1
|
Nita A, Abraham SP, Elrefaay ER, Fafilek B, Cizkova E, Ursachi VC, Gudernova I, Koudelka A, Dudeja P, Gregor T, Feketova Z, Rico G, Svozilova K, Celiker C, Czyrek AA, Barta T, Trantirek L, Wiedlocha A, Krejci P, Bosakova M. FGFR2 residence in primary cilia is necessary for epithelial cell signaling. J Cell Biol 2025; 224:e202311030. [PMID: 40257378 PMCID: PMC12010920 DOI: 10.1083/jcb.202311030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 11/21/2024] [Accepted: 03/21/2025] [Indexed: 04/22/2025] Open
Abstract
Primary cilium projects from cells to provide a communication platform with neighboring cells and the surrounding environment. This is ensured by the selective entry of membrane receptors and signaling molecules, producing fine-tuned and effective responses to the extracellular cues. In this study, we focused on one family of signaling molecules, the fibroblast growth factor receptors (FGFRs), their residence within cilia, and its role in FGFR signaling. We show that FGFR1 and FGFR2, but not FGFR3 and FGFR4, localize to primary cilia of the developing mouse tissues and in vitro cells. For FGFR2, we demonstrate that the ciliary residence is necessary for its signaling and expression of target morphogenic genes. We also show that the pathogenic FGFR2 variants have minimal cilium presence, which can be rescued for the p.P253R variant associated with the Apert syndrome by using the RLY-4008 kinase inhibitor. Finally, we determine the molecular regulators of FGFR2 trafficking to cilia, including IFT144, BBS1, and the conserved T429V430 motif within FGFR2.
Collapse
Affiliation(s)
- Alexandru Nita
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Institute of Animal Physiology and Genetics of the CAS, Brno, Czech Republic
| | - Sara P. Abraham
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Institute of Animal Physiology and Genetics of the CAS, Brno, Czech Republic
| | - Eman R. Elrefaay
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Institute of Animal Physiology and Genetics of the CAS, Brno, Czech Republic
| | - Bohumil Fafilek
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Eliska Cizkova
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Vlad Constantin Ursachi
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- International Clinical Research Center, St. Anne’s University Hospital, Brno, Czech Republic
| | - Iva Gudernova
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Institute of Animal Physiology and Genetics of the CAS, Brno, Czech Republic
| | - Adolf Koudelka
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Pooja Dudeja
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- International Clinical Research Center, St. Anne’s University Hospital, Brno, Czech Republic
| | - Tomas Gregor
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Zuzana Feketova
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- International Clinical Research Center, St. Anne’s University Hospital, Brno, Czech Republic
| | - Gustavo Rico
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- International Clinical Research Center, St. Anne’s University Hospital, Brno, Czech Republic
| | - Katerina Svozilova
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Institute of Animal Physiology and Genetics of the CAS, Brno, Czech Republic
| | - Canan Celiker
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Aleksandra A. Czyrek
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- International Clinical Research Center, St. Anne’s University Hospital, Brno, Czech Republic
| | - Tomas Barta
- Institute of Animal Physiology and Genetics of the CAS, Brno, Czech Republic
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Lukas Trantirek
- CEITEC - Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Antoni Wiedlocha
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Centre for Cancer Cell Reprograming, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Pavel Krejci
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Institute of Animal Physiology and Genetics of the CAS, Brno, Czech Republic
- International Clinical Research Center, St. Anne’s University Hospital, Brno, Czech Republic
| | - Michaela Bosakova
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Institute of Animal Physiology and Genetics of the CAS, Brno, Czech Republic
| |
Collapse
|
2
|
Deraniyagala AS, Maier W, Parra M, Nanista E, Sowunmi DO, Hassan M, Chasen N, Sharma S, Lechtreck KF, Cole ES, Bernardes N, Chook YM, Gaertig J. Importin-9 and a TPR domain protein MpH drive periodic patterning of ciliary arrays in Tetrahymena. J Cell Biol 2025; 224:e202409057. [PMID: 40152790 PMCID: PMC11951933 DOI: 10.1083/jcb.202409057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 01/11/2025] [Accepted: 02/27/2025] [Indexed: 03/29/2025] Open
Abstract
We explored how the number of structures is determined in an intracellular organelle series. In Tetrahymena, the oral apparatus contains three diagonal ciliary rows: M1, M2, and M3. During development, the M rows emerge by sequential segmentation of a group of basal bodies, starting with the longest and most anterior M1 and ending with the shortest and most posterior M3. The mpD-1 and mpH-1 alleles increase and decrease the number of M rows, respectively. We identify MpH as a TPR protein and MpD as an importin-9. Both proteins localize to the M rows and form concentration gradients. MpH is a row elongation factor whose loss shortens all M rows and often prevents the formation of M3. MpD limits row initiation after the emergence of M2. MpD could be a part of a negative feedback loop that limits row initiation when M1 assembly is properly advanced. We conclude that the forming oral apparatus has properties of a semi-autonomous intracellular developmental field.
Collapse
Affiliation(s)
| | - Wolfgang Maier
- Bioinformatics, University of Freiburg, Freiburg, Germany
| | - Mireya Parra
- Department of Cellular Biology, University of Georgia, Athens, GA, USA
| | - Elise Nanista
- Department of Cellular Biology, University of Georgia, Athens, GA, USA
| | | | - Michael Hassan
- Department of Cellular Biology, University of Georgia, Athens, GA, USA
| | - Nathan Chasen
- Department of Cellular Biology, University of Georgia, Athens, GA, USA
| | - Sunita Sharma
- Department of Cellular Biology, University of Georgia, Athens, GA, USA
| | - Karl F. Lechtreck
- Department of Cellular Biology, University of Georgia, Athens, GA, USA
| | - Eric S. Cole
- Biology Department, St. Olaf College, Northfield, MN, USA
| | - Natalia Bernardes
- Departments of Pharmacology and Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yuh Min Chook
- Departments of Pharmacology and Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jacek Gaertig
- Department of Cellular Biology, University of Georgia, Athens, GA, USA
| |
Collapse
|
3
|
Chen Z, He J, Guo Y, Hao Y, Lv W, Chen Z, Wang J, Yang Y, Wang K, Liu Z, Ouyang Q, Su Z, Hu P, Xiao G. Adherent junctions: Physiology, role in hydrocephalus and potential therapeutic targets. IBRO Neurosci Rep 2025; 18:283-292. [PMID: 39995568 PMCID: PMC11849119 DOI: 10.1016/j.ibneur.2025.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 01/14/2025] [Accepted: 02/03/2025] [Indexed: 02/26/2025] Open
Abstract
In all epithelial cells, the adherent junctions (AJs) with cadherin as the core play an important role in the maintenance of the connection and the formation of apical-basal polarity. The ependymal cells close to the ventricular system rely on AJs with N-cadherin at the core to maintain their normal morphology and function. Therefore, it has an important impact on the function and disease of the central nervous system. Hydrocephalus is a pathological phenomenon of excessive cerebrospinal fluid accumulating in the ventricular system accompanied by continuous ventricular dilatation, which can be divided into obstructive hydrocephalus and communicating hydrocephalus according to the pathogenesis. Obstructive hydrocephalus is often associated with excessive ependymal cells produced by differentiation of radial glial cells. The etiology of communicating hydrocephalus is mainly related to the dyskinesia of cerebrospinal fluid. In addition, the damage of the brain barrier can lead to brain edema and aggravate the symptoms. At present, the researches on the pathogenesis of hydrocephalus are mainly focused on the development of ependymal cells and cilia, while less attention has been paid to molecules such as AJs, which play an important role in maintaining the polarity of ependymal cells. This paper discusses the formation and function of AJs and their role in preventing hydrocephalus by preserving the polarity of ependymal cilia, regulating the number of ependymal cells, and upholding the brain barrier integrity to impede hydrocephalus exacerbation, which provides a new direction for the study of hydrocephalus.
Collapse
Affiliation(s)
- Zhiye Chen
- Department of Diagnostic Radiology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan 410008, PR China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Jian He
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Yating Guo
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Yue Hao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Wentao Lv
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Zexin Chen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Junqiang Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Yijian Yang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Kaiyue Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Zhikun Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Qian Ouyang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- Department of Neurosurgery, Zhuzhou Hospital, Central South University Xiangya School of Medicine, Zhuzhou, Hunan 412000, PR China
| | - Zhangjie Su
- Department of Neurosurgery, Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge CB21 2QQ, UK
| | - Pingsheng Hu
- Department of Diagnostic Radiology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan 410008, PR China
| | - Gelei Xiao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| |
Collapse
|
4
|
Sheridan J, Grata A, Dorr J, Suva EE, Bresteau E, Mitchell LR, Hassan O, Mitchell B. Centriolar defects underlie a primary ciliary dyskinesia phenotype in an adenylate kinase 7 deficient ciliated epithelium. Dev Biol 2025; 524:152-161. [PMID: 40381709 DOI: 10.1016/j.ydbio.2025.05.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 04/01/2025] [Accepted: 05/13/2025] [Indexed: 05/20/2025]
Abstract
The skin of Xenopus embryos contains numerous multiciliated cells (MCCs), which collectively generate a directed fluid flow across the epithelial surface essential for distributing the overlaying mucous. MCCs develop into highly specialized cells to generate this flow, containing approximately 150 evenly spaced centrioles that give rise to motile cilia. MCC-driven fluid flow can be impaired when ciliary dysfunction occurs, resulting in primary ciliary dyskinesia (PCD) in humans. Mutations in a large number of genes (∼50) have been found to be causative to PCD. Recently, studies have linked low levels of Adenylate Kinase 7 (AK7) gene expression to patients with PCD; however, the mechanism for this link remains unclear. Additionally, AK7 mutations have been linked to multiple PCD patients. Adenylate kinases modulate ATP production and consumption, with AK7 explicitly associated with motile cilia. Here we reproduce an AK7 PCD-like phenotype in Xenopus and describe the cellular consequences that occur with manipulation of AK7 levels. We show that AK7 localizes throughout the cilia in a DPY30 domain-dependent manner, suggesting a ciliary function. Additionally, we find that AK7 overexpression increases centriole number, suggesting a role in regulating centriole biogenesis. We find that in AK7-depleted embryos, cilia number, length, and beat frequency are all reduced, which in turn significantly decreases the tissue-wide mucociliary flow. Additionally, we find a decrease in centriole number and an increase in sub-apical centrioles, implying that AK7 influences both centriole biogenesis and docking, which we propose underlie its defect in ciliogenesis. We find that both the AK domain and the DPY30 domain are required for proper centriole regulation. We propose that AK7 plays a role in PCD by impacting centriole biogenesis and apical docking, ultimately leading to ciliogenesis defects that impair mucociliary clearance.
Collapse
Affiliation(s)
- Jennifer Sheridan
- Northwestern University, Feinberg School of Medicine, Department of Cell and Developmental Biology, USA
| | - Aline Grata
- Northwestern University, Feinberg School of Medicine, Department of Cell and Developmental Biology, USA
| | - Julia Dorr
- Northwestern University, Feinberg School of Medicine, Department of Cell and Developmental Biology, USA
| | - Eve E Suva
- Northwestern University, Feinberg School of Medicine, Department of Cell and Developmental Biology, USA
| | - Enzo Bresteau
- Northwestern University, Feinberg School of Medicine, Department of Cell and Developmental Biology, USA
| | - Linus R Mitchell
- Northwestern University, Feinberg School of Medicine, Department of Cell and Developmental Biology, USA
| | - Osama Hassan
- Northwestern University, Feinberg School of Medicine, Department of Cell and Developmental Biology, USA
| | - Brian Mitchell
- Northwestern University, Feinberg School of Medicine, Department of Cell and Developmental Biology, USA; Northwestern University, Lurie Cancer Center, USA.
| |
Collapse
|
5
|
Wiegel J, Helmstädter M, Walz G, Bergen MD. Spontaneous Calcium Bursts Organize the Apical Actin Cytoskeleton of Multiciliated Cells. Int J Mol Sci 2025; 26:2507. [PMID: 40141151 PMCID: PMC11942550 DOI: 10.3390/ijms26062507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 03/05/2025] [Accepted: 03/07/2025] [Indexed: 03/28/2025] Open
Abstract
Motile cilia perform crucial functions during embryonic development and in adult tissues. They are anchored by an apical actin network that forms microridge-like structures on the surface of multiciliated cells. Using Xenopus as a model system to investigate the mechanisms underlying the formation of these specialized actin structures, we observed stochastic bursts of intracellular calcium concentration in developing multiciliated cells. Through optogenetic manipulation of calcium signaling, we found that individual calcium bursts triggered the fusion and extension of actin structures by activating non-muscle myosin. Repeated cycles of calcium activation promoted assembly and coherence of the maturing apical actin network. Inhibition of the endogenous inositol triphosphate-calcium pathway disrupted the formation of apical actin/microridge-like structures by reducing local centriolar RhoA signaling. This disruption was rescued by transient expression of constitutively active RhoA in multiciliated cells. Our findings identify repetitive calcium bursts as a driving force that promotes the self-organization of the highly specialized actin cytoskeleton of multiciliated cells.
Collapse
Affiliation(s)
- Johannes Wiegel
- Department of Medicine IV, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106 Freiburg, Germany; (J.W.); (M.H.); (G.W.)
| | - Martin Helmstädter
- Department of Medicine IV, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106 Freiburg, Germany; (J.W.); (M.H.); (G.W.)
- EMcore, Renal Division, Department of Medicine, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Gerd Walz
- Department of Medicine IV, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106 Freiburg, Germany; (J.W.); (M.H.); (G.W.)
- BIOSS and CIBSS Centre for Integrative Biological Signalling, University of Freiburg, Schänzlestrasse 18, 79104 Freiburg, Germany
| | - Max D. Bergen
- Department of Medicine IV, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106 Freiburg, Germany; (J.W.); (M.H.); (G.W.)
| |
Collapse
|
6
|
Narayanan V, Rao VG, Arrigo A, Kulkarni SS. Multiciliated cells adapt the mechanochemical Piezo1-Erk1/2-Yap1 cell proliferation axis to fine-tune centriole number. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.21.634139. [PMID: 39896494 PMCID: PMC11785133 DOI: 10.1101/2025.01.21.634139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Multiciliated cells (MCCs) are specialized epithelial cells that undergo massive amplification of centrioles, constructing several motile cilia to propel fluid flow. The abundance of cilia is critical for efficient fluid flow, yet how MCCs regulate centriole/cilia numbers remains a major knowledge gap. We have shown that mechanical tension plays a central role in regulating apical area and centriole number in MCCs. Here, we demonstrate that centriole amplification is controlled by a mechanochemical pathway essential for cell proliferation in cycling cells. Specifically, MCCs under tension use Piezo1-mediated calcium signaling to drive Erk½ phosphorylation via PKC and subsequent Yap1 activation. Remarkably, MCCs use this pathway to activate a cilia-specific transcription program, influencing the expression of Foxj1, a master regulator of motile ciliogenesis. Our work is the first to identify a novel function for an important mechanochemical pathway in centriole amplification in MCCs, offering new insights into ciliopathies and cancer, where aberrant centriole numbers are implicated. Teaser This study demonstrates that multiciliated cells utilize the mechanochemical Piezo1-Erk1/2-Yap1 cell proliferation axis to activate the cilia-specific transcriptional factor Foxj1 and amplify centrioles in a tension- dependent manner.
Collapse
|
7
|
Prasai A, Ivashchenko O, Maskova K, Bykova S, Schmidt Cernohorska M, Stepanek O, Huranova M. BBSome-deficient cells activate intraciliary CDC42 to trigger actin-dependent ciliary ectocytosis. EMBO Rep 2025; 26:36-60. [PMID: 39587330 PMCID: PMC11724091 DOI: 10.1038/s44319-024-00326-z] [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: 09/04/2023] [Revised: 10/20/2024] [Accepted: 10/25/2024] [Indexed: 11/27/2024] Open
Abstract
Bardet-Biedl syndrome (BBS) is a pleiotropic ciliopathy caused by dysfunction of the BBSome, a cargo adaptor essential for export of transmembrane receptors from cilia. Although actin-dependent ectocytosis has been proposed to compensate defective cargo retrieval, its molecular basis remains unclear, especially in relation to BBS pathology. In this study, we investigated how actin polymerization and ectocytosis are regulated within the cilium. Our findings reveal that ciliary CDC42, a RHO-family GTPase triggers in situ actin polymerization, ciliary ectocytosis, and cilia shortening in BBSome-deficient cells. Activation of the Sonic Hedgehog pathway further enhances CDC42 activity specifically in BBSome-deficient cilia. Inhibition of CDC42 in BBSome-deficient cells decreases the frequency and duration of ciliary actin polymerization events, causing buildup of G protein coupled receptor 161 (GPR161) in bulges along the axoneme during Sonic Hedgehog signaling. Overall, our study identifies CDC42 as a key trigger of ciliary ectocytosis. Hyperactive ciliary CDC42 and ectocytosis and the resulting loss of ciliary material might contribute to BBS disease severity.
Collapse
Affiliation(s)
- Avishek Prasai
- Laboratory of Adaptive Immunity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
- Faculty of Science, Department of Developmental and Cell Biology, Charles University, Prague, Czech Republic
- Center for Molecular Signaling (PZMS), Department of Medical Biochemistry and Molecular Biology, Saarland University School of Medicine, Homburg, Germany
| | - Olha Ivashchenko
- Laboratory of Adaptive Immunity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
- Faculty of Science, Department of Developmental and Cell Biology, Charles University, Prague, Czech Republic
| | - Kristyna Maskova
- Laboratory of Adaptive Immunity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Sofiia Bykova
- Laboratory of Adaptive Immunity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Marketa Schmidt Cernohorska
- Laboratory of Adaptive Immunity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
- Max Perutz Labs, University of Vienna, Vienna Biocenter (VBC), Vienna, Austria
| | - Ondrej Stepanek
- Laboratory of Adaptive Immunity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Martina Huranova
- Laboratory of Adaptive Immunity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic.
| |
Collapse
|
8
|
Lee M, Carpenter C, Hwang YS, Yoon J, Lu Q, Westlake CJ, Moody SA, Yamaguchi TP, Daar IO. Proliferation associated 2G4 is required for the ciliation of vertebrate motile cilia. Commun Biol 2024; 7:1430. [PMID: 39496919 PMCID: PMC11535434 DOI: 10.1038/s42003-024-07150-0] [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: 05/28/2024] [Accepted: 10/25/2024] [Indexed: 11/06/2024] Open
Abstract
Motile cilia are critical structures that regulate early embryonic development and tissue homeostasis through synchronized ciliary motility. The formation of motile cilia is dependent on precisely controlled sequential processes including the generation, migration, and docking of centrioles/basal bodies as well as ciliary growth. Using the published proteomics data from various organisms, we identified proliferation-associated 2G4 as a novel regulator of ciliogenesis. Loss-of-function studies using Xenopus laevis as a model system reveal that Pa2G4 is essential for proper ciliogenesis and synchronized movement of cilia in multiciliated cells (MCCs) and the gastrocoel roof plate (GRP). Pa2G4 morphant MCCs exhibit defective basal body docking to the surface as a result of compromised Rac1 activity, apical actin network formation, and immature distal appendage generation. Interestingly, the regions that include the RNA-binding domain and the C-terminus of Pa2G4 are necessary for ciliogenesis in both MCCs and GRP cells. Our findings may provide insights into motile cilia-related genetic diseases such as Primary Ciliary Dyskinesia.
Collapse
Affiliation(s)
- Moonsup Lee
- Cancer & Developmental Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Christina Carpenter
- Electron Microscopy Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Yoo-Seok Hwang
- Cancer & Developmental Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Jaeho Yoon
- Cancer & Developmental Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Quanlong Lu
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Christopher J Westlake
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Sally A Moody
- Department of Anatomy and Cell Biology, George Washington University, School of Medicine and Health Sciences, Washington, USA
| | - Terry P Yamaguchi
- Cancer & Developmental Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA.
| | - Ira O Daar
- Cancer & Developmental Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, USA.
| |
Collapse
|
9
|
Kulkarni SS, Stephenson RE, Amalraj S, Arrigo A, Betleja E, Moresco JJ, Yates JR, Mahjoub MR, Miller AL, Khokha MK. The Heterotaxy Gene CCDC11 Is Important for Cytokinesis via RhoA Regulation. Cytoskeleton (Hoboken) 2024. [PMID: 39479942 DOI: 10.1002/cm.21952] [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: 10/23/2023] [Revised: 10/10/2024] [Accepted: 10/15/2024] [Indexed: 11/02/2024]
Abstract
Mutations in CCDC11 (cfap53) have been identified in multiple patients with heterotaxy (Htx), a disorder of left-right (LR) patterning of the internal organs. In Xenopus, depletion of Ccdc11 causes defects in LR patterning, recapitulating the patient phenotype. Upon Ccdc11 depletion, monociliated cells of the Left-Right Organizer (LRO) exhibit multiple cilia per cell. Unexpectedly, we found that Ccdc11 is necessary for successful cytokinesis, explaining the multiciliation phenotype observed in Ccdc11-depleted cells. The small GTPase RhoA is critical for cytokinesis, and our Ccdc11 depletion phenotypes are reminiscent of RhoA loss of function. Here, we demonstrate that during cytokinesis CCDC11 is localized to the cytokinetic contractile ring overlapping with RhoA, and CCDC11 regulates total RhoA protein levels. Our results connect CCDC11 to cytokinesis and LR patterning via RhoA regulation, providing a potential mechanism for heterotaxy disease pathogenesis.
Collapse
Affiliation(s)
- Saurabh S Kulkarni
- Department of Cell Biology and Biology, University of Virginia, Charlottesville, Virginia, USA
| | - Rachel E Stephenson
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Sarah Amalraj
- Pediatric Genomics Discovery Program, Department of Pediatrics and Genetics, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Angelo Arrigo
- Department of Cell Biology and Biology, University of Virginia, Charlottesville, Virginia, USA
| | - Ewelina Betleja
- Department of Medicine (Nephrology), Washington University in St. Louis, St. Louis, Missouri, USA
| | - James J Moresco
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, California, USA
| | - John R Yates
- Department of Molecular Medicine, Scripps Research Institute, La Jolla, California, USA
| | - Moe R Mahjoub
- Department of Medicine (Nephrology), Washington University in St. Louis, St. Louis, Missouri, USA
- Department of Cell Biology and Physiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Ann L Miller
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Mustafa K Khokha
- Pediatric Genomics Discovery Program, Department of Pediatrics and Genetics, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
10
|
Nguyen TK, Baker S, Rodriguez JM, Arceri L, Wingert RA. Using Zebrafish to Study Multiciliated Cell Development and Disease States. Cells 2024; 13:1749. [PMID: 39513856 PMCID: PMC11545745 DOI: 10.3390/cells13211749] [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: 10/04/2024] [Revised: 10/20/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Multiciliated cells (MCCs) serve many important functions, including fluid propulsion and chemo- and mechanosensing. Diseases ranging from rare conditions to the recent COVID-19 global health pandemic have been linked to MCC defects. In recent years, the zebrafish has emerged as a model to investigate the biology of MCCs. Here, we review the major events in MCC formation including centriole biogenesis and basal body docking. Then, we discuss studies on the role of MCCs in diseases of the brain, respiratory, kidney and reproductive systems, as well as recent findings about the link between MCCs and SARS-CoV-2. Next, we explore why the zebrafish is a useful model to study MCCs and provide a comprehensive overview of previous studies of genetic components essential for MCC development and motility across three major tissues in the zebrafish: the pronephros, brain ependymal cells and nasal placode. Taken together, here we provide a cohesive summary of MCC research using the zebrafish and its future potential for expanding our understanding of MCC-related disease states.
Collapse
Affiliation(s)
- Thanh Khoa Nguyen
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA; (S.B.); (J.-M.R.); (L.A.)
| | | | | | | | - Rebecca A. Wingert
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA; (S.B.); (J.-M.R.); (L.A.)
| |
Collapse
|
11
|
Wesselman HM, Arceri L, Nguyen TK, Lara CM, Wingert RA. Genetic mechanisms of multiciliated cell development: from fate choice to differentiation in zebrafish and other models. FEBS J 2024; 291:4159-4192. [PMID: 37997009 DOI: 10.1111/febs.17012] [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] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 10/17/2023] [Accepted: 11/21/2023] [Indexed: 11/25/2023]
Abstract
Multiciliated cells (MCCS) form bundles of cilia and their activities are essential for the proper development and physiology of many organ systems. Not surprisingly, defects in MCCs have profound consequences and are associated with numerous disease states. Here, we discuss the current understanding of MCC formation, with a special focus on the genetic and molecular mechanisms of MCC fate choice and differentiation. Furthermore, we cast a spotlight on the use of zebrafish to study MCC ontogeny and several recent advances made in understanding MCCs using this vertebrate model to delineate mechanisms of MCC emergence in the developing kidney.
Collapse
Affiliation(s)
| | - Liana Arceri
- Department of Biological Sciences, University of Notre Dame, IN, USA
| | - Thanh Khoa Nguyen
- Department of Biological Sciences, University of Notre Dame, IN, USA
| | - Caroline M Lara
- Department of Biological Sciences, University of Notre Dame, IN, USA
| | - Rebecca A Wingert
- Department of Biological Sciences, University of Notre Dame, IN, USA
| |
Collapse
|
12
|
Kim Y, Lee HK, Park KY, Ismail T, Lee H, Lee HS. Actin Depolymerizing Factor Destrin Regulates Cilia Development and Function during Vertebrate Embryogenesis. Dev Reprod 2024; 28:109-119. [PMID: 39444639 PMCID: PMC11495882 DOI: 10.12717/dr.2024.28.3.109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/05/2024] [Accepted: 08/22/2024] [Indexed: 10/25/2024]
Abstract
The actin cytoskeleton plays fundamental roles in ciliogenesis and the actin depolymerizing factor destrin regulates actin dynamics by treadmilling actin filaments and increasing globular actin pools. However, the specific developmental roles of destrin in ciliogenesis have not been fully elucidated. Here, we investigated the function of destrin in ciliogenesis using Xenopus laevis and human retinal pigmented epithelial (hRPE1) cells. We discovered the loss of destrin increased the number of multiciliated cells in the Xenopus epithelium and impeded cilia motility. Additionally, destrin depletion remarkably reduced the length of primary cilia in the Xenopus neural tube and hRPE1 cells by affecting actin dynamics. Immunofluorescence using markers of ciliary components indicated that destrin controls the directionality and polarity of basal bodies and axonemal elongation by modulating actin dynamics, independent of basal body docking. In conclusion, destrin plays a significant role during vertebrate ciliogenesis regulating both primary and multicilia development. Our data suggest new insights for understanding the roles of actin dynamics in cilia development.
Collapse
Affiliation(s)
- Youni Kim
- KNU G-LAMP Project Group, KNU Institute
of Basic Sciences, School of Life Sciences, BK21 FOUR KNU Creative
BioResearch Group, College of Natural Sciences, Kyungpook National
University, Daegu 41566, Korea
| | - Hyun-Kyung Lee
- KNU G-LAMP Project Group, KNU Institute
of Basic Sciences, School of Life Sciences, BK21 FOUR KNU Creative
BioResearch Group, College of Natural Sciences, Kyungpook National
University, Daegu 41566, Korea
| | - Kyeong-Yeon Park
- KNU G-LAMP Project Group, KNU Institute
of Basic Sciences, School of Life Sciences, BK21 FOUR KNU Creative
BioResearch Group, College of Natural Sciences, Kyungpook National
University, Daegu 41566, Korea
| | - Tayaba Ismail
- KNU G-LAMP Project Group, KNU Institute
of Basic Sciences, School of Life Sciences, BK21 FOUR KNU Creative
BioResearch Group, College of Natural Sciences, Kyungpook National
University, Daegu 41566, Korea
| | - Hongchan Lee
- KNU G-LAMP Project Group, KNU Institute
of Basic Sciences, School of Life Sciences, BK21 FOUR KNU Creative
BioResearch Group, College of Natural Sciences, Kyungpook National
University, Daegu 41566, Korea
| | - Hyun-Shik Lee
- KNU G-LAMP Project Group, KNU Institute
of Basic Sciences, School of Life Sciences, BK21 FOUR KNU Creative
BioResearch Group, College of Natural Sciences, Kyungpook National
University, Daegu 41566, Korea
| |
Collapse
|
13
|
Howes A, Rogerson C, Belyaev N, Karagyozova T, Rapiteanu R, Fradique R, Pellicciotta N, Mayhew D, Hurd C, Crotta S, Singh T, Dingwell K, Myatt A, Arad N, Hasan H, Bijlsma H, Panjwani A, Vijayan V, Young G, Bridges A, Petit-Frere S, Betts J, Larminie C, Smith JC, Hessel EM, Michalovich D, Walport L, Cicuta P, Powell AJ, Beinke S, Wack A. The FAM13A Long Isoform Regulates Cilia Movement and Coordination in Airway Mucociliary Transport. Am J Respir Cell Mol Biol 2024; 71:282-293. [PMID: 38691660 PMCID: PMC11376246 DOI: 10.1165/rcmb.2024-0063oc] [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: 02/15/2024] [Accepted: 05/01/2024] [Indexed: 05/03/2024] Open
Abstract
Single nucelotide polymorphisms (SNPs) at the FAM13A locus are among the most commonly reported risk alleles associated with chronic obstructive pulmonary disease (COPD) and other respiratory diseases; however, the physiological role of FAM13A is unclear. In humans, two major protein isoforms are expressed at the FAM13A locus: "long" and "short," but their functions remain unknown, partly because of a lack of isoform conservation in mice. We performed in-depth characterization of organotypic primary human airway epithelial cell subsets and show that multiciliated cells predominantly express the FAM13A long isoform containing a putative N-terminal Rho GTPase-activating protein (RhoGAP) domain. Using purified proteins, we directly demonstrate the RhoGAP activity of this domain. In Xenopus laevis, which conserve the long-isoform, Fam13a deficiency impaired cilia-dependent embryo motility. In human primary epithelial cells, long-isoform deficiency did not affect multiciliogenesis but reduced cilia coordination in mucociliary transport assays. This is the first demonstration that FAM13A isoforms are differentially expressed within the airway epithelium, with implications for the assessment and interpretation of SNP effects on FAM13A expression levels. We also show that the long FAM13A isoform coordinates cilia-driven movement, suggesting that FAM13A risk alleles may affect susceptibility to respiratory diseases through deficiencies in mucociliary clearance.
Collapse
Affiliation(s)
| | - Clare Rogerson
- Immunoregulation Laboratory
- Crick-GSK Biomedical LinkLabs
| | | | | | | | - Ricardo Fradique
- Cavendish Laboratory, University of Cambridge, Cambridge, United Kingdom
| | | | | | - Catherine Hurd
- Protein-Protein Interaction Laboratory
- Crick-GSK Biomedical LinkLabs
| | | | | | | | - Anniek Myatt
- Capgemini Engineering, Capgemini UK, Stevenage, United Kingdom; and
| | - Navot Arad
- Capgemini Engineering, Capgemini UK, Stevenage, United Kingdom; and
| | - Hikmatyar Hasan
- Capgemini Engineering, Capgemini UK, Stevenage, United Kingdom; and
| | - Hielke Bijlsma
- Capgemini Engineering, Capgemini UK, Stevenage, United Kingdom; and
| | | | - Vinaya Vijayan
- Development Digital and Tech, GSK, Collegeville, Pennsylvania
| | - George Young
- Bioinformatics and Biostatistics, The Francis Crick Institute, London, United Kingdom
| | | | | | | | | | | | - Edith M. Hessel
- Refractory Respiratory Inflammation Discovery Performance Unit, GSK R&D, Stevenage, United Kingdom
| | | | | | - Pietro Cicuta
- Cavendish Laboratory, University of Cambridge, Cambridge, United Kingdom
| | | | | | | |
Collapse
|
14
|
Hannaford MR, Rusan NM. Positioning centrioles and centrosomes. J Cell Biol 2024; 223:e202311140. [PMID: 38512059 PMCID: PMC10959756 DOI: 10.1083/jcb.202311140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 02/23/2024] [Accepted: 02/26/2024] [Indexed: 03/22/2024] Open
Abstract
Centrosomes are the primary microtubule organizer in eukaryotic cells. In addition to shaping the intracellular microtubule network and the mitotic spindle, centrosomes are responsible for positioning cilia and flagella. To fulfill these diverse functions, centrosomes must be properly located within cells, which requires that they undergo intracellular transport. Importantly, centrosome mispositioning has been linked to ciliopathies, cancer, and infertility. The mechanisms by which centrosomes migrate are diverse and context dependent. In many cells, centrosomes move via indirect motor transport, whereby centrosomal microtubules engage anchored motor proteins that exert forces on those microtubules, resulting in centrosome movement. However, in some cases, centrosomes move via direct motor transport, whereby the centrosome or centriole functions as cargo that directly binds molecular motors which then walk on stationary microtubules. In this review, we summarize the mechanisms of centrosome motility and the consequences of centrosome mispositioning and identify key questions that remain to be addressed.
Collapse
Affiliation(s)
- Matthew R. Hannaford
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Nasser M. Rusan
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
15
|
Brody SL, Pan J, Huang T, Xu J, Xu H, Koenitizer J, Brennan SK, Nanjundappa R, Saba TG, Berical A, Hawkins FJ, Wang X, Zhang R, Mahjoub MR, Horani A, Dutcher SK. Loss of an extensive ciliary connectome induces proteostasis and cell fate switching in a severe motile ciliopathy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.20.585965. [PMID: 38562900 PMCID: PMC10983967 DOI: 10.1101/2024.03.20.585965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Motile cilia have essential cellular functions in development, reproduction, and homeostasis. Genetic causes for motile ciliopathies have been identified, but the consequences on cellular functions beyond impaired motility remain unknown. Variants in CCDC39 and CCDC40 cause severe disease not explained by loss of motility. Using human cells with pathological variants in these genes, Chlamydomonas genetics, cryo-electron microscopy, single cell RNA transcriptomics, and proteomics, we identified perturbations in multiple cilia-independent pathways. Absence of the axonemal CCDC39/CCDC40 heterodimer results in loss of a connectome of over 90 proteins. The undocked connectome activates cell quality control pathways, switches multiciliated cell fate, impairs microtubule architecture, and creates a defective periciliary barrier. Both cilia-dependent and independent defects are likely responsible for the disease severity. Our findings provide a foundation for reconsidering the broad cellular impact of pathologic variants in ciliopathies and suggest new directions for therapies.
Collapse
Affiliation(s)
- Steven L. Brody
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Jiehong Pan
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Tao Huang
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Jian Xu
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Huihui Xu
- Department of Pediatrics, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Jeffrey Koenitizer
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Steven K. Brennan
- Department of Pediatrics, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Rashmi Nanjundappa
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Thomas G. Saba
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, 48108, USA
| | - Andrew Berical
- Center for Regenerative Medicine, Boston University, Boston, MA, 02118, USA
| | - Finn J. Hawkins
- Center for Regenerative Medicine, Boston University, Boston, MA, 02118, USA
| | - Xiangli Wang
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Rui Zhang
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Moe R. Mahjoub
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, 63110, USA
- Department of Cell Biology and Physisology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Amjad Horani
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, 48108, USA
- Department of Cell Biology and Physisology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Susan K. Dutcher
- Department of Cell Biology and Physisology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| |
Collapse
|
16
|
Kalot R, Sentell Z, Kitzler TM, Torban E. Primary cilia and actin regulatory pathways in renal ciliopathies. FRONTIERS IN NEPHROLOGY 2024; 3:1331847. [PMID: 38292052 PMCID: PMC10824913 DOI: 10.3389/fneph.2023.1331847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 12/20/2023] [Indexed: 02/01/2024]
Abstract
Ciliopathies are a group of rare genetic disorders caused by defects to the structure or function of the primary cilium. They often affect multiple organs, leading to brain malformations, congenital heart defects, and anomalies of the retina or skeletal system. Kidney abnormalities are among the most frequent ciliopathic phenotypes manifesting as smaller, dysplastic, and cystic kidneys that are often accompanied by renal fibrosis. Many renal ciliopathies cause chronic kidney disease and often progress to end-stage renal disease, necessitating replacing therapies. There are more than 35 known ciliopathies; each is a rare hereditary condition, yet collectively they account for a significant proportion of chronic kidney disease worldwide. The primary cilium is a tiny microtubule-based organelle at the apex of almost all vertebrate cells. It serves as a "cellular antenna" surveying environment outside the cell and transducing this information inside the cell to trigger multiple signaling responses crucial for tissue morphogenesis and homeostasis. Hundreds of proteins and unique cellular mechanisms are involved in cilia formation. Recent evidence suggests that actin remodeling and regulation at the base of the primary cilium strongly impacts ciliogenesis. In this review, we provide an overview of the structure and function of the primary cilium, focusing on the role of actin cytoskeleton and its regulators in ciliogenesis. We then describe the key clinical, genetic, and molecular aspects of renal ciliopathies. We highlight what is known about actin regulation in the pathogenesis of these diseases with the aim to consider these recent molecular findings as potential therapeutic targets for renal ciliopathies.
Collapse
Affiliation(s)
- Rita Kalot
- Department of Medicine and Department of Physiology, McGill University, Montreal, QC, Canada
- The Research Institute of the McGill University Health Center, Montreal, QC, Canada
| | - Zachary Sentell
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Thomas M. Kitzler
- Department of Human Genetics, McGill University, Montreal, QC, Canada
- McGill University Health Center, Montreal, QC, Canada
| | - Elena Torban
- Department of Medicine and Department of Physiology, McGill University, Montreal, QC, Canada
- The Research Institute of the McGill University Health Center, Montreal, QC, Canada
| |
Collapse
|
17
|
Lyu Q, Li Q, Zhou J, Zhao H. Formation and function of multiciliated cells. J Cell Biol 2024; 223:e202307150. [PMID: 38032388 PMCID: PMC10689204 DOI: 10.1083/jcb.202307150] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/29/2023] [Accepted: 11/14/2023] [Indexed: 12/01/2023] Open
Abstract
In vertebrates, multiciliated cells (MCCs) are terminally differentiated cells that line the airway tracts, brain ventricles, and reproductive ducts. Each MCC contains dozens to hundreds of motile cilia that beat in a synchronized manner to drive fluid flow across epithelia, the dysfunction of which is associated with a group of human diseases referred to as motile ciliopathies, such as primary cilia dyskinesia. Given the dynamic and complex process of multiciliogenesis, the biological events essential for forming multiple motile cilia are comparatively unelucidated. Thanks to advancements in genetic tools, omics technologies, and structural biology, significant progress has been achieved in the past decade in understanding the molecular mechanism underlying the regulation of multiple motile cilia formation. In this review, we discuss recent studies with ex vivo culture MCC and animal models, summarize current knowledge of multiciliogenesis, and particularly highlight recent advances and their implications.
Collapse
Affiliation(s)
- Qian Lyu
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Qingchao Li
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Jun Zhou
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
- State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Cell Ecosystem, College of Life Sciences, Nankai University, Tianjin, China
| | - Huijie Zhao
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| |
Collapse
|
18
|
Rayamajhi D, Ege M, Ukhanov K, Ringers C, Zhang Y, Jung I, D’Gama PP, Li SS, Cosacak MI, Kizil C, Park HC, Yaksi E, Martens JR, Brody SL, Jurisch-Yaksi N, Roy S. The forkhead transcription factor Foxj1 controls vertebrate olfactory cilia biogenesis and sensory neuron differentiation. PLoS Biol 2024; 22:e3002468. [PMID: 38271330 PMCID: PMC10810531 DOI: 10.1371/journal.pbio.3002468] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 12/12/2023] [Indexed: 01/27/2024] Open
Abstract
In vertebrates, olfactory receptors localize on multiple cilia elaborated on dendritic knobs of olfactory sensory neurons (OSNs). Although olfactory cilia dysfunction can cause anosmia, how their differentiation is programmed at the transcriptional level has remained largely unexplored. We discovered in zebrafish and mice that Foxj1, a forkhead domain-containing transcription factor traditionally linked with motile cilia biogenesis, is expressed in OSNs and required for olfactory epithelium (OE) formation. In keeping with the immotile nature of olfactory cilia, we observed that ciliary motility genes are repressed in zebrafish, mouse, and human OSNs. Strikingly, we also found that besides ciliogenesis, Foxj1 controls the differentiation of the OSNs themselves by regulating their cell type-specific gene expression, such as that of olfactory marker protein (omp) involved in odor-evoked signal transduction. In line with this, response to bile acids, odors detected by OMP-positive OSNs, was significantly diminished in foxj1 mutant zebrafish. Taken together, our findings establish how the canonical Foxj1-mediated motile ciliogenic transcriptional program has been repurposed for the biogenesis of immotile olfactory cilia, as well as for the development of the OSNs.
Collapse
Affiliation(s)
- Dheeraj Rayamajhi
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | - Mert Ege
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Kirill Ukhanov
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, Florida, United States of America
| | - Christa Ringers
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, Norwegian University of Science and Technology, Trondheim, Norway
| | - Yiliu Zhang
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | - Inyoung Jung
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Biomedical Sciences, Korea University, Ansan, South Korea
| | - Percival P. D’Gama
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Summer Shijia Li
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | - Mehmet Ilyas Cosacak
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Helmholtz Association, Dresden, Germany
| | - Caghan Kizil
- Department of Neurology and The Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Hae-Chul Park
- Department of Biomedical Sciences, Korea University, Ansan, South Korea
| | - Emre Yaksi
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, Norwegian University of Science and Technology, Trondheim, Norway
- Koç University Research Center for Translational Medicine, Koç University School of Medicine, Istanbul, Turkey
| | - Jeffrey R. Martens
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, Florida, United States of America
| | - Steven L. Brody
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Nathalie Jurisch-Yaksi
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, Norwegian University of Science and Technology, Trondheim, Norway
| | - Sudipto Roy
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore
- Department of Paediatrics, National University of Singapore, Singapore
| |
Collapse
|
19
|
Wang S, Wang X, Pan C, Liu Y, Lei M, Guo X, Chen Q, Yang X, Ouyang C, Ren Z. Functions of actin-binding proteins in cilia structure remodeling and signaling. Biol Cell 2023; 115:e202300026. [PMID: 37478133 DOI: 10.1111/boc.202300026] [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: 03/22/2023] [Revised: 07/04/2023] [Accepted: 07/11/2023] [Indexed: 07/23/2023]
Abstract
Cilia are microtubule-based organelles found on the surfaces of many types of cells, including cardiac fibroblasts, vascular endothelial cells, human retinal pigmented epithelial-1 (RPE-1) cells, and alveolar epithelial cells. These organelles can be classified as immotile cilia, referred to as primary cilia in mammalian cells, and motile cilia. Primary cilia are cellular sensors that detect extracellular signals; this is a critical function associated with ciliopathies, which are characterized by the typical clinical features of developmental disorders. Cilia are extensively studied organelles of the microtubule cytoskeleton. However, the ciliary actin cytoskeleton has rarely been studied. Clear evidence has shown that highly regulated actin cytoskeleton dynamics contribute to normal ciliary function. Actin-binding proteins (ABPs) play vital roles in filamentous actin (F-actin) morphology. Here, we discuss recent progress in understanding the roles of ABPs in ciliary structural remodeling and further downstream ciliary signaling with a focus on the molecular mechanisms underlying actin cytoskeleton-related ciliopathies.
Collapse
Affiliation(s)
- Siqi Wang
- Hubei Key Laboratory of Diabetes and Angiopathy, Medicine Research Institute, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Xin Wang
- School of Mathematics and Statistics, Hubei University of Science and Technology, Xianning, China
| | - Congbin Pan
- Hubei Key Laboratory of Diabetes and Angiopathy, Medicine Research Institute, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Ying Liu
- College of Life Sciences, Shandong Provincial Key Laboratory of Animal Resistance Biology, Shandong Normal University, Jinan, China
| | - Min Lei
- Hubei Key Laboratory of Diabetes and Angiopathy, Medicine Research Institute, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Xiying Guo
- Hubei Key Laboratory of Diabetes and Angiopathy, Medicine Research Institute, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Qingjie Chen
- Hubei Key Laboratory of Diabetes and Angiopathy, Medicine Research Institute, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Xiaosong Yang
- Hubei Key Laboratory of Diabetes and Angiopathy, Medicine Research Institute, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Changhan Ouyang
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Zhanhong Ren
- Hubei Key Laboratory of Diabetes and Angiopathy, Medicine Research Institute, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| |
Collapse
|
20
|
Sheridan J, Grata A, Suva EE, Bresteau E, Mitchell LR, Hassan O, Mitchell B. Novel centriolar defects underlie a primary ciliary dyskinesia phenotype in an adenylate kinase 7 deficient ciliated epithelium. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.25.550535. [PMID: 37546962 PMCID: PMC10402086 DOI: 10.1101/2023.07.25.550535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
The skin of Xenopus embryos contains numerous multiciliated cells (MCCs), which collectively generate a directed fluid flow across the epithelial surface essential for distributing the overlaying mucous. MCCs develop into highly specialized cells to generate this flow, containing approximately 150 evenly spaced centrioles that give rise to motile cilia. MCC-driven fluid flow can be impaired when ciliary dysfunction occurs, resulting in primary ciliary dyskinesia (PCD) in humans. Mutations in a large number of genes (~50) have been found to be causative to PCD. Recently, studies have linked low levels of Adenylate Kinase 7 (AK7) gene expression to patients with PCD; however, the mechanism for this link remains unclear. Additionally, AK7 mutations have been linked to multiple PCD patients. Adenylate kinases modulate ATP production and consumption, with AK7 explicitly associated with motile cilia. Here we reproduce an AK7 PCD-like phenotype in Xenopus and describe the cellular consequences that occur with manipulation of AK7 levels. We show that AK7 localizes throughout the cilia in a DPY30 domain-dependent manner, suggesting a ciliary function. Additionally, we find that AK7 overexpression increases centriole number, suggesting a role in regulating centriole biogenesis. We find that in AK7-depleted embryos, cilia number, length, and beat frequency are all reduced, which in turn, significantly decreases the tissue-wide mucociliary flow. Additionally, we find a decrease in centriole number and an increase in sub-apical centrioles, implying that AK7 influences both centriole biogenesis and docking, which we propose underlie its defect in ciliogenesis. We propose that AK7 plays a role in PCD by impacting centriole biogenesis and apical docking, ultimately leading to ciliogenesis defects that impair mucociliary clearance.
Collapse
Affiliation(s)
- Jennifer Sheridan
- Northwestern University, Feinberg School of Medicine, Department of Cell and Developmental Biology
| | - Aline Grata
- Northwestern University, Feinberg School of Medicine, Department of Cell and Developmental Biology
| | - Eve E. Suva
- Northwestern University, Feinberg School of Medicine, Department of Cell and Developmental Biology
| | - Enzo Bresteau
- Northwestern University, Feinberg School of Medicine, Department of Cell and Developmental Biology
| | - Linus R. Mitchell
- Northwestern University, Feinberg School of Medicine, Department of Cell and Developmental Biology
| | - Osama Hassan
- Northwestern University, Feinberg School of Medicine, Department of Cell and Developmental Biology
| | - Brian Mitchell
- Northwestern University, Feinberg School of Medicine, Department of Cell and Developmental Biology
- Northwestern University, Lurie Cancer Center
| |
Collapse
|
21
|
Horani A, Gupta DK, Xu J, Xu H, del Carmen Puga-Molina L, Santi CM, Ramagiri S, Brennan SK, Pan J, Koenitzer JR, Huang T, Hyland RM, Gunsten SP, Tzeng SC, Strahle JM, Mill P, Mahjoub MR, Dutcher SK, Brody SL. The effect of Dnaaf5 gene dosage on primary ciliary dyskinesia phenotypes. JCI Insight 2023; 8:e168836. [PMID: 37104040 PMCID: PMC10393236 DOI: 10.1172/jci.insight.168836] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 04/20/2023] [Indexed: 04/28/2023] Open
Abstract
DNAAF5 is a dynein motor assembly factor associated with the autosomal heterogenic recessive condition of motile cilia, primary ciliary dyskinesia (PCD). The effects of allele heterozygosity on motile cilia function are unknown. We used CRISPR-Cas9 genome editing in mice to recreate a human missense variant identified in patients with mild PCD and a second, frameshift-null deletion in Dnaaf5. Litters with Dnaaf5 heteroallelic variants showed distinct missense and null gene dosage effects. Homozygosity for the null Dnaaf5 alleles was embryonic lethal. Compound heterozygous animals with the missense and null alleles showed severe disease manifesting as hydrocephalus and early lethality. However, animals homozygous for the missense mutation had improved survival, with partially preserved cilia function and motor assembly observed by ultrastructure analysis. Notably, the same variant alleles exhibited divergent cilia function across different multiciliated tissues. Proteomic analysis of isolated airway cilia from mutant mice revealed reduction in some axonemal regulatory and structural proteins not previously reported in DNAAF5 variants. Transcriptional analysis of mouse and human mutant cells showed increased expression of genes coding for axonemal proteins. These findings suggest allele-specific and tissue-specific molecular requirements for cilia motor assembly that may affect disease phenotypes and clinical trajectory in motile ciliopathies.
Collapse
Affiliation(s)
- Amjad Horani
- Department of Pediatrics
- Department of Cell Biology and Physiology
| | | | | | | | | | | | - Sruthi Ramagiri
- Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | | | | | | | | | | | | | - Jennifer M. Strahle
- Department of Neurosurgery, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Pleasantine Mill
- MRC Human Genetics Unit, University of Edinburgh, Edinburgh, United Kingdom
| | - Moe R. Mahjoub
- Department of Cell Biology and Physiology
- Department of Medicine
| | - Susan K. Dutcher
- Department of Cell Biology and Physiology
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, USA
| | | |
Collapse
|
22
|
Safavian D, Kim MS, Xie H, El-Zeiry M, Palander O, Dai L, Collins RF, Froese C, Shannon R, Nagata KI, Trimble WS. Septin-mediated RhoA activation engages the exocyst complex to recruit the cilium transition zone. J Cell Biol 2023; 222:e201911062. [PMID: 36912772 PMCID: PMC10039714 DOI: 10.1083/jcb.201911062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 01/25/2022] [Accepted: 01/05/2023] [Indexed: 03/14/2023] Open
Abstract
Septins are filamentous GTPases that play important but poorly characterized roles in ciliogenesis. Here, we show that SEPTIN9 regulates RhoA signaling at the base of cilia by binding and activating the RhoA guanine nucleotide exchange factor, ARHGEF18. GTP-RhoA is known to activate the membrane targeting exocyst complex, and suppression of SEPTIN9 causes disruption of ciliogenesis and mislocalization of an exocyst subunit, SEC8. Using basal body-targeted proteins, we show that upregulating RhoA signaling at the cilium can rescue ciliary defects and mislocalization of SEC8 caused by global SEPTIN9 depletion. Moreover, we demonstrate that the transition zone components, RPGRIP1L and TCTN2, fail to accumulate at the transition zone in cells lacking SEPTIN9 or depleted of the exocyst complex. Thus, SEPTIN9 regulates the recruitment of transition zone proteins on Golgi-derived vesicles by activating the exocyst via RhoA to allow the formation of primary cilia.
Collapse
Affiliation(s)
- Darya Safavian
- Cell Biology Program, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Moshe S. Kim
- Cell Biology Program, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Hong Xie
- Cell Biology Program, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Maha El-Zeiry
- Cell Biology Program, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Oliva Palander
- Cell Biology Program, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Lu Dai
- Cell Biology Program, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Richard F. Collins
- Cell Biology Program, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Carol Froese
- Cell Biology Program, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Rachel Shannon
- Cell Biology Program, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Koh-ichi Nagata
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Human Service Center, Kasugai, Aichi, Japan
| | - William S. Trimble
- Cell Biology Program, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
23
|
Konjikusic MJ, Lee C, Yue Y, Shrestha BD, Nguimtsop AM, Horani A, Brody S, Prakash VN, Gray RS, Verhey KJ, Wallingford JB. Kif9 is an active kinesin motor required for ciliary beating and proximodistal patterning of motile axonemes. J Cell Sci 2023; 136:jcs259535. [PMID: 35531639 PMCID: PMC9357393 DOI: 10.1242/jcs.259535] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 04/27/2022] [Indexed: 03/19/2024] Open
Abstract
Most motile cilia have a stereotyped structure of nine microtubule outer doublets and a single central pair of microtubules. The central pair of microtubules are surrounded by a set of proteins, termed the central pair apparatus. A specific kinesin, Klp1 projects from the central pair and contributes to ciliary motility in Chlamydomonas. The vertebrate ortholog, Kif9, is required for beating in mouse sperm flagella, but the mechanism of Kif9/Klp1 function remains poorly defined. Here, using Xenopus epidermal multiciliated cells, we show that Kif9 is necessary for ciliary motility and the proper distal localization of not only central pair proteins, but also radial spokes and dynein arms. In addition, single-molecule assays in vitro reveal that Xenopus Kif9 is a long-range processive motor, although it does not mediate long-range movement in ciliary axonemes in vivo. Together, our data suggest that Kif9 is integral for ciliary beating and is necessary for proper axonemal distal end integrity.
Collapse
Affiliation(s)
- Mia J. Konjikusic
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
- Department of Pediatrics, Dell Pediatric Research Institute, 1400 Barbara Jordan Blvd, The University of Texas at Austin, Dell Medical School, Austin, TX 78712, USA
- Department of Nutritional Sciences, 200 W 24th Street, The University of Texas at Austin, Austin, TX 78712, USA
| | - Chanjae Lee
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Yang Yue
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | | | - Ange M. Nguimtsop
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Amjad Horani
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63130, USA
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Steven Brody
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Vivek N. Prakash
- Department of Physics, University of Miami, Coral Gables, FL 33146, USA
- Department of Biology and Department of Marine Biology and Ecology, University of Miami, Coral Gables, FL 33146,USA
| | - Ryan S. Gray
- Department of Pediatrics, Dell Pediatric Research Institute, 1400 Barbara Jordan Blvd, The University of Texas at Austin, Dell Medical School, Austin, TX 78712, USA
- Department of Nutritional Sciences, 200 W 24th Street, The University of Texas at Austin, Austin, TX 78712, USA
| | - Kristen J. Verhey
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - John B. Wallingford
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
24
|
Zhao H, Khan Z, Westlake CJ. Ciliogenesis membrane dynamics and organization. Semin Cell Dev Biol 2023; 133:20-31. [PMID: 35351373 PMCID: PMC9510604 DOI: 10.1016/j.semcdb.2022.03.021] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 12/28/2022]
Abstract
Ciliogenesis is a complex multistep process used to describe assembly of cilia and flagella. These organelles play essential roles in motility and signaling on the surface of cells. Cilia are built at the distal ends of centrioles through the formation of an axoneme that is surrounded by the ciliary membrane. As is the case in the biogenesis of other cellular organelles, regulators of membrane trafficking play essential roles in ciliogenesis, albeit with a unique feature that membranes are organized around microtubule-based structures. Membrane association with the distal end of the centriole is a critical initiating step for ciliogenesis. Studies of this process in different cell types suggests that a singular mechanism may not be utilized to initiate cilium assembly. In this review, we focus on recent insights into cilium biogenesis and the roles membrane trafficking regulators play in described ciliogenesis mechanisms with relevance to human disease.
Collapse
Affiliation(s)
- Huijie Zhao
- Center for Cancer Research, NCI Frederick, Laboratory of Cellular and Developmental, Signaling, Frederick, MD 21702, USA
| | - Ziam Khan
- Center for Cancer Research, NCI Frederick, Laboratory of Cellular and Developmental, Signaling, Frederick, MD 21702, USA
| | - Christopher J Westlake
- Center for Cancer Research, NCI Frederick, Laboratory of Cellular and Developmental, Signaling, Frederick, MD 21702, USA.
| |
Collapse
|
25
|
Horani A, Gupta DK, Xu J, Xu H, Del Carmen Puga-Molina L, Santi CM, Ramagiri S, Brennen SK, Pan J, Huang T, Hyland RM, Gunsten SP, Tzeng SC, Strahle JM, Mill P, Mahjoub MR, Dutcher SK, Brody SL. The effect of Dnaaf5 gene dosage on primary ciliary dyskinesia phenotypes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.13.523966. [PMID: 36712068 PMCID: PMC9882222 DOI: 10.1101/2023.01.13.523966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
DNAAF5 is a dynein motor assembly factor associated with the autosomal heterogenic recessive condition of motile cilia, primary ciliary dyskinesia (PCD). The effects of allele heterozygosity on motile cilia function are unknown. We used CRISPR-Cas9 genome editing in mice to recreate a human missense variant identified in patients with mild PCD and a second, frameshift null deletion in Dnaaf5 . Litters with Dnaaf5 heteroallelic variants showed distinct missense and null gene dosage effects. Homozygosity for the null Dnaaf5 alleles was embryonic lethal. Compound heterozygous animals with the missense and null alleles showed severe disease manifesting as hydrocephalus and early lethality. However, animals homozygous for the missense mutation had improved survival, with partial preserved cilia function and motor assembly observed by ultrastructure analysis. Notably, the same variant alleles exhibited divergent cilia function across different multiciliated tissues. Proteomic analysis of isolated airway cilia from mutant mice revealed reduction in some axonemal regulatory and structural proteins not previously reported in DNAAF5 variants. While transcriptional analysis of mouse and human mutant cells showed increased expression of genes coding for axonemal proteins. Together, these findings suggest allele-specific and tissue-specific molecular requirements for cilia motor assembly that may affect disease phenotypes and clinical trajectory in motile ciliopathies. Brief Summary A mouse model of human DNAAF5 primary ciliary dyskinesia variants reveals gene dosage effects of mutant alleles and tissue-specific molecular requirements for cilia motor assembly.
Collapse
|
26
|
Amack JD. Structures and functions of cilia during vertebrate embryo development. Mol Reprod Dev 2022; 89:579-596. [PMID: 36367893 PMCID: PMC9805515 DOI: 10.1002/mrd.23650] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/05/2022] [Accepted: 10/28/2022] [Indexed: 11/13/2022]
Abstract
Cilia are hair-like structures that project from the surface of cells. In vertebrates, most cells have an immotile primary cilium that mediates cell signaling, and some specialized cells assemble one or multiple cilia that are motile and beat synchronously to move fluids in one direction. Gene mutations that alter cilia structure or function cause a broad spectrum of disorders termed ciliopathies that impact virtually every system in the body. A wide range of birth defects associated with ciliopathies underscores critical functions for cilia during embryonic development. In many cases, the mechanisms underlying cilia functions during development and disease remain poorly understood. This review describes different types of cilia in vertebrate embryos and discusses recent research results from diverse model systems that provide novel insights into how cilia form and function during embryo development. The work discussed here not only expands our understanding of in vivo cilia biology, but also opens new questions about cilia and their roles in establishing healthy embryos.
Collapse
Affiliation(s)
- Jeffrey D. Amack
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, New York, USA,,BioInspired Syracuse: Institute for Material and Living Systems, Syracuse, New York, USA
| |
Collapse
|
27
|
Mahjoub MR, Nanjundappa R, Harvey MN. Development of a multiciliated cell. Curr Opin Cell Biol 2022; 77:102105. [PMID: 35716530 DOI: 10.1016/j.ceb.2022.102105] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 05/09/2022] [Accepted: 05/17/2022] [Indexed: 11/30/2022]
Abstract
Multiciliated cells (MCC) are evolutionary conserved, highly specialized cell types that contain dozens to hundreds of motile cilia that they use to propel fluid directionally. To template these cilia, each MCC produces between 30 and 500 basal bodies via a process termed centriole amplification. Much progress has been made in recent years in understanding the pathways involved in MCC fate determination, differentiation, and ciliogenesis. Recent studies using mammalian cell culture systems, mice, Xenopus, and other model organisms have started to uncover the mechanisms involved in centriole and cilia biogenesis. Yet, how MCC progenitor cells regulate the precise number of centrioles and cilia during their differentiation remains largely unknown. In this review, we will examine recent findings that address this fundamental question.
Collapse
Affiliation(s)
- Moe R Mahjoub
- Department of Medicine (Nephrology Division), Washington University, St Louis, MO, USA; Department of Cell Biology and Physiology, Washington University, St Louis, MO, USA.
| | - Rashmi Nanjundappa
- Department of Medicine (Nephrology Division), Washington University, St Louis, MO, USA
| | - Megan N Harvey
- Department of Medicine (Nephrology Division), Washington University, St Louis, MO, USA
| |
Collapse
|
28
|
Abstract
Primary cilia play a key role in the ability of cells to respond to extracellular stimuli, such as signaling molecules and environmental cues. These sensory organelles are crucial to the development of many organ systems, and defects in primary ciliogenesis lead to multisystemic genetic disorders, known as ciliopathies. Here, we review recent advances in the understanding of several key aspects of the regulation of ciliogenesis. Primary ciliogenesis is thought to take different pathways depending on cell type, and some recent studies shed new light on the cell-type-specific mechanisms regulating ciliogenesis at the apical surface in polarized epithelial cells, which are particularly relevant for many ciliopathies. Furthermore, recent findings have demonstrated the importance of actin cytoskeleton dynamics in positively and negatively regulating multiple stages of ciliogenesis, including the vesicular trafficking of ciliary components and the positioning and docking of the basal body. Finally, studies on the formation of motile cilia in multiciliated epithelial cells have revealed requirements for actin remodeling in this process too, as well as showing evidence of an additional alternative ciliogenesis pathway.
Collapse
Affiliation(s)
| | - Rytis Prekeris
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
29
|
Yasunaga T, Wiegel J, Bergen MD, Helmstädter M, Epting D, Paolini A, Çiçek Ö, Radziwill G, Engel C, Brox T, Ronneberger O, Walentek P, Ulbrich MH, Walz G. Microridge-like structures anchor motile cilia. Nat Commun 2022; 13:2056. [PMID: 35440631 PMCID: PMC9018822 DOI: 10.1038/s41467-022-29741-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 03/30/2022] [Indexed: 01/11/2023] Open
Abstract
Several tissues contain cells with multiple motile cilia that generate a fluid or particle flow to support development and organ functions; defective motility causes human disease. Developmental cues orient motile cilia, but how cilia are locked into their final position to maintain a directional flow is not understood. Here we find that the actin cytoskeleton is highly dynamic during early development of multiciliated cells (MCCs). While apical actin bundles become increasingly more static, subapical actin filaments are nucleated from the distal tip of ciliary rootlets. Anchorage of these subapical actin filaments requires the presence of microridge-like structures formed during MCC development, and the activity of Nonmuscle Myosin II. Optogenetic manipulation of Ezrin, a core component of the microridge actin-anchoring complex, or inhibition of Myosin Light Chain Kinase interfere with rootlet anchorage and orientation. These observations identify microridge-like structures as an essential component of basal body rootlet anchoring in MCCs.
Collapse
Affiliation(s)
- Takayuki Yasunaga
- Department of Medicine IV, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106, Freiburg, Germany
| | - Johannes Wiegel
- Department of Medicine IV, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106, Freiburg, Germany
| | - Max D Bergen
- Department of Medicine IV, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106, Freiburg, Germany
| | - Martin Helmstädter
- Department of Medicine IV, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106, Freiburg, Germany
| | - Daniel Epting
- Department of Medicine IV, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106, Freiburg, Germany
| | - Andrea Paolini
- Department of Medicine IV, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Schaenzlestrasse 1, 79104, Freiburg, Germany
| | - Özgün Çiçek
- Pattern Recognition and Image Processing, Department of Computer Science, University of Freiburg, Georges-Köhler-Allee 52, 79110, Freiburg, Germany
- BIOSS Centre for Biological Signalling Studies, University of Freiburg, Schänzlestrasse 18, 79104, Freiburg, Germany
| | - Gerald Radziwill
- BIOSS Centre for Biological Signalling Studies, University of Freiburg, Schänzlestrasse 18, 79104, Freiburg, Germany
- CIBSS Centre for Integrative Biological Signalling Studies, University of Freiburg, Schänzlestrasse 18, 79104, Freiburg, Germany
| | - Christina Engel
- Department of Medicine IV, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106, Freiburg, Germany
| | - Thomas Brox
- Pattern Recognition and Image Processing, Department of Computer Science, University of Freiburg, Georges-Köhler-Allee 52, 79110, Freiburg, Germany
- BIOSS Centre for Biological Signalling Studies, University of Freiburg, Schänzlestrasse 18, 79104, Freiburg, Germany
- CIBSS Centre for Integrative Biological Signalling Studies, University of Freiburg, Schänzlestrasse 18, 79104, Freiburg, Germany
| | - Olaf Ronneberger
- Pattern Recognition and Image Processing, Department of Computer Science, University of Freiburg, Georges-Köhler-Allee 52, 79110, Freiburg, Germany
- BIOSS Centre for Biological Signalling Studies, University of Freiburg, Schänzlestrasse 18, 79104, Freiburg, Germany
| | - Peter Walentek
- Department of Medicine IV, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106, Freiburg, Germany
- CIBSS Centre for Integrative Biological Signalling Studies, University of Freiburg, Schänzlestrasse 18, 79104, Freiburg, Germany
| | - Maximilian H Ulbrich
- Department of Medicine IV, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106, Freiburg, Germany
- BIOSS Centre for Biological Signalling Studies, University of Freiburg, Schänzlestrasse 18, 79104, Freiburg, Germany
| | - Gerd Walz
- Department of Medicine IV, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106, Freiburg, Germany.
- BIOSS Centre for Biological Signalling Studies, University of Freiburg, Schänzlestrasse 18, 79104, Freiburg, Germany.
- CIBSS Centre for Integrative Biological Signalling Studies, University of Freiburg, Schänzlestrasse 18, 79104, Freiburg, Germany.
| |
Collapse
|
30
|
Nommick A, Boutin C, Rosnet O, Schirmer C, Bazellières E, Thomé V, Loiseau E, Viallat A, Kodjabachian L. Lrrcc1 and Ccdc61 are conserved effectors of multiciliated cell function. J Cell Sci 2022; 135:274401. [DOI: 10.1242/jcs.258960] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 01/05/2022] [Indexed: 11/20/2022] Open
Abstract
Ciliated epithelia perform essential functions across animal evolution, ranging from locomotion of marine organisms to mucociliary clearance of airways in mammals. These epithelia are composed of multiciliated cells (MCCs) harbouring myriads of motile cilia, which rest on modified centrioles called basal bodies (BBs), and beat coordinately to generate directed fluid flows. Thus, BB biogenesis and organization is central to MCC function. In basal eukaryotes, the coiled-coil domain proteins Lrrcc1 and Ccdc61 were shown to be required for proper BB construction and function. Here, we used the Xenopus embryonic ciliated epidermis to characterize Lrrcc1 and Ccdc61 in vertebrate MCCs. We found that they both encode BB components, localized proximally at the junction with striated rootlets. Knocking down either gene caused defects in BB docking, spacing, and polarization. Moreover, their depletion impaired the apical cytoskeleton, and altered ciliary beating. Consequently, cilia-powered fluid flow was greatly reduced in morphant tadpoles, which displayed enhanced mortality when exposed to pathogenic bacteria. This work illustrates how integration across organizational scales make elementary BB components essential for the emergence of the physiological function of ciliated epithelia.
Collapse
Affiliation(s)
- Aude Nommick
- Aix Marseille Univ, CNRS, IBDM, Turing Center for Living Systems, Marseille, France
| | - Camille Boutin
- Aix Marseille Univ, CNRS, IBDM, Turing Center for Living Systems, Marseille, France
| | - Olivier Rosnet
- Aix Marseille Univ, CNRS, IBDM, Turing Center for Living Systems, Marseille, France
| | - Claire Schirmer
- Aix Marseille Univ, CNRS, IBDM, Turing Center for Living Systems, Marseille, France
| | - Elsa Bazellières
- Aix Marseille Univ, CNRS, IBDM, Turing Center for Living Systems, Marseille, France
| | - Virginie Thomé
- Aix Marseille Univ, CNRS, IBDM, Turing Center for Living Systems, Marseille, France
| | - Etienne Loiseau
- Aix Marseille Univ, CNRS, CINaM, Turing Center for Living Systems, Marseille, France
| | - Annie Viallat
- Aix Marseille Univ, CNRS, CINaM, Turing Center for Living Systems, Marseille, France
| | - Laurent Kodjabachian
- Aix Marseille Univ, CNRS, IBDM, Turing Center for Living Systems, Marseille, France
| |
Collapse
|
31
|
Pei J, Beri NR, Zou AJ, Hubel P, Dorando HK, Bergant V, Andrews RD, Pan J, Andrews JM, Sheehan KCF, Pichlmair A, Amarasinghe GK, Brody SL, Payton JE, Leung DW. Nuclear-localized human respiratory syncytial virus NS1 protein modulates host gene transcription. Cell Rep 2021; 37:109803. [PMID: 34644581 PMCID: PMC8609347 DOI: 10.1016/j.celrep.2021.109803] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 04/28/2021] [Accepted: 09/16/2021] [Indexed: 12/13/2022] Open
Abstract
Human respiratory syncytial virus (RSV) is a common cause of lower respiratory tract infections in the pediatric, elderly, and immunocompromised individuals. RSV non-structural protein NS1 is a known cytosolic immune antagonist, but how NS1 modulates host responses remains poorly defined. Here, we observe NS1 partitioning into the nucleus of RSV-infected cells, including the human airway epithelium. Nuclear NS1 coimmunoprecipitates with Mediator complex and is chromatin associated. Chromatin-immunoprecipitation demonstrates enrichment of NS1 that overlaps Mediator and transcription factor binding within the promoters and enhancers of differentially expressed genes during RSV infection. Mutation of the NS1 C-terminal helix reduces NS1 impact on host gene expression. These data suggest that nuclear NS1 alters host responses to RSV infection by binding at regulatory elements of immune response genes and modulating host gene transcription. Our study identifies another layer of regulation by virally encoded proteins that shapes host response and impacts immunity to RSV.
Collapse
Affiliation(s)
- Jingjing Pei
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Nina R Beri
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Angela J Zou
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Philipp Hubel
- Innate Immunity Laboratory, Max-Planck Institute of Biochemistry, Martinsried/Munich 82152, Germany
| | - Hannah K Dorando
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Valter Bergant
- Institute for Virology, Technical University of Munich, School of Medicine, 81675 Munich, Germany
| | - Rebecca D Andrews
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jiehong Pan
- Department of Medicine, Division of Pulmonary and Critical Care, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jared M Andrews
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Kathleen C F Sheehan
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Andreas Pichlmair
- Innate Immunity Laboratory, Max-Planck Institute of Biochemistry, Martinsried/Munich 82152, Germany; Institute for Virology, Technical University of Munich, School of Medicine, 81675 Munich, Germany
| | - Gaya K Amarasinghe
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Steven L Brody
- Department of Medicine, Division of Pulmonary and Critical Care, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jacqueline E Payton
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Daisy W Leung
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
32
|
Mecklenburg N, Kowalczyk I, Witte F, Görne J, Laier A, Mamo TM, Gonschior H, Lehmann M, Richter M, Sporbert A, Purfürst B, Hübner N, Hammes A. Identification of disease-relevant modulators of the SHH pathway in the developing brain. Development 2021; 148:272000. [PMID: 34463328 DOI: 10.1242/dev.199307] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 07/19/2021] [Indexed: 12/13/2022]
Abstract
Pathogenic gene variants in humans that affect the sonic hedgehog (SHH) pathway lead to severe brain malformations with variable penetrance due to unknown modifier genes. To identify such modifiers, we established novel congenic mouse models. LRP2-deficient C57BL/6N mice suffer from heart outflow tract defects and holoprosencephaly caused by impaired SHH activity. These defects are fully rescued on a FVB/N background, indicating a strong influence of modifier genes. Applying comparative transcriptomics, we identified Pttg1 and Ulk4 as candidate modifiers upregulated in the rescue strain. Functional analyses showed that ULK4 and PTTG1, both microtubule-associated proteins, are positive regulators of SHH signaling, rendering the pathway more resilient to disturbances. In addition, we characterized ULK4 and PTTG1 as previously unidentified components of primary cilia in the neuroepithelium. The identification of genes that powerfully modulate the penetrance of genetic disturbances affecting the brain and heart is likely relevant to understanding the variability in human congenital disorders.
Collapse
Affiliation(s)
- Nora Mecklenburg
- Disorders of the Nervous System, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Izabela Kowalczyk
- Disorders of the Nervous System, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Franziska Witte
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Jessica Görne
- Disorders of the Nervous System, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Alena Laier
- Disorders of the Nervous System, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Tamrat M Mamo
- Disorders of the Nervous System, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Hannes Gonschior
- Cellular Imaging, Light Microscopy, Leibniz-Research Institute for Molecular Pharmacology (FMP), 13125 Berlin, Germany
| | - Martin Lehmann
- Cellular Imaging, Light Microscopy, Leibniz-Research Institute for Molecular Pharmacology (FMP), 13125 Berlin, Germany
| | - Matthias Richter
- Advanced Light Microscopy Technology Platform, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Anje Sporbert
- Advanced Light Microscopy Technology Platform, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Bettina Purfürst
- Electron microscopy technology platform, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| | - Norbert Hübner
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Berlin, 10785 Berlin, Germany.,Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany.,Berlin Institute of Health (BIH), 10178 Berlin, Germany
| | - Annette Hammes
- Disorders of the Nervous System, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), 13125 Berlin, Germany
| |
Collapse
|
33
|
Mikhailik A, Michurina TV, Dikranian K, Hearn S, Maxakov VI, Siller SS, Takemaru KI, Enikolopov G, Peunova N. nNOS regulates ciliated cell polarity, ciliary beat frequency, and directional flow in mouse trachea. Life Sci Alliance 2021; 4:4/5/e202000981. [PMID: 33653689 PMCID: PMC8008965 DOI: 10.26508/lsa.202000981] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 11/24/2022] Open
Abstract
Clearance of the airway is dependent on directional mucus flow across the mucociliary epithelium, and deficient flow is implicated in a range of human disorders. Efficient flow relies on proper polarization of the multiciliated cells and sufficient ciliary beat frequency. We show that NO, produced by nNOS in the multiciliated cells of the mouse trachea, controls both the planar polarity and the ciliary beat frequency and is thereby necessary for the generation of the robust flow. The effect of nNOS on the polarity of ciliated cells relies on its interactions with the apical networks of actin and microtubules and involves RhoA activation. The action of nNOS on the beat frequency is mediated by guanylate cyclase; both NO donors and cGMP can augment fluid flow in the trachea and rescue the deficient flow in nNOS mutants. Our results link insufficient availability of NO in ciliated cells to defects in flow and ciliary activity and may thereby explain the low levels of exhaled NO in ciliopathies.
Collapse
Affiliation(s)
- Anatoly Mikhailik
- Center for Developmental Genetics, Stony Brook University, Stony Brook, NY, USA.,Department of Anesthesiology, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Tatyana V Michurina
- Center for Developmental Genetics, Stony Brook University, Stony Brook, NY, USA.,Department of Anesthesiology, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Krikor Dikranian
- Department of Neuroscience, Washington University, St. Louis, MO, USA
| | - Stephen Hearn
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Vladimir I Maxakov
- Center for Developmental Genetics, Stony Brook University, Stony Brook, NY, USA.,Department of Anesthesiology, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Saul S Siller
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, USA
| | - Ken-Ichi Takemaru
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, USA
| | - Grigori Enikolopov
- Center for Developmental Genetics, Stony Brook University, Stony Brook, NY, USA.,Department of Anesthesiology, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| | - Natalia Peunova
- Center for Developmental Genetics, Stony Brook University, Stony Brook, NY, USA .,Department of Anesthesiology, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY, USA
| |
Collapse
|
34
|
Lewis M, Stracker TH. Transcriptional regulation of multiciliated cell differentiation. Semin Cell Dev Biol 2021; 110:51-60. [DOI: 10.1016/j.semcdb.2020.04.007] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/25/2020] [Accepted: 04/13/2020] [Indexed: 01/01/2023]
|
35
|
Hawkins FJ, Suzuki S, Beermann ML, Barillà C, Wang R, Villacorta-Martin C, Berical A, Jean JC, Le Suer J, Matte T, Simone-Roach C, Tang Y, Schlaeger TM, Crane AM, Matthias N, Huang SXL, Randell SH, Wu J, Spence JR, Carraro G, Stripp BR, Rab A, Sorsher EJ, Horani A, Brody SL, Davis BR, Kotton DN. Derivation of Airway Basal Stem Cells from Human Pluripotent Stem Cells. Cell Stem Cell 2021; 28:79-95.e8. [PMID: 33098807 PMCID: PMC7796997 DOI: 10.1016/j.stem.2020.09.017] [Citation(s) in RCA: 137] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 08/03/2020] [Accepted: 09/26/2020] [Indexed: 12/26/2022]
Abstract
The derivation of tissue-specific stem cells from human induced pluripotent stem cells (iPSCs) would have broad reaching implications for regenerative medicine. Here, we report the directed differentiation of human iPSCs into airway basal cells ("iBCs"), a population resembling the stem cell of the airway epithelium. Using a dual fluorescent reporter system (NKX2-1GFP;TP63tdTomato), we track and purify these cells as they first emerge as developmentally immature NKX2-1GFP+ lung progenitors and subsequently augment a TP63 program during proximal airway epithelial patterning. In response to primary basal cell medium, NKX2-1GFP+/TP63tdTomato+ cells display the molecular and functional phenotype of airway basal cells, including the capacity to self-renew or undergo multi-lineage differentiation in vitro and in tracheal xenografts in vivo. iBCs and their differentiated progeny model perturbations that characterize acquired and genetic airway diseases, including the mucus metaplasia of asthma, chloride channel dysfunction of cystic fibrosis, and ciliary defects of primary ciliary dyskinesia.
Collapse
Affiliation(s)
- Finn J Hawkins
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA; The Pulmonary Center and Department of Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA
| | - Shingo Suzuki
- Center for Stem Cell and Regenerative Medicine, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Mary Lou Beermann
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA
| | - Cristina Barillà
- Center for Stem Cell and Regenerative Medicine, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Ruobing Wang
- Pulmonary and Respiratory Diseases, Boston Children's Hospital, Boston, MA 02115, USA
| | - Carlos Villacorta-Martin
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA
| | - Andrew Berical
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA; The Pulmonary Center and Department of Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA
| | - J C Jean
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA
| | - Jake Le Suer
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA; The Pulmonary Center and Department of Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA
| | - Taylor Matte
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA
| | | | - Yang Tang
- Boston Children's Hospital Stem Cell Program, Boston, MA 02115, USA
| | - Thorsten M Schlaeger
- Boston Children's Hospital Stem Cell Program, Boston, MA 02115, USA; Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Ana M Crane
- Center for Stem Cell and Regenerative Medicine, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Nadine Matthias
- Center for Stem Cell and Regenerative Medicine, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Sarah X L Huang
- Center for Stem Cell and Regenerative Medicine, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Scott H Randell
- Marsico Lung Institute and Cystic Fibrosis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Joshua Wu
- Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Jason R Spence
- Department of Internal Medicine, Gastroenterology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, MI 48109, USA
| | - Gianni Carraro
- Department of Medicine, Lung and Regenerative Medicine Institutes, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Barry R Stripp
- Department of Medicine, Lung and Regenerative Medicine Institutes, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Andras Rab
- Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Eric J Sorsher
- Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Amjad Horani
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Steven L Brody
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Brian R Davis
- Center for Stem Cell and Regenerative Medicine, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX 77030, USA.
| | - Darrell N Kotton
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA; The Pulmonary Center and Department of Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA.
| |
Collapse
|
36
|
Smith CEL, Lake AVR, Johnson CA. Primary Cilia, Ciliogenesis and the Actin Cytoskeleton: A Little Less Resorption, A Little More Actin Please. Front Cell Dev Biol 2020; 8:622822. [PMID: 33392209 PMCID: PMC7773788 DOI: 10.3389/fcell.2020.622822] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 11/24/2020] [Indexed: 12/13/2022] Open
Abstract
Primary cilia are microtubule-based organelles that extend from the apical surface of most mammalian cells, forming when the basal body (derived from the mother centriole) docks at the apical cell membrane. They act as universal cellular "antennae" in vertebrates that receive and integrate mechanical and chemical signals from the extracellular environment, serving diverse roles in chemo-, mechano- and photo-sensation that control developmental signaling, cell polarity and cell proliferation. Mutations in ciliary genes cause a major group of inherited developmental disorders called ciliopathies. There are very few preventative treatments or new therapeutic interventions that modify disease progression or the long-term outlook of patients with these conditions. Recent work has identified at least four distinct but interrelated cellular processes that regulate cilia formation and maintenance, comprising the cell cycle, cellular proteostasis, signaling pathways and structural influences of the actin cytoskeleton. The actin cytoskeleton is composed of microfilaments that are formed from filamentous (F) polymers of globular G-actin subunits. Actin filaments are organized into bundles and networks, and are attached to the cell membrane, by diverse cross-linking proteins. During cell migration, actin filament bundles form either radially at the leading edge or as axial stress fibers. Early studies demonstrated that loss-of-function mutations in ciliopathy genes increased stress fiber formation and impaired ciliogenesis whereas pharmacological inhibition of actin polymerization promoted ciliogenesis. These studies suggest that polymerization of the actin cytoskeleton, F-actin branching and the formation of stress fibers all inhibit primary cilium formation, whereas depolymerization or depletion of actin enhance ciliogenesis. Here, we review the mechanistic basis for these effects on ciliogenesis, which comprise several cellular processes acting in concert at different timescales. Actin polymerization is both a physical barrier to both cilia-targeted vesicle transport and to the membrane remodeling required for ciliogenesis. In contrast, actin may cause cilia loss by localizing disassembly factors at the ciliary base, and F-actin branching may itself activate the YAP/TAZ pathway to promote cilia disassembly. The fundamental role of actin polymerization in the control of ciliogenesis may present potential new targets for disease-modifying therapeutic approaches in treating ciliopathies.
Collapse
Affiliation(s)
| | | | - Colin A. Johnson
- Leeds Institute of Medical Research at St. James’s, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
37
|
Lee C, Cox RM, Papoulas O, Horani A, Drew K, Devitt CC, Brody SL, Marcotte EM, Wallingford JB. Functional partitioning of a liquid-like organelle during assembly of axonemal dyneins. eLife 2020; 9:e58662. [PMID: 33263282 PMCID: PMC7785291 DOI: 10.7554/elife.58662] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 12/01/2020] [Indexed: 12/20/2022] Open
Abstract
Ciliary motility is driven by axonemal dyneins that are assembled in the cytoplasm before deployment to cilia. Motile ciliopathy can result from defects in the dyneins themselves or from defects in factors required for their cytoplasmic pre-assembly. Recent work demonstrates that axonemal dyneins, their specific assembly factors, and broadly-acting chaperones are concentrated in liquid-like organelles in the cytoplasm called DynAPs (Dynein Axonemal Particles). Here, we use in vivo imaging in Xenopus to show that inner dynein arm (IDA) and outer dynein arm (ODA) subunits are partitioned into non-overlapping sub-regions within DynAPs. Using affinity- purification mass-spectrometry of in vivo interaction partners, we also identify novel partners for inner and outer dynein arms. Among these, we identify C16orf71/Daap1 as a novel axonemal dynein regulator. Daap1 interacts with ODA subunits, localizes specifically to the cytoplasm, is enriched in DynAPs, and is required for the deployment of ODAs to axonemes. Our work reveals a new complexity in the structure and function of a cell-type specific liquid-like organelle that is directly relevant to human genetic disease.
Collapse
Affiliation(s)
- Chanjae Lee
- Department of Molecular Biosciences, University of TexasAustinUnited States
| | - Rachael M Cox
- Department of Molecular Biosciences, University of TexasAustinUnited States
| | - Ophelia Papoulas
- Department of Molecular Biosciences, University of TexasAustinUnited States
| | - Amjad Horani
- Department of Pediatrics, Washington University School of MedicineSt. LouisUnited States
| | - Kevin Drew
- Department of Molecular Biosciences, University of TexasAustinUnited States
| | - Caitlin C Devitt
- Department of Molecular Biosciences, University of TexasAustinUnited States
| | - Steven L Brody
- Department of Medicine, Washington University School of MedicineSt. LouisUnited States
| | - Edward M Marcotte
- Department of Molecular Biosciences, University of TexasAustinUnited States
| | - John B Wallingford
- Department of Molecular Biosciences, University of TexasAustinUnited States
| |
Collapse
|
38
|
Brücker L, Kretschmer V, May-Simera HL. The entangled relationship between cilia and actin. Int J Biochem Cell Biol 2020; 129:105877. [PMID: 33166678 DOI: 10.1016/j.biocel.2020.105877] [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: 08/25/2020] [Revised: 10/23/2020] [Accepted: 10/26/2020] [Indexed: 12/14/2022]
Abstract
Primary cilia are microtubule-based sensory cell organelles that are vital for tissue and organ development. They act as an antenna, receiving and transducing signals, enabling communication between cells. Defects in ciliogenesis result in severe genetic disorders collectively termed ciliopathies. In recent years, the importance of the direct and indirect involvement of actin regulators in ciliogenesis came into focus as it was shown that F-actin polymerisation impacts ciliation. The ciliary basal body was further identified as both a microtubule and actin organising centre. In the current review, we summarize recent studies on F-actin in and around primary cilia, focusing on different actin regulators and their effect on ciliogenesis, from the initial steps of basal body positioning and regulation of ciliary assembly and disassembly. Since primary cilia are also involved in several intracellular signalling pathways such as planar cell polarity (PCP), subsequently affecting actin rearrangements, the multiple effectors of this pathway are highlighted in more detail with a focus on the feedback loops connecting actin networks and cilia proteins. Finally, we elucidate the role of actin regulators in the development of ciliopathy symptoms and cancer.
Collapse
Affiliation(s)
- Lena Brücker
- Cilia Cell Biology, Institute of Molecular Physiology, Johannes-Gutenberg University, Mainz, Germany
| | - Viola Kretschmer
- Cilia Cell Biology, Institute of Molecular Physiology, Johannes-Gutenberg University, Mainz, Germany
| | - Helen Louise May-Simera
- Cilia Cell Biology, Institute of Molecular Physiology, Johannes-Gutenberg University, Mainz, Germany.
| |
Collapse
|
39
|
Palander O, Trimble WS. DIAPH1 regulates ciliogenesis and trafficking in primary cilia. FASEB J 2020; 34:16516-16535. [PMID: 33124112 DOI: 10.1096/fj.202001178r] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 09/08/2020] [Accepted: 10/12/2020] [Indexed: 01/29/2023]
Abstract
Primary cilia are critical hubs for several signaling pathways, and defects in ciliogenesis or cilia maintenance produce a range of diseases collectively known as ciliopathies. Ciliogenesis requires vesicle trafficking along a network of microtubules and actin filaments to the basal body. The DIAPH1 (Diaphanous-related formin) family of formins promotes both actin polymerization and EB1-dependent microtubule (MT) stability. EB1 and EB3 have previously been implicated in cilia biogenesis to carry out centrosome-related functions. However, the role of DIAPH1 proteins had not been examined. Here we show that the depletion of DIAPH1 decreased ciliogenesis, cilia length, and reduced trafficking within cilia. Additionally, both actin nucleating and microtubule-stabilizing properties of DIAPH1 are important for their cilia functions. To assess their roles in ciliogenesis in isolation, we targeted DIAPH1 specifically to the basal body, which caused an increase in cilia length and increased trafficking within cilia. Intriguingly, expression of DIAPH1 mutants associated with human deafness and microcephaly impaired ciliation and caused cilia elongation and bulb formation. These results suggest that the actin and microtubule functions of DIAPH1 proteins regulate cilia maintenance in part by regulating vesicular trafficking to the base of the primary cilia.
Collapse
Affiliation(s)
- Oliva Palander
- Cell Biology Program, Hospital for Sick Children, Toronto, ON, Canada.,Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - William S Trimble
- Cell Biology Program, Hospital for Sick Children, Toronto, ON, Canada.,Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
40
|
Collins C, Ventrella R, Mitchell BJ. Building a ciliated epithelium: Transcriptional regulation and radial intercalation of multiciliated cells. Curr Top Dev Biol 2020; 145:3-39. [PMID: 34074533 DOI: 10.1016/bs.ctdb.2020.08.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The epidermis of the Xenopus embryo has emerged as a powerful tool for studying the development of a ciliated epithelium. Interspersed throughout the epithelium are multiciliated cells (MCCs) with 100+ motile cilia that beat in a coordinated manner to generate fluid flow over the surface of the cell. MCCs are essential for various developmental processes and, furthermore, ciliary dysfunction is associated with numerous pathologies. Therefore, understanding the cellular mechanisms involved in establishing a ciliated epithelium are of particular interest. MCCs originate in the inner epithelial layer of Xenopus skin, where Notch signaling plays a critical role in determining which progenitors will adopt a ciliated cell fate. Then, activation of various transcriptional regulators, such as GemC1 and MCIDAS, initiate the MCC transcriptional program, resulting in centriole amplification and the formation of motile cilia. Following specification and differentiation, MCCs undergo the process of radial intercalation, where cells apically migrate from the inner layer to the outer epithelial layer. This process involves the cooperation of various cytoskeletal networks, activation of various signaling molecules, and changes in cell-ECM and cell-cell adhesion. Coordination of these cellular processes is required for complete incorporation into the outer epithelial layer and generation of a functional ciliated epithelium. Here, we highlight recent advances made in understanding the transcriptional cascades required for MCC specification and differentiation and the coordination of cellular processes that facilitate radial intercalation. Proper regulation of these signaling pathways and processes are the foundation for developing a ciliated epithelium.
Collapse
Affiliation(s)
- Caitlin Collins
- Department of Cell and Developmental Biology, Lurie Comprehensive Cancer Center, Northwestern University, Feinberg School of Medicine, Chicago, IL, United States
| | - Rosa Ventrella
- Department of Cell and Developmental Biology, Lurie Comprehensive Cancer Center, Northwestern University, Feinberg School of Medicine, Chicago, IL, United States
| | - Brian J Mitchell
- Department of Cell and Developmental Biology, Lurie Comprehensive Cancer Center, Northwestern University, Feinberg School of Medicine, Chicago, IL, United States.
| |
Collapse
|
41
|
Streets AJ, Prosseda PP, Ong AC. Polycystin-1 regulates ARHGAP35-dependent centrosomal RhoA activation and ROCK signaling. JCI Insight 2020; 5:135385. [PMID: 32663194 PMCID: PMC7455122 DOI: 10.1172/jci.insight.135385] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 07/08/2020] [Indexed: 11/17/2022] Open
Abstract
Mutations in PKD1 (encoding for polycystin-1 [PC1]) are found in 80%–85% of patients with autosomal dominant polycystic kidney disease (ADPKD). We tested the hypothesis that changes in actin dynamics result from PKD1 mutations through dysregulation of compartmentalized centrosomal RhoA signaling mediated by specific RhoGAP (ARHGAP) proteins resulting in the complex cellular cystic phenotype. Initial studies revealed that the actin cytoskeleton was highly disorganized in cystic cells derived from patients with PKD1 and was associated with an increase in total and centrosomal active RhoA and ROCK signaling. Using cilia length as a phenotypic readout for centrosomal RhoA activity, we identified ARHGAP5, -29, and -35 as essential regulators of ciliation in normal human renal tubular cells. Importantly, a specific decrease in centrosomal ARHGAP35 was observed in PKD1-null cells using a centrosome-targeted proximity ligation assay and by dual immunofluorescence labeling. Finally, the ROCK inhibitor hydroxyfasudil reduced cyst expansion in both human PKD1 3D cyst assays and an inducible Pkd1 mouse model. In summary, we report a potentially novel interaction between PC1 and ARHGAP35 in the regulation of centrosomal RhoA activation and ROCK signaling. Targeting the RhoA/ROCK pathway inhibited cyst formation in vitro and in vivo, indicating its relevance to ADPKD pathogenesis and for developing new therapies to inhibit cyst initiation. Polycystin-1, the major protein mutated in autosomal dominant polycystic kidney disease, activates centrosomal RhoA activity via interaction with the Rho-GAP protein ARHGAP35, resulting in shorter cilia.
Collapse
|
42
|
Bustamante-Marin XM, Horani A, Stoyanova M, Charng WL, Bottier M, Sears PR, Yin WN, Daniels LA, Bowen H, Conrad DF, Knowles MR, Ostrowski LE, Zariwala MA, Dutcher SK. Mutation of CFAP57, a protein required for the asymmetric targeting of a subset of inner dynein arms in Chlamydomonas, causes primary ciliary dyskinesia. PLoS Genet 2020; 16:e1008691. [PMID: 32764743 PMCID: PMC7444499 DOI: 10.1371/journal.pgen.1008691] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 08/19/2020] [Accepted: 02/22/2020] [Indexed: 01/10/2023] Open
Abstract
Primary ciliary dyskinesia (PCD) is characterized by chronic airway disease, reduced fertility, and randomization of the left/right body axis. It is caused by defects of motile cilia and sperm flagella. We screened a cohort of affected individuals that lack an obvious axonemal defect for pathogenic variants using whole exome capture, next generation sequencing, and bioinformatic analysis assuming an autosomal recessive trait. We identified one subject with an apparently homozygous nonsense variant [(c.1762C>T), p.(Arg588*)] in the uncharacterized CFAP57 gene. Interestingly, the variant results in the skipping of exon 11 (58 amino acids), which may be due to disruption of an exonic splicing enhancer. In normal human nasal epithelial cells, CFAP57 localizes throughout the ciliary axoneme. Nasal cells from the PCD patient express a shorter, mutant version of CFAP57 and the protein is not incorporated into the axoneme. The missing 58 amino acids include portions of WD repeats that may be important for loading onto the intraflagellar transport (IFT) complexes for transport or docking onto the axoneme. A reduced beat frequency and an alteration in ciliary waveform was observed. Knockdown of CFAP57 in human tracheobronchial epithelial cells (hTECs) recapitulates these findings. Phylogenetic analysis showed that CFAP57 is highly conserved in organisms that assemble motile cilia. CFAP57 is allelic with the BOP2/IDA8/FAP57 gene identified previously in Chlamydomonas reinhardtii. Two independent, insertional fap57 Chlamydomonas mutant strains show reduced swimming velocity and altered waveforms. Tandem mass tag (TMT) mass spectroscopy shows that FAP57 is missing, and the "g" inner dyneins (DHC7 and DHC3) and the "d" inner dynein (DHC2) are reduced, but the FAP57 paralog FBB7 is increased. Together, our data identify a homozygous variant in CFAP57 that causes PCD that is likely due to a defect in the inner dynein arm assembly process.
Collapse
Affiliation(s)
- Ximena M. Bustamante-Marin
- Department of Medicine, Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Amjad Horani
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Mihaela Stoyanova
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Wu-Lin Charng
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Mathieu Bottier
- Department of Mechanical Engineering, Washington University, St. Louis, Missouri, United States of America
| | - Patrick R. Sears
- Department of Medicine, Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Wei-Ning Yin
- Department of Medicine, Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Leigh Anne Daniels
- Department of Medicine, Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Hailey Bowen
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Donald F. Conrad
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Division of Genetics, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States of America
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Michael R. Knowles
- Department of Medicine, Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Lawrence E. Ostrowski
- Department of Medicine, Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Maimoona A. Zariwala
- Department of Pathology and Laboratory Medicine and the Marsico Lung Institute, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Susan K. Dutcher
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| |
Collapse
|
43
|
Lee M, Hwang YS, Yoon J, Sun J, Harned A, Nagashima K, Daar IO. Developmentally regulated GTP-binding protein 1 modulates ciliogenesis via an interaction with Dishevelled. J Cell Biol 2019; 218:2659-2676. [PMID: 31270137 PMCID: PMC6683737 DOI: 10.1083/jcb.201811147] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 04/25/2019] [Accepted: 06/10/2019] [Indexed: 12/11/2022] Open
Abstract
Our study reveals Drg1 as a new binding partner of Dishevelled. The Drg1–Dishevelled association regulates Daam1 and RhoA interactions and activity, leading to polymerization and stability of the actin cytoskeleton, a process that is essential for proper multiciliation. Cilia are critical for proper embryonic development and maintaining homeostasis. Although extensively studied, there are still significant gaps regarding the proteins involved in regulating ciliogenesis. Using the Xenopus laevis embryo, we show that Dishevelled (Dvl), a key Wnt signaling scaffold that is critical to proper ciliogenesis, interacts with Drg1 (developmentally regulated GTP-binding protein 1). The loss of Drg1 or disruption of the interaction with Dvl reduces the length and number of cilia and displays defects in basal body migration and docking to the apical surface of multiciliated cells (MCCs). Moreover, Drg1 morphants display abnormal rotational polarity of basal bodies and a decrease in apical actin and RhoA activity that can be attributed to disruption of the protein complex between Dvl and Daam1, as well as between Daam1 and RhoA. These results support the concept that the Drg1–Dvl interaction regulates apical actin polymerization and stability in MCCs. Thus, Drg1 is a newly identified partner of Dvl in regulating ciliogenesis.
Collapse
Affiliation(s)
| | | | - Jaeho Yoon
- National Cancer Institute, Frederick, MD
| | - Jian Sun
- National Cancer Institute, Frederick, MD
| | - Adam Harned
- Electron Microscope Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Kunio Nagashima
- Electron Microscope Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD
| | - Ira O Daar
- National Cancer Institute, Frederick, MD
| |
Collapse
|
44
|
Kumar D, Mains RE, Eipper BA, King SM. Ciliary and cytoskeletal functions of an ancient monooxygenase essential for bioactive amidated peptide synthesis. Cell Mol Life Sci 2019; 76:2329-2348. [PMID: 30879092 PMCID: PMC6529398 DOI: 10.1007/s00018-019-03065-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 02/28/2019] [Accepted: 03/07/2019] [Indexed: 02/07/2023]
Abstract
Many secreted peptides used for cell-cell communication require conversion of a C-terminal glycine to an amide for bioactivity. This reaction is catalyzed only by the integral membrane protein peptidylglycine α-amidating monooxygenase (PAM). PAM has been highly conserved and is found throughout the metazoa; PAM-like sequences are also present in choanoflagellates, filastereans, unicellular and colonial chlorophyte green algae, dinoflagellates and haptophytes. Recent studies have revealed that in addition to playing a key role in peptidergic signaling, PAM also regulates ciliogenesis in vertebrates, planaria and chlorophyte algae, and is required for the stability of actin-based microvilli. Here we briefly introduce the basic principles involved in ciliogenesis, the sequential reactions catalyzed by PAM and the trafficking of PAM through the secretory and endocytic pathways. We then discuss the multi-faceted roles this enzyme plays in the formation and maintenance of cytoskeleton-based cellular protrusions and propose models for how PAM protein and amidating activity might contribute to ciliogenesis. Finally, we consider why some ciliated organisms lack PAM, and discuss the potential ramifications of ciliary localized PAM for the endocrine features commonly observed in patients with ciliopathies.
Collapse
Affiliation(s)
- Dhivya Kumar
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT, 06030, USA
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Richard E Mains
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Betty A Eipper
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT, 06030, USA
- Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Stephen M King
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT, 06030, USA.
| |
Collapse
|
45
|
Jack B, Mueller DM, Fee AC, Tetlow AL, Avasthi P. Partially Redundant Actin Genes in Chlamydomonas Control Transition Zone Organization and Flagellum-Directed Traffic. Cell Rep 2019; 27:2459-2467.e3. [PMID: 31116988 PMCID: PMC6541019 DOI: 10.1016/j.celrep.2019.04.087] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 12/03/2018] [Accepted: 04/18/2019] [Indexed: 11/16/2022] Open
Abstract
The unicellular green alga Chlamydomonas reinhardtii is a biflagellated cell with two actin genes: one encoding a conventional actin (IDA5) and the other encoding a divergent novel actin-like protein (NAP1). Here, we probe how actin redundancy contributes to flagellar assembly. Disrupting a single actin allows complete flagellar assembly. However, when disrupting both actins using latrunculin B (LatB) treatment on the nap1 mutant background, we find that actins are necessary for flagellar growth from newly synthesized limiting flagellar proteins. Under total actin disruption, transmission electron microscopy identified an accumulation of Golgi-adjacent vesicles. We also find that there is a mislocalization of a key transition zone gating and ciliopathy protein, NPHP-4. Our experiments demonstrate that each stage of flagellar biogenesis requires redundant actin function to varying degrees, with an absolute requirement for these actins in transport of Golgi-adjacent vesicles and flagellar incorporation of newly synthesized proteins.
Collapse
Affiliation(s)
- Brittany Jack
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - David M Mueller
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Ann C Fee
- University of Missouri-Kansas City, School of Medicine, Kansas City, MO 64110, USA
| | - Ashley L Tetlow
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Prachee Avasthi
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA; Department of Ophthalmology, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| |
Collapse
|
46
|
Nanjundappa R, Kong D, Shim K, Stearns T, Brody SL, Loncarek J, Mahjoub MR. Regulation of cilia abundance in multiciliated cells. eLife 2019; 8:e44039. [PMID: 31025935 PMCID: PMC6504233 DOI: 10.7554/elife.44039] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 04/25/2019] [Indexed: 12/14/2022] Open
Abstract
Multiciliated cells (MCC) contain hundreds of motile cilia used to propel fluid over their surface. To template these cilia, each MCC produces between 100-600 centrioles by a process termed centriole amplification. Yet, how MCC regulate the precise number of centrioles and cilia remains unknown. Airway progenitor cells contain two parental centrioles (PC) and form structures called deuterosomes that nucleate centrioles during amplification. Using an ex vivo airway culture model, we show that ablation of PC does not perturb deuterosome formation and centriole amplification. In contrast, loss of PC caused an increase in deuterosome and centriole abundance, highlighting the presence of a compensatory mechanism. Quantification of centriole abundance in vitro and in vivo identified a linear relationship between surface area and centriole number. By manipulating cell size, we discovered that centriole number scales with surface area. Our results demonstrate that a cell-intrinsic surface area-dependent mechanism controls centriole and cilia abundance in multiciliated cells.
Collapse
Affiliation(s)
- Rashmi Nanjundappa
- Nephrology Division, Department of MedicineWashington UniversitySt LouisUnited States
| | - Dong Kong
- Center for Cancer Research, National Cancer InstituteFrederickUnited States
| | - Kyuhwan Shim
- Nephrology Division, Department of MedicineWashington UniversitySt LouisUnited States
| | - Tim Stearns
- Department of BiologyStanford UniversityStanfordUnited States
| | - Steven L Brody
- Pulmonary Division, Department of MedicineWashington UniversitySt LouisUnited States
| | - Jadranka Loncarek
- Center for Cancer Research, National Cancer InstituteFrederickUnited States
| | - Moe R Mahjoub
- Nephrology Division, Department of MedicineWashington UniversitySt LouisUnited States
- Department of Cell Biology and PhysiologyWashington UniversitySt LouisUnited States
| |
Collapse
|
47
|
Inoue D, Obino D, Pineau J, Farina F, Gaillard J, Guerin C, Blanchoin L, Lennon-Duménil AM, Théry M. Actin filaments regulate microtubule growth at the centrosome. EMBO J 2019; 38:embj.201899630. [PMID: 30902847 DOI: 10.15252/embj.201899630] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 02/05/2019] [Accepted: 02/21/2019] [Indexed: 12/22/2022] Open
Abstract
The centrosome is the main microtubule-organizing centre. It also organizes a local network of actin filaments. However, the precise function of the actin network at the centrosome is not well understood. Here, we show that increasing densities of actin filaments at the centrosome of lymphocytes are correlated with reduced amounts of microtubules. Furthermore, lymphocyte activation resulted in disassembly of centrosomal actin and an increase in microtubule number. To further investigate the direct crosstalk between actin and microtubules at the centrosome, we performed in vitro reconstitution assays based on (i) purified centrosomes and (ii) on the co-micropatterning of microtubule seeds and actin filaments. These two assays demonstrated that actin filaments constitute a physical barrier blocking elongation of nascent microtubules. Finally, we showed that cell adhesion and cell spreading lead to lower densities of centrosomal actin, thus resulting in higher microtubule growth. We therefore propose a novel mechanism, by which the number of centrosomal microtubules is regulated by cell adhesion and actin-network architecture.
Collapse
Affiliation(s)
- Daisuke Inoue
- CEA, CNRS, INRA, Biosciences & Biotechnology Institute of Grenoble, UMR5168, CytoMorpho Lab, Univ. Grenoble-Alpes, Grenoble, France
| | - Dorian Obino
- INSERM, U932 Immunité et Cancer, Institut Curie, PSL Research University, Paris, France
| | - Judith Pineau
- INSERM, U932 Immunité et Cancer, Institut Curie, PSL Research University, Paris, France
| | - Francesca Farina
- CEA, CNRS, INRA, Biosciences & Biotechnology Institute of Grenoble, UMR5168, CytoMorpho Lab, Univ. Grenoble-Alpes, Grenoble, France
| | - Jérémie Gaillard
- CEA, CNRS, INRA, Biosciences & Biotechnology Institute of Grenoble, UMR5168, CytoMorpho Lab, Univ. Grenoble-Alpes, Grenoble, France.,INSERM, CEA, Hôpital Saint Louis, Institut Universitaire d'Hematologie, UMRS1160, CytoMorpho Lab, Univ. Paris Diderot, Paris, France
| | - Christophe Guerin
- CEA, CNRS, INRA, Biosciences & Biotechnology Institute of Grenoble, UMR5168, CytoMorpho Lab, Univ. Grenoble-Alpes, Grenoble, France.,INSERM, CEA, Hôpital Saint Louis, Institut Universitaire d'Hematologie, UMRS1160, CytoMorpho Lab, Univ. Paris Diderot, Paris, France
| | - Laurent Blanchoin
- CEA, CNRS, INRA, Biosciences & Biotechnology Institute of Grenoble, UMR5168, CytoMorpho Lab, Univ. Grenoble-Alpes, Grenoble, France .,INSERM, CEA, Hôpital Saint Louis, Institut Universitaire d'Hematologie, UMRS1160, CytoMorpho Lab, Univ. Paris Diderot, Paris, France
| | | | - Manuel Théry
- CEA, CNRS, INRA, Biosciences & Biotechnology Institute of Grenoble, UMR5168, CytoMorpho Lab, Univ. Grenoble-Alpes, Grenoble, France .,INSERM, CEA, Hôpital Saint Louis, Institut Universitaire d'Hematologie, UMRS1160, CytoMorpho Lab, Univ. Paris Diderot, Paris, France
| |
Collapse
|
48
|
Mukherjee I, Roy S, Chakrabarti S. Identification of Important Effector Proteins in the FOXJ1 Transcriptional Network Associated With Ciliogenesis and Ciliary Function. Front Genet 2019; 10:23. [PMID: 30881373 PMCID: PMC6405523 DOI: 10.3389/fgene.2019.00023] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 01/15/2019] [Indexed: 12/17/2022] Open
Abstract
Developmental defects in motile cilia, arising from genetic abnormalities in one or more ciliary genes, can lead to a common ciliopathy known as primary ciliary dyskinesia (PCD). Functional studies in model organisms undertaken to understand PCD or cilia biogenesis have identified 100s of genes regulated by Foxj1, the master regulator of motile ciliogenesis. However, limited systems based studies have been performed to elucidate proteins or network/s crucial to the motile ciliary interactome, although this approach holds promise for identification of multiple cilia-associated genes, which, in turn, could be utilized for screening and early diagnosis of the disease. Here, based on the assumption that FOXJ1-mediated regulatory and signaling networks are representative of the motile cilia interactome, we have constructed and analyzed the gene regulatory and protein–protein interaction network (PPIN) mediated by FOXJ1. The predicted FOXJ1 regulatory network comprises of 424 directly and 148 indirectly regulated genes. Additionally, based on gene ontology analysis, we have associated 17 directly and 6 indirectly regulated genes with possible ciliary roles. Topological and perturbation analyses of the PPIN (6927 proteins, 40,608 interactions) identified 121 proteins expressed in ciliated cells, which interact with multiple proteins encoded by FoxJ1 induced genes (FIG) as important interacting proteins (IIP). However, it is plausible that IIP transcriptionally regulated by FOXJ1 and/or differentially expressed in PCD are likely to have crucial roles in motile cilia. We have found 20 de-regulated topologically important effector proteins in the FOXJ1 regulatory network, among which some (PLSCR1, SSX2IP, ACTN2, CDC42, HSP90AA1, PIAS4) have previously reported ciliary roles. Furthermore, based on pathway enrichment of these proteins and their primary interactors, we have rationalized their possible roles in the ciliary interactome. For instance, 5 among these novel proteins that are involved in cilia associated signaling pathways (like Notch, Wnt, Hedgehog, Toll-like receptor etc.) could be ‘topologically important signaling proteins.’ Therefore, based on this FOXJ1 network study we have predicted important effectors in the motile cilia interactome, which are possibly associated with ciliary biology and/or function and are likely to further our understanding of the pathophysiology in ciliopathies like PCD.
Collapse
Affiliation(s)
- Ishita Mukherjee
- Translational Research Unit of Excellence, Structural Biology and Bioinformatics Division, Council for Scientific and Industrial Research - Indian Institute of Chemical Biology, Kolkata, India
| | - Sudipto Roy
- Institute of Molecular and Cell Biology, Singapore, Singapore.,Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Saikat Chakrabarti
- Translational Research Unit of Excellence, Structural Biology and Bioinformatics Division, Council for Scientific and Industrial Research - Indian Institute of Chemical Biology, Kolkata, India
| |
Collapse
|
49
|
Zhang Y, Mao D, Keeler SP, Wang X, Wu K, Gerovac BJ, Shornick LL, Agapov EV, Holtzman MJ. Respiratory Enterovirus (like Parainfluenza Virus) Can Cause Chronic Lung Disease if Protection by Airway Epithelial STAT1 Is Lost. THE JOURNAL OF IMMUNOLOGY 2019; 202:2332-2347. [PMID: 30804041 DOI: 10.4049/jimmunol.1801491] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 02/11/2019] [Indexed: 12/11/2022]
Abstract
Epithelial barrier cells are proposed to be critical for host defense, and airway epithelial cell capacity for IFN signal transduction is presumed to protect against respiratory viral infection. However, it has been difficult to fully test these concepts given the absence of tools to analyze IFN signaling specific to airway epithelial cells in vivo. To address these issues, we generated a new line of transgenic mice with Cre-driver genes (Foxj1 and Scgb1a1) for a floxed-Stat1 allele (designated Foxj1-Scgb1a1-Cre-Stat1f/f mice) to target the master IFN signal regulator STAT1 in airway epithelial cells and tested these mice for control of infection because of mouse parainfluenza (Sendai) virus and human enterovirus D68 (EV-D68). Indeed, both types of infections showed increases in viral titers and severity of acute illness in Foxj1-Scgb1a1-Cre-Stat1f/f mice and conventional Stat1-/- mice compared with wild-type mice. In concert, the chronic lung disease that develops after Sendai virus infection was also increased in Foxj1-Scgb1a1-Cre-Stat1f/f and Stat1-/ - mice, marked by airway and adjacent parenchymal immune cell infiltration and mucus production for at least 7 wk postinfection. Unexpectedly, relatively mild EV-D68 infection also progressed to chronic lung disease in Foxj1-Scgb1a1-Cre-Stat1f/f and Stat1 -/- mice but was limited (like viral replication) to airways. The results thereby provide proof-of-concept for a critical role of barrier epithelial cells in protection from acute illness and chronic disease after viral infection and suggest a specific role for airway epithelial cells given the limitation of EV-D68 replication and acute and chronic manifestations of disease primarily to airway tissue.
Collapse
Affiliation(s)
- Yong Zhang
- Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Dailing Mao
- Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Shamus P Keeler
- Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Xinyu Wang
- Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Kangyun Wu
- Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Benjamin J Gerovac
- Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Laurie L Shornick
- Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Eugene V Agapov
- Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Michael J Holtzman
- Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
50
|
Rangel L, Bernabé-Rubio M, Fernández-Barrera J, Casares-Arias J, Millán J, Alonso MA, Correas I. Caveolin-1α regulates primary cilium length by controlling RhoA GTPase activity. Sci Rep 2019; 9:1116. [PMID: 30718762 PMCID: PMC6362014 DOI: 10.1038/s41598-018-38020-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 10/26/2018] [Indexed: 11/08/2022] Open
Abstract
The primary cilium is a single non-motile protrusion of the plasma membrane of most types of mammalian cell. The structure, length and function of the primary cilium must be tightly controlled because their dysfunction is associated with disease. Caveolin 1 (Cav1), which is best known as a component of membrane invaginations called caveolae, is also present in non-caveolar membrane domains whose function is beginning to be understood. We show that silencing of α and β Cav1 isoforms in different cell lines increases ciliary length regardless of the route of primary ciliogenesis. The sole expression of Cav1α, which is distributed at the apical membrane, restores normal cilium size in Cav1 KO MDCK cells. Cells KO for only Cav1α, which also show long cilia, have a disrupted actin cytoskeleton and reduced RhoA GTPase activity at the apical membrane, and a greater accumulation of Rab11 vesicles at the centrosome. Subsequent experiments showed that DIA1 and ROCK help regulate ciliary length. Since MDCK cells lack apical caveolae, our results imply that non-caveolar apical Cav1α is an important regulator of ciliary length, exerting its effect via RhoA and its effectors, ROCK and DIA1.
Collapse
Affiliation(s)
- Laura Rangel
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain
- Department of Molecular Biology, Universidad Autónoma de Madrid, Madrid, Spain
| | - Miguel Bernabé-Rubio
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain
| | - Jaime Fernández-Barrera
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain
| | - Javier Casares-Arias
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain
| | - Jaime Millán
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain
| | - Miguel A Alonso
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain.
| | - Isabel Correas
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain.
- Department of Molecular Biology, Universidad Autónoma de Madrid, Madrid, Spain.
| |
Collapse
|