China
| | - Gengyuan Zhang
- The First Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, People's Republic of China
| | - Chen Wang
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, People's Republic of China.,The First Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, People's Republic of China
| | - Zuoyi Jiao
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, People's Republic of China.,The First Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, People's Republic of China
| |
Collapse
|
22
|
Pruski M, Lang B. Primary Cilia-An Underexplored Topic in Major Mental Illness. Front Psychiatry 2019; 10:104. [PMID: 30886591 PMCID: PMC6409319 DOI: 10.3389/fpsyt.2019.00104] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 02/12/2019] [Indexed: 12/20/2022] Open
Abstract
Though much progress has been made in recent years towards understanding the function and physiology of primary cilia, they remain a somewhat elusive organelle. Some studies have explored the role of primary cilia in the developing nervous system, and their dysfunction has been linked with several neurosensory deficits. Yet, very little has been written on their potential role in psychiatric disorders. This article provides an overview of some of the functions of primary cilia in signalling pathways, and demonstrates that they are a worthy candidate in psychiatric research. The links between primary cilia and major mental illness have been demonstrated to exist at several levels, spanning genetics, signalling pathways, and pharmacology as well as cell division and migration. The primary focus of this review is on the sensory role of the primary cilium and the neurodevelopmental hypothesis of psychiatric disease. As such, the primary cilium is demonstrated to be a key link between the cellular environment and cell behaviour, and hence of key importance in the considerations of the nature and nurture debate in psychiatric research.
Collapse
Affiliation(s)
- Michal Pruski
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China
- Critical Care Laboratory, Critical Care Directorate, Manchester Royal Infirmary, Manchester University NHS Foundation Trust, Manchester, United Kingdom
- School of Healthcare Science, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, United Kingdom
| | - Bing Lang
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen, United Kingdom
| |
Collapse
|