1
|
Sun S, Liang B, Koplas A, Tikhonenko I, Nachury M, Khodjakov A, Sui H. Intraflagellar transport trains can switch rails and move along multiple microtubules in intact primary cilia. Proc Natl Acad Sci U S A 2025; 122:e2413968122. [PMID: 40249775 PMCID: PMC12037007 DOI: 10.1073/pnas.2413968122] [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: 07/12/2024] [Accepted: 02/20/2025] [Indexed: 04/20/2025] Open
Abstract
Structural homeostasis and proper distributions of signaling molecules in cilia require a constant flow of cargoes carried by intraflagellar transport (IFT) trains in both anterograde and retrograde directions within the thin, long ciliary shafts. In the motile cilium framework, the nine microtubule doublets of the same length serve as the transportation rails, and a preferential association to the two subtubules of the microtubule doublets prevents collisions among the IFT trains that move in opposite directions. However, this mechanism is incompatible with the primary cilia structure, where most of the nine microtubule doublets terminate in the ciliary shafts-only several of them reach the ciliary tip and only in a singlet form. Here, we demonstrate that anterograde and retrograde trains in primary cilia interact with both subtubules of the microtubule doublets without apparent preference. They can switch microtubules, and they may simultaneously interact with multiple microtubules to facilitate their movement. This architecture makes the collisions inevitable, and live-cell recordings reveal that anterograde and retrograde trains tend to pause when they come into direct contact. We also find that the velocity of the train's movement often changes after the pause. Thus, the motion behaviors of IFT trains in primary cilia are distinctive from those of motile cilia, and our data offer an essential foundation for understanding proper signaling molecule distributions in primary cilia.
Collapse
Affiliation(s)
- Shufeng Sun
- Wadsworth Center, New York State Department of Health, Albany, NY12237
| | - Biqing Liang
- Department of Biomedical Sciences, School of Public Health, University at Albany, Albany, NY12237
| | - Adam Koplas
- Wadsworth Center, New York State Department of Health, Albany, NY12237
| | - Irina Tikhonenko
- Wadsworth Center, New York State Department of Health, Albany, NY12237
| | - Maxence Nachury
- Department of Ophthalmology, School of Medicine, University of California at San Francisco, San Francisco, CA94158
| | - Alexey Khodjakov
- Wadsworth Center, New York State Department of Health, Albany, NY12237
| | - Haixin Sui
- Wadsworth Center, New York State Department of Health, Albany, NY12237
- Department of Biomedical Sciences, School of Public Health, University at Albany, Albany, NY12237
| |
Collapse
|
2
|
Tasaki K, Satoda Y, Chiba S, Shin HW, Katoh Y, Nakayama K. Mutually independent and cilia-independent assembly of IFT-A and IFT-B complexes at mother centriole. Mol Biol Cell 2025; 36:ar48. [PMID: 40020180 PMCID: PMC12005097 DOI: 10.1091/mbc.e24-11-0509] [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: 11/18/2024] [Revised: 02/06/2025] [Accepted: 02/10/2025] [Indexed: 03/05/2025] Open
Abstract
The intraflagellar transport (IFT) machinery, containing the IFT-A and IFT-B complexes and powered by dynein-2 and kinesin-2 motors, is crucial for bidirectional trafficking of ciliary proteins and their import/export across the transition zone (TZ). Stepwise assembly of anterograde IFT trains was proposed previously; that is, the IFT-B complex first forms a TZ-tethered scaffold with sequential incorporation of IFT-A, dynein-2, and finally kinesin-2. However, IFT-A and IFT-B complexes also demonstrate distinct localization to the basal body/mother centriole. We show that IFT-A, IFT-B, and dynein-2 complexes are recruited to the mother centriole independently of ciliogenesis. Furthermore, mother centriole recruitment of IFT-A and IFT-B can occur in the absence of IFT-B and IFT-A, respectively, and dynein-2 recruitment is independent of IFT-A and IFT-B. Expansion microscopy revealed that the IFT-A/IFT-B pool at the basal body is distinct from that at the TZ. We conclude that IFT-A and IFT-B are recruited to the mother centriole in a mutually independent and ciliogenesis-independent manner before IFT train assembly.
Collapse
Affiliation(s)
- Koshi Tasaki
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Yuuki Satoda
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Shuhei Chiba
- Laboratory of Molecular and Cellular Biology, Tohoku University, Aobayama, Sendai, Miyagi 980-8578, Japan
| | - Hye-Won Shin
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Yohei Katoh
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Kazuhisa Nakayama
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| |
Collapse
|
3
|
Yamamoto R, Sahashi Y, Shimo-Kon R, Sakato-Antoku M, Suzuki M, Luo L, Tanaka H, Ishikawa T, Yagi T, King SM, Kurisu G, Kon T. Chlamydomonas FBB18 is a ubiquitin-like protein essential for the cytoplasmic preassembly of various ciliary dyneins. Proc Natl Acad Sci U S A 2025; 122:e2423948122. [PMID: 40106351 PMCID: PMC11962417 DOI: 10.1073/pnas.2423948122] [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: 11/21/2024] [Accepted: 02/14/2025] [Indexed: 03/22/2025] Open
Abstract
Motile cilia are organelles found on many eukaryotic cells that play critical roles in development and fertility. Human CFAP298 has been implicated in the transport/assembly of ciliary dyneins, and defects in this protein cause primary ciliary dyskinesia. However, neither the exact function nor the structure of CFAP298 have been elucidated. Here, we took advantage of Chlamydomonas, a ciliated alga, to study the structure and function of FBB18, an ortholog of CFAP298. Multiple ciliary dyneins were greatly reduced in cilia of Chlamydomonas fbb18 mutants. In addition, we found that both the stability of ciliary dynein heavy chains (HCs) and the association between HCs and intermediate/light chains (IC/LCs) are greatly reduced in fbb18 cytoplasm, strongly suggesting that FBB18 functions in the cytoplasmic assembly (the so-called "preassembly") of dynein complexes from HC/IC/LCs. Furthermore, X-ray crystallography revealed that FBB18 forms a bilobed structure with globular domains at both ends of the molecule, connected by an α-helical bundle. Unexpectedly, one globular domain shows high similarity to ubiquitin, a small protein critical for the modification of a variety of protein complexes, and this ubiquitin-like domain is indispensable for the molecular function of FBB18. Our results demonstrate that FBB18, a specialized member of the ubiquitin-like protein family, plays a critical role in dynein preassembly, most likely by mediating diverse interactions between dynein HCs, molecular chaperone(s), and other preassembly factor(s) using the ubiquitin-like domain as well as other regions, and by facilitating the proper folding of dynein HCs.
Collapse
Affiliation(s)
- Ryosuke Yamamoto
- Department of Biological Sciences, Graduate School of Science, Osaka University, Osaka560-0043, Japan
| | - Yui Sahashi
- Department of Biological Sciences, Graduate School of Science, Osaka University, Osaka560-0043, Japan
| | - Rieko Shimo-Kon
- Department of Biological Sciences, Graduate School of Science, Osaka University, Osaka560-0043, Japan
| | - Miho Sakato-Antoku
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT06030-3305
| | - Miyuka Suzuki
- Department of Biological Sciences, Graduate School of Science, Osaka University, Osaka560-0043, Japan
| | - Leo Luo
- Department of Biology and Chemistry, Paul Scherrer Institute, Villigen5232, Switzerland
- Department of Biology, ETH Zurich, Zurich8093, Switzerland
| | - Hideaki Tanaka
- Institute for Protein Research, Osaka University, Osaka565-0871, Japan
| | - Takashi Ishikawa
- Department of Biology and Chemistry, Paul Scherrer Institute, Villigen5232, Switzerland
- Department of Biology, ETH Zurich, Zurich8093, Switzerland
| | - Toshiki Yagi
- Department of Life and Environmental Sciences, Faculty of Bioresource Sciences, Prefectural University of Hiroshima, Hiroshima727-0023, Japan
| | - Stephen M. King
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT06030-3305
| | - Genji Kurisu
- Department of Biological Sciences, Graduate School of Science, Osaka University, Osaka560-0043, Japan
- Institute for Protein Research, Osaka University, Osaka565-0871, Japan
| | - Takahide Kon
- Department of Biological Sciences, Graduate School of Science, Osaka University, Osaka560-0043, Japan
| |
Collapse
|
4
|
Alshriem LA, Buqaileh R, Alorjani Q, AbouAlaiwi W. Ciliary Ion Channels in Polycystic Kidney Disease. Cells 2025; 14:459. [PMID: 40136708 PMCID: PMC11941060 DOI: 10.3390/cells14060459] [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: 02/07/2025] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 03/27/2025] Open
Abstract
Polycystic kidney disease (PKD) is the most common hereditary disorder that disrupts renal function and frequently progresses to end-stage renal disease. Recent advances have elucidated the critical role of primary cilia and ciliary ion channels, including transient receptor potential (TRP) channels, cystic fibrosis transmembrane conductance regulator (CFTR), and polycystin channels, in the pathogenesis of PKD. While some channels primarily function as chloride conductance channels (e.g., CFTR), others primarily regulate calcium (Ca+2) homeostasis. These ion channels are essential for cellular signaling and maintaining the normal kidney architecture. Dysregulation of these pathways due to genetic mutations in PKD1 and PKD2 leads to disrupted Ca+2 and cAMP signaling, aberrant fluid secretion, and uncontrolled cellular proliferation, resulting in tubular cystogenesis. Understanding the molecular mechanisms underlying these dysfunctions has opened the door for innovative therapeutic strategies, including TRPV4 activators, CFTR inhibitors, and calcimimetics, to mitigate cyst growth and preserve renal function. This review summarizes the current knowledge on the roles of ciliary ion channels in PKD pathophysiology, highlights therapeutic interventions targeting these channels, and identifies future research directions for improving patient outcomes.
Collapse
Affiliation(s)
- Lubna A. Alshriem
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, Toledo, OH 43614, USA; (L.A.A.); (R.B.); (Q.A.)
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, P.O. Box 3030, Irbid 22110, Jordan
| | - Raghad Buqaileh
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, Toledo, OH 43614, USA; (L.A.A.); (R.B.); (Q.A.)
| | - Qasim Alorjani
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, Toledo, OH 43614, USA; (L.A.A.); (R.B.); (Q.A.)
| | - Wissam AbouAlaiwi
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, Toledo, OH 43614, USA; (L.A.A.); (R.B.); (Q.A.)
| |
Collapse
|
5
|
Ku PI, Sreeja JS, Chadha A, Williams DS, Engelke MF, Subramanian R. Collaborative role of two distinct cilium-specific cytoskeletal systems in driving Hedgehog-responsive transcription factor trafficking. SCIENCE ADVANCES 2025; 11:eadt5439. [PMID: 40073114 PMCID: PMC11900865 DOI: 10.1126/sciadv.adt5439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 02/05/2025] [Indexed: 03/14/2025]
Abstract
In vertebrate Hedgehog (Hh) signaling, the precise output of the final effectors, GLI (glioma-associated oncogene) transcription factors, depends on the primary cilium. Upon pathway initiation, generating the precise levels of the activator form of GLI depends on its concentration at the cilium tip. The mechanisms underlying this critical step in Hh signaling are unclear. We developed an assay to visualize GLI2, the primary GLI activator isoform, at single-particle resolution within the cilium. We found that GLI2 is a cargo of intraflagellar transport (IFT) machinery. Anterograde-biased IFT loading of GLI2 in a restricted time window following pathway activation results in the tip accumulation of GLI2. Unexpectedly, we found that the conserved Hh regulator KIF7, a nonmotile kinesin, is important for the temporal control of IFT-mediated GLI2 transport and retention of GLI2 at the cilium tip. Our findings underscore a design principle where a cilia-specific cytoskeletal transport system and an Hh pathway-specific cytoskeletal protein collaboratively regulate GLI2 trafficking for Hh signaling.
Collapse
Affiliation(s)
- Pei-I Ku
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Jamuna S. Sreeja
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Abhishek Chadha
- Departments of Ophthalmology and Neurobiology, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - David S. Williams
- Departments of Ophthalmology and Neurobiology, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Martin F. Engelke
- Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, TN, USA
| | - Radhika Subramanian
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
6
|
De Decker M, Zelina P, Moens TG, Beckers J, Contardo M, Dittlau KS, Van Schoor E, Ronisz A, Eggermont K, Moisse M, Chandran S, Veldink JH, Thal DR, Van Den Bosch L, Pasterkamp RJ, Van Damme P. C21ORF2 mutations point towards primary cilia dysfunction in amyotrophic lateral sclerosis. Brain 2025; 148:803-816. [PMID: 39703094 PMCID: PMC11884758 DOI: 10.1093/brain/awae331] [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: 04/21/2024] [Revised: 08/09/2024] [Accepted: 09/08/2024] [Indexed: 12/21/2024] Open
Abstract
Progressive loss of motor neurons is the hallmark of the neurodegenerative disease amyotrophic lateral sclerosis (ALS), but the underlying disease mechanisms remain incompletely understood. In this study, we investigate the effects of C21ORF2 mutations, a gene recently linked to ALS, and find that primary cilia are dysfunctional. Human patient-derived mutant C21ORF2 motor neurons have a reduced ciliary frequency and length. We report that C21ORF2 is located at the basal body of the primary cilium, and mutations associated with ALS alter this localization. Furthermore, we show that a reduction of C21ORF2 levels in cell lines and motor neurons is sufficient to cause fewer primary cilia and reduced cilial length. This ciliary dysfunction leads to defective downstream sonic hedgehog signalling and reduces the expression of cellular retinoic acid binding protein 1 (CRABP1), a protein involved in motor neuron maintenance and survival. In a compartmentalized co-culture system of motor neurons and muscle cells, these ciliary defects were associated with a reduced ability of neuromuscular junction formation. Interestingly, these cilia defects are seemingly not restricted to C21ORF2 ALS, as we also observed perturbed primary cilia in cultured motor neurons and post-mortem motor cortex from patients with the most common genetic subtype of ALS caused by repeat expansions in the C9ORF72 gene. Finally, overexpression of C21ORF2 in mutant C21ORF2 motor neurons rescued the ciliary frequency and length, CRAPBP1 expression and neuromuscular junction formation, confirming the importance of primary cilia for motor neuron function. These results point towards primary cilia dysfunction contributing to motor neuron degeneration in ALS and open new avenues for further research and interventions for this as yet untreatable disease.
Collapse
Affiliation(s)
- Mathias De Decker
- Department of Neurosciences, Laboratory of Neurobiology and Leuven Brain Institute (LBI), KU Leuven—University of Leuven, 3000 Leuven, Belgium
- VIB, Center for Brain & Disease Research, 3000 Leuven, Belgium
| | - Pavol Zelina
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Thomas G Moens
- Department of Neurosciences, Laboratory of Neurobiology and Leuven Brain Institute (LBI), KU Leuven—University of Leuven, 3000 Leuven, Belgium
- VIB, Center for Brain & Disease Research, 3000 Leuven, Belgium
| | - Jimmy Beckers
- Department of Neurosciences, Laboratory of Neurobiology and Leuven Brain Institute (LBI), KU Leuven—University of Leuven, 3000 Leuven, Belgium
| | - Matilde Contardo
- Department of Neurosciences, Laboratory of Neurobiology and Leuven Brain Institute (LBI), KU Leuven—University of Leuven, 3000 Leuven, Belgium
- VIB, Center for Brain & Disease Research, 3000 Leuven, Belgium
| | - Katarina Stoklund Dittlau
- Department of Neurosciences, Laboratory of Neurobiology and Leuven Brain Institute (LBI), KU Leuven—University of Leuven, 3000 Leuven, Belgium
- VIB, Center for Brain & Disease Research, 3000 Leuven, Belgium
| | - Evelien Van Schoor
- Department of Neurosciences, Laboratory of Neurobiology and Leuven Brain Institute (LBI), KU Leuven—University of Leuven, 3000 Leuven, Belgium
- VIB, Center for Brain & Disease Research, 3000 Leuven, Belgium
- Laboratory of Neuropathology, Department of Imaging and Pathology, Leuven Brain Institute (LBI), KU Leuven (University of Leuven), 3000 Leuven, Belgium
| | - Alicja Ronisz
- Laboratory of Neuropathology, Department of Imaging and Pathology, Leuven Brain Institute (LBI), KU Leuven (University of Leuven), 3000 Leuven, Belgium
| | - Kristel Eggermont
- Department of Neurosciences, Laboratory of Neurobiology and Leuven Brain Institute (LBI), KU Leuven—University of Leuven, 3000 Leuven, Belgium
- VIB, Center for Brain & Disease Research, 3000 Leuven, Belgium
| | - Matthieu Moisse
- Department of Neurosciences, Laboratory of Neurobiology and Leuven Brain Institute (LBI), KU Leuven—University of Leuven, 3000 Leuven, Belgium
- VIB, Center for Brain & Disease Research, 3000 Leuven, Belgium
| | - Siddharthan Chandran
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh EH16 4SB, UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Jan H Veldink
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
| | - Dietmar Rudolf Thal
- Laboratory of Neuropathology, Department of Imaging and Pathology, Leuven Brain Institute (LBI), KU Leuven (University of Leuven), 3000 Leuven, Belgium
- Department of Pathology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Ludo Van Den Bosch
- Department of Neurosciences, Laboratory of Neurobiology and Leuven Brain Institute (LBI), KU Leuven—University of Leuven, 3000 Leuven, Belgium
- VIB, Center for Brain & Disease Research, 3000 Leuven, Belgium
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Philip Van Damme
- Department of Neurosciences, Laboratory of Neurobiology and Leuven Brain Institute (LBI), KU Leuven—University of Leuven, 3000 Leuven, Belgium
- Department of Neurology, University Hospitals Leuven, 3000 Leuven, Belgium
| |
Collapse
|
7
|
Wang J, Kidmose RT, Boegholm N, Zacharia NK, Thomsen MB, Christensen A, Malik T, Lechtreck K, Lorentzen E. Integrative in silico and biochemical analyses demonstrate direct Arl3-mediated ODA16 release from the intraflagellar transport machinery. J Biol Chem 2025; 301:108237. [PMID: 39880089 PMCID: PMC11879689 DOI: 10.1016/j.jbc.2025.108237] [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: 09/19/2024] [Revised: 12/27/2024] [Accepted: 01/17/2025] [Indexed: 01/31/2025] Open
Abstract
Outer dynein arms (ODAs) are essential for ciliary motility and are preassembled in the cytoplasm before trafficking into cilia by intraflagellar transport (IFT). ODA16 is a key adaptor protein that links ODAs to the IFT machinery via direct interaction with the IFT46 protein. However, the molecular mechanisms regulating the assembly, transport, and release of ODAs remain poorly understood. Here, we employ AlphaPulldown, an in silico screening method, to identify direct interactors of ODA16, including the dynein adaptor IDA3 and the small GTPase Arl3. We use structural modeling, biochemical, and biophysical assays on Chlamydomonas and human proteins to elucidate the interactions and regulatory mechanisms governing the IFT of ODAs. We identify a conserved N-terminal motif in Chlamydomonas IFT46 that mediates its binding to one side of the ODA16 structure. Biochemical dissection reveals that IDA3 and Arl3 bind to the same surface of ODA16 (the C-terminal β-propeller face), which is opposite to the IFT46 binding site, enabling them to dissociate ODA16 from IFT46, likely through an allosteric mechanism. Our findings provide mechanistic insights into the concerted actions of IFT and adaptor proteins in ODA transport and regulation.
Collapse
Affiliation(s)
- Jiaolong Wang
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus C, Denmark
| | - Rune T Kidmose
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus C, Denmark
| | - Niels Boegholm
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus C, Denmark
| | - Nevin K Zacharia
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus C, Denmark
| | - Mads B Thomsen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus C, Denmark
| | - Anni Christensen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus C, Denmark
| | - Tara Malik
- Department of Cellular Biology, University of Georgia, Athens, Georgia, USA
| | - Karl Lechtreck
- Department of Cellular Biology, University of Georgia, Athens, Georgia, USA
| | - Esben Lorentzen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus C, Denmark.
| |
Collapse
|
8
|
Yamaguchi H, Meyer MD, Barrell WB, Faisal M, Berdeaux R, Liu KJ, Komatsu Y. The primary cilia: Orchestrating cranial neural crest cell development. Differentiation 2025; 142:100818. [PMID: 39500655 PMCID: PMC11911094 DOI: 10.1016/j.diff.2024.100818] [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: 08/20/2024] [Revised: 10/28/2024] [Accepted: 10/28/2024] [Indexed: 03/17/2025]
Abstract
Primary cilia (hereafter "cilia") are microtubule-based antenna-like organelles projecting from the surface of vertebrate cells. Cilia can serve as cellular antennae controlling cell growth and differentiation. Absent or dysfunctional cilia frequently lead to craniofacial anomalies known as craniofacial ciliopathies. However, the detailed pathological mechanisms of craniofacial ciliopathies remain unclear. This perspective discusses our current understanding of the role of cilia in cranial neural crest cells. We also describe potential mechanisms of ciliogenesis in cranial neural crest cells, which may contribute to unraveling the complex pathogenesis of craniofacial ciliopathies.
Collapse
Affiliation(s)
- Hiroyuki Yamaguchi
- Department of Pediatrics, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Matthew D Meyer
- Shared Equipment Authority, Rice University, Houston, TX, USA
| | - William B Barrell
- Centre for Craniofacial and Regenerative Biology, King's College London, SE1 9RT, London, UK
| | - Maryam Faisal
- Department of Pediatrics, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, USA; Department of Bioengineering, Rice University George R. Brown School of Engineering, 77005, Houston, TX, USA
| | - Rebecca Berdeaux
- Department of Integrative Biology and Pharmacology, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, USA; CellChorus INC, Houston, TX, USA
| | - Karen J Liu
- Centre for Craniofacial and Regenerative Biology, King's College London, SE1 9RT, London, UK
| | - Yoshihiro Komatsu
- Department of Pediatrics, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, USA; Graduate Program in Genetics & Epigenetics, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, 77030, Houston, TX, USA.
| |
Collapse
|
9
|
Yang S, Wang X, Gao H, Yuan S. Motile cilia: Key developmental and functional roles in reproductive systems. Andrology 2025. [PMID: 39895399 DOI: 10.1111/andr.70007] [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: 11/20/2024] [Revised: 01/13/2025] [Accepted: 01/23/2025] [Indexed: 02/04/2025]
Abstract
BACKGROUND Cilia are specialized microtubule-based organelles that extend from the cell surface and are classified into non-motile and motile types. The assembly and function of cilia are regulated by a complex molecular network that enables motile cilia to generate fluid flow across epithelial surfaces through coordinated beating. These motile cilia are found in the respiratory, nervous, and reproductive systems. In males, motile cilia are found in the efferent ducts and facilitate the transport of sperm from the testis to the epididymis. In females, they are mainly found in the oviducts, where they help to transport, nourish and fertilize eggs, and are also present in the endometrial epithelium. MATERIAL-METHODS This review compares the common factors that affect motile cilia in both male and female reproductive tracts, discusses the origin and development of multiciliated cell and cilia within the efferent ducts and oviducts, and enumerates the infertility or related reproductive diseases that may arise due to motile cilia defects. RESULTS-DISCUSSION In males, motile cilia in the efferent ducts create turbulence through their beating, which keeps semen suspended and prevents ductal obstruction. In females, motile cilia are distributed on the epithelia of the oviducts and the endometrium. Specifically, motile cilia in the infundibulum of the oviduct aid in capturing oocytes, while cilia in the isthmus region have been found to bind to sperm heads, facilitating the formation of the sperm reservoir. Several common factors, such as miR-34b/c and miR-449, TAp73, Gemc1, and estrogen, etc., have been shown to play crucial regulatory roles in motile cilia within the efferent ducts and oviducts, thereby further influencing fertility outcomes. CONCLUSIONS Pathogenic mutations that disrupt ciliary function can impair ciliogenesis or alter the structure of sperm flagella, potentially resulting in infertility. Consequently, motile cilia in both the male and female reproductive tracts are crucial for fertility. There are still numerous unresolved mysteries surrounding these cilia that merit further investigation by researchers, as they hold great significance for the clinical diagnosis and treatment of infertility and related reproductive disorders.
Collapse
Affiliation(s)
- Shiyu Yang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoli Wang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huihui Gao
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuiqiao Yuan
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Laboratory of Animal Center, Huazhong University of Science and Technology, Wuhan, China
- Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, China
| |
Collapse
|
10
|
Harima R, Hara K, Tanemura K. TCTEX1D2 is essential for sperm flagellum formation in mice. Sci Rep 2025; 15:2413. [PMID: 39827215 PMCID: PMC11743150 DOI: 10.1038/s41598-024-83424-1] [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: 10/10/2024] [Accepted: 12/16/2024] [Indexed: 01/22/2025] Open
Abstract
Flagella and cilia are widely conserved motile structures, in mammalian, sperm possess flagella. Large protein complexes called dynein, including cytoplasmic dynein 2 and axonemal dynein, play a role in the formation of cilia and flagella. The function of each subunit component of dynein complexes in sperm flagellum formation remains unclear. One such subunit is TCTEX1D2. Co-immunoprecipitation studies showed that TCTEX1D2 interacted with cytoplasmic dynein 2 subunits WDR34, WDR60, and DYNLT1 in the testes. Furthermore, TCTEX1D2 also interacted with WDR63 and WDR78, subunits of inner dynein arm, which is axonemal dynein. Tctex1d2-/- mice generated in this study exhibited male infertility due to flagellar dysplasia, and the axonemal structures were disrupted inside the flagella. Further, the localization of cytoplasmic dynein 2 subunits was abnormal in in Tctex1d2-/- mice. In contrast, the motile cilia of Tctex1d2-/- mice were normal. Overall, we revealed that TCTEX1D2 is important for the assembly of cytoplasmic dynein 2 and inner dynein arm and functions in two distinct dynein complexes during mouse sperm flagellum formation. This is only in sperm flagellum formation, not in cilia formation.
Collapse
Affiliation(s)
- Ryua Harima
- Laboratory of Animal Reproduction and Development, Graduate School of Agricultural Science, Tohoku University, 468-1 Aramaki-Aoba, Aoba-Ku, Sendai, Miyagi, 980-8572, Japan
| | - Kenshiro Hara
- Laboratory of Animal Reproduction and Development, Graduate School of Agricultural Science, Tohoku University, 468-1 Aramaki-Aoba, Aoba-Ku, Sendai, Miyagi, 980-8572, Japan
| | - Kentaro Tanemura
- Laboratory of Animal Reproduction and Development, Graduate School of Agricultural Science, Tohoku University, 468-1 Aramaki-Aoba, Aoba-Ku, Sendai, Miyagi, 980-8572, Japan.
- Laboratory of Reproductive Technology (Repro-SOLEIL), Graduate School of Agricultural Science, Tohoku University, 468-1 Aramaki-Aoba, Aoba-Ku, Sendai, Miyagi, 980-8572, Japan.
| |
Collapse
|
11
|
Jun JH, Cha H, Ko JY, Kim HS, Yoo KH, Park JH. Loss of Kat2b impairs intraflagellar transport and the Hedgehog signaling pathway in primary cilia. Sci Rep 2025; 15:2127. [PMID: 39820844 PMCID: PMC11739504 DOI: 10.1038/s41598-025-86292-5] [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: 08/14/2024] [Accepted: 01/09/2025] [Indexed: 01/19/2025] Open
Abstract
Primary cilia are sensory organelles that regulate various signaling pathways. When microtubules are compared to a highway, motor proteins carry and transport cargo proteins, which are tuned by post-translational modifications, such as acetylation. However, the role of acetylation in primary cilia regulation remains unclear. In this study, histone K (lysine) acetyltransferase 2 B (Kat2b) was identified as a novel regulator of primary cilia. Kat2b, which mainly regulates transcription as a p300/CBP associated factor, is localized to the cytosol, centrosome, and cilium basal body. In addition, basal Kat2b expression gradually increased during ciliogenesis. Kat2b regulates the rate of cilia assembly, particularly in the early stages, and loss of Kat2b reduces the recruitment of intraflagellar transport (IFT) components to the ciliary axoneme and impairs Hedgehog (Hh) signaling activation. In addition, Kat2b-knockout mice showed mild abnormalities and ciliary IFT defects in the kidneys. These results establish a link between acetylation regulated by Kat2b and its relevance to ciliary assembly and function.
Collapse
Affiliation(s)
- Jae Hee Jun
- Department of Biological Science, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Hwayeon Cha
- Department of Biological Science, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Je Yeong Ko
- Department of Biological Science, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Ho-Shik Kim
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Kyung Hyun Yoo
- Department of Biological Science, Sookmyung Women's University, Seoul, 04310, Republic of Korea.
| | - Jong Hoon Park
- Department of Biological Science, Sookmyung Women's University, Seoul, 04310, Republic of Korea.
| |
Collapse
|
12
|
Liu Y, Fang Y, Dhikhirullahi O, Zhang L, Zhang Z. Intraflagellar Transport (IFT) and Sperm Formation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1469:395-409. [PMID: 40301266 DOI: 10.1007/978-3-031-82990-1_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/01/2025]
Abstract
Intraflagellar transport (IFT) is a conserved mechanism for cilia formation. Twenty-two IFT components form the IFT-A complex (six components) and IFT-B complex (sixteen components). Driven by kinesin and dynein motor proteins, these IFT complexes are involved in the trafficking of proteins needed for cilia assembly by anterograde transport and retrograde transport. IFT core components also associate with other proteins for cilia formation. Mutations in IFT core components result in ciliogenesis defects and human diseases, including male infertility. Sperm flagella are specialized motile cilia that not only have core axoneme structure but also possess accessory structures. IFT is required to assemble these structures to form functional sperm. This summary highlights the regulatory roles of specific IFT proteins in spermatogenesis. A deeper understanding of IFT-related mechanisms can shed light on the etiology and pathophysiology of certain male infertility cases, as well as provide insights for the development of novel male contraceptives.
Collapse
Affiliation(s)
- Yunhao Liu
- School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Yu Fang
- School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | | | - Ling Zhang
- School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Zhibing Zhang
- Department of Physiology, Wayne State University, Detroit, MI, USA.
- Department of Obstetrics & Gynecology, Wayne State University, Detroit, MI, USA.
| |
Collapse
|
13
|
Palicharla VR, Badgandi HB, Hwang SH, Legué E, Liem KF, Mukhopadhyay S. A defined tubby domain β-barrel surface region of TULP3 mediates ciliary trafficking of diverse cargoes. Mol Biol Cell 2025; 36:ar1. [PMID: 39565681 PMCID: PMC11742108 DOI: 10.1091/mbc.e24-09-0426] [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: 09/27/2024] [Revised: 11/05/2024] [Accepted: 11/12/2024] [Indexed: 11/22/2024] Open
Abstract
The primary cilium is a paradigmatic subcellular compartment at the nexus of numerous cellular and morphogenetic pathways. The tubby family protein TULP3 acts as an adapter of the intraflagellar transport complex A in transporting integral membrane and membrane-associated lipidated proteins into cilia. However, the mechanisms by which TULP3 coordinates ciliary transport of diverse cargoes is not well understood. Here, we provide molecular insights into TULP3-mediated ciliary cargo recognition. We screened for critical TULP3 residues by proximity biotinylation-mass spectrometry, structural analysis, and testing TULP3 variants in human patients with hepatorenal fibrocystic disease and spina bifida. The TULP3 residues we identified 1) were located on one side of the β-barrel of the tubby domain away from the phosphoinositide binding site, 2) mediated ciliary trafficking of lipidated and transmembrane cargoes, and 3) determined proximity with these cargoes in vivo without affecting ciliary localization, phosphoinositide binding or hydrodynamic properties of TULP3. Overall, these findings implicate a specific region of one of the surfaces of the TULP3 β-barrel in ciliary trafficking of diverse cargoes. This region overlooks the β-strands 8-12 of the β-barrel and is away from the membrane anchoring phosphoinositide binding site. Targeting the TULP3-cargo interactions could provide therapeutics in ciliary trafficking diseases.
Collapse
Affiliation(s)
- Vivek Reddy Palicharla
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Hemant B. Badgandi
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Sun-Hee Hwang
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Emilie Legué
- Vertebrate Developmental Biology Program, Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06520
| | - Karel F. Liem
- Vertebrate Developmental Biology Program, Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06520
| | - Saikat Mukhopadhyay
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| |
Collapse
|
14
|
Constable S, Ott CM, Lemire AL, White K, Xun Y, Lim A, Lippincott-Schwartz J, Mukhopadhyay S. Permanent cilia loss during cerebellar granule cell neurogenesis involves withdrawal of cilia maintenance and centriole capping. Proc Natl Acad Sci U S A 2024; 121:e2408083121. [PMID: 39705308 DOI: 10.1073/pnas.2408083121] [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: 04/22/2024] [Accepted: 11/09/2024] [Indexed: 12/22/2024] Open
Abstract
Brain neurons utilize the primary cilium as a privileged compartment to detect and respond to extracellular ligands such as Sonic hedgehog (SHH). However, cilia in cerebellar granule cell (GC) neurons disassemble during differentiation through ultrastructurally unique intermediates, a process we refer to as cilia deconstruction. In addition, mature neurons do not reciliate despite having docked centrioles. Here, we identify molecular changes that accompany cilia deconstruction and centriole docking in GC neurons. We used single cell transcriptomic and immunocytological analyses to compare the transcript levels and subcellular localization of proteins between progenitor, differentiating, and mature GCs. Differentiating GCs lacked transcripts for key activators of premitotic cilia resorption, indicating that cilia disassembly in differentiating cells is distinct from premitotic cilia resorption. Instead, during differentiation, transcripts of many genes required for cilia maintenance-specifically those encoding components of intraflagellar transport, pericentrosomal material, and centriolar satellites-decreased. The abundance of several corresponding proteins in and around cilia and centrosomes also decreased. These changes coincided with downregulation of SHH signaling prior to differentiation, even in a mutant with excessive SHH activation. Finally, mother centrioles in maturing granule neurons recruited the cap complex protein, CEP97. These data suggest that a global, developmentally programmed decrease in cilium maintenance in differentiating GCs mediates cilia deconstruction, while capping of docked mother centrioles prevents cilia regrowth and dysregulated SHH signaling. Our study provides mechanistic insights expanding our understanding of permanent cilia loss in multiple tissue-specific contexts.
Collapse
Affiliation(s)
- Sandii Constable
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Carolyn M Ott
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147
| | - Andrew L Lemire
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147
| | - Kevin White
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Yu Xun
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Amin Lim
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | | | - Saikat Mukhopadhyay
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| |
Collapse
|
15
|
Hufft-Martinez BM, Wang HH, Saadi I, Tran PV. Actin cytoskeletal regulation of ciliogenesis in development and disease. Dev Dyn 2024; 253:1076-1093. [PMID: 38958410 PMCID: PMC11611694 DOI: 10.1002/dvdy.724] [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: 04/07/2024] [Revised: 05/29/2024] [Accepted: 06/15/2024] [Indexed: 07/04/2024] Open
Abstract
Primary cilia are antenna-like sensory organelles that are evolutionarily conserved in nearly all modern eukaryotes, from the single-celled green alga, Chlamydomonas reinhardtii, to vertebrates and mammals. Cilia are microtubule-based cellular projections that have adapted to perform a broad range of species-specific functions, from cell motility to detection of light and the transduction of extracellular mechanical and chemical signals. These functions render cilia essential for organismal development and survival. The high conservation of cilia has allowed for discoveries in C. reinhardtii to inform our understanding of the basic biology of mammalian primary cilia, and to provide insight into the genetic etiology of ciliopathies. Over the last two decades, a growing number of studies has revealed that multiple aspects of ciliary homeostasis are regulated by the actin cytoskeleton, including centrosome migration and positioning, vesicle transport to the basal body, ectocytosis, and ciliary-mediated signaling. Here, we review actin regulation of ciliary homeostasis, and highlight conserved and divergent mechanisms in C. reinhardtii and mammalian cells. Further, we compare the disease manifestations of patients with ciliopathies to those with mutations in actin and actin-associated genes, and propose that primary cilia defects caused by genetic alteration of the actin cytoskeleton may underlie certain birth defects.
Collapse
Affiliation(s)
| | - Henry H Wang
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS
| | - Irfan Saadi
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS
- Institute of Reproductive and Developmental Sciences, University of Kansas Medical Center, Kansas City, KS
| | - Pamela V Tran
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS
- Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS
| |
Collapse
|
16
|
Esser H, Kilpatrick AM, Man TY, Aird R, Rodrigo-Torres D, Buch ML, Boulter L, Walmsley S, Oniscu GC, Schneeberger S, Ferreira-Gonzalez S, Forbes SJ. Primary cilia as a targetable node between biliary injury, senescence and regeneration in liver transplantation. J Hepatol 2024; 81:1005-1022. [PMID: 38879173 DOI: 10.1016/j.jhep.2024.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/05/2024] [Accepted: 06/01/2024] [Indexed: 09/27/2024]
Abstract
BACKGROUND & AIMS Biliary complications are a major cause of morbidity and mortality in liver transplantation. Up to 25% of patients that develop biliary complications require additional surgical procedures, re-transplantation or die in the absence of a suitable regraft. Here, we investigate the role of the primary cilium, a highly specialised sensory organelle, in biliary injury leading to post-transplant biliary complications. METHODS Human biopsies were used to study the structure and function of primary cilia in liver transplant recipients that develop biliary complications (n = 7) in comparison with recipients without biliary complications (n = 12). To study the biological effects of the primary cilia during transplantation, we generated murine models that recapitulate liver procurement and cold storage, and assessed the elimination of the primary cilia in biliary epithelial cells in the K19CreERTKif3afl/fl mouse model. To explore the molecular mechanisms responsible for the observed phenotypes we used in vitro models of ischemia, cellular senescence and primary cilia ablation. Finally, we used pharmacological and genetic approaches to target cellular senescence and the primary cilia, both in mouse models and discarded human donor livers. RESULTS Prolonged ischemic periods before transplantation result in ciliary shortening and cellular senescence, an irreversible cell cycle arrest that blocks regeneration. Our results indicate that primary cilia damage results in biliary injury and a loss of regenerative potential. Senescence negatively impacts primary cilia structure and triggers a negative feedback loop that further impairs regeneration. Finally, we explore how targeted interventions for cellular senescence and/or the stabilisation of the primary cilia improve biliary regeneration following ischemic injury. CONCLUSIONS Primary cilia play an essential role in biliary regeneration and we demonstrate that senolytics and cilia-stabilising treatments provide a potential therapeutic opportunity to reduce the rate of biliary complications and improve clinical outcomes in liver transplantation. IMPACT AND IMPLICATIONS Up to 25% of liver transplants result in biliary complications, leading to additional surgery, retransplants, or death. We found that the incidence of biliary complications is increased by damage to the primary cilium, an antenna that protrudes from the cell and is key to regeneration. Here, we show that treatments that preserve the primary cilia during the transplant process provide a potential solution to reduce the rates of biliary complications.
Collapse
Affiliation(s)
- Hannah Esser
- MRC Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK; Department of Visceral, Transplant and Thoracic Surgery, OrganLife Laboratory, Centre of Operative Medicine, Innsbruck Medical University. Anichstrasse 35, 6020 Innsbruck, Austria
| | - Alastair Morris Kilpatrick
- MRC Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Tak Yung Man
- MRC Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Rhona Aird
- MRC Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Daniel Rodrigo-Torres
- MRC Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Madita Lina Buch
- MRC Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK; Department of Visceral, Transplant and Thoracic Surgery, OrganLife Laboratory, Centre of Operative Medicine, Innsbruck Medical University. Anichstrasse 35, 6020 Innsbruck, Austria
| | - Luke Boulter
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh; Edinburgh EH4 2XU, UK
| | - Sarah Walmsley
- Centre for Inflammation Research (CIR), University of Edinburgh. The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Gabriel Corneliu Oniscu
- Edinburgh Transplant Centre, Royal Infirmary of Edinburgh; 51 Little France Crescent, Edinburgh EH16 4SA, UK; Division of Transplantation, CLINTEC, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Stefan Schneeberger
- Department of Visceral, Transplant and Thoracic Surgery, OrganLife Laboratory, Centre of Operative Medicine, Innsbruck Medical University. Anichstrasse 35, 6020 Innsbruck, Austria
| | - Sofia Ferreira-Gonzalez
- MRC Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK; Centre for Inflammation Research (CIR), University of Edinburgh. The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, UK.
| | - Stuart John Forbes
- MRC Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK.
| |
Collapse
|
17
|
Rodríguez-González D, Guillén-Sánchez G, Del Pozo V, Cañas JA. Single-Cell Analysis: A Method for In-Depth Phenotyping of Cells Involved in Asthma. Int J Mol Sci 2024; 25:12633. [PMID: 39684345 DOI: 10.3390/ijms252312633] [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: 11/01/2024] [Revised: 11/20/2024] [Accepted: 11/23/2024] [Indexed: 12/18/2024] Open
Abstract
Asthma is a chronic inflammatory lung disease with high prevalence, making it one of the most common chronic conditions worldwide. Its pathophysiology is influenced by a range of genetic and environmental factors, resulting in a complex and heterogeneous disease profile. Asthma is primarily associated with a type 2 (T2) immune response, though non-T2 endotypes also contribute to disease pathology. Generally, asthma is characterized by the infiltration and activation of various cell types, including dendritic cells, eosinophils, innate lymphoid cells, lymphocytes, mast cells, and neutrophils, which participate in T1, T2, and T17 immune responses. Despite advances in understanding, many questions remain unresolved. Therefore, emerging omic techniques, such as single-cell RNA sequencing (scRNA-seq), offer novel insights into the underlying mechanisms of asthma and the roles of these immune cells. Recent scRNA-seq studies in asthma have identified multiple novel immune cell subtypes and clusters, suggesting their potential functions in disease pathology. The rapid advancement of scRNA-seq technology now enables in-depth investigation of individual cells within tissues, allowing for precise cell-type classification and detailed molecular profiling. Nonetheless, certain limitations persist, which require further refinement in future studies.
Collapse
Affiliation(s)
- Daniel Rodríguez-González
- Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Universidad Autónoma de Madrid (UAM), 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Respiratorias (CIBERES), 28029 Madrid, Spain
| | - Gema Guillén-Sánchez
- Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Universidad Autónoma de Madrid (UAM), 28040 Madrid, Spain
| | - Victoria Del Pozo
- Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Universidad Autónoma de Madrid (UAM), 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Respiratorias (CIBERES), 28029 Madrid, Spain
- Medicine Department, School of Medicine, Faculty of Medicine, Campus of Medicine, Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain
| | - José Antonio Cañas
- Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Universidad Autónoma de Madrid (UAM), 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Respiratorias (CIBERES), 28029 Madrid, Spain
| |
Collapse
|
18
|
Fitzsimons LA, Staurengo-Ferrari L, Khomula EV, Bogen O, Araldi D, Bonet IJM, Green PG, Jordan EE, Sclafani F, Nowak CE, Moulton JK, Ganter GK, Levine JD, Tucker KL. The Nociceptor Primary Cilium Contributes to Mechanical Nociceptive Threshold and Inflammatory and Neuropathic Pain. J Neurosci 2024; 44:e1265242024. [PMID: 39349056 PMCID: PMC11580782 DOI: 10.1523/jneurosci.1265-24.2024] [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: 07/02/2024] [Revised: 08/16/2024] [Accepted: 09/20/2024] [Indexed: 10/02/2024] Open
Abstract
The primary cilium, a single microtubule-based organelle protruding from the cell surface and critical for neural development, also functions in adult neurons. While some dorsal root ganglion neurons elaborate a primary cilium, whether it is expressed by and functional in nociceptors is unknown. Recent studies have shown the role of Hedgehog, whose canonical signaling is primary cilium dependent, in nociceptor sensitization. We establish the presence of primary cilia in soma of rat nociceptors, where they contribute to mechanical threshold, prostaglandin E2 (PGE2)-induced hyperalgesia, and chemotherapy-induced neuropathic pain (CIPN). Intrathecal administration of siRNA targeting Ift88, a primary cilium-specific intraflagellar transport (IFT) protein required for ciliary integrity, resulted in attenuation of Ift88 mRNA and nociceptor primary cilia. Attenuation of primary cilia was associated with an increase in mechanical nociceptive threshold in vivo and decrease in nociceptor excitability in vitro, abrogation of hyperalgesia, and nociceptor sensitization induced by both a prototypical pronociceptive inflammatory mediator PGE2 and paclitaxel CIPN, in a sex-specific fashion. siRNA targeting Ift52, another IFT protein, and knockdown of NompB, the Drosophila Ift88 ortholog, also abrogated CIPN and reduced baseline mechanosensitivity, respectively, providing independent confirmation for primary cilia control of nociceptor function. Hedgehog-induced hyperalgesia is attenuated by Ift88 siRNA, supporting the role for primary cilia in Hedgehog-induced hyperalgesia. Attenuation of CIPN by cyclopamine (intradermal and intraganglion), which inhibits Hedgehog signaling, supports the role of Hedgehog in CIPN. Our findings support the role of the nociceptor primary cilium in control of mechanical nociceptive threshold and inflammatory and neuropathic pain, the latter Hedgehog-dependent.
Collapse
Affiliation(s)
- Lindsey A Fitzsimons
- Deparment of Biomedical Sciences, College of Osteopathic Medicine, University of New England, Biddeford, Maine 04005
- Center for Excellence in the Neurosciences, University of New England, Biddeford, Maine 04005
| | - Larissa Staurengo-Ferrari
- Department of Oral and Maxillofacial Surgery, UCSF Pain and Addiction Research Center, University of California San Francisco, San Francisco 94115
| | - Eugen V Khomula
- Department of Oral and Maxillofacial Surgery, UCSF Pain and Addiction Research Center, University of California San Francisco, San Francisco 94115
| | - Oliver Bogen
- Department of Oral and Maxillofacial Surgery, UCSF Pain and Addiction Research Center, University of California San Francisco, San Francisco 94115
| | - Dionéia Araldi
- Department of Oral and Maxillofacial Surgery, UCSF Pain and Addiction Research Center, University of California San Francisco, San Francisco 94115
| | - Ivan J M Bonet
- Department of Oral and Maxillofacial Surgery, UCSF Pain and Addiction Research Center, University of California San Francisco, San Francisco 94115
| | - Paul G Green
- Department of Oral and Maxillofacial Surgery, UCSF Pain and Addiction Research Center, University of California San Francisco, San Francisco 94115
- Department of Preventative and Restorative Dental Sciences, University of California San Francisco, San Francisco 94115
| | - Ethan E Jordan
- Deparment of Biomedical Sciences, College of Osteopathic Medicine, University of New England, Biddeford, Maine 04005
- Center for Excellence in the Neurosciences, University of New England, Biddeford, Maine 04005
| | - Finn Sclafani
- Center for Excellence in the Neurosciences, University of New England, Biddeford, Maine 04005
- School of Biological Sciences, College of Arts and Sciences, University of New England, Biddeford, Maine 04005
| | - Connor E Nowak
- Center for Excellence in the Neurosciences, University of New England, Biddeford, Maine 04005
- School of Biological Sciences, College of Arts and Sciences, University of New England, Biddeford, Maine 04005
| | - Julie K Moulton
- Center for Excellence in the Neurosciences, University of New England, Biddeford, Maine 04005
- School of Biological Sciences, College of Arts and Sciences, University of New England, Biddeford, Maine 04005
| | - Geoffrey K Ganter
- Center for Excellence in the Neurosciences, University of New England, Biddeford, Maine 04005
- School of Biological Sciences, College of Arts and Sciences, University of New England, Biddeford, Maine 04005
| | - Jon D Levine
- Department of Oral and Maxillofacial Surgery, UCSF Pain and Addiction Research Center, University of California San Francisco, San Francisco 94115
- Department of Medicine, Division of Neuroscience, University of California San Francisco, San Francisco 94115
| | - Kerry L Tucker
- Deparment of Biomedical Sciences, College of Osteopathic Medicine, University of New England, Biddeford, Maine 04005
- Center for Excellence in the Neurosciences, University of New England, Biddeford, Maine 04005
| |
Collapse
|
19
|
Spallotta F, Illi B. The Role of HDAC6 in Glioblastoma Multiforme: A New Avenue to Therapeutic Interventions? Biomedicines 2024; 12:2631. [PMID: 39595195 PMCID: PMC11591585 DOI: 10.3390/biomedicines12112631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/14/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
Despite the great advances in basic research results, glioblastoma multiforme (GBM) still remains an incurable tumour. To date, a GBM diagnosis is a death sentence within 15-18 months, due to the high recurrence rate and resistance to conventional radio- and chemotherapy approaches. The effort the scientific community is lavishing on the never-ending battle against GBM is reflected by the huge number of clinical trials launched, about 2003 on 10 September 2024. However, we are still far from both an in-depth comprehension of the biological and molecular processes leading to GBM onset and progression and, importantly, a cure. GBM is provided with high intratumoral heterogeneity, immunosuppressive capacity, and infiltrative ability due to neoangiogenesis. These features impact both tumour aggressiveness and therapeutic vulnerability, which is further limited by the presence in the tumour core of niches of glioblastoma stem cells (GSCs) that are responsible for the relapse of this brain neoplasm. Epigenetic alterations may both drive and develop along GBM progression and also rely on changes in the expression of the genes encoding histone-modifying enzymes, including histone deacetylases (HDACs). Among them, HDAC6-a cytoplasmic HDAC-has recently gained attention because of its role in modulating several biological aspects of GBM, including DNA repair ability, massive growth, radio- and chemoresistance, and de-differentiation through primary cilia disruption. In this review article, the available information related to HDAC6 function in GBM will be presented, with the aim of proposing its inhibition as a valuable therapeutic route for this deadly brain tumour.
Collapse
Affiliation(s)
- Francesco Spallotta
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, 00185 Rome, Italy;
- Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, 00185 Rome, Italy
| | - Barbara Illi
- Institute of Molecular Biology and Pathology, National Research Council (IBPM-CNR), 00185 Rome, Italy
| |
Collapse
|
20
|
Riparbelli MG, Pratelli A, Callaini G. The cilium like region of the Drosophila bifurca spermatocyte: Elongation of a giant axoneme without intraflagellar transport. Cytoskeleton (Hoboken) 2024; 81:529-538. [PMID: 38073091 DOI: 10.1002/cm.21816] [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: 11/01/2023] [Revised: 11/26/2023] [Accepted: 11/27/2023] [Indexed: 11/20/2024]
Abstract
The growth of the ciliary axonemes mainly depends on the evolutionary conserved intraflagellar transport (IFT) machinery. However, insect spermatocytes are characterized by cilium-like regions (CLRs) that elongate in the absence of IFT. It is generally believed that the dynamics of these structures relies on the free diffusion of soluble tubulin from the cytoplasm. However, this passive process could allow the elongation of short ciliary axonemes, but it is unclear whether simple diffusion of tubulin molecules can ensure the correct assembly of elongated ciliary structures. To decipher this point we analyzed the assembly of the CLRs held by the primary spermatocytes of Drosophila bifurca. These ciliary structures consist of a very elongated axoneme that grows without IFT and, therefore, could represent a good model in which to evaluate the role played by the free diffusion of soluble tubulin. The observation of wavy microtubules in the axonemal lumen of fully elongated CLRs of D. bifurca may be consistent with the diffusion of tubulin within the axonemal lumen. Progressive consumption of soluble tubulin used for axoneme growth at the apical tip of the CLRs could result in a gradient sufficient to move tubulin from the cytoplasm to the apical end of the forming ciliary structure. When the axoneme reaches its full length, tubulin molecules are not drawn to the tip of the CLRs and accumulate at the base of the axoneme, where its concentration may exceed the threshold need for microtubule polymerization. The presence of γ-TuRCs at the proximal ends of the supernumerary microtubules could enhance their nucleation.
Collapse
Affiliation(s)
| | - Ambra Pratelli
- Department of Life Sciences, University of Siena, Siena, Italy
| | | |
Collapse
|
21
|
Patel MB, Griffin PJ, Olson SF, Dai J, Hou Y, Malik T, Das P, Zhang G, Zhao W, Witman GB, Lechtreck KF. Distribution and bulk flow analyses of the intraflagellar transport (IFT) motor kinesin-2 support an "on-demand" model for Chlamydomonas ciliary length control. Cytoskeleton (Hoboken) 2024; 81:586-604. [PMID: 38456596 PMCID: PMC11380706 DOI: 10.1002/cm.21851] [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: 10/30/2023] [Revised: 02/22/2024] [Accepted: 02/23/2024] [Indexed: 03/09/2024]
Abstract
Most cells tightly control the length of their cilia. The regulation likely involves intraflagellar transport (IFT), a bidirectional motility of multi-subunit particles organized into trains that deliver building blocks into the organelle. In Chlamydomonas, the anterograde IFT motor kinesin-2 consists of the motor subunits FLA8 and FLA10 and the nonmotor subunit KAP. KAP dissociates from IFT at the ciliary tip and diffuses back to the cell body. This observation led to the diffusion-as-a-ruler model of ciliary length control, which postulates that KAP is progressively sequestered into elongating cilia because its return to the cell body will require increasingly more time, limiting motor availability at the ciliary base, train assembly, building block supply, and ciliary growth. Here, we show that Chlamydomonas FLA8 also returns to the cell body by diffusion. However, more than 95% of KAP and FLA8 are present in the cell body and, at a given time, just ~1% of the motor participates in IFT. After repeated photobleaching of both cilia, IFT of fluorescent kinesin subunits continued indicating that kinesin-2 cycles from the large cell-body pool through the cilia and back. Furthermore, growing and full-length cilia contained similar amounts of kinesin-2 subunits and the size of the motor pool at the base changed only slightly with ciliary length. These observations are incompatible with the diffusion-as-a-ruler model, but rather support an "on-demand model," in which the cargo load of the trains is regulated to assemble cilia of the desired length.
Collapse
Affiliation(s)
- Mansi B Patel
- Department of Cellular Biology, University of Georgia, Athens, Georgia, USA
| | - Paul J Griffin
- Department of Cellular Biology, University of Georgia, Athens, Georgia, USA
| | - Spencer F Olson
- Department of Cellular Biology, University of Georgia, Athens, Georgia, USA
| | - Jin Dai
- Department of Cellular Biology, University of Georgia, Athens, Georgia, USA
| | - Yuqing Hou
- Department of Radiology, UMass Chan Medical School, Worcester, Massachusetts, USA
| | - Tara Malik
- Department of Cellular Biology, University of Georgia, Athens, Georgia, USA
| | - Poulomi Das
- Department of Cellular Biology, University of Georgia, Athens, Georgia, USA
| | - Gui Zhang
- Department of Cellular Biology, University of Georgia, Athens, Georgia, USA
| | - Winston Zhao
- Department of Radiology, UMass Chan Medical School, Worcester, Massachusetts, USA
| | - George B Witman
- Department of Radiology, UMass Chan Medical School, Worcester, Massachusetts, USA
| | - Karl F Lechtreck
- Department of Cellular Biology, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
22
|
Philbrook A, O’Donnell MP, Grunenkovaite L, Sengupta P. Cilia structure and intraflagellar transport differentially regulate sensory response dynamics within and between C. elegans chemosensory neurons. PLoS Biol 2024; 22:e3002892. [PMID: 39591402 PMCID: PMC11593760 DOI: 10.1371/journal.pbio.3002892] [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: 05/10/2024] [Accepted: 10/10/2024] [Indexed: 11/28/2024] Open
Abstract
Sensory neurons contain morphologically diverse primary cilia that are built by intraflagellar transport (IFT) and house sensory signaling molecules. Since both ciliary structural and signaling proteins are trafficked via IFT, it has been challenging to decouple the contributions of IFT and cilia structure to neuronal responses. By acutely inhibiting IFT without altering cilia structure and vice versa, here we describe the differential roles of ciliary trafficking and sensory ending morphology in shaping chemosensory responses in Caenorhabditis elegans. We show that a minimum cilium length but not continuous IFT is necessary for a subset of responses in the ASH nociceptive neurons. In contrast, neither cilia nor continuous IFT are necessary for odorant responses in the AWA olfactory neurons. Instead, continuous IFT differentially modulates response dynamics in AWA. Upon acute inhibition of IFT, cilia-destined odorant receptors are shunted to ectopic branches emanating from the AWA cilia base. Spatial segregation of receptors in these branches from a cilia-restricted regulatory kinase results in odorant desensitization defects, highlighting the importance of precise organization of signaling molecules at sensory endings in regulating response dynamics. We also find that adaptation of AWA responses upon repeated exposure to an odorant is mediated by IFT-driven removal of its cognate receptor, whereas adaptation to a second odorant is regulated via IFT-independent mechanisms. Our results reveal unexpected complexity in the contribution of IFT and cilia organization to the regulation of responses even within a single chemosensory neuron type and establish a critical role for these processes in the precise modulation of olfactory behaviors.
Collapse
Affiliation(s)
- Alison Philbrook
- Department of Biology, Brandeis University, Waltham, Massachusetts, United States of America
| | - Michael P. O’Donnell
- Department of Molecular, Cellular, and Developmental Biology, Yale University, Connecticut, United States of America
| | - Laura Grunenkovaite
- Department of Biology, Brandeis University, Waltham, Massachusetts, United States of America
| | - Piali Sengupta
- Department of Biology, Brandeis University, Waltham, Massachusetts, United States of America
| |
Collapse
|
23
|
Chaya T, Maeda Y, Tsutsumi R, Ando M, Ma Y, Kajimura N, Tanaka T, Furukawa T. Ccrk-Mak/Ick signaling is a ciliary transport regulator essential for retinal photoreceptor survival. Life Sci Alliance 2024; 7:e202402880. [PMID: 39293864 PMCID: PMC11412320 DOI: 10.26508/lsa.202402880] [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: 06/08/2024] [Revised: 08/27/2024] [Accepted: 08/27/2024] [Indexed: 09/20/2024] Open
Abstract
Primary cilia are microtubule-based sensory organelles whose dysfunction causes ciliopathies in humans. The formation, function, and maintenance of primary cilia depend crucially on intraflagellar transport (IFT); however, the regulatory mechanisms of IFT at ciliary tips are poorly understood. Here, we identified that the ciliopathy kinase Mak is a ciliary tip-localized IFT regulator that cooperatively acts with the ciliopathy kinase Ick, an IFT regulator. Simultaneous disruption of Mak and Ick resulted in loss of photoreceptor ciliary axonemes and severe retinal degeneration. Gene delivery of Ick and pharmacological inhibition of FGF receptors, Ick negative regulators, ameliorated retinal degeneration in Mak -/- mice. We also identified that Ccrk kinase is an upstream activator of Mak and Ick in retinal photoreceptor cells. Furthermore, the overexpression of Mak, Ick, and Ccrk and pharmacological inhibition of FGF receptors suppressed ciliopathy-related phenotypes caused by cytoplasmic dynein inhibition in cultured cells. Collectively, our results show that the Ccrk-Mak/Ick axis is an IFT regulator essential for retinal photoreceptor maintenance and present activation of Ick as a potential therapeutic approach for retinitis pigmentosa caused by MAK mutations.
Collapse
Affiliation(s)
- Taro Chaya
- Laboratory for Molecular and Developmental Biology, Institute for Protein Research, Osaka University, Osaka, Japan
| | - Yamato Maeda
- Laboratory for Molecular and Developmental Biology, Institute for Protein Research, Osaka University, Osaka, Japan
| | - Ryotaro Tsutsumi
- Laboratory for Molecular and Developmental Biology, Institute for Protein Research, Osaka University, Osaka, Japan
| | - Makoto Ando
- Laboratory for Molecular and Developmental Biology, Institute for Protein Research, Osaka University, Osaka, Japan
| | - Yujie Ma
- Laboratory for Molecular and Developmental Biology, Institute for Protein Research, Osaka University, Osaka, Japan
| | - Naoko Kajimura
- Research Center for Ultra-High Voltage Electron Microscopy, Osaka University, Osaka, Japan
| | - Teruyuki Tanaka
- Department of Developmental Medical Sciences, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takahisa Furukawa
- Laboratory for Molecular and Developmental Biology, Institute for Protein Research, Osaka University, Osaka, Japan
| |
Collapse
|
24
|
Kawasaki M, Kawasaki K, Sari FT, Kudo T, Nihara J, Kitamura M, Nagai T, Utama V, Ishida Y, Meguro F, Kesuma A, Fujita A, Nishimura T, Kogure Y, Maruyama S, Tanuma JI, Kakihara Y, Maeda T, Ghafoor S, Khonsari RH, Corre P, Sharpe PT, Cobourne M, Franco B, Ohazama A. Cell-cell interaction determines cell fate of mesoderm-derived cell in tongue development through Hh signaling. eLife 2024; 13:e85042. [PMID: 39392396 PMCID: PMC11469673 DOI: 10.7554/elife.85042] [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: 11/19/2022] [Accepted: 09/03/2024] [Indexed: 10/12/2024] Open
Abstract
Dysfunction of primary cilia leads to genetic disorder, ciliopathies, which shows various malformations in many vital organs such as brain. Multiple tongue deformities including cleft, hamartoma, and ankyloglossia are also seen in ciliopathies, which yield difficulties in fundamental functions such as mastication and vocalization. Here, we found these tongue anomalies in mice with mutation of ciliary protein. Abnormal cranial neural crest-derived cells (CNCC) failed to evoke Hh signal for differentiation of mesoderm-derived cells into myoblasts, which resulted in abnormal differentiation of mesoderm-derived cells into adipocytes. The ectopic adipose subsequently arrested tongue swelling formation. Ankyloglossia was caused by aberrant cell migration due to lack of non-canonical Wnt signaling. In addition to ciliopathies, these tongue anomalies are often observed as non-familial condition in human. We found that these tongue deformities could be reproduced in wild-type mice by simple mechanical manipulations to disturb cellular processes which were disrupted in mutant mice. Our results provide hints for possible future treatment in ciliopathies.
Collapse
Affiliation(s)
- Maiko Kawasaki
- Division of Oral Anatomy, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata UniversityNiigataJapan
| | - Katsushige Kawasaki
- Division of Oral Anatomy, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata UniversityNiigataJapan
- Center for Advanced Oral Science, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata UniversityNiigataJapan
| | - Finsa Tisna Sari
- Division of Oral Anatomy, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata UniversityNiigataJapan
| | - Takehisa Kudo
- Division of Oral Anatomy, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata UniversityNiigataJapan
- Division of Orthodontics, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata UniversityNiigataJapan
| | - Jun Nihara
- Division of Orthodontics, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata UniversityNiigataJapan
| | - Madoka Kitamura
- Division of Oral Anatomy, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata UniversityNiigataJapan
- Division of Orthodontics, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata UniversityNiigataJapan
| | - Takahiro Nagai
- Division of Oral Anatomy, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata UniversityNiigataJapan
| | - Vanessa Utama
- Division of Oral Anatomy, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata UniversityNiigataJapan
| | - Yoko Ishida
- Center for Advanced Oral Science, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata UniversityNiigataJapan
| | - Fumiya Meguro
- Division of Oral Anatomy, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata UniversityNiigataJapan
| | - Alex Kesuma
- Division of Oral Anatomy, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata UniversityNiigataJapan
| | - Akira Fujita
- Division of Orthodontics, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata UniversityNiigataJapan
| | - Takayuki Nishimura
- Division of Oral Anatomy, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata UniversityNiigataJapan
| | - Yuan Kogure
- Division of Oral Anatomy, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata UniversityNiigataJapan
| | - Satoshi Maruyama
- Division of Oral Pathology, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata UniversityNiigataJapan
| | - Jun-ichi Tanuma
- Division of Oral Pathology, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata UniversityNiigataJapan
| | - Yoshito Kakihara
- Division of Dental Pharmacology, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata UniversityNiigataJapan
| | - Takeyasu Maeda
- Center for Advanced Oral Science, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata UniversityNiigataJapan
| | - Sarah Ghafoor
- Centre for Craniofacial & Regenerative Biology, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, Guy’s HospitalLondonUnited Kingdom
| | - Roman H Khonsari
- Centre for Craniofacial & Regenerative Biology, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, Guy’s HospitalLondonUnited Kingdom
| | - Pierre Corre
- Service de Chirurgie Maxillofaciale et tomatology, Centre Hospitalier Universitaire de Nantes,1 place Alexis Ricordeau 44000NantesFrance
| | - Paul T Sharpe
- Centre for Craniofacial & Regenerative Biology, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, Guy’s HospitalLondonUnited Kingdom
| | - Martyn Cobourne
- Centre for Craniofacial & Regenerative Biology, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, Guy’s HospitalLondonUnited Kingdom
| | - Brunella Franco
- Telethon Institute of Genetics and Medicine (TIGEM), PozzuoliNaplesItaly
- Medical Genetics, Department of Translational Medical Sciences, Federico II University of Naples, ItalyNaplesItaly
- Scuola Superiore Meridionale, School for Advanced Studies, Genomics and Experimental Medicine program,NaplesItaly
| | - Atsushi Ohazama
- Division of Oral Anatomy, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata UniversityNiigataJapan
| |
Collapse
|
25
|
Ott CM, Constable S, Nguyen TM, White K, Lee WCA, Lippincott-Schwartz J, Mukhopadhyay S. Permanent deconstruction of intracellular primary cilia in differentiating granule cell neurons. J Cell Biol 2024; 223:e202404038. [PMID: 39137043 PMCID: PMC11320830 DOI: 10.1083/jcb.202404038] [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: 04/09/2024] [Revised: 06/03/2024] [Accepted: 06/26/2024] [Indexed: 08/15/2024] Open
Abstract
Primary cilia on granule cell neuron progenitors in the developing cerebellum detect sonic hedgehog to facilitate proliferation. Following differentiation, cerebellar granule cells become the most abundant neuronal cell type in the brain. While granule cell cilia are essential during early developmental stages, they become infrequent upon maturation. Here, we provide nanoscopic resolution of cilia in situ using large-scale electron microscopy volumes and immunostaining of mouse cerebella. In many granule cells, we found intracellular cilia, concealed from the external environment. Cilia were disassembled in differentiating granule cell neurons-in a process we call cilia deconstruction-distinct from premitotic cilia resorption in proliferating progenitors. In differentiating granule cells, cilia deconstruction involved unique disassembly intermediates, and, as maturation progressed, mother centriolar docking at the plasma membrane. Unlike ciliated neurons in other brain regions, our results show the deconstruction of concealed cilia in differentiating granule cells, which might prevent mitogenic hedgehog responsiveness. Ciliary deconstruction could be paradigmatic of cilia removal during differentiation in other tissues.
Collapse
Affiliation(s)
- Carolyn M Ott
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Sandii Constable
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Tri M Nguyen
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Kevin White
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Wei-Chung Allen Lee
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Saikat Mukhopadhyay
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
26
|
Ku PI, Sreeja JS, Chadha A, Williams DS, Engelke MF, Subramanian R. Collaborative role of two distinct cilium-specific cytoskeletal systems in driving Hedgehog-responsive transcription factor trafficking. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.26.615198. [PMID: 39386719 PMCID: PMC11463396 DOI: 10.1101/2024.09.26.615198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Calibrated transcriptional outputs in cellular signaling require fine regulation of transcription factor activity. In vertebrate Hedgehog (Hh) signaling, the precise output of the final effectors, the GLI (Glioma-associated-oncogene) transcription factors, depends on the primary cilium. In particular, the formation of the activator form of GLI upon pathway initiation requires its concentration at the distal cilium tip. However, the mechanisms underlying this critical step in Hh signaling are unclear. We developed a real-time imaging assay to visualize GLI2, the primary GLI activator isoform, at single particle resolution within the cilium. We observed that GLI2 is a cargo of Intraflagellar Transport (IFT) machinery and is transported with anterograde bias during a restricted time window following pathway activation. Our findings position IFT as a crucial mediator of transcription factor transport within the cilium for vertebrate Hh signaling, in addition to IFT's well-established role in ciliogenesis. Surprisingly, a conserved Hh pathway regulator, the atypical non-motile kinesin KIF7, is critical for the temporal control of GLI2 transport by IFT and the spatial control of GLI2 localization at the cilium tip. This discovery underscores the collaborative role of a motile and a non-motile cilium-specific cytoskeletal system in determining the transcriptional output during Hh signaling.
Collapse
|
27
|
Ewerling A, May-Simera HL. Evolutionary trajectory for nuclear functions of ciliary transport complex proteins. Microbiol Mol Biol Rev 2024; 88:e0000624. [PMID: 38995044 PMCID: PMC11426024 DOI: 10.1128/mmbr.00006-24] [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] [Indexed: 07/13/2024] Open
Abstract
SUMMARYCilia and the nucleus were two defining features of the last eukaryotic common ancestor. In early eukaryotic evolution, these structures evolved through the diversification of a common membrane-coating ancestor, the protocoatomer. While in cilia, the descendants of this protein complex evolved into parts of the intraflagellar transport complexes and BBSome, the nucleus gained its selectivity by recruiting protocoatomer-like proteins to the nuclear envelope to form the selective nuclear pore complexes. Recent studies show a growing number of proteins shared between the proteomes of the respective organelles, and it is currently unknown how ciliary transport proteins could acquire nuclear functions and vice versa. The nuclear functions of ciliary proteins are still observable today and remain relevant for the understanding of the disease mechanisms behind ciliopathies. In this work, we review the evolutionary history of cilia and nucleus and their respective defining proteins and integrate current knowledge into theories for early eukaryotic evolution. We postulate a scenario where both compartments co-evolved and that fits current models of eukaryotic evolution, explaining how ciliary proteins and nucleoporins acquired their dual functions.
Collapse
Affiliation(s)
- Alexander Ewerling
- Faculty of Biology, Institute of Molecular Physiology, Johannes Gutenberg-University, Mainz, Germany
| | - Helen Louise May-Simera
- Faculty of Biology, Institute of Molecular Physiology, Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
28
|
Thuy PX, Jang TK, Moon EY. Vinblastine Resistance Is Associated with Nephronophthisis 3-Mediated Primary Cilia via Intraflagellar Transport Protein 88 and Apoptosis-Antagonizing Transcription Factor. Int J Mol Sci 2024; 25:10369. [PMID: 39408701 PMCID: PMC11477320 DOI: 10.3390/ijms251910369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/15/2024] [Accepted: 09/17/2024] [Indexed: 10/20/2024] Open
Abstract
Primary cilia (PC) are microtubule-based organelles that function as cellular antennae to sense and transduce extracellular signals. Nephronophthisis 3 (NPHP3) is localized in the inversin compartment of PC. Mutations in NPHP3 are associated with renal-hepatic-pancreatic dysplasia. In this study, we investigated whether vinblastine (VBL), a microtubule destabilizer, induces anticancer drug resistance through NPHP3-associated PC formation in HeLa human cervical cancer cells. A considerable increase in PC frequency was observed in HeLa cells under serum-deprived (SD) conditions, which led to the inhibition of VBL-induced cell death. VBL-resistant cells were established by repetitive treatments with VBL and showed an increase in PC frequency. NPHP3 expression was also increased by VBL treatment under serum starvation as well as in VBL-resistant cells. NPHP3 expression and PC-associated resistance were positively correlated with apoptosis-antagonizing transcription factor (AATF) and negatively correlated with inhibition of NPHP3. In addition, AATF-mediated NPHP3 expression is associated with PC formation via the regulation of intraflagellar transport protein 88 (IFT88). VBL resistance ability was reduced by treating with ciliobrevin A, a well-known ciliogenesis inhibitor. Collectively, cancer cell survival following VBL treatment is regulated by PC formation via AATF-mediated expression of IFT88 and NPHP3. Our data suggest that the activation of AATF and IFT88 could be a novel regulator to induce anticancer drug resistance through NPHP3-associated PC formation.
Collapse
Affiliation(s)
| | | | - Eun-Yi Moon
- Department of Bioscience and Biotechnology, Sejong University, Seoul 05006, Republic of Korea; (P.X.T.); (T.-K.J.)
| |
Collapse
|
29
|
Huang Y, Dong X, Sun SY, Lim TK, Lin Q, He CY. ARL3 GTPases facilitate ODA16 unloading from IFT in motile cilia. SCIENCE ADVANCES 2024; 10:eadq2950. [PMID: 39231220 PMCID: PMC11373600 DOI: 10.1126/sciadv.adq2950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 07/30/2024] [Indexed: 09/06/2024]
Abstract
Eukaryotic cilia and flagella are essential for cell motility and sensory functions. Their biogenesis and maintenance rely on the intraflagellar transport (IFT). Several cargo adapters have been identified to aid IFT cargo transport, but how ciliary cargos are discharged from the IFT remains largely unknown. During our explorations of small GTPases ARL13 and ARL3 in Trypanosoma brucei, we found that ODA16, a known IFT cargo adapter present exclusively in motile cilia, is a specific effector of ARL3. In the cilia, active ARL3 GTPases bind to ODA16 and dissociate ODA16 from the IFT complex. Depletion of ARL3 GTPases stabilizes ODA16 interaction with the IFT, leading to ODA16 accumulation in cilia and defects in axonemal assembly. The interactions between human ODA16 homolog HsDAW1 and ARL GTPases are conserved, and these interactions are altered in HsDAW1 disease variants. These findings revealed a conserved function of ARL GTPases in IFT transport of motile ciliary components, and a mechanism of cargo unloading from the IFT.
Collapse
Affiliation(s)
- Yameng Huang
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Xiaoduo Dong
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Stella Y Sun
- Department of Structural Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Teck-Kwang Lim
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Qingsong Lin
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Cynthia Y He
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
- The Centre for BioImaging Sciences, National University of Singapore, Singapore, Singapore
| |
Collapse
|
30
|
Gray S, Fort C, Wheeler RJ. Intraflagellar transport speed is sensitive to genetic and mechanical perturbations to flagellar beating. J Cell Biol 2024; 223:e202401154. [PMID: 38829962 PMCID: PMC11148470 DOI: 10.1083/jcb.202401154] [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: 01/31/2024] [Revised: 05/01/2024] [Accepted: 05/13/2024] [Indexed: 06/05/2024] Open
Abstract
Two sets of motor proteins underpin motile cilia/flagella function. The axoneme-associated inner and outer dynein arms drive sliding of adjacent axoneme microtubule doublets to periodically bend the flagellum for beating, while intraflagellar transport (IFT) kinesins and dyneins carry IFT trains bidirectionally along the axoneme. Despite assembling motile cilia and flagella, IFT train speeds have only previously been quantified in immobilized flagella-mechanical immobilization or genetic paralysis. This has limited investigation of the interaction between IFT and flagellar beating. Here, in uniflagellate Leishmania parasites, we use high-frequency, dual-color fluorescence microscopy to visualize IFT train movement in beating flagella. We discovered that adhesion of flagella to a microscope slide is detrimental, reducing IFT train speed and increasing train stalling. In flagella free to move, IFT train speed is not strongly dependent on flagella beat type; however, permanent disruption of flagella beating by deletion of genes necessary for formation or regulation of beating showed an inverse correlation of beat frequency and IFT train speed.
Collapse
Affiliation(s)
- Sophie Gray
- Nuffield Department of Medicine, Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, UK
| | - Cecile Fort
- Nuffield Department of Medicine, Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, UK
| | - Richard John Wheeler
- Nuffield Department of Medicine, Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, UK
| |
Collapse
|
31
|
Mizuno A, Takeuchi K, Nagata Y, Harada H, Yamamoto T, Ishikawa T, Maeda S, Ohka F, Ueno H, Saito R. Isolation of ependymal cilia from mouse brain. J Neurosci Methods 2024; 409:110198. [PMID: 38878975 DOI: 10.1016/j.jneumeth.2024.110198] [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: 05/18/2024] [Accepted: 06/11/2024] [Indexed: 06/25/2024]
Abstract
BACKGROUND Ependymal cilia play a major role in the circulation of cerebrospinal fluid. Although isolation of cilia is an essential technique for investigating ciliary structure, to the best of our knowledge, no report on the isolation and structural analysis of ependymal cilia from mouse brain is available. NEW METHOD We developed a novel method for isolating ependymal cilia from mouse brain ventricles. We isolated ependymal cilia by partially opening the lateral ventricles and gently applying shear stress, followed by pipetting and ultracentrifugation. RESULTS Using this new method, we were able to observe cilia separately. The results demonstrated that our method successfully isolated intact ependymal cilia with preserved morphology and ultrastructure. In this procedure, the ventricular ependymal cell layer was partially detached. COMPARISON WITH EXISTING METHODS Compared to existing methods for isolating cilia from other tissues, our method is meticulously tailored for extracting ependymal cilia from the mouse brain. Designed with a keen understanding of the fragility of the ventricular ependyma, our method prioritizes minimizing tissue damage during the isolation procedure. CONCLUSIONS We isolated ependymal cilia from mouse brain by applying shear stress selectively to the ventricles. Our method can be used to conduct more detailed studies on the structure of ependymal cilia.
Collapse
Affiliation(s)
- Akihiro Mizuno
- Department of Neurosurgery, Komaki City Hospital, Aichi, Japan
| | | | - Yuichi Nagata
- Department of Neurosurgery, Nagoya University, Nagoya, Japan
| | - Hideyuki Harada
- Department of Neurosurgery, Nagoya University, Nagoya, Japan
| | - Taiki Yamamoto
- Department of Neurosurgery, Gifu Prefectural Tajimi Hospital, Gifu, Japan
| | - Takayuki Ishikawa
- Department of Neurosurgery, Japanese Red Cross Aichi Medical Center Nagoya Daini Hospital, Aichi, Japan
| | - Sachi Maeda
- Department of Neurosurgery, Nagoya University, Nagoya, Japan
| | - Fumiharu Ohka
- Department of Neurosurgery, Nagoya University, Nagoya, Japan
| | - Hironori Ueno
- Natural Science, Aichi University of Education, Aichi, Japan
| | - Ryuta Saito
- Department of Neurosurgery, Nagoya University, Nagoya, Japan
| |
Collapse
|
32
|
Putnová I, Putnová BM, Hurník P, Štembírek J, Buchtová M, Kolísková P. Primary cilia-associated signalling in squamous cell carcinoma of head and neck region. Front Oncol 2024; 14:1413255. [PMID: 39234399 PMCID: PMC11372790 DOI: 10.3389/fonc.2024.1413255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 07/29/2024] [Indexed: 09/06/2024] Open
Abstract
Squamous cell carcinoma (SCC) of the head and neck originates from the mucosal lining of the upper aerodigestive tract, including the lip, tongue, nasopharynx, oropharynx, larynx and hypopharynx. In this review, we summarise what is currently known about the potential function of primary cilia in the pathogenesis of this disease. As primary cilia represent a key cellular structure for signal transduction and are related to cell proliferation, an understanding of their role in carcinogenesis is necessary for the design of new treatment approaches. Here, we introduce cilia-related signalling in head and neck squamous cell carcinoma (HNSCC) and its possible association with HNSCC tumorigenesis. From this point of view, PDGF, EGF, Wnt and Hh signalling are discussed as all these pathways were found to be dysregulated in HNSCC. Moreover, we review the clinical potential of small molecules affecting primary cilia signalling to target squamous cell carcinoma of the head and neck area.
Collapse
Affiliation(s)
- Iveta Putnová
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czechia
- Department of Anatomy, Histology and Embryology, University of Veterinary Sciences Brno, Brno, Czechia
| | - Barbora Moldovan Putnová
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czechia
- Department of Pathological Morphology and Parasitology, University of Veterinary Sciences Brno, Brno, Czechia
| | - Pavel Hurník
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czechia
- Institute of Molecular and Clinical Pathology and Medical Genetics, University Hospital Ostrava, Ostrava, Czechia
- Institute of Molecular and Clinical Pathology and Medical Genetics, Faculty of Medicine, University of Ostrava, Ostrava, Czechia
| | - Jan Štembírek
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czechia
- Department of Maxillofacial Surgery, University Hospital Ostrava, Ostrava, Czechia
| | - Marcela Buchtová
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czechia
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
| | - Petra Kolísková
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czechia
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
| |
Collapse
|
33
|
Cai Y, Zhang X, Yang C, Jiang Y, Chen Y. Melatonin alleviates high-fat-diet-induced dry eye by regulating macrophage polarization via IFT27 and lowering ERK/JNK phosphorylation. iScience 2024; 27:110367. [PMID: 39100927 PMCID: PMC11294704 DOI: 10.1016/j.isci.2024.110367] [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: 01/11/2024] [Revised: 03/03/2024] [Accepted: 06/21/2024] [Indexed: 08/06/2024] Open
Abstract
Dry eye disease is the most common ocular surface disease globally, requiring a more effective treatment. We observed that a high-fat diet induced macrophage polarization to M1 and further induced inflammation in the meibomian and lacrimal glands. A four-week treatment with melatonin (MLT) eye drops can regulate macrophage polarization and alleviate dry eye signs. To investigate the therapeutic effects and mechanisms of action of MLT on high-fat-diet-induced dry eye disease in mice, RAW 264.7 cells pretreated with LPS and/or MLT underwent digital RNA with the perturbation of genes sequencing (DRUG-seq). Results showed that IFT27 was up-regulated, and MAPK pathways were suppressed after MLT pre-treatment. ERK/JNK phosphorylation was reduced in meibomian glands of MLT-treated dry eye mice and increased in IFT27 knockdown RAW 264.7 cells. In summary, MLT regulated macrophage polarization via IFT27 and reduced ERK/JNK phosphorylation. These results support that MLT is a promising medication for dry eye disease.
Collapse
Affiliation(s)
- Yuying Cai
- Department of Ophthalmology, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xin Zhang
- Department of Ophthalmology, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chuanxi Yang
- Department of Cardiology, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yaping Jiang
- Department of Ophthalmology, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yihui Chen
- Department of Ophthalmology, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
34
|
Zhou Z, Yang X, Mao A, Xu H, Lin C, Yang M, Hu W, Shao J, Xu P, Li Y, Li W, Lin R, Zhang R, Xie Q, Xu Z, Meng W. Deficiency of CAMSAP2 impairs olfaction and the morphogenesis of mitral cells. EMBO Rep 2024; 25:2861-2877. [PMID: 38839944 PMCID: PMC11239855 DOI: 10.1038/s44319-024-00166-x] [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: 10/06/2023] [Revised: 04/07/2024] [Accepted: 04/22/2024] [Indexed: 06/07/2024] Open
Abstract
In developing olfactory bulb (OB), mitral cells (MCs) remodel their dendrites to establish the precise olfactory circuit, and these circuits are critical for individuals to sense odors and elicit behaviors for survival. However, how microtubules (MTs) participate in the process of dendritic remodeling remains elusive. Here, we reveal that calmodulin-regulated spectrin-associated proteins (CAMSAPs), a family of proteins that bind to the minus-end of the noncentrosomal MTs, play a crucial part in the development of MC dendrites. We observed that Camsap2 knockout (KO) males are infertile while the reproductive tract is normal. Further study showed that the infertility was due to the severe defects of mating behavior in male mice. Besides, mice with loss-of-function displayed defects in the sense of smell. Furthermore, we found that the deficiency of CAMSAP2 impairs the classical morphology of MCs, and the CAMSAP2-dependent dendritic remodeling process is responsible for this defect. Thus, our findings demonstrate that CAMSAP2 plays a vital role in regulating the development of MCs.
Collapse
Affiliation(s)
- Zhengrong Zhou
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 100101, Beijing, China.
- Neuroscience Center, Department of Basic Medical Sciences, Shantou University Medical College, 515041, Shantou, Guangdong, China.
| | - Xiaojuan Yang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Aihua Mao
- Biology Department, College of Sciences, Shantou University, 515063, Shantou, China
| | - Honglin Xu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Chunnuan Lin
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Mengge Yang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Weichang Hu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Jinhui Shao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Peipei Xu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Yuejia Li
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Wenguang Li
- Animal Laboratory Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Ruifan Lin
- Chinese Academy of Chinese Medical Sciences, 100700, Beijing, China
| | - Rui Zhang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Qi Xie
- Chinese Academy of Chinese Medical Sciences, 100700, Beijing, China
| | - Zhiheng Xu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
- Innovation Academy for Seed Design, Chinese Academy of Sciences, 100101, Beijing, China
| | - Wenxiang Meng
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 100101, Beijing, China.
- University of Chinese Academy of Sciences, 100049, Beijing, China.
- Innovation Academy for Seed Design, Chinese Academy of Sciences, 100101, Beijing, China.
| |
Collapse
|
35
|
Patel K, Smith NJ. Primary cilia, A-kinase anchoring proteins and constitutive activity at the orphan G protein-coupled receptor GPR161: A tale about a tail. Br J Pharmacol 2024; 181:2182-2196. [PMID: 36772847 DOI: 10.1111/bph.16053] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/22/2022] [Accepted: 02/04/2023] [Indexed: 02/12/2023] Open
Abstract
Primary cilia are non-motile antennae-like structures responsible for sensing environmental changes in most mammalian cells. Ciliary signalling is largely mediated by the Sonic Hedgehog (Shh) pathway, which acts as a master regulator of ciliary protein transit and is essential for normal embryonic development. One particularly important player in primary cilia is the orphan G protein-coupled receptor, GPR161. In this review, we introduce GPR161 in the context of Shh signalling and describe the unique features on its C-terminus such as PKA phosphorylation sites and an A-kinase anchoring protein motif, which may influence the function of the receptor, cAMP compartmentalisation and/or trafficking within primary cilia. We discuss the recent putative pairing of GPR161 and spexin-1, highlighting the additional steps needed before GPR161 could be considered 'deorphanised'. Finally, we speculate that the marked constitutive activity and unconventional regulation of GPR161 may indicate that the receptor may not require an endogenous ligand. LINKED ARTICLES: This article is part of a themed issue Therapeutic Targeting of G Protein-Coupled Receptors: hot topics from the Australasian Society of Clinical and Experimental Pharmacologists and Toxicologists 2021 Virtual Annual Scientific Meeting. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v181.14/issuetoc.
Collapse
Affiliation(s)
- Kinjal Patel
- Orphan Receptor Laboratory, School of Biomedical Sciences, Faculty of Medicine & Health, UNSW Sydney, Kensington, New South Wales, Australia
| | - Nicola J Smith
- Orphan Receptor Laboratory, School of Biomedical Sciences, Faculty of Medicine & Health, UNSW Sydney, Kensington, New South Wales, Australia
| |
Collapse
|
36
|
King SM, Sakato-Antoku M, Patel-King RS, Balsbaugh JL. The methylome of motile cilia. Mol Biol Cell 2024; 35:ar89. [PMID: 38696262 PMCID: PMC11244166 DOI: 10.1091/mbc.e24-03-0130] [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: 03/25/2024] [Revised: 04/17/2024] [Accepted: 04/24/2024] [Indexed: 05/04/2024] Open
Abstract
Cilia are highly complex motile, sensory, and secretory organelles that contain perhaps 1000 or more distinct protein components, many of which are subject to various posttranslational modifications such as phosphorylation, N-terminal acetylation, and proteolytic processing. Another common modification is the addition of one or more methyl groups to the side chains of arginine and lysine residues. These tunable additions delocalize the side-chain charge, decrease hydrogen bond capacity, and increase both bulk and hydrophobicity. Methylation is usually mediated by S-adenosylmethionine (SAM)-dependent methyltransferases and reversed by demethylases. Previous studies have identified several ciliary proteins that are subject to methylation including axonemal dynein heavy chains that are modified by a cytosolic methyltransferase. Here, we have performed an extensive proteomic analysis of multiple independently derived cilia samples to assess the potential for SAM metabolism and the extent of methylation in these organelles. We find that cilia contain all the enzymes needed for generation of the SAM methyl donor and recycling of the S-adenosylhomocysteine and tetrahydrofolate byproducts. In addition, we find that at least 155 distinct ciliary proteins are methylated, in some cases at multiple sites. These data provide a comprehensive resource for studying the consequences of methyl marks on ciliary biology.
Collapse
Affiliation(s)
- Stephen M. King
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT 3305
| | - Miho Sakato-Antoku
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT 3305
| | - Ramila S. Patel-King
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT 3305
| | - Jeremy L. Balsbaugh
- Proteomics and Metabolomics Facility, Center for Open Research Resources & Equipment, University of Connecticut, Storrs, CT 06269
| |
Collapse
|
37
|
Reddy Palicharla V, Mukhopadhyay S. Molecular and structural perspectives on protein trafficking to the primary cilium membrane. Biochem Soc Trans 2024; 52:1473-1487. [PMID: 38864436 PMCID: PMC11346432 DOI: 10.1042/bst20231403] [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: 03/20/2024] [Revised: 05/23/2024] [Accepted: 05/24/2024] [Indexed: 06/13/2024]
Abstract
The primary cilium is a dynamic subcellular compartment templated from the mother centriole or basal body. Cilia are solitary and tiny, but remarkably consequential in cellular pathways regulating proliferation, differentiation, and maintenance. Multiple transmembrane proteins such as G-protein-coupled receptors, channels, enzymes, and membrane-associated lipidated proteins are enriched in the ciliary membrane. The precise regulation of ciliary membrane content is essential for effective signal transduction and maintenance of tissue homeostasis. Surprisingly, a few conserved molecular factors, intraflagellar transport complex A and the tubby family adapter protein TULP3, mediate the transport of most membrane cargoes into cilia. Recent advances in cryogenic electron microscopy provide fundamental insights into these molecular players. Here, we review the molecular players mediating cargo delivery into the ciliary membrane through the lens of structural biology. These mechanistic insights into ciliary transport provide a framework for understanding of disease variants in ciliopathies, enable precise manipulation of cilia-mediated pathways, and provide a platform for the development of targeted therapeutics.
Collapse
Affiliation(s)
- Vivek Reddy Palicharla
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, U.S.A
| | - Saikat Mukhopadhyay
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, U.S.A
| |
Collapse
|
38
|
Kuwasako K, Dang W, He F, Takahashi M, Tsuda K, Nagata T, Tanaka A, Kobayashi N, Kigawa T, Güntert P, Shirouzu M, Yokoyama S, Muto Y. 1H, 13C, and 15N resonance assignments and solution structure of the N-terminal divergent calponin homology (NN-CH) domain of human intraflagellar transport protein 54. BIOMOLECULAR NMR ASSIGNMENTS 2024; 18:71-78. [PMID: 38551798 DOI: 10.1007/s12104-024-10170-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 03/13/2024] [Indexed: 05/12/2024]
Abstract
The intraflagellar transport (IFT) machinery plays a crucial role in the bidirectional trafficking of components necessary for ciliary signaling, such as the Hedgehog, Wnt/PCR, and cAMP/PKA systems. Defects in some components of the IFT machinery cause dysfunction, leading to a wide range of human diseases and developmental disorders termed ciliopathies, such as nephronophthisis. The IFT machinery comprises three sub-complexes: BBsome, IFT-A, and IFT-B. The IFT protein 54 (IFT54) is an important component of the IFT-B sub-complex. In anterograde movement, IFT54 binds to active kinesin-II, walking along the cilia microtubule axoneme and carrying the dynein-2 complex in an inactive state, which works for retrograde movement. Several mutations in IFT54 are known to cause Senior-Loken syndrome, a ciliopathy. IFT54 possesses a divergent Calponin Homology (CH) domain termed as NN-CH domain at its N-terminus. However, several aspects of the function of the NN-CH domain of IFT54 are still obscure. Here, we report the 1H, 15N, and 13C resonance assignments of the NN-CH domain of human IFT54 and its solution structure. The NN-CH domain of human IFT54 adopts essentially the α1-α2-α3-α4-α5 topology as that of mouse IFT54, whose structure was determined by X-ray crystallographic study. The structural information and assignments obtained in this study shed light on the molecular function of the NN-CH domain in IFT54.
Collapse
Affiliation(s)
- Kanako Kuwasako
- RIKEN, Systems and Structural Biology Center, 1-7-22 Suehiro-cho, Tsurumi, Yokohama, 230-0045, Japan
- RIKEN Center for Biosystems Dynamics Research, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, 230- 0045, Japan
- Faculty of Pharmacy and Research Institute of Pharmaceutical Sciences, Musashino University, Tokyo, 202-8585, Japan
| | - Weirong Dang
- RIKEN, Systems and Structural Biology Center, 1-7-22 Suehiro-cho, Tsurumi, Yokohama, 230-0045, Japan
| | - Fahu He
- RIKEN, Systems and Structural Biology Center, 1-7-22 Suehiro-cho, Tsurumi, Yokohama, 230-0045, Japan
| | - Mari Takahashi
- RIKEN, Systems and Structural Biology Center, 1-7-22 Suehiro-cho, Tsurumi, Yokohama, 230-0045, Japan
- RIKEN Center for Biosystems Dynamics Research, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, 230- 0045, Japan
| | - Kengo Tsuda
- RIKEN, Systems and Structural Biology Center, 1-7-22 Suehiro-cho, Tsurumi, Yokohama, 230-0045, Japan
| | - Takashi Nagata
- RIKEN, Systems and Structural Biology Center, 1-7-22 Suehiro-cho, Tsurumi, Yokohama, 230-0045, Japan
- Institute of Advanced Energy, Graduate School of Energy Science, Kyoto University, Gokasho, Kyoto, Uji, 611-0011, Japan
| | - Akiko Tanaka
- RIKEN, Systems and Structural Biology Center, 1-7-22 Suehiro-cho, Tsurumi, Yokohama, 230-0045, Japan
| | - Naohiro Kobayashi
- RIKEN, Systems and Structural Biology Center, 1-7-22 Suehiro-cho, Tsurumi, Yokohama, 230-0045, Japan
- RIKEN Yokohama NMR Facility, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, 230-0045, Japan
| | - Takanori Kigawa
- RIKEN, Systems and Structural Biology Center, 1-7-22 Suehiro-cho, Tsurumi, Yokohama, 230-0045, Japan
- RIKEN Center for Biosystems Dynamics Research, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, 230- 0045, Japan
| | - Peter Güntert
- Tatsuo Miyazawa Memorial Program, RIKEN Genomic Sciences Center, Yokohama, 230-0045, Japan
- Institute of Biophysical Chemistry, Goethe-University Frankfurt am Main, Max-von-Laue-Str. 9, Frankfurt am Main, 60438, Germany
- Institute of Molecular Physical Science, ETH Zurich, Vladimir-Prelog-Weg 2, Zurich, 8093, Switzerland
- Department of Chemistry, Tokyo Metropolitan University, 1-1 Minami-Ohsawa, Hachioji, Tokyo, 192- 0397, Japan
| | - Mikako Shirouzu
- RIKEN, Systems and Structural Biology Center, 1-7-22 Suehiro-cho, Tsurumi, Yokohama, 230-0045, Japan
- RIKEN Center for Biosystems Dynamics Research, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, 230- 0045, Japan
| | - Shigeyuki Yokoyama
- RIKEN, Systems and Structural Biology Center, 1-7-22 Suehiro-cho, Tsurumi, Yokohama, 230-0045, Japan.
- RIKEN Structural Biology Laboratory, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, 230-0045, Japan.
- RIKEN Cluster for Science, Technology and Innovation Hub, 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Yokohama, 230-0045, Japan.
| | - Yutaka Muto
- RIKEN, Systems and Structural Biology Center, 1-7-22 Suehiro-cho, Tsurumi, Yokohama, 230-0045, Japan.
- RIKEN Center for Biosystems Dynamics Research, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, 230- 0045, Japan.
- Faculty of Pharmacy and Research Institute of Pharmaceutical Sciences, Musashino University, Tokyo, 202-8585, Japan.
| |
Collapse
|
39
|
Chen H, Wu Z, Yan Z, Chen C, Zhang Y, Wang Q, Gao Y, Ling K, Hu J, Wei Q. The ARPKD Protein DZIP1L Regulates Ciliary Protein Entry by Modulating the Architecture and Function of Ciliary Transition Fibers. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308820. [PMID: 38634253 PMCID: PMC11200010 DOI: 10.1002/advs.202308820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 03/13/2024] [Indexed: 04/19/2024]
Abstract
Serving as the cell's sensory antennae, primary cilia are linked to numerous human genetic diseases when they malfunction. DZIP1L, identified as one of the genetic causes of human autosomal recessive polycystic kidney disease (ARPKD), is an evolutionarily conserved ciliary basal body protein. Although it has been reported that DZIP1L is involved in the ciliary entry of PKD proteins, the underlying mechanism remains elusive. Here, an uncharacterized role of DZIP1L is reported in modulating the architecture and function of transition fibers (TFs), striking ciliary base structures essential for selective cilia gating. Using C. elegans as a model, C01G5.7 (hereafter termed DZIP-1) is identified as the sole homolog of DZIP1L, which specifically localizes to TFs. While DZIP-1 or ANKR-26 (the ortholog of ANKRD26) deficiency shows subtle impact on TFs, co-depletion of DZIP-1 and ANKR-26 disrupts TF assembly and cilia gating for soluble and membrane proteins, including the ortholog of ADPKD protein polycystin-2. Notably, the synergistic role for DZIP1L and ANKRD26 in the formation and function of TFs is highly conserved in mammalian cilia. Hence, the findings illuminate an evolutionarily conserved role of DZIP1L in TFs architecture and function, highlighting TFs as a vital part of the ciliary gate implicated in ciliopathies ARPKD.
Collapse
Affiliation(s)
- Huicheng Chen
- CAS Key Laboratory of Insect Developmental and Evolutionary BiologyCAS Center for Excellence in Molecular Plant SciencesChinese Academy of SciencesShanghai200032China
- University of Chinese Academy of SciencesBeijing100039China
- Center for Energy Metabolism and ReproductionInstitute of Biomedicine and BiotechnologyShenzhen Institutes of Advanced TechnologyChinese Academy of Sciences (CAS)Shenzhen518055China
| | - Zhimao Wu
- Center for Energy Metabolism and ReproductionInstitute of Biomedicine and BiotechnologyShenzhen Institutes of Advanced TechnologyChinese Academy of Sciences (CAS)Shenzhen518055China
| | - Ziwei Yan
- CAS Key Laboratory of Insect Developmental and Evolutionary BiologyCAS Center for Excellence in Molecular Plant SciencesChinese Academy of SciencesShanghai200032China
- University of Chinese Academy of SciencesBeijing100039China
| | - Chuan Chen
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMN55905USA
| | - Yingying Zhang
- Center for Energy Metabolism and ReproductionInstitute of Biomedicine and BiotechnologyShenzhen Institutes of Advanced TechnologyChinese Academy of Sciences (CAS)Shenzhen518055China
| | - Qiaoling Wang
- Institute of Medicine and Pharmaceutical SciencesZhengzhou UniversityZhengzhou430000China
| | - Yuqing Gao
- Center for Energy Metabolism and ReproductionInstitute of Biomedicine and BiotechnologyShenzhen Institutes of Advanced TechnologyChinese Academy of Sciences (CAS)Shenzhen518055China
| | - Kun Ling
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMN55905USA
| | - Jinghua Hu
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMN55905USA
| | - Qing Wei
- Center for Energy Metabolism and ReproductionInstitute of Biomedicine and BiotechnologyShenzhen Institutes of Advanced TechnologyChinese Academy of Sciences (CAS)Shenzhen518055China
- School of Synthetic BiologyShanxi Key Laboratory of Nucleic Acid BiopesticidesShanxi UniversityTaiyuan030006China
| |
Collapse
|
40
|
Philbrook A, O'Donnell MP, Grunenkovaite L, Sengupta P. Differential modulation of sensory response dynamics by cilia structure and intraflagellar transport within and across chemosensory neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.16.594529. [PMID: 38798636 PMCID: PMC11118401 DOI: 10.1101/2024.05.16.594529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Sensory neurons contain morphologically diverse primary cilia that are built by intraflagellar transport (IFT) and house sensory signaling molecules. Since both ciliary structural and signaling proteins are trafficked via IFT, it has been challenging to decouple the contributions of IFT and cilia structure to neuronal responses. By acutely inhibiting IFT without altering cilia structure and vice versa , here we describe the differential roles of ciliary trafficking and sensory ending morphology in shaping chemosensory responses in C. elegans. We show that a minimum cilium length but not continuous IFT is necessary for a subset of responses in the ASH nociceptive neurons. In contrast, neither cilia nor continuous IFT are necessary for odorant responses in the AWA olfactory neurons. Instead, continuous IFT differentially modulates response dynamics in AWA. Upon acute inhibition of IFT, cilia-destined odorant receptors are shunted to ectopic branches emanating from the cilia base. Spatial segregation of receptors in these branches from a cilia-restricted regulatory kinase results in odorant desensitization defects, highlighting the importance of precise organization of signaling molecules at sensory endings in regulating response dynamics. We also find that adaptation of AWA responses upon repeated exposure to an odorant is mediated by IFT-driven removal of its cognate receptor, whereas adaptation to a second odorant is regulated via IFT-independent mechanisms. Our results reveal unexpected complexity in the contribution of IFT and cilia organization to the regulation of responses even within a single chemosensory neuron type, and establish a critical role for these processes in the precise modulation of olfactory behaviors.
Collapse
|
41
|
De Mori R, Tardivo S, Pollara L, Giliani SC, Ali E, Giordano L, Leuzzi V, Fischetto R, Gener B, Diprima S, Morelli MJ, Monti MC, Sottile V, Valente EM. Joubert syndrome-derived induced pluripotent stem cells show altered neuronal differentiation in vitro. Cell Tissue Res 2024; 396:255-267. [PMID: 38502237 PMCID: PMC11055696 DOI: 10.1007/s00441-024-03876-9] [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: 08/04/2023] [Accepted: 02/06/2024] [Indexed: 03/21/2024]
Abstract
Joubert syndrome (JS) is a recessively inherited congenital ataxia characterized by hypotonia, psychomotor delay, abnormal ocular movements, intellectual disability, and a peculiar cerebellar and brainstem malformation, the "molar tooth sign." Over 40 causative genes have been reported, all encoding for proteins implicated in the structure or functioning of the primary cilium, a subcellular organelle widely present in embryonic and adult tissues. In this paper, we developed an in vitro neuronal differentiation model using patient-derived induced pluripotent stem cells (iPSCs), to evaluate possible neurodevelopmental defects in JS. To this end, iPSCs from four JS patients harboring mutations in distinct JS genes (AHI1, CPLANE1, TMEM67, and CC2D2A) were differentiated alongside healthy control cells to obtain mid-hindbrain precursors and cerebellar granule cells. Differentiation was monitored over 31 days through the detection of lineage-specific marker expression by qRT-PCR, immunofluorescence, and transcriptomics analysis. All JS patient-derived iPSCs, regardless of the mutant gene, showed a similar impairment to differentiate into mid-hindbrain and cerebellar granule cells when compared to healthy controls. In addition, analysis of primary cilium count and morphology showed notable ciliary defects in all differentiating JS patient-derived iPSCs compared to controls. These results confirm that patient-derived iPSCs are an accessible and relevant in vitro model to analyze cellular phenotypes connected to the presence of JS gene mutations in a neuronal context.
Collapse
Affiliation(s)
- Roberta De Mori
- Induced Pluripotent Stem Cells Unit, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Silvia Tardivo
- Neurogenetics Lab, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Lidia Pollara
- Neurogenetics Research Unit, IRCCS Mondino Foundation, Pavia, Italy
| | - Silvia Clara Giliani
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Eltahir Ali
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Lucio Giordano
- Paediatric Neurology and Psychiatry Unit, Spedali Civili Children's Hospital, University of Brescia, Brescia, Italy
| | - Vincenzo Leuzzi
- Unit of Child Neurology and Psychiatry, Department of Human Neuroscience, University of Rome La Sapienza, Rome, Italy
| | - Rita Fischetto
- Clinical Genetics Unit, Department of Pediatric Medicine, XXIII Children's Hospital, Bari, Giovanni, Italy
| | - Blanca Gener
- Department of Genetics, Cruces University Hospital, BioBizkaia Health Research Institute, 48903 Barakaldo, Cruces PlazaBizkaia, Spain
| | - Santo Diprima
- IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Maria Cristina Monti
- Unit of Biostatistics and Clinical Epidemiology, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Pavia, Italy
| | - Virginie Sottile
- Department of Molecular Medicine, University of Pavia, Pavia, Italy.
| | - Enza Maria Valente
- Neurogenetics Research Unit, IRCCS Mondino Foundation, Pavia, Italy.
- Department of Molecular Medicine, University of Pavia, Pavia, Italy.
| |
Collapse
|
42
|
Leung M, Steinman J, Li D, Lor A, Gruesen A, Sadah A, van Kuijk FJ, Montezuma SR, Kondkar AA, Radhakrishnan R, Lobo GP. The Logistical Backbone of Photoreceptor Cell Function: Complementary Mechanisms of Dietary Vitamin A Receptors and Rhodopsin Transporters. Int J Mol Sci 2024; 25:4278. [PMID: 38673863 PMCID: PMC11050646 DOI: 10.3390/ijms25084278] [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: 03/12/2024] [Revised: 04/02/2024] [Accepted: 04/04/2024] [Indexed: 04/28/2024] Open
Abstract
In this review, we outline our current understanding of the mechanisms involved in the absorption, storage, and transport of dietary vitamin A to the eye, and the trafficking of rhodopsin protein to the photoreceptor outer segments, which encompasses the logistical backbone required for photoreceptor cell function. Two key mechanisms of this process are emphasized in this manuscript: ocular and systemic vitamin A membrane transporters, and rhodopsin transporters. Understanding the complementary mechanisms responsible for the generation and proper transport of the retinylidene protein to the photoreceptor outer segment will eventually shed light on the importance of genes encoded by these proteins, and their relationship on normal visual function and in the pathophysiology of retinal degenerative diseases.
Collapse
Affiliation(s)
- Matthias Leung
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN 55455, USA; (M.L.); (J.S.); (D.L.); (A.L.); (A.G.); (A.S.); (F.J.v.K.); (S.R.M.)
| | - Jeremy Steinman
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN 55455, USA; (M.L.); (J.S.); (D.L.); (A.L.); (A.G.); (A.S.); (F.J.v.K.); (S.R.M.)
| | - Dorothy Li
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN 55455, USA; (M.L.); (J.S.); (D.L.); (A.L.); (A.G.); (A.S.); (F.J.v.K.); (S.R.M.)
| | - Anjelynt Lor
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN 55455, USA; (M.L.); (J.S.); (D.L.); (A.L.); (A.G.); (A.S.); (F.J.v.K.); (S.R.M.)
| | - Andrew Gruesen
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN 55455, USA; (M.L.); (J.S.); (D.L.); (A.L.); (A.G.); (A.S.); (F.J.v.K.); (S.R.M.)
| | - Ahmed Sadah
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN 55455, USA; (M.L.); (J.S.); (D.L.); (A.L.); (A.G.); (A.S.); (F.J.v.K.); (S.R.M.)
| | - Frederik J. van Kuijk
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN 55455, USA; (M.L.); (J.S.); (D.L.); (A.L.); (A.G.); (A.S.); (F.J.v.K.); (S.R.M.)
| | - Sandra R. Montezuma
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN 55455, USA; (M.L.); (J.S.); (D.L.); (A.L.); (A.G.); (A.S.); (F.J.v.K.); (S.R.M.)
| | - Altaf A. Kondkar
- Department of Ophthalmology, College of Medicine, King Saud University, Riyadh 12271, Saudi Arabia;
| | - Rakesh Radhakrishnan
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN 55455, USA; (M.L.); (J.S.); (D.L.); (A.L.); (A.G.); (A.S.); (F.J.v.K.); (S.R.M.)
| | - Glenn P. Lobo
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, MN 55455, USA; (M.L.); (J.S.); (D.L.); (A.L.); (A.G.); (A.S.); (F.J.v.K.); (S.R.M.)
| |
Collapse
|
43
|
Lin Z, Shen Y, Li Y, Lu C, Zhu Y, He R, Cao Z, Yin Z, Gao H, Guo B, Ma X, Cao M, Luo M. Novel compound heterozygous variants in ARL13B lead to Joubert syndrome. J Cell Physiol 2024; 239:e31189. [PMID: 38219074 DOI: 10.1002/jcp.31189] [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: 08/04/2023] [Revised: 12/07/2023] [Accepted: 12/22/2023] [Indexed: 01/15/2024]
Abstract
Joubert syndrome (JBTS) is a systematic developmental disorder mainly characterized by a pathognomonic mid-hindbrain malformation. All known JBTS-associated genes encode proteins involved in the function of antenna-like cellular organelle, primary cilium, which plays essential roles in cellular signal transduction and development. Here, we identified four unreported variants in ARL13B in two patients with the classical features of JBTS. ARL13B is a member of the Ras GTPase family and functions in ciliogenesis and cilia-related signaling. The two missense variants in ARL13B harbored the substitutions of amino acids at evolutionarily conserved positions. Using model cell lines, we found that the accumulations of the missense variants in cilia were impaired and the variants showed attenuated functions in ciliogenesis or the trafficking of INPP5E. Overall, these findings expanded the ARL13B pathogenetic variant spectrum of JBTS.
Collapse
Affiliation(s)
- Zaisheng Lin
- International Peace Maternity and Child Health Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yue Shen
- National Human Genetic Resources Center, National Research Institute for Family Planning, Beijing, China
| | - Yan Li
- International Peace Maternity and Child Health Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chao Lu
- National Human Genetic Resources Center, National Research Institute for Family Planning, Beijing, China
| | - Ying Zhu
- International Peace Maternity and Child Health Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruida He
- International Peace Maternity and Child Health Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zongfu Cao
- National Human Genetic Resources Center, National Research Institute for Family Planning, Beijing, China
| | - Zhe Yin
- National Human Genetic Resources Center, National Research Institute for Family Planning, Beijing, China
| | - Huafang Gao
- National Human Genetic Resources Center, National Research Institute for Family Planning, Beijing, China
| | - Bin Guo
- International Peace Maternity and Child Health Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xu Ma
- National Human Genetic Resources Center, National Research Institute for Family Planning, Beijing, China
| | - Muqing Cao
- International Peace Maternity and Child Health Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Pathophysiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Minna Luo
- National Human Genetic Resources Center, National Research Institute for Family Planning, Beijing, China
| |
Collapse
|
44
|
Zhang X, Yao S, Zhang L, Yang L, Yang M, Guo Q, Li Y, Wang Z, Lei B, Jin X. Mechanisms underlying morphological and functional changes of cilia in fibroblasts derived from patients bearing ARL3 T31A and ARL3 T31A/C118F mutations. FASEB J 2024; 38:e23519. [PMID: 38457249 DOI: 10.1096/fj.202301906r] [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: 09/18/2023] [Revised: 01/31/2024] [Accepted: 02/15/2024] [Indexed: 03/10/2024]
Abstract
ARL3 is essential for cilia development, and mutations in ARL3 are closely associated with ciliopathies. In a previous study, we observed distinct phenotypes of retinal dystrophy in patients with heterozygous ARL3T31A and compound heterozygous ARL3T31A/C118F mutations, indicating that different mutation types may exert diverse effects on their functions. Here, we generated transformed immortal fibroblast cells from patients carrying heterozygous ARL3T31A and compound heterozygous ARL3T31A/C118F mutations, and systematically evaluated their cilia morphology and function, which were further validated in ARPE-19 cells. Results showed that both ARL3T31A and ARL3T31A/C118F mutations led to a decrease in cilium formation. The ARL3T31A/C118F mutations caused significantly elongated cilia and impaired retrograde transport, whereas the ARL3T31A mutation did not induce significant changes in fibroblasts. RNA-sequencing results indicated that compared to ARL3T31A , ARL3T31A/C118F fibroblasts exhibited a higher enrichment of biological processes related to neuron projection development, tissue morphogenesis, and extracellular matrix (ECM) organization, with noticeable alterations in pathways such as ECM-receptor interaction, focal adhesion, and TGF-β signaling. Similar changes were observed in the proteomic results in ARPE-19 cells. Core regulated genes including IQUB, UNC13D, RAB3IP, and GRIP1 were specifically downregulated in the ARL3T31A/C118F group, and expressions of IQUB, NPM2, and SLC38A4 were further validated. Additionally, IQUB showed a rescuing effect on the overlong cilia observed in ARL3T31A/C118F fibroblasts. Our results not only enhance our understanding of ARL3-related diseases but also provide new insights into the analysis of heterozygous and compound heterozygous mutations in genetics.
Collapse
Affiliation(s)
- Xiaoli Zhang
- Henan Eye Institute, Henan Eye Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, China
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Institute of Neuroscience and The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Eye Institute, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, China
| | - Shun Yao
- Henan Eye Institute, Henan Eye Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Lujia Zhang
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Institute of Neuroscience and The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lin Yang
- Henan Eye Institute, Henan Eye Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, China
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Mingzhu Yang
- Henan Eye Institute, Henan Eye Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Qingge Guo
- Henan Eye Institute, Henan Eye Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Yan Li
- Henan Eye Institute, Henan Eye Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, China
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Zhongfeng Wang
- Institute of Neuroscience and The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Bo Lei
- Henan Eye Institute, Henan Eye Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, China
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Eye Institute, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, China
| | - Xiuxiu Jin
- Henan Eye Institute, Henan Eye Hospital, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, China
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Eye Institute, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, China
| |
Collapse
|
45
|
Brinzer RA, Winter AD, Page AP. The relationship between intraflagellar transport and upstream protein trafficking pathways and macrocyclic lactone resistance in Caenorhabditis elegans. G3 (BETHESDA, MD.) 2024; 14:jkae009. [PMID: 38227795 PMCID: PMC10917524 DOI: 10.1093/g3journal/jkae009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 01/08/2024] [Accepted: 01/08/2024] [Indexed: 01/18/2024]
Abstract
Parasitic nematodes are globally important and place a heavy disease burden on infected humans, crops, and livestock, while commonly administered anthelmintics used for treatment are being rendered ineffective by increasing levels of resistance. It has recently been shown in the model nematode Caenorhabditis elegans that the sensory cilia of the amphid neurons play an important role in resistance toward macrocyclic lactones such as ivermectin (an avermectin) and moxidectin (a milbemycin) either through reduced uptake or intertissue signaling pathways. This study interrogated the extent to which ciliary defects relate to macrocyclic lactone resistance and dye-filling defects using a combination of forward genetics and targeted resistance screening approaches and confirmed the importance of intraflagellar transport in this process. This approach also identified the protein trafficking pathways used by the downstream effectors and the components of the ciliary basal body that are required for effector entry into these nonmotile structures. In total, 24 novel C. elegans anthelmintic survival-associated genes were identified in this study. When combined with previously known resistance genes, there are now 46 resistance-associated genes that are directly involved in amphid, cilia, and intraflagellar transport function.
Collapse
Affiliation(s)
- Robert A Brinzer
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Scotland G61 1QH, UK
| | - Alan D Winter
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Scotland G61 1QH, UK
| | - Antony P Page
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Scotland G61 1QH, UK
| |
Collapse
|
46
|
Pazour GJ. Cilia Structure and Function in Human Disease. CURRENT OPINION IN ENDOCRINE AND METABOLIC RESEARCH 2024; 34:100509. [PMID: 38836197 PMCID: PMC11147146 DOI: 10.1016/j.coemr.2024.100509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
Ciliary dysfunction causes a large group of developmental and degenerative human diseases known as ciliopathies. These diseases reflect the critical roles that cilia play in sensing the environment and in force generation for motility. Sensory functions include our senses of vision and olfaction. In addition, primary and motile cilia throughout our body monitor the environment allowing cells to coordinate their biology with the cells around them. This coordination is critical to organ development and maintenance, and ciliary dysfunction causes diverse structural birth defects and degenerative diseases. Defects in motility cause lung disease due to the failure of mucociliary clearance, male infertility due to the failure of sperm motility and the ability of sperm to move through the efferent ducts, and disturbances of the left-right axis due to a failure of nodal cilia to establish proper left-right cues.
Collapse
Affiliation(s)
- Gregory J Pazour
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Biotech II, Worcester, Massachusetts, USA
| |
Collapse
|
47
|
Fitzsimons LA, Tasouri E, Willaredt MA, Stetson D, Gojak C, Kirsch J, Gardner HAR, Gorgas K, Tucker KL. Primary cilia are critical for tracheoesophageal septation. Dev Dyn 2024; 253:312-332. [PMID: 37776236 PMCID: PMC10922539 DOI: 10.1002/dvdy.660] [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: 03/13/2023] [Revised: 08/14/2023] [Accepted: 09/09/2023] [Indexed: 10/02/2023] Open
Abstract
INTRODUCTION Primary cilia play pivotal roles in the patterning and morphogenesis of a wide variety of organs during mammalian development. Here we examined murine foregut septation in the cobblestone mutant, a hypomorphic allele of the gene encoding the intraflagellar transport protein IFT88, a protein essential for normal cilia function. RESULTS We reveal a crucial role for primary cilia in foregut division, since their dramatic decrease in cilia in both the foregut endoderm and mesenchyme of mutant embryos resulted in a proximal tracheoesophageal septation defects and in the formation of distal tracheo(broncho)esophageal fistulae similar to the most common congenital tracheoesophageal malformations in humans. Interestingly, the dorsoventral patterning determining the dorsal digestive and the ventral respiratory endoderm remained intact, whereas Hedgehog signaling was aberrantly activated. CONCLUSIONS Our results demonstrate the cobblestone mutant to represent one of the very few mouse models that display both correct endodermal dorsoventral specification but defective compartmentalization of the proximal foregut. It stands exemplary for a tracheoesophageal ciliopathy, offering the possibility to elucidate the molecular mechanisms how primary cilia orchestrate the septation process. The plethora of malformations observed in the cobblestone embryo allow for a deeper insight into a putative link between primary cilia and human VATER/VACTERL syndromes.
Collapse
Affiliation(s)
- Lindsey Avery Fitzsimons
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, Maine, U.S.A
- Dept. of Biomedical Sciences, Center for Excellence in the Neurosciences, College of Osteopathic Medicine, University of New England, Biddeford, Maine 04005, U.S.A
| | - Evangelia Tasouri
- Interdisciplinary Center for Neurosciences, University of Heidelberg, 69120 Heidelberg, Germany
- Institute of Anatomy and Cell Biology, University of Heidelberg, 69120 Heidelberg, Germany
| | - Marc August Willaredt
- Interdisciplinary Center for Neurosciences, University of Heidelberg, 69120 Heidelberg, Germany
- Institute of Anatomy and Cell Biology, University of Heidelberg, 69120 Heidelberg, Germany
| | - Daniel Stetson
- AstraZeneca Pharmaceuticals LP, 35 Gatehouse Drive, Waltham, Massachusetts 02451, U.S.A
| | - Christian Gojak
- Interdisciplinary Center for Neurosciences, University of Heidelberg, 69120 Heidelberg, Germany
- Institute of Anatomy and Cell Biology, University of Heidelberg, 69120 Heidelberg, Germany
| | - Joachim Kirsch
- Institute of Anatomy and Cell Biology, University of Heidelberg, 69120 Heidelberg, Germany
| | | | - Karin Gorgas
- Institute of Anatomy and Cell Biology, University of Heidelberg, 69120 Heidelberg, Germany
| | - Kerry L. Tucker
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, Maine, U.S.A
- Dept. of Biomedical Sciences, Center for Excellence in the Neurosciences, College of Osteopathic Medicine, University of New England, Biddeford, Maine 04005, U.S.A
| |
Collapse
|
48
|
Leggere JC, Hibbard JV, Papoulas O, Lee C, Pearson CG, Marcotte EM, Wallingford JB. Label-free proteomic comparison reveals ciliary and nonciliary phenotypes of IFT-A mutants. Mol Biol Cell 2024; 35:ar39. [PMID: 38170584 PMCID: PMC10916875 DOI: 10.1091/mbc.e23-03-0084] [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: 03/10/2023] [Revised: 12/11/2023] [Accepted: 12/18/2023] [Indexed: 01/05/2024] Open
Abstract
DIFFRAC is a powerful method for systematically comparing proteome content and organization between samples in a high-throughput manner. By subjecting control and experimental protein extracts to native chromatography and quantifying the contents of each fraction using mass spectrometry, it enables the quantitative detection of alterations to protein complexes and abundances. Here, we applied DIFFRAC to investigate the consequences of genetic loss of Ift122, a subunit of the intraflagellar transport-A (IFT-A) protein complex that plays a vital role in the formation and function of cilia and flagella, on the proteome of Tetrahymena thermophila. A single DIFFRAC experiment was sufficient to detect changes in protein behavior that mirrored known effects of IFT-A loss and revealed new biology. We uncovered several novel IFT-A-regulated proteins, which we validated through live imaging in Xenopus multiciliated cells, shedding new light on both the ciliary and non-ciliary functions of IFT-A. Our findings underscore the robustness of DIFFRAC for revealing proteomic changes in response to genetic or biochemical perturbation.
Collapse
Affiliation(s)
- Janelle C. Leggere
- Department of Molecular Biosciences, University of Texas at Austin, TX 78712
| | - Jaime V.K. Hibbard
- Department of Molecular Biosciences, University of Texas at Austin, TX 78712
| | - Ophelia Papoulas
- Department of Molecular Biosciences, University of Texas at Austin, TX 78712
| | - Chanjae Lee
- Department of Molecular Biosciences, University of Texas at Austin, TX 78712
| | - Chad G. Pearson
- Anschutz Medical Campus, Department of Cell and Developmental Biology, University of Colorado, Aurora, CO 80045
| | - Edward M. Marcotte
- Department of Molecular Biosciences, University of Texas at Austin, TX 78712
| | - John B. Wallingford
- Department of Molecular Biosciences, University of Texas at Austin, TX 78712
| |
Collapse
|
49
|
Jayarajan RO, Chakraborty S, Raghu KG, Purushothaman J, Veleri S. Joubert syndrome causing mutation in C2 domain of CC2D2A affects structural integrity of cilia and cellular signaling molecules. Exp Brain Res 2024; 242:619-637. [PMID: 38231387 DOI: 10.1007/s00221-023-06762-y] [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: 09/28/2023] [Accepted: 12/08/2023] [Indexed: 01/18/2024]
Abstract
Cilia are organelles extend from cells to sense external signals for tuning intracellular signaling for optimal cellular functioning. They have evolved sensory and motor roles in various cells for tissue organization and homeostasis in development and post-development. More than a thousand genes are required for cilia function. Mutations in them cause multisystem disorders termed ciliopathies. The null mutations in CC2D2A result in Meckel syndrome (MKS), which is embryonic lethal, whereas patients who have missense mutations in the C2 domain of CC2D2A display Joubert syndrome (JBTS). They survive with blindness and mental retardation. How C2 domain defects cause disease conditions is not understood. To answer this question, C2 domain of Cc2d2a (mice gene) was knocked down (KD) in IMCD-3 cells by shRNA. This resulted in defective cilia morphology observed by immunofluorescence analysis. To further probe the cellular signaling alteration in affected cells, gene expression profiling was done by RNAseq and compared with the controls. Bioinformatics analysis revealed that the differentially expressed genes (DEGs) have functions in cilia. Among the 61 cilia DEGs identified, 50 genes were downregulated and 11 genes were upregulated. These cilia genes are involved in cilium assembly, protein trafficking to the cilium, intraflagellar transport (IFT), cellular signaling like polarity patterning, and Hedgehog signaling pathway. This suggests that the C2 domain of CC2D2A plays a critical role in cilia assembly and molecular signaling hosted in cilia for cellular homeostasis. Taken together, the missense mutations in the C2 domain of CC2D2A seen in JBTS might have affected cilia-mediated signaling in neurons of the retina and brain.
Collapse
Affiliation(s)
- Roopasree O Jayarajan
- Agro-processing and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Thiruvananthapuram, Kerala, 695019, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Soura Chakraborty
- Department of Pathology, University of Cambridge, Cambridge, CB2 1QP, UK
| | - Kozhiparambil Gopalan Raghu
- Agro-processing and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Thiruvananthapuram, Kerala, 695019, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Jayamurthy Purushothaman
- Agro-processing and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Thiruvananthapuram, Kerala, 695019, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Shobi Veleri
- Drug Safety Division, National Institute of Nutrition, Indian Council of Medical Research, Department of Health Research, Ministry of Health and Family Welfare, Govt. of India, Hyderabad, 500007, India.
| |
Collapse
|
50
|
Luxmi R, King SM. Cilia Provide a Platform for the Generation, Regulated Secretion, and Reception of Peptidergic Signals. Cells 2024; 13:303. [PMID: 38391915 PMCID: PMC10886904 DOI: 10.3390/cells13040303] [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: 01/15/2024] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 02/24/2024] Open
Abstract
Cilia are microtubule-based cellular projections that act as motile, sensory, and secretory organelles. These structures receive information from the environment and transmit downstream signals to the cell body. Cilia also release vesicular ectosomes that bud from the ciliary membrane and carry an array of bioactive enzymes and peptide products. Peptidergic signals represent an ancient mode of intercellular communication, and in metazoans are involved in the maintenance of cellular homeostasis and various other physiological processes and responses. Numerous peptide receptors, subtilisin-like proteases, the peptide-amidating enzyme, and bioactive amidated peptide products have been localized to these organelles. In this review, we detail how cilia serve as specialized signaling organelles and act as a platform for the regulated processing and secretion of peptidergic signals. We especially focus on the processing and trafficking pathways by which a peptide precursor from the green alga Chlamydomonas reinhardtii is converted into an amidated bioactive product-a chemotactic modulator-and released from cilia in ectosomes. Biochemical dissection of this complex ciliary secretory pathway provides a paradigm for understanding cilia-based peptidergic signaling in mammals and other eukaryotes.
Collapse
Affiliation(s)
| | - Stephen M. King
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT 06030-3305, USA;
| |
Collapse
|