1
|
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] [Download PDF] [Figures] [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
|
2
|
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
|
3
|
Sakinah-Syed G, Liew JS, Abdul Majid N, Inche Zainal Abidin SA. Alteration of primary cilia and intraflagellar transport 20 (IFT20) expression in oral squamous cell carcinoma (OSCC) cell lines. PeerJ 2025; 13:e18931. [PMID: 40017656 PMCID: PMC11867036 DOI: 10.7717/peerj.18931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 01/13/2025] [Indexed: 03/01/2025] Open
Abstract
Background Aberrations in primary cilia expression and intraflagellar transport (IFT) protein function have been implicated in tumourigenesis. This study explores the relationship between the alteration of primary cilia and tumourigenesis by investigating primary cilia expression and the role of IFT20 in regulating matrix metalloproteinase 9 (MMP-9) expression in oral squamous cell carcinoma (OSCC) cell lines. Methods The frequency and length of primary cilia were determined in OKF6-TERT2 cells, HSC-2 cells, and HSC-3 cells using immunofluorescence. Additionally, primary cilia presence in non-proliferating OSCC cells was examined. OSCC cells were treated with either small interfering RNA (siRNA) negative control or siRNA targeting IFT20 for functional analysis. mRNA expression levels of IFT20 and MMP-9 were quantified using quantitative reverse transcription polymerase chain reaction (qRT-PCR). Results Results showed that HSC-2 cells exhibit abundant primary cilia when cultured in low serum media (2% serum) for 48 h, followed by serum starvation for over 72 h. No significant changes in cilia expression were observed in HSC-3 cells compared to OKF6-TERT2 cells. Ciliated cells were found in non-proliferating HSC-2 and HSC-3 cells. OSCC cells showed longer cilia than OKF6-TERT2 cells, indicating ciliary abnormalities. Changes in ciliation and cilium length of OSCC cells were accompanied by increased expression of IFT20, an intraflagellar transport protein crucial for the primary cilia assembly. However, IFT20 knockdown did not affect MMP-9 at the mRNA level in these cells. Conclusions This study reveals the differences in primary cilia expression among OSCC cells. Furthermore, the increased abundance and elongation of primary cilia in OSCC cells are accompanied by elevated expression of IFT20. Nonetheless, IFT20 did not affect MMP-9 mRNA expression in OSCC cells.
Collapse
Affiliation(s)
- Gulam Sakinah-Syed
- Institute of Biological Sciences, Faculty of Science, Universiti Malaya, Kuala Lumpur, WP Kuala Lumpur, Malaysia
- Department of Oral & Craniofacial Sciences, Faculty of Dentistry, Universiti Malaya, Kuala Lumpur, WP Kuala Lumpur, Malaysia
| | - Jia Shi Liew
- Institute of Biological Sciences, Faculty of Science, Universiti Malaya, Kuala Lumpur, WP Kuala Lumpur, Malaysia
| | - Nazia Abdul Majid
- Institute of Biological Sciences, Faculty of Science, Universiti Malaya, Kuala Lumpur, WP Kuala Lumpur, Malaysia
| | - Siti Amalina Inche Zainal Abidin
- Department of Oral & Craniofacial Sciences, Faculty of Dentistry, Universiti Malaya, Kuala Lumpur, WP Kuala Lumpur, Malaysia
- Oral Cancer Research & Coordinating Center, Faculty of Dentistry, Universiti Malaya, Kuala Lumpur, WP Kuala Lumpur, Malaysia
| |
Collapse
|
4
|
Li W, Niu C, Yap YT, Li T, Zheng C, Goswami M, Kandiraju S, Dhikhirullahi O, Xu J, Zhang J, Kelly CV, Zhang Z. Two-directional trafficking of the IFT25 protein in the developing mouse sperm flagella. Biol Reprod 2025; 112:309-318. [PMID: 39561113 PMCID: PMC12032603 DOI: 10.1093/biolre/ioae171] [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: 03/17/2024] [Revised: 10/02/2024] [Accepted: 11/18/2024] [Indexed: 11/21/2024] Open
Abstract
Intraflagellar transport 25 is a component of the intraflagellar transport 25-B complex. In mice, even though this intraflagellar transport component is not required for cilia formation in somatic cells, it is essential for sperm formation. However, the intracellular localization of this protein in male germ cells is not known given no reliable antibodies are available for histologic studies, and the dynamic trafficking in the developing sperm flagella is not clear. To examine localization of the protein in male germ cells and further investigate the mechanism of intraflagellar transport in sperm formation, particularly to look into the dynamic trafficking of the protein, we generated a mouse intraflagellar transport 25-green fluorescent protein knock-in mouse model using the clustered regularly interspaced short palindromic repeats/clustered regularly interspaced short palindromic repeats associated protein 9 system, with the mouse intraflagellar transport 25 protein fused with a green fluorescent protein tag in the C-terminus. Three independent lines were analyzed. Western blotting using both anti-intraflagellar transport 25 and anti-green fluorescent protein antibodies showed that the intraflagellar transport 25-green fluorescent protein fusion protein was highly abundant only in the testis, which is consistent with the endogenous intraflagellar transport 25 protein. Examination of localization of the intraflagellar transport 25-green fluorescent protein in isolated germ cells revealed that the fusion protein was present in the cytoplasm of spermatocytes and round spermatids and a strong signal was present in the developing sperm flagellar. The homozygous knock-in mice had normal spermatogenesis, fertility and sperm parameters. Diffusion analysis of intraflagellar transport 25 within the developing flagellar revealed the presence of both mobile and immobile fractions as revealed by fluorescence recovery after photobleaching. Kymograph and fluorescence recovery after photobleaching analyses demonstrate the transport of intraflagellar transport 25-green fluorescent protein within the developing tail demonstrate no apparent preference for trafficking toward and away from the cell body. The speed of trafficking depends on the stage of sperm development, ranging from highly mobile unrestricted diffusion initially, mobile punctate structures in developing sperm, and immobile punctate structures in mature sperm. Our studies demonstrate that mouse intraflagellar transport 25 travels along the developing sperm flagella in two directions that might be essential for functional sperm formation.
Collapse
Affiliation(s)
- Wei Li
- Department of Physiology, Wayne State University, Detroit, MI, USA
| | - Changmin Niu
- Department of Physiology, Wayne State University, Detroit, MI, USA
- School of Nursing School of Public Health, Yangzhou University, Yangzhou, Jiangsu, China
| | - Yi Tian Yap
- Department of Physiology, Wayne State University, Detroit, MI, USA
| | - Tao Li
- Department of Physiology, Wayne State University, Detroit, MI, USA
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Cheng Zheng
- Department of Physiology, Wayne State University, Detroit, MI, USA
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University of Science and Technology, Wuhan, Hubei, PR China
| | | | | | | | - Jie Xu
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Jifeng Zhang
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Christopher V Kelly
- Department of Physics and Astronomy, Wayne State University, Detroit, MI, USA
| | - Zhibing Zhang
- Department of Physiology, Wayne State University, Detroit, MI, USA
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, USA
| |
Collapse
|
5
|
Formstone C, Aldeiri B, Davenport M, Francis‐West P. Ventral body wall closure: Mechanistic insights from mouse models and translation to human pathology. Dev Dyn 2025; 254:102-141. [PMID: 39319771 PMCID: PMC11809137 DOI: 10.1002/dvdy.735] [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: 12/19/2023] [Revised: 08/17/2024] [Accepted: 08/21/2024] [Indexed: 09/26/2024] Open
Abstract
The ventral body wall (VBW) that encloses the thoracic and abdominal cavities arises by extensive cell movements and morphogenetic changes during embryonic development. These morphogenetic processes include embryonic folding generating the primary body wall; the initial ventral cover of the embryo, followed by directed mesodermal cell migrations, contributing to the secondary body wall. Clinical anomalies in VBW development affect approximately 1 in 3000 live births. However, the cell interactions and critical cellular behaviors that control VBW development remain little understood. Here, we describe the embryonic origins of the VBW, the cellular and morphogenetic processes, and key genes, that are essential for VBW development. We also provide a clinical overview of VBW anomalies, together with environmental and genetic influences, and discuss the insight gained from over 70 mouse models that exhibit VBW defects, and their relevance, with respect to human pathology. In doing so we propose a phenotypic framework for researchers in the field which takes into account the clinical picture. We also highlight cases where there is a current paucity of mouse models for particular clinical defects and key gaps in knowledge about embryonic VBW development that need to be addressed to further understand mechanisms of human VBW pathologies.
Collapse
Affiliation(s)
- Caroline Formstone
- Department of Clinical, Pharmaceutical and Biological SciencesUniversity of HertfordshireHatfieldUK
| | - Bashar Aldeiri
- Department of Paediatric SurgeryChelsea and Westminster HospitalLondonUK
| | - Mark Davenport
- Department of Paediatric SurgeryKing's College HospitalLondonUK
| | | |
Collapse
|
6
|
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] [Download PDF] [Figures] [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
|
7
|
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
|
8
|
Gupta M, Lewis TR, Stuck MW, Spencer WJ, Klementieva NV, Arshavsky VY, Pazour GJ. Inpp5e Is Critical for Photoreceptor Outer Segment Maintenance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.27.609873. [PMID: 39253441 PMCID: PMC11383302 DOI: 10.1101/2024.08.27.609873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
In humans, inositol polyphosphate-5-phosphatase e (INPP5E) mutations cause retinal degeneration as part of Joubert and MORM syndromes and can also cause non-syndromic blindness. In mice, mutations cause a spectrum of brain, kidney, and other anomalies and prevent the formation of photoreceptor outer segments. To further explore the function of Inpp5e in photoreceptors, we generated conditional and inducible knockouts of mouse Inpp5e where the gene was deleted either during outer segment formation or after outer segments were fully formed. In both cases, the loss of Inpp5e led to severe defects in photoreceptor outer segment morphology and ultimately photoreceptor cell loss. The primary morphological defect consisted of outer segment shortening and reduction in the number of newly forming discs at the outer segment base. This was accompanied by structural abnormalities of the Golgi apparatus, mislocalized rhodopsin, and an accumulation of extracellular vesicles. In addition, knockout cells showed a reduction in the size and prevalence of the actin network at the site of new disc morphogenesis and the occasional formation of membrane whorls instead of discs in a subset of cells. Together, these data demonstrate that Inpp5e plays a critical role in maintaining the outer segment and the normal process of outer segment renewal depends on the activity of this enzyme.
Collapse
|
9
|
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
|
10
|
Kodzik N, Ciereszko A, Judycka S, Słowińska M, Szczepkowska B, Świderska B, Dietrich MA. Cryoprotectant-specific alterations in the proteome of Siberian sturgeon spermatozoa induced by cryopreservation. Sci Rep 2024; 14:17707. [PMID: 39085328 PMCID: PMC11291920 DOI: 10.1038/s41598-024-68395-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 07/23/2024] [Indexed: 08/02/2024] Open
Abstract
Cryopreservation is crucial for conserving genetic diversity in endangered species including the critically endangered group of sturgeons (Acipenseridae), but it can compromise sperm quality and protein profiles. Although cryopreservation with dimethyl sulfoxide (DMSO) and methanol (MeOH) results in the recovery of good post-thaw motility, DMSO-preserved sperm show reduced fertilization ability. This study was conducted in Siberian sturgeon as a model for Acipenserid fishes to explore the effects of DMSO and MeOH on the proteome of semen using advanced proteomics methods-liquid chromatography‒mass spectrometry and two-dimensional difference gel electrophoresis. We analyzed the proteomic profiles of fresh and cryopreserved spermatozoa and their extracellular medium and showed that cryopreservation decreases motility and viability and increases reactive oxygen species levels, membrane fluidity, and acrosome damage. Despite having similar post-thaw semen motility, sperm treated with DMSO had significantly lower fertilization success (6.2%) than those treated with MeOH (51.2%). A total of 224 and 118 differentially abundant proteins were identified in spermatozoa preserved with MeOH and DMSO, respectively. MeOH-related proteins were linked to chromosomal structure and mitochondrial functionality, while DMSO-related proteins impacted fertilization by altering the acrosome reaction and binding of sperm to the zona pellucida and nuclear organization. Additionally, cryopreservation led to alterations in the proacrosin/acrosin system in both cryoprotectants. This study provides the first comprehensive proteomic characterization of Siberian sturgeon sperm after cryopreservation, offering insights into how cryoprotectants impact fertilization ability.
Collapse
Affiliation(s)
- Natalia Kodzik
- Department of Gamete and Embryo Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland
| | - Andrzej Ciereszko
- Department of Gamete and Embryo Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland
| | - Sylwia Judycka
- Department of Gamete and Embryo Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland
| | - Mariola Słowińska
- Department of Gamete and Embryo Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland
| | - Bożena Szczepkowska
- Department of Sturgeon Fish Breeding, National Inland Fisheries Research Institute in Olsztyn, 11-610, Pozezdrze, Pieczarki, Poland
| | - Bianka Świderska
- Mass Spectrometry Laboratory, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Pawinskiego 5a, 02-106, Warsaw, Poland
| | - Mariola A Dietrich
- Department of Gamete and Embryo Biology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland.
| |
Collapse
|
11
|
Long X, Chen L, Xiao X, Min X, Wu Y, Yang Z, Wen X. Structure, function, and research progress of primary cilia in reproductive physiology and reproductive diseases. Front Cell Dev Biol 2024; 12:1418928. [PMID: 38887518 PMCID: PMC11180893 DOI: 10.3389/fcell.2024.1418928] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 05/16/2024] [Indexed: 06/20/2024] Open
Abstract
Primary cilia, serving as the central hub for cellular signal transduction, possess the remarkable ability to translate diverse extracellular signals, both chemical and mechanical, into intracellular responses. Their ubiquitous presence in the reproductive system underscores their pivotal roles in various cellular processes including development, differentiation, and migration. Emerging evidence suggests primary cilia as key players in reproductive physiology and associated pathologies. Notably, primary cilia have been identified in granulosa cells within mouse ovaries and uterine stromal cells, and perturbations in their structure and function have been implicated in a spectrum of reproductive dysfunctions and ciliary-related diseases. Furthermore, disruptions in primary cilia-mediated signal transduction pathways under pathological conditions exacerbate the onset and progression of reproductive disorders. This review provides a comprehensive overview of current research progress on primary cilia and their associated signaling pathways in reproductive physiology and diseases, with the aim of furnishing theoretical groundwork for the prevention and management of primary cilia-related structural and functional abnormalities contributing to reproductive system pathologies.
Collapse
Affiliation(s)
- Xiaochuan Long
- Clinical Veterinary Laboratory, College of Animal Science, Guizhou University, Guizhou, China
- Key Laboratory of Animal Genetic, Breeding and Reproduction in the plateau Mountainous Region, Ministry of Education, Guizhou University, Guizhou, China
| | - Li Chen
- Clinical Veterinary Laboratory, College of Animal Science, Guizhou University, Guizhou, China
- Key Laboratory of Animal Genetic, Breeding and Reproduction in the plateau Mountainous Region, Ministry of Education, Guizhou University, Guizhou, China
| | - Xinyao Xiao
- Clinical Veterinary Laboratory, College of Animal Science, Guizhou University, Guizhou, China
- Key Laboratory of Animal Genetic, Breeding and Reproduction in the plateau Mountainous Region, Ministry of Education, Guizhou University, Guizhou, China
| | - Xiayu Min
- Clinical Veterinary Laboratory, College of Animal Science, Guizhou University, Guizhou, China
- Key Laboratory of Animal Genetic, Breeding and Reproduction in the plateau Mountainous Region, Ministry of Education, Guizhou University, Guizhou, China
| | - Yao Wu
- Clinical Veterinary Laboratory, College of Animal Science, Guizhou University, Guizhou, China
- Key Laboratory of Animal Genetic, Breeding and Reproduction in the plateau Mountainous Region, Ministry of Education, Guizhou University, Guizhou, China
| | - Zengming Yang
- Key Laboratory of Animal Genetic, Breeding and Reproduction in the plateau Mountainous Region, Ministry of Education, Guizhou University, Guizhou, China
- Basic Veterinary Laboratory, College of Animal Science, Guizhou University, Guizhou, China
| | - Xin Wen
- Clinical Veterinary Laboratory, College of Animal Science, Guizhou University, Guizhou, China
- Key Laboratory of Animal Genetic, Breeding and Reproduction in the plateau Mountainous Region, Ministry of Education, Guizhou University, Guizhou, China
| |
Collapse
|
12
|
Tingey M, Ruba A, Jiang Z, Yang W. Deciphering vesicle-assisted transport mechanisms in cytoplasm to cilium trafficking. Front Cell Neurosci 2024; 18:1379976. [PMID: 38860265 PMCID: PMC11163138 DOI: 10.3389/fncel.2024.1379976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 05/13/2024] [Indexed: 06/12/2024] Open
Abstract
The cilium, a pivotal organelle crucial for cell signaling and proper cell function, relies on meticulous macromolecular transport from the cytoplasm for its formation and maintenance. While the intraflagellar transport (IFT) pathway has traditionally been the focus of extensive study concerning ciliogenesis and ciliary maintenance, recent research highlights a complementary and alternative mechanism-vesicle-assisted transport (VAT) in cytoplasm to cilium trafficking. Despite its potential significance, the VAT pathway remains largely uncharacterized. This review explores recent studies providing evidence for the dynamics of vesicle-related diffusion and transport within the live primary cilium, employing high-speed super-resolution light microscopy. Additionally, we analyze the spatial distribution of vesicles in the cilium, mainly relying on electron microscopy data. By scrutinizing the VAT pathways that facilitate cargo transport into the cilium, with a specific emphasis on recent advancements and imaging data, our objective is to synthesize a comprehensive model of ciliary transport through the integration of IFT-VAT mechanisms.
Collapse
Affiliation(s)
| | | | | | - Weidong Yang
- Department of Biology, Temple University, Philadelphia, PA, United States
| |
Collapse
|
13
|
Han S, Hu Y, Jia D, Lv Y, Liu M, Wang D, Chao J, Xia X, Wang Q, Liu P, Cai Y, Ren X. IFT27 regulates the long-term maintenance of photoreceptor outer segments in zebrafish. Gene 2024; 905:148237. [PMID: 38310983 DOI: 10.1016/j.gene.2024.148237] [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: 11/30/2023] [Revised: 01/22/2024] [Accepted: 01/30/2024] [Indexed: 02/06/2024]
Abstract
Approximately a quarter of Retinitis Pigmentosa (RP) is caused by mutations in transport-related genes in cilia. IFT27 (Intraflagellar Transport 27), a core component of the ciliary intraflagellar transport (IFT) system, has been implicated as a significant pathogenic gene in RP. The pathogenic mechanisms and subsequent pathology related to IFT27 mutations in RP are largely obscure. Here, we utilized TALEN technology to create an ift27 knockout (ift27-/-) zebrafish model. Electroretinography (ERG) detection showed impaired vision in this model. Histopathological examinations disclosed that ift27 mutations cause progressive degeneration of photoreceptors in zebrafish, and this degeneration was late-onset. Immunofluorescence labeling of outer segments showed that rods degenerated before cones, aligning with the conventional characterization of RP. In cultured human retinal pigment epithelial cells, we found that IFT27 was involved in maintaining ciliary morphology. Furthermore, decreased IFT27 expression resulted in the inhibition of the Hedgehog (Hh) signaling pathway, including decreased expression of key factors in the Hh pathway and abnormal localization of the ciliary mediator Gli2. In summary, we generated an ift27-/- zebrafish line with retinal degeneration which mimicked the symptoms of RP patients, highlighting IFT27's integral role in the long-term maintenance of cilia via the Hh signaling pathway. This work may furnish new insights into the treatment or delay of RP caused by IFT27 mutations.
Collapse
Affiliation(s)
- Shanshan Han
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, Hubei, China; College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, Hubei, China.
| | - Yue Hu
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, Hubei, China; College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, Hubei, China
| | - Danna Jia
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China
| | - Yuexia Lv
- Prenatal Diagnosis Center, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Mugen Liu
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Decheng Wang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, Hubei, China; College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, Hubei, China
| | - Jin Chao
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, Hubei, China; College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, Hubei, China
| | - Xuan Xia
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, Hubei, China; College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, Hubei, China
| | - Qiong Wang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, Hubei, China; College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, Hubei, China
| | - Pei Liu
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, Hubei, China; College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, Hubei, China
| | - Yu Cai
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, Hubei, China; College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, Hubei, China
| | - Xiang Ren
- Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
14
|
Mukhopadhyay AG, Toropova K, Daly L, Wells JN, Vuolo L, Mladenov M, Seda M, Jenkins D, Stephens DJ, Roberts AJ. Structure and tethering mechanism of dynein-2 intermediate chains in intraflagellar transport. EMBO J 2024; 43:1257-1272. [PMID: 38454149 PMCID: PMC10987677 DOI: 10.1038/s44318-024-00060-1] [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: 09/05/2023] [Revised: 02/06/2024] [Accepted: 02/09/2024] [Indexed: 03/09/2024] Open
Abstract
Dynein-2 is a large multiprotein complex that powers retrograde intraflagellar transport (IFT) of cargoes within cilia/flagella, but the molecular mechanism underlying this function is still emerging. Distinctively, dynein-2 contains two identical force-generating heavy chains that interact with two different intermediate chains (WDR34 and WDR60). Here, we dissect regulation of dynein-2 function by WDR34 and WDR60 using an integrative approach including cryo-electron microscopy and CRISPR/Cas9-enabled cell biology. A 3.9 Å resolution structure shows how WDR34 and WDR60 use surprisingly different interactions to engage equivalent sites of the two heavy chains. We show that cilia can assemble in the absence of either WDR34 or WDR60 individually, but not both subunits. Dynein-2-dependent distribution of cargoes depends more strongly on WDR60, because the unique N-terminal extension of WDR60 facilitates dynein-2 targeting to cilia. Strikingly, this N-terminal extension can be transplanted onto WDR34 and retain function, suggesting it acts as a flexible tether to the IFT "trains" that assemble at the ciliary base. We discuss how use of unstructured tethers represents an emerging theme in IFT train interactions.
Collapse
Affiliation(s)
- Aakash G Mukhopadhyay
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
- Institute of Structural and Molecular Biology, Department of Biological Sciences, Birkbeck, University of London, London, UK
| | - Katerina Toropova
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
- Institute of Structural and Molecular Biology, Department of Biological Sciences, Birkbeck, University of London, London, UK
| | - Lydia Daly
- Institute of Structural and Molecular Biology, Department of Biological Sciences, Birkbeck, University of London, London, UK
- Randall Centre of Cell & Molecular Biophysics, King's College London, London, UK
| | - Jennifer N Wells
- Institute of Structural and Molecular Biology, Department of Biological Sciences, Birkbeck, University of London, London, UK
- MRC London Institute of Medical Sciences (LMS), London, UK
| | - Laura Vuolo
- Cell Biology Laboratories, School of Biochemistry, University of Bristol, Bristol, UK
| | - Miroslav Mladenov
- Institute of Structural and Molecular Biology, Department of Biological Sciences, Birkbeck, University of London, London, UK
- Cellular Signalling and Cytoskeletal Function Laboratory, The Francis Crick Institute, London, UK
| | - Marian Seda
- UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Dagan Jenkins
- UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | - David J Stephens
- Cell Biology Laboratories, School of Biochemistry, University of Bristol, Bristol, UK
| | - Anthony J Roberts
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK.
- Institute of Structural and Molecular Biology, Department of Biological Sciences, Birkbeck, University of London, London, UK.
| |
Collapse
|
15
|
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
|
16
|
Francis RJB, San Agustin JT, Szabo Rogers HL, Cui C, Jonassen JA, Eguether T, Follit JA, Lo CW, Pazour GJ. Autonomous and non-cell autonomous role of cilia in structural birth defects in mice. PLoS Biol 2023; 21:e3002425. [PMID: 38079449 PMCID: PMC10735189 DOI: 10.1371/journal.pbio.3002425] [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: 06/26/2023] [Revised: 12/21/2023] [Accepted: 11/09/2023] [Indexed: 12/21/2023] Open
Abstract
Ciliopathies are associated with wide spectrum of structural birth defects (SBDs), indicating important roles for cilia in development. Here, we provide novel insights into the temporospatial requirement for cilia in SBDs arising from deficiency in Ift140, an intraflagellar transport (IFT) protein regulating ciliogenesis. Ift140-deficient mice exhibit cilia defects accompanied by wide spectrum of SBDs including macrostomia (craniofacial defects), exencephaly, body wall defects, tracheoesophageal fistula (TEF), randomized heart looping, congenital heart defects (CHDs), lung hypoplasia, renal anomalies, and polydactyly. Tamoxifen inducible CAGGCre-ER deletion of a floxed Ift140 allele between E5.5 to 9.5 revealed early requirement for Ift140 in left-right heart looping regulation, mid to late requirement for cardiac outflow septation and alignment, and late requirement for craniofacial development and body wall closure. Surprisingly, CHD were not observed with 4 Cre drivers targeting different lineages essential for heart development, but craniofacial defects and omphalocele were observed with Wnt1-Cre targeting neural crest and Tbx18-Cre targeting epicardial lineage and rostral sclerotome through which trunk neural crest cells migrate. These findings revealed cell autonomous role of cilia in cranial/trunk neural crest-mediated craniofacial and body wall closure defects, while non-cell autonomous multi-lineage interactions underlie CHD pathogenesis, revealing unexpected developmental complexity for CHD associated with ciliopathies.
Collapse
Affiliation(s)
- Richard J. B. Francis
- Department of Developmental Biology, University of Pittsburgh, Rangos Research Center, Pittsburgh, Pennsylvania, United States of America
- Discipline of Biomedical Sciences and Molecular Biology; College of Public Health, Medical and Veterinary Science, James Cook University, Townsville, Australia
| | - Jovenal T. San Agustin
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| | - Heather L. Szabo Rogers
- Department of Developmental Biology, University of Pittsburgh, Rangos Research Center, Pittsburgh, Pennsylvania, United States of America
- Center for Craniofacial Regeneration, Department of Oral Biology, School of Dental Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Cheng Cui
- Department of Developmental Biology, University of Pittsburgh, Rangos Research Center, Pittsburgh, Pennsylvania, United States of America
| | - Julie A. Jonassen
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| | - Thibaut Eguether
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| | - John A. Follit
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| | - Cecilia W. Lo
- Department of Developmental Biology, University of Pittsburgh, Rangos Research Center, Pittsburgh, Pennsylvania, United States of America
| | - Gregory J. Pazour
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| |
Collapse
|
17
|
Boegholm N, Petriman NA, Loureiro‐López M, Wang J, Vela MIS, Liu B, Kanie T, Ng R, Jackson PK, Andersen JS, Lorentzen E. The IFT81-IFT74 complex acts as an unconventional RabL2 GTPase-activating protein during intraflagellar transport. EMBO J 2023; 42:e111807. [PMID: 37606072 PMCID: PMC10505919 DOI: 10.15252/embj.2022111807] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 07/24/2023] [Accepted: 08/03/2023] [Indexed: 08/23/2023] Open
Abstract
Cilia are important cellular organelles for signaling and motility and are constructed via intraflagellar transport (IFT). RabL2 is a small GTPase that localizes to the basal body of cilia via an interaction with the centriolar protein CEP19 before downstream association with the IFT machinery, which is followed by initiation of IFT. We reconstituted and purified RabL2 with CEP19 or IFT proteins to show that a reconstituted pentameric IFT complex containing IFT81/74 enhances the GTP hydrolysis rate of RabL2. The binding site on IFT81/74 that promotes GTP hydrolysis in RabL2 was mapped to a 70-amino-acid-long coiled-coil region of IFT81/74. We present structural models for RabL2-containing IFT complexes that we validate in vitro and in cellulo and demonstrate that Chlamydomonas IFT81/74 enhances GTP hydrolysis of human RabL2, suggesting an ancient evolutionarily conserved activity. Our results provide an architectural understanding of how RabL2 is incorporated into the IFT complex and a molecular rationale for why RabL2 dissociates from anterograde IFT trains soon after departure from the ciliary base.
Collapse
Affiliation(s)
- Niels Boegholm
- Department of Molecular Biology and GeneticsAarhus UniversityAarhus CDenmark
| | - Narcis A Petriman
- Department of Molecular Biology and GeneticsAarhus UniversityAarhus CDenmark
| | - Marta Loureiro‐López
- Department for Biochemistry and Molecular BiologyUniversity of Southern DenmarkOdense MDenmark
| | - Jiaolong Wang
- Department of Molecular Biology and GeneticsAarhus UniversityAarhus CDenmark
| | | | - Beibei Liu
- Department of Cell BiologyUniversity of Oklahoma Health Science CenterOklahomaOKUSA
| | - Tomoharu Kanie
- Department of Cell BiologyUniversity of Oklahoma Health Science CenterOklahomaOKUSA
- Baxter Laboratory, Department of Microbiology & ImmunologyStanford University School of MedicineStanfordCAUSA
| | - Roy Ng
- Baxter Laboratory, Department of Microbiology & ImmunologyStanford University School of MedicineStanfordCAUSA
| | - Peter K Jackson
- Baxter Laboratory, Department of Microbiology & ImmunologyStanford University School of MedicineStanfordCAUSA
- Department of PathologyStanford University School of MedicineStanfordCAUSA
| | - Jens S Andersen
- Department for Biochemistry and Molecular BiologyUniversity of Southern DenmarkOdense MDenmark
| | - Esben Lorentzen
- Department of Molecular Biology and GeneticsAarhus UniversityAarhus CDenmark
| |
Collapse
|
18
|
Xu J, Iyyanar PPR, Lan Y, Jiang R. Sonic hedgehog signaling in craniofacial development. Differentiation 2023; 133:60-76. [PMID: 37481904 PMCID: PMC10529669 DOI: 10.1016/j.diff.2023.07.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 07/04/2023] [Accepted: 07/12/2023] [Indexed: 07/25/2023]
Abstract
Mutations in SHH and several other genes encoding components of the Hedgehog signaling pathway have been associated with holoprosencephaly syndromes, with craniofacial anomalies ranging in severity from cyclopia to facial cleft to midfacial and mandibular hypoplasia. Studies in animal models have revealed that SHH signaling plays crucial roles at multiple stages of craniofacial morphogenesis, from cranial neural crest cell survival to growth and patterning of the facial primordia to organogenesis of the palate, mandible, tongue, tooth, and taste bud formation and homeostasis. This article provides a summary of the major findings in studies of the roles of SHH signaling in craniofacial development, with emphasis on recent advances in the understanding of the molecular and cellular mechanisms regulating the SHH signaling pathway activity and those involving SHH signaling in the formation and patterning of craniofacial structures.
Collapse
Affiliation(s)
- Jingyue Xu
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA.
| | - Paul P R Iyyanar
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Yu Lan
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA; Division of Plastic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA; Departments of Pediatrics and Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Rulang Jiang
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA; Division of Plastic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA; Departments of Pediatrics and Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA.
| |
Collapse
|
19
|
Tsang TH, Wiese M, Helmstädter M, Stehle T, Seyfferth J, Shvedunova M, Holz H, Walz G, Akhtar A. Transcriptional regulation by the NSL complex enables diversification of IFT functions in ciliated versus nonciliated cells. SCIENCE ADVANCES 2023; 9:eadh5598. [PMID: 37624894 PMCID: PMC10456878 DOI: 10.1126/sciadv.adh5598] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 07/25/2023] [Indexed: 08/27/2023]
Abstract
Members of the NSL histone acetyltransferase complex are involved in multiorgan developmental syndromes. While the NSL complex is known for its importance in early development, its role in fully differentiated cells remains enigmatic. Using a kidney-specific model, we discovered that deletion of NSL complex members KANSL2 or KANSL3 in postmitotic podocytes led to catastrophic kidney dysfunction. Systematic comparison of two primary differentiated cell types reveals the NSL complex as a master regulator of intraciliary transport genes in both dividing and nondividing cells. NSL complex ablation led to loss of cilia and impaired sonic hedgehog pathway in ciliated fibroblasts. By contrast, nonciliated podocytes responded with altered microtubule dynamics and obliterated podocyte functions. Finally, overexpression of wild-type but not a double zinc finger (ZF-ZF) domain mutant of KANSL2 rescued the transcriptional defects, revealing a critical function of this domain in NSL complex assembly and function. Thus, the NSL complex exhibits bifurcation of functions to enable diversity of specialized outcomes in differentiated cells.
Collapse
Affiliation(s)
- Tsz Hong Tsang
- Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
- International Max Planck Research School for Molecular and Cellular Biology (IMPRS-MCB), 79108 Freiburg, Germany
| | - Meike Wiese
- Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Martin Helmstädter
- Department of Medicine IV, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106 Freiburg, Germany
| | - Thomas Stehle
- Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Janine Seyfferth
- Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Maria Shvedunova
- Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Herbert Holz
- Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Gerd Walz
- Department of Medicine IV, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106 Freiburg, Germany
- BIOSS Centre for Biological Signalling Studies, University of Freiburg, Schänzlestrasse 18, 79104 Freiburg, Germany
- CIBSS Centre for Integrative Biological Signalling Studies, University of Freiburg, Schänzlestrasse 18, 79104 Freiburg, Germany
| | - Asifa Akhtar
- Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
- CIBSS Centre for Integrative Biological Signalling Studies, University of Freiburg, Schänzlestrasse 18, 79104 Freiburg, Germany
| |
Collapse
|
20
|
Zhang RK, Sun WY, Liu YX, Zhang EY, Fan ZC. RABL2 promotes the outward transition zone passage of signaling proteins in cilia via ARL3. Proc Natl Acad Sci U S A 2023; 120:e2302603120. [PMID: 37579161 PMCID: PMC10450674 DOI: 10.1073/pnas.2302603120] [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: 02/20/2023] [Accepted: 07/10/2023] [Indexed: 08/16/2023] Open
Abstract
Certain transmembrane and membrane-tethered signaling proteins export from cilia as BBSome cargoes via the outward BBSome transition zone (TZ) diffusion pathway, indispensable for maintaining their ciliary dynamics to enable cells to sense and transduce extracellular stimuli inside the cell. Murine Rab-like 2 (Rabl2) GTPase resembles Chlamydomonas Arf-like 3 (ARL3) GTPase in promoting outward TZ passage of the signaling protein cargo-laden BBSome. During this process, ARL3 binds to and recruits the retrograde IFT train-dissociated BBSome as its effector to diffuse through the TZ for ciliary retrieval, while how RABL2 and ARL3 cross talk in this event remains uncertain. Here, we report that Chlamydomonas RABL2 in a GTP-bound form (RABL2GTP) cycles through cilia via IFT as an IFT-B1 cargo, dissociates from retrograde IFT trains at a ciliary region right above the TZ, and converts to RABL2GDP for activating ARL3GDP as an ARL3 guanine nucleotide exchange factor. This confers ARL3GTP to detach from the ciliary membrane and become available for binding and recruiting the phospholipase D (PLD)-laden BBSome, autonomous of retrograde IFT association, to diffuse through the TZ for ciliary retrieval. Afterward, RABL2GDP exits cilia by being bound to the ARL3GTP/BBSome entity as a BBSome cargo. Our data identify ciliary signaling proteins exported from cilia via the RABL2-ARL3 cascade-mediated outward BBSome TZ diffusion pathway. According to this model, hedgehog signaling defect-induced Bardet-Biedl syndrome caused by RABL2 mutations in humans could be well explained in a mutation-specific manner, providing us with a mechanistic understanding behind the outward BBSome TZ passage required for proper ciliary signaling.
Collapse
Affiliation(s)
- Rui-Kai Zhang
- State Key Laboratory of Food Nutrition and Safety, Institute of Health Biotechnology, Tianjin University of Science and Technology, Tianjin300457, China
| | - Wei-Yue Sun
- State Key Laboratory of Food Nutrition and Safety, Institute of Health Biotechnology, Tianjin University of Science and Technology, Tianjin300457, China
| | - Yan-Xia Liu
- State Key Laboratory of Food Nutrition and Safety, Institute of Health Biotechnology, Tianjin University of Science and Technology, Tianjin300457, China
| | | | - Zhen-Chuan Fan
- State Key Laboratory of Food Nutrition and Safety, Institute of Health Biotechnology, Tianjin University of Science and Technology, Tianjin300457, China
| |
Collapse
|
21
|
Liu J, Hermo L, Ding D, Wei C, Mann JM, Yan X, Melnick AF, Wu Y, Withrow A, Cibelli J, Hess RA, Chen C. SYPL1 defines a vesicular pathway essential for sperm cytoplasmic droplet formation and male fertility. Nat Commun 2023; 14:5113. [PMID: 37607933 PMCID: PMC10444883 DOI: 10.1038/s41467-023-40862-1] [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: 03/18/2023] [Accepted: 08/14/2023] [Indexed: 08/24/2023] Open
Abstract
The cytoplasmic droplet is a conserved dilated area of cytoplasm situated at the neck of the sperm flagellum. Viewed as residual cytoplasm inherited from late spermatids, the cytoplasmic droplet contains numerous saccular elements as its key content. However, the origin of these saccules and the function of the cytoplasmic droplet have long been speculative. Here, we identify the molecular origin of these cytoplasmic droplet components by uncovering a vesicle pathway essential for formation and sequestration of saccules within the cytoplasmic droplet. This process is governed by a transmembrane protein SYPL1 and its interaction with VAMP3. Genetic ablation of SYPL1 in mice reveals that SYPL1 dictates the formation and accumulation of saccular elements in the forming cytoplasmic droplet. Derived from the Golgi, SYPL1 vesicles are critical for segregation of key metabolic enzymes within the forming cytoplasmic droplet of late spermatids and epididymal sperm, which are required for sperm development and male fertility. Our results uncover a mechanism to actively form and segregate saccules within the cytoplasmic droplet to promote sperm fertility.
Collapse
Affiliation(s)
- Jiali Liu
- Department of Animal Science, Michigan State University, East Lansing, MI, USA
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Louis Hermo
- Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec, Canada
| | - Deqiang Ding
- Department of Animal Science, Michigan State University, East Lansing, MI, USA
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Chao Wei
- Department of Animal Science, Michigan State University, East Lansing, MI, USA
| | - Jeffrey M Mann
- Department of Animal Science, Michigan State University, East Lansing, MI, USA
| | - Xiaoyuan Yan
- Department of Animal Science, Michigan State University, East Lansing, MI, USA
| | - Ashley F Melnick
- Department of Animal Science, Michigan State University, East Lansing, MI, USA
| | - Yingjie Wu
- Department of Animal Science, Michigan State University, East Lansing, MI, USA
- College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Alicia Withrow
- Center for Advanced Microscopy, Michigan State University, East Lansing, MI, USA
| | - Jose Cibelli
- Department of Animal Science, Michigan State University, East Lansing, MI, USA
| | - Rex A Hess
- Department of Comparative Biosciences, University of Illinois, Urbana, Illinois, USA
| | - Chen Chen
- Department of Animal Science, Michigan State University, East Lansing, MI, USA.
- Reproductive and Developmental Sciences Program, Michigan State University, East Lansing, MI, USA.
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, Grand Rapids, MI, USA.
| |
Collapse
|
22
|
Tian X, Zhao H, Zhou J. Organization, functions, and mechanisms of the BBSome in development, ciliopathies, and beyond. eLife 2023; 12:e87623. [PMID: 37466224 DOI: 10.7554/elife.87623] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 07/06/2023] [Indexed: 07/20/2023] Open
Abstract
The BBSome is an octameric protein complex that regulates ciliary transport and signaling. Mutations in BBSome subunits are closely associated with ciliary defects and lead to ciliopathies, notably Bardet-Biedl syndrome. Over the past few years, there has been significant progress in elucidating the molecular organization and functions of the BBSome complex. An improved understanding of BBSome-mediated biological events and molecular mechanisms is expected to help advance the development of diagnostic and therapeutic approaches for BBSome-related diseases. Here, we review the current literature on the structural assembly, transport regulation, and molecular functions of the BBSome, emphasizing its roles in cilium-related processes. We also provide perspectives on the pathological role of the BBSome in ciliopathies as well as how these can be exploited for therapeutic benefit.
Collapse
Affiliation(s)
- Xiaoyu Tian
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Huijie Zhao
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Jun Zhou
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
- State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Cell Ecosystem, College of Life Sciences, Nankai University, Tianjin, China
| |
Collapse
|
23
|
Bakey Z, Cabrera OA, Hoefele J, Antony D, Wu K, Stuck MW, Micha D, Eguether T, Smith AO, van der Wel NN, Wagner M, Strittmatter L, Beales PL, Jonassen JA, Thiffault I, Cadieux-Dion M, Boyes L, Sharif S, Tüysüz B, Dunstheimer D, Niessen HWM, Devine W, Lo CW, Mitchison HM, Schmidts M, Pazour GJ. IFT74 variants cause skeletal ciliopathy and motile cilia defects in mice and humans. PLoS Genet 2023; 19:e1010796. [PMID: 37315079 DOI: 10.1371/journal.pgen.1010796] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 05/23/2023] [Indexed: 06/16/2023] Open
Abstract
Motile and non-motile cilia play critical roles in mammalian development and health. These organelles are composed of a 1000 or more unique proteins, but their assembly depends entirely on proteins synthesized in the cell body and transported into the cilium by intraflagellar transport (IFT). In mammals, malfunction of non-motile cilia due to IFT dysfunction results in complex developmental phenotypes that affect most organs. In contrast, disruption of motile cilia function causes subfertility, disruption of the left-right body axis, and recurrent airway infections with progressive lung damage. In this work, we characterize allele specific phenotypes resulting from IFT74 dysfunction in human and mice. We identified two families carrying a deletion encompassing IFT74 exon 2, the first coding exon, resulting in a protein lacking the first 40 amino acids and two individuals carrying biallelic splice site mutations. Homozygous exon 2 deletion cases presented a ciliary chondrodysplasia with narrow thorax and progressive growth retardation along with a mucociliary clearance disorder phenotype with severely shorted cilia. Splice site variants resulted in a lethal skeletal chondrodysplasia phenotype. In mice, removal of the first 40 amino acids likewise results in a motile cilia phenotype but with little effect on primary cilia structure. Mice carrying this allele are born alive but are growth restricted and developed hydrocephaly in the first month of life. In contrast, a strong, likely null, allele of Ift74 in mouse completely blocks ciliary assembly and causes severe heart defects and midgestational lethality. In vitro studies suggest that the first 40 amino acids of IFT74 are dispensable for binding of other IFT subunits but are important for tubulin binding. Higher demands on tubulin transport in motile cilia compared to primary cilia resulting from increased mechanical stress and repair needs could account for the motile cilia phenotype observed in human and mice.
Collapse
Affiliation(s)
- Zeineb Bakey
- Center for Pediatrics and Adolescent Medicine, University Hospital Freiburg, Freiburg University Faculty of Medicine, Freiburg, Germany
- Human Genetics Department, Radboud University Medical Center Nijmegen and Radboud Institute for Molecular Life Sciences (RIMLS), Nijmegen, The Netherlands
| | - Oscar A Cabrera
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Biotech II, Worcester, Massachusetts, United States of America
| | - Julia Hoefele
- Institute for Human Genetics, Technical University Munich (TUM), School of Medicine, Munich, Germany
| | - Dinu Antony
- Center for Pediatrics and Adolescent Medicine, University Hospital Freiburg, Freiburg University Faculty of Medicine, Freiburg, Germany
- Human Genetics Department, Radboud University Medical Center Nijmegen and Radboud Institute for Molecular Life Sciences (RIMLS), Nijmegen, The Netherlands
| | - Kaman Wu
- Human Genetics Department, Radboud University Medical Center Nijmegen and Radboud Institute for Molecular Life Sciences (RIMLS), Nijmegen, The Netherlands
| | - Michael W Stuck
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Biotech II, Worcester, Massachusetts, United States of America
| | - Dimitra Micha
- Department of Human Genetics, Amsterdam Movement Sciences, Amsterdam UMC, Amsterdam, The Netherlands
| | - Thibaut Eguether
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Biotech II, Worcester, Massachusetts, United States of America
| | - Abigail O Smith
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Biotech II, Worcester, Massachusetts, United States of America
| | - Nicole N van der Wel
- Electron microscopy Center Amsterdam, Department of Medical Biology, VUMC, Amsterdam, The Netherlands
| | - Matias Wagner
- Institute for Human Genetics, Technical University Munich (TUM), School of Medicine, Munich, Germany
| | - Lara Strittmatter
- Electron Microscopy Core, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| | - Philip L Beales
- Genetics and Genomic Medicine Programme, University College London, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Julie A Jonassen
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, Massachusetts, United States of America
| | - Isabelle Thiffault
- Genomic Medicine Center, Children's Mercy Hospital, Kansas City, Missouri, United States of America
| | - Maxime Cadieux-Dion
- Genomic Medicine Center, Children's Mercy Hospital, Kansas City, Missouri, United States of America
| | - Laura Boyes
- West Midlands Genomic Medicine Hub, Birmingham Women's Hospital, Birmingham, United Kingdom
| | - Saba Sharif
- West Midlands Genomic Medicine Hub, Birmingham Women's Hospital, Birmingham, United Kingdom
| | - Beyhan Tüysüz
- Department of Pediatrics, Division of Pediatric Genetics, Cerrahpasa Medical Faculty, University-Cerrahpasa, Istanbul, Turkey
| | - Desiree Dunstheimer
- Center for Pediatrics and Adolescent Medicine, University Hospital Augsburg, Augsburg, Germany
| | - Hans W M Niessen
- Department of Pathology, Amsterdam University Medical Center (AUMC), Amsterdam, The Netherlands
| | - William Devine
- Department of Developmental Biology, University of Pittsburgh, 8111 Rangos Research Center, Pittsburgh, Pennsylvania, United States of America
| | - Cecilia W Lo
- Department of Developmental Biology, University of Pittsburgh, 8111 Rangos Research Center, Pittsburgh, Pennsylvania, United States of America
| | - Hannah M Mitchison
- Genetics and Genomic Medicine Programme, University College London, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Miriam Schmidts
- Center for Pediatrics and Adolescent Medicine, University Hospital Freiburg, Freiburg University Faculty of Medicine, Freiburg, Germany
- Human Genetics Department, Radboud University Medical Center Nijmegen and Radboud Institute for Molecular Life Sciences (RIMLS), Nijmegen, The Netherlands
- CIBSS-Center for Integrative Biological Signaling Studies, University of Freiburg, Freiburg, Germany
| | - Gregory J Pazour
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Biotech II, Worcester, Massachusetts, United States of America
| |
Collapse
|
24
|
Francis R, San Agustin JT, Szabo Rogers HL, Cui C, Jonassen JA, Eguether T, Follit JA, Lo CW, Pazour GJ. Autonomous and non-cell autonomous etiology of ciliopathy associated structural birth defects. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.07.544132. [PMID: 37333142 PMCID: PMC10274801 DOI: 10.1101/2023.06.07.544132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Ciliopathies are associated with wide spectrum of structural birth defects (SBD), indicating important roles for cilia in development. Here we provide novel insights into the temporospatial requirement for cilia in SBDs arising from deficiency in Ift140 , an intraflagellar transport protein regulating ciliogenesis. Ift140 deficient mice exhibit cilia defects accompanied by wide spectrum of SBDs including macrostomia (craniofacial defects), exencephaly, body wall defects, tracheoesophageal fistula, randomized heart looping, congenital heart defects (CHD), lung hypoplasia, renal anomalies, and polydactyly. Tamoxifen inducible CAG-Cre deletion of a floxed Ift140 allele between E5.5 to 9.5 revealed early requirement for Ift140 in left-right heart looping regulation, mid to late requirement for cardiac outflow septation and alignment, and late requirement for craniofacial development and body wall closure. Surprisingly, CHD was not observed with four Cre drivers targeting different lineages essential for heart development, but craniofacial defects and omphalocele were observed with Wnt1-Cre targeting neural crest and Tbx18-Cre targeting epicardial lineage and rostral sclerotome through which trunk neural crest cells migrate. These findings revealed cell autonomous role of cilia in cranial/trunk neural crest mediated craniofacial and body wall closure defects, while non-cell autonomous multi-lineage interactions underlie CHD pathogenesis, revealing unexpected developmental complexity for CHD associated with ciliopathy.
Collapse
|
25
|
Liu YX, Li WJ, Zhang RK, Sun SN, Fan ZC. Unraveling the intricate cargo-BBSome coupling mechanism at the ciliary tip. Proc Natl Acad Sci U S A 2023; 120:e2218819120. [PMID: 36943875 PMCID: PMC10068815 DOI: 10.1073/pnas.2218819120] [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: 11/03/2022] [Accepted: 02/13/2023] [Indexed: 03/23/2023] Open
Abstract
Certain ciliary transmembrane and membrane-tethered signaling proteins migrate from the ciliary tip to base via retrograde intraflagellar transport (IFT), essential for maintaining their ciliary dynamics to enable cells to sense and transduce extracellular stimuli inside the cell. During this process, the BBSome functions as an adaptor between retrograde IFT trains and these signaling protein cargoes. The Arf-like 13 (ARL13) small GTPase resembles ARL6/BBS3 in facilitating these signaling cargoes to couple with the BBSome at the ciliary tip prior to loading onto retrograde IFT trains for transporting towards the ciliary base, while the molecular basis for how this intricate coupling event happens remains elusive. Here, we report that Chlamydomonas ARL13 only in a GTP-bound form (ARL13GTP) anchors to the membrane for diffusing into cilia. Upon entering cilia, ARL13 undergoes GTPase cycle for shuttling between the ciliary membrane (ARL13GTP) and matrix (ARL13GDP). To achieve this goal, the ciliary membrane-anchored BBS3GTP binds the ciliary matrix-residing ARL13GDP to activate the latter as an ARL13 guanine nucleotide exchange factor. At the ciliary tip, ARL13GTP recruits the ciliary matrix-residing and post-remodeled BBSome as an ARL13 effector to anchor to the ciliary membrane. This makes the BBSome spatiotemporally become available for the ciliary membrane-tethered phospholipase D (PLD) to couple with. Afterward, ARL13GTP hydrolyzes GTP for releasing the PLD-laden BBSome to load onto retrograde IFT trains. According to this model, hedgehog signaling defects associated with ARL13b and BBS3 mutations in humans could be satisfactorily explained, providing us a mechanistic understanding behind BBSome-cargo coupling required for proper ciliary signaling.
Collapse
Affiliation(s)
- Yan-Xia Liu
- State Key Laboratory of Food Nutrition and Safety, Institute of Health Biotechnology, Tianjin University of Science and Technology, Tianjin300457, China
| | - Wen-Juan Li
- State Key Laboratory of Food Nutrition and Safety, Institute of Health Biotechnology, Tianjin University of Science and Technology, Tianjin300457, China
| | - Rui-Kai Zhang
- State Key Laboratory of Food Nutrition and Safety, Institute of Health Biotechnology, Tianjin University of Science and Technology, Tianjin300457, China
| | - Sheng-Nan Sun
- State Key Laboratory of Food Nutrition and Safety, Institute of Health Biotechnology, Tianjin University of Science and Technology, Tianjin300457, China
| | - Zhen-Chuan Fan
- State Key Laboratory of Food Nutrition and Safety, Institute of Health Biotechnology, Tianjin University of Science and Technology, Tianjin300457, China
| |
Collapse
|
26
|
Holroyd NA, Walsh C, Gourmet L, Walker-Samuel S. Quantitative Image Processing for Three-Dimensional Episcopic Images of Biological Structures: Current State and Future Directions. Biomedicines 2023; 11:909. [PMID: 36979887 PMCID: PMC10045950 DOI: 10.3390/biomedicines11030909] [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: 01/24/2023] [Revised: 03/03/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023] Open
Abstract
Episcopic imaging using techniques such as High Resolution Episcopic Microscopy (HREM) and its variants, allows biological samples to be visualized in three dimensions over a large field of view. Quantitative analysis of episcopic image data is undertaken using a range of methods. In this systematic review, we look at trends in quantitative analysis of episcopic images and discuss avenues for further research. Papers published between 2011 and 2022 were analyzed for details about quantitative analysis approaches, methods of image annotation and choice of image processing software. It is shown that quantitative processing is becoming more common in episcopic microscopy and that manual annotation is the predominant method of image analysis. Our meta-analysis highlights where tools and methods require further development in this field, and we discuss what this means for the future of quantitative episcopic imaging, as well as how annotation and quantification may be automated and standardized across the field.
Collapse
Affiliation(s)
| | - Claire Walsh
- Centre for Computational Medicine, University College London, London WC1E 6DD, UK
- Department of Mechanical Engineering, University College London, London WC1E 7JE, UK
| | - Lucie Gourmet
- Centre for Computational Medicine, University College London, London WC1E 6DD, UK
| | - Simon Walker-Samuel
- Centre for Computational Medicine, University College London, London WC1E 6DD, UK
| |
Collapse
|
27
|
Liu YX, Zhang RK, Fan ZC. RABL4/IFT27 in a nucleotide-independent manner promotes phospholipase D ciliary retrieval via facilitating BBSome reassembly at the ciliary tip. J Cell Physiol 2023; 238:549-565. [PMID: 36852649 DOI: 10.1002/jcp.30945] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 12/24/2022] [Accepted: 01/02/2023] [Indexed: 03/01/2023]
Abstract
Certain ciliary transmembrane and membrane-associated signaling proteins export from cilia as intraflagellar transport (IFT) cargoes in a BBSome-dependent manner. Upon reaching the ciliary tip via anterograde IFT, the BBSome disassembles before being reassembled to form an intact entity for cargo phospholipase D (PLD) coupling. During this BBSome remodeling process, Chlamydomonas Rab-like 4 GTPase IFT27, by binding its partner IFT25 to form the heterodimeric IFT25/27, is indispensable for BBSome reassembly. Here, we show that IFT27 binds IFT25 in an IFT27 nucleotide-independent manner. IFT25/27 and the IFT subcomplexes IFT-A and -B are irrelevant for maintaining the stability of one another. GTP-loading onto IFT27 enhances the IFT25/27 affinity for binding to the IFT-B subcomplex core IFT-B1 entity in cytoplasm, while GDP-bound IFT27 does not prevent IFT25/27 from entering and cycling through cilia by integrating into IFT-B1. Upon at the ciliary tip, IFT25/27 cycles on and off IFT-B1 and this process is irrelevant with the nucleotide state of IFT27. During BBSome remodeling at the ciliary tip, IFT25/27 promotes BBSome reassembly independent of IFT27 nucleotide state, making postremodeled BBSomes available for PLD to interact with. Thus, IFT25/27 facilitates BBSome-dependent PLD export from cilia via controlling availability of intact BBSomes at the ciliary tip, while IFT27 nucleotide state does not participate in this regulatory event.
Collapse
Affiliation(s)
- Yan-Xia Liu
- State Key Laboratory of Food Nutrition and Safety, Institute of Health Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Rui-Kai Zhang
- State Key Laboratory of Food Nutrition and Safety, Institute of Health Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Zhen-Chuan Fan
- State Key Laboratory of Food Nutrition and Safety, Institute of Health Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| |
Collapse
|
28
|
Bakey Z, Cabrera OA, Hoefele J, Antony D, Wu K, Stuck MW, Micha D, Eguether T, Smith AO, van der Wel NN, Wagner M, Strittmatter L, Beales PL, Jonassen JA, Thiffault I, Cadieux-Dion M, Boyes L, Sharif S, Tüysüz B, Dunstheimer D, Niessen HW, Devine W, Lo CW, Mitchison HM, Schmidts M, Pazour GJ. IFT74 variants cause skeletal ciliopathy and motile cilia defects in mice and humans. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.02.23.23286106. [PMID: 36865301 PMCID: PMC9980244 DOI: 10.1101/2023.02.23.23286106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2023]
Abstract
Motile and non-motile cilia are critical to mammalian development and health. Assembly of these organelles depends on proteins synthesized in the cell body and transported into the cilium by intraflagellar transport (IFT). A series of human and mouse IFT74 variants were studied to understand the function of this IFT subunit. Humans missing exon 2, which codes for the first 40 residues, presented an unusual combination of ciliary chondrodysplasia and mucociliary clearance disorders while individuals carrying biallelic splice site variants developed a lethal skeletal chondrodysplasia. In mice, variants thought to remove all Ift74 function, completely block ciliary assembly and result in midgestational lethality. A mouse allele that removes the first 40 amino acids, analogous to the human exon 2 deletion, results in a motile cilia phenotype with mild skeletal abnormalities. In vitro studies suggest that the first 40 amino acids of IFT74 are dispensable for binding of other IFT subunits but are important for tubulin binding. Higher demands on tubulin transport in motile cilia compared to primary cilia could account for the motile cilia phenotype observed in human and mice.
Collapse
|
29
|
Petriman NA, Loureiro‐López M, Taschner M, Zacharia NK, Georgieva MM, Boegholm N, Wang J, Mourão A, Russell RB, Andersen JS, Lorentzen E. Biochemically validated structural model of the 15-subunit intraflagellar transport complex IFT-B. EMBO J 2022; 41:e112440. [PMID: 36354106 PMCID: PMC9753473 DOI: 10.15252/embj.2022112440] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/17/2022] [Accepted: 10/20/2022] [Indexed: 11/11/2022] Open
Abstract
Cilia are ubiquitous eukaryotic organelles impotant for cellular motility, signaling, and sensory reception. Cilium formation requires intraflagellar transport of structural and signaling components and involves 22 different proteins organized into intraflagellar transport (IFT) complexes IFT-A and IFT-B that are transported by molecular motors. The IFT-B complex constitutes the backbone of polymeric IFT trains carrying cargo between the cilium and the cell body. Currently, high-resolution structures are only available for smaller IFT-B subcomplexes leaving > 50% structurally uncharacterized. Here, we used Alphafold to structurally model the 15-subunit IFT-B complex. The model was validated using cross-linking/mass-spectrometry data on reconstituted IFT-B complexes, X-ray scattering in solution, diffraction from crystals as well as site-directed mutagenesis and protein-binding assays. The IFT-B structure reveals an elongated and highly flexible complex consistent with cryo-electron tomographic reconstructions of IFT trains. The IFT-B complex organizes into IFT-B1 and IFT-B2 parts with binding sites for ciliary cargo and the inactive IFT dynein motor, respectively. Interestingly, our results are consistent with two different binding sites for IFT81/74 on IFT88/70/52/46 suggesting the possibility of different structural architectures for the IFT-B1 complex. Our data present a structural framework to understand IFT-B complex assembly, function, and ciliopathy variants.
Collapse
Affiliation(s)
- Narcis A Petriman
- Department of Molecular Biology and GeneticsAarhus UniversityAarhus CDenmark
| | - Marta Loureiro‐López
- Department for Biochemistry and Molecular BiologyUniversity of Southern DenmarkOdense MDenmark
| | - Michael Taschner
- Department of Fundamental MicrobiologyUniversity of LausanneLausanneSwitzerland
| | - Nevin K Zacharia
- Department of Molecular Biology and GeneticsAarhus UniversityAarhus CDenmark
| | | | - Niels Boegholm
- Department of Molecular Biology and GeneticsAarhus UniversityAarhus CDenmark
| | - Jiaolong Wang
- Department of Molecular Biology and GeneticsAarhus UniversityAarhus CDenmark
| | - André Mourão
- Institute of Structural BiologyHelmholtz Zentrum MünchenNeuherbergGermany
| | | | - Jens S Andersen
- Department for Biochemistry and Molecular BiologyUniversity of Southern DenmarkOdense MDenmark
| | - Esben Lorentzen
- Department of Molecular Biology and GeneticsAarhus UniversityAarhus CDenmark
| |
Collapse
|
30
|
Yan X, Shen Y. Rab-like small GTPases in the regulation of ciliary Bardet-Biedl syndrome (BBS) complex transport. FEBS J 2022; 289:7359-7367. [PMID: 34655445 DOI: 10.1111/febs.16232] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/13/2021] [Accepted: 10/15/2021] [Indexed: 01/13/2023]
Abstract
Primary cilia, microtubule-based hair-like structures protruding from most cells, contain membranes enriched in signaling molecules and function as sensory and regulatory organelles critical for development and tissue homeostasis. Intraflagellar transport (IFT), cilia-specific bidirectional transport, is required for the assembly, maintenance, and function of cilia. BBSome, the coat complex, acts as the adaptor between the IFT complex and membrane proteins and is therefore essential for establishing the specific compartmentalization of signaling molecules in the cilia. Recent findings have revealed that three ciliary Rab-like small GTPases, IFT27, IFT22, and Rabl2, play critical regulatory roles in ciliary BBSome transport. In this review, we provide an overview of these three Rab-like small GTPases and their relationship with BBSome.
Collapse
Affiliation(s)
- Xiumin Yan
- Ministry of Education-Shanghai Key Laboratory of Children's Environmental Health, Institute of Early Life Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Yidong Shen
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
31
|
Amack JD. Structures and functions of cilia during vertebrate embryo development. Mol Reprod Dev 2022; 89:579-596. [PMID: 36367893 PMCID: PMC9805515 DOI: 10.1002/mrd.23650] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/05/2022] [Accepted: 10/28/2022] [Indexed: 11/13/2022]
Abstract
Cilia are hair-like structures that project from the surface of cells. In vertebrates, most cells have an immotile primary cilium that mediates cell signaling, and some specialized cells assemble one or multiple cilia that are motile and beat synchronously to move fluids in one direction. Gene mutations that alter cilia structure or function cause a broad spectrum of disorders termed ciliopathies that impact virtually every system in the body. A wide range of birth defects associated with ciliopathies underscores critical functions for cilia during embryonic development. In many cases, the mechanisms underlying cilia functions during development and disease remain poorly understood. This review describes different types of cilia in vertebrate embryos and discusses recent research results from diverse model systems that provide novel insights into how cilia form and function during embryo development. The work discussed here not only expands our understanding of in vivo cilia biology, but also opens new questions about cilia and their roles in establishing healthy embryos.
Collapse
Affiliation(s)
- Jeffrey D. Amack
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, New York, USA,,BioInspired Syracuse: Institute for Material and Living Systems, Syracuse, New York, USA
| |
Collapse
|
32
|
Chen Y, Lu C, Shang X, Wu K, Chen K. Primary cilia: The central role in the electromagnetic field induced bone healing. Front Pharmacol 2022; 13:1062119. [DOI: 10.3389/fphar.2022.1062119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 11/07/2022] [Indexed: 12/03/2022] Open
Abstract
Primary cilia have emerged as the cellular “antenna” that can receive and transduce extracellular chemical/physical signals, thus playing an important role in regulating cellular activities. Although the electromagnetic field (EMF) is an effective treatment for bone fractures since 1978, however, the detailed mechanisms leading to such positive effects are still unclear. Primary cilia may play a central role in receiving EMF signals, translating physical signals into biochemical information, and initiating various signalingsignaling pathways to transduce signals into the nucleus. In this review, we elucidated the process of bone healing, the structure, and function of primary cilia, as well as the application and mechanism of EMF in treating fracture healing. To comprehensively understand the process of bone healing, we used bioinformatics to analyze the molecular change and associated the results with other studies. Moreover, this review summarizedsummarized some limitations in EMFs-related research and provides an outlook for ongoing studies. In conclusion, this review illustrated the primary cilia and related molecular mechanisms in the EMF-induced bone healing process, and it may shed light on future research.
Collapse
|
33
|
Zhou Z, Katoh Y, Nakayama K. CEP19-RABL2-IFT-B axis controls BBSome-mediated ciliary GPCR export. Mol Biol Cell 2022; 33:ar126. [PMID: 36074075 PMCID: PMC9634966 DOI: 10.1091/mbc.e22-05-0161] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The intraflagellar transport (IFT) machinery mediates the import and export of ciliary proteins across the ciliary gate, as well as bidirectional protein trafficking within cilia. In addition to ciliary anterograde protein trafficking, the IFT-B complex participates in the export of membrane proteins together with the BBSome, which consists of eight subunits encoded by the causative genes of Bardet-Biedl syndrome (BBS). The IFT25-IFT27/BBS19 dimer in the IFT-B complex constitutes its interface with the BBSome. We show here that IFT25-IFT27 and the RABL2 GTPase bind the IFT74/BBS22-IFT81 dimer of the IFT-B complex in a mutually exclusive manner. Cells expressing GTP-locked RABL2 [RABL2(Q80L)], but not wild-type RABL2, phenocopied IFT27-knockout cells, that is, they demonstrated BBS-associated ciliary defects, including accumulation of LZTFL1/BBS17 and the BBSome within cilia and the suppression of export of the ciliary GPCRs GPR161 and Smoothened. RABL2(Q80L) enters cilia in a manner dependent on the basal body protein CEP19, but its entry into cilia is not necessary for causing BBS-associated ciliary defects. These observations suggest that GTP-bound RABL2 is likely to be required for recruitment of the IFT-B complex to the ciliary base, where it is replaced with IFT25-IFT27.
Collapse
Affiliation(s)
| | - Yohei Katoh
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan,*Address correspondence to: Kazuhisa Nakayama (); Yohei Katoh ()
| | - Kazuhisa Nakayama
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan,*Address correspondence to: Kazuhisa Nakayama (); Yohei Katoh ()
| |
Collapse
|
34
|
Finetti F, Onnis A, Baldari CT. IFT20: An Eclectic Regulator of Cellular Processes beyond Intraflagellar Transport. Int J Mol Sci 2022; 23:ijms232012147. [PMID: 36292997 PMCID: PMC9603483 DOI: 10.3390/ijms232012147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 10/06/2022] [Accepted: 10/10/2022] [Indexed: 11/16/2022] Open
Abstract
Initially discovered as the smallest component of the intraflagellar transport (IFT) system, the IFT20 protein has been found to be implicated in several unconventional mechanisms beyond its essential role in the assembly and maintenance of the primary cilium. IFT20 is now considered a key player not only in ciliogenesis but also in vesicular trafficking of membrane receptors and signaling proteins. Moreover, its ability to associate with a wide array of interacting partners in a cell-type specific manner has expanded the function of IFT20 to the regulation of intracellular degradative and secretory pathways. In this review, we will present an overview of the multifaceted role of IFT20 in both ciliated and non-ciliated cells.
Collapse
|
35
|
Rah G, Cha H, Kim J, Song J, Kim H, Oh YK, Ahn C, Kang M, Kim J, Yoo KH, Kim MJ, Ko HW, Ko JY, Park JH. KLC3 Regulates Ciliary Trafficking and Cyst Progression in CILK1 Deficiency-Related Polycystic Kidney Disease. J Am Soc Nephrol 2022; 33:1726-1741. [PMID: 35961787 PMCID: PMC9529174 DOI: 10.1681/asn.2021111455] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 05/23/2022] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Ciliogenesis-associated kinase 1 (CILK1) is a ciliary gene that localizes in primary cilia and regulates ciliary transport. Mutations in CILK1 cause various ciliopathies. However, the pathogenesis of CILK1-deficient kidney disease is unknown. METHODS To examine whether CILK1 deficiency causes PKD accompanied by abnormal cilia, we generated mice with deletion of Cilk1 in cells of the renal collecting duct. A yeast two-hybrid system and coimmunoprecipitation (co-IP) were used to identify a novel regulator, kinesin light chain-3 (KLC3), of ciliary trafficking and cyst progression in the Cilk1-deficient model. Immunocytochemistry and co-IP were used to examine the effect of KLC3 on ciliary trafficking of the IFT-B complex and EGFR. We evaluated the effects of these genes on ciliary trafficking and cyst progression by modulating CILK1 and KLC3 expression levels. RESULTS CILK1 deficiency leads to PKD accompanied by abnormal ciliary trafficking. KLC3 interacts with CILK1 at cilia bases and is increased in cyst-lining cells of CILK1-deficient mice. KLC3 overexpression promotes ciliary recruitment of IFT-B and EGFR in the CILK1 deficiency condition, which contributes to the ciliary defect in cystogenesis. Reduction in KLC3 rescued the ciliary defects and inhibited cyst progression caused by CILK1 deficiency. CONCLUSIONS Our findings suggest that CILK1 deficiency in renal collecting ducts leads to PKD and promotes ciliary trafficking via increased KLC3.
Collapse
Affiliation(s)
- Gyuyeong Rah
- Department of Biological Science, Sookmyung Women’s University, Seoul, Korea
| | - Hwayeon Cha
- Department of Biological Science, Sookmyung Women’s University, Seoul, Korea
| | - Joohee Kim
- Department of Biological Science, Sookmyung Women’s University, Seoul, Korea
| | - Jieun Song
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Hyunho Kim
- Center for Medical Innovation, Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Yun Kyu Oh
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Korea
| | - Curie Ahn
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Minyong Kang
- Department of Urology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jongmin Kim
- Department of Biological Science, Sookmyung Women’s University, Seoul, Korea
| | - Kyung Hyun Yoo
- Department of Biological Science, Sookmyung Women’s University, Seoul, Korea
| | - Min Jung Kim
- Department of Biological Science, Sookmyung Women’s University, Seoul, Korea
| | - Hyuk Wan Ko
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Je Yeong Ko
- Department of Biological Science, Sookmyung Women’s University, Seoul, Korea
| | - Jong Hoon Park
- Department of Biological Science, Sookmyung Women’s University, Seoul, Korea
| |
Collapse
|
36
|
Schembs L, Willems A, Hasenpusch-Theil K, Cooper JD, Whiting K, Burr K, Bøstrand SMK, Selvaraj BT, Chandran S, Theil T. The ciliary gene INPP5E confers dorsal telencephalic identity to human cortical organoids by negatively regulating Sonic hedgehog signaling. Cell Rep 2022; 39:110811. [PMID: 35584663 PMCID: PMC9620745 DOI: 10.1016/j.celrep.2022.110811] [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: 06/09/2021] [Revised: 02/07/2022] [Accepted: 04/20/2022] [Indexed: 12/02/2022] Open
Abstract
Defects in primary cilia, cellular antennas that control multiple intracellular signaling pathways, underlie several neurodevelopmental disorders, but it remains unknown how cilia control essential steps in human brain formation. Here, we show that cilia are present on the apical surface of radial glial cells in human fetal forebrain. Interfering with cilia signaling in human organoids by mutating the INPP5E gene leads to the formation of ventral telencephalic cell types instead of cortical progenitors and neurons. INPP5E mutant organoids also show increased Sonic hedgehog (SHH) signaling, and cyclopamine treatment partially rescues this ventralization. In addition, ciliary expression of SMO, GLI2, GPR161, and several intraflagellar transport (IFT) proteins is increased. Overall, these findings establish the importance of primary cilia for dorsal and ventral patterning in human corticogenesis, indicate a tissue-specific role of INPP5E as a negative regulator of SHH signaling, and have implications for the emerging roles of cilia in the pathogenesis of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Leah Schembs
- Centre for Discovery Brain Sciences, Hugh Robson Building, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Ariane Willems
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK; UK Dementia Research Institute at University of Edinburgh, University of Edinburgh, Edinburgh EH16 4SB, UK; Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
| | - Kerstin Hasenpusch-Theil
- Centre for Discovery Brain Sciences, Hugh Robson Building, University of Edinburgh, Edinburgh EH8 9XD, UK; Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK
| | - James D Cooper
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK; UK Dementia Research Institute at University of Edinburgh, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Katie Whiting
- Centre for Discovery Brain Sciences, Hugh Robson Building, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Karen Burr
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK; UK Dementia Research Institute at University of Edinburgh, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Sunniva M K Bøstrand
- Centre for Discovery Brain Sciences, Hugh Robson Building, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Bhuvaneish T Selvaraj
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK; UK Dementia Research Institute at University of Edinburgh, University of Edinburgh, Edinburgh EH16 4SB, UK; Anne Rowling Regenerative Neurology Clinic, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Siddharthan Chandran
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK; UK Dementia Research Institute at University of Edinburgh, University of Edinburgh, Edinburgh EH16 4SB, UK; Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK; Anne Rowling Regenerative Neurology Clinic, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Thomas Theil
- Centre for Discovery Brain Sciences, Hugh Robson Building, University of Edinburgh, Edinburgh EH8 9XD, UK; Simons Initiative for the Developing Brain, University of Edinburgh, Hugh Robson Building, Edinburgh EH8 9XD, UK.
| |
Collapse
|
37
|
Chandra B, Tung ML, Hsu Y, Scheetz T, Sheffield VC. Retinal ciliopathies through the lens of Bardet-Biedl Syndrome: Past, present and future. Prog Retin Eye Res 2021; 89:101035. [PMID: 34929400 DOI: 10.1016/j.preteyeres.2021.101035] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 12/10/2021] [Accepted: 12/13/2021] [Indexed: 12/15/2022]
Abstract
The primary cilium is a highly specialized and evolutionary conserved organelle in eukaryotes that plays a significant role in cell signaling and trafficking. Over the past few decades tremendous progress has been made in understanding the physiology of cilia and the underlying pathomechanisms of various ciliopathies. Syndromic ciliopathies consist of a group of disorders caused by ciliary dysfunction or abnormal ciliogenesis. These disorders have multiorgan involvement in addition to retinal degeneration underscoring the ubiquitous distribution of primary cilia in different cell types. Genotype-phenotype correlation is often challenging due to the allelic heterogeneity and pleiotropy of these disorders. In this review, we discuss the clinical and genetic features of syndromic ciliopathies with a focus on Bardet-Biedl syndrome (BBS) as a representative disorder. We discuss the structure and function of primary cilia and their role in retinal photoreceptors. We describe the progress made thus far in understanding the functional and genetic characterization including expression quantitative trait locus (eQTL) analysis of BBS genes. In the future directions section, we discuss the emerging technologies, such as gene therapy, as well as anticipated challenges and their implications in therapeutic development for ciliopathies.
Collapse
Affiliation(s)
- Bharatendu Chandra
- Stead Family Department of Pediatrics, Division of Medical Genetics and Genomics, University of Iowa Carver College of Medicine, Iowa City, IA, USA; Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Moon Ley Tung
- Stead Family Department of Pediatrics, Division of Medical Genetics and Genomics, University of Iowa Carver College of Medicine, Iowa City, IA, USA; Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Ying Hsu
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, Iowa City, IA, USA
| | - Todd Scheetz
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, Iowa City, IA, USA
| | - Val C Sheffield
- Stead Family Department of Pediatrics, Division of Medical Genetics and Genomics, University of Iowa Carver College of Medicine, Iowa City, IA, USA; Department of Ophthalmology and Visual Sciences, Carver College of Medicine, Iowa City, IA, USA.
| |
Collapse
|
38
|
Zhou Z, Qiu H, Castro-Araya RF, Takei R, Nakayama K, Katoh Y. Impaired cooperation between IFT74/BBS22-IFT81 and IFT25-IFT27/BBS19 causes Bardet-Biedl syndrome. Hum Mol Genet 2021; 31:1681-1693. [PMID: 34888642 DOI: 10.1093/hmg/ddab354] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/19/2021] [Accepted: 12/06/2021] [Indexed: 02/03/2023] Open
Abstract
The IFT-B complex mediates ciliary anterograde protein trafficking and membrane protein export together with the BBSome. Bardet-Biedl syndrome (BBS) is caused by mutations in not only all BBSome subunits, but also in some IFT-B subunits, including IFT74/BBS22 and IFT27/BBS19, which form heterodimers with IFT81 and IFT25, respectively. We found that the IFT25-IFT27 dimer bind the C-terminal region of the IFT74-IFT81 dimer, and that the IFT25-IFT27-binding region encompasses the region deleted in the BBS variants of IFT74. In addition, we found that the missense BBS variants of IFT27 are impaired in IFT74-IFT81 binding, and are unable to rescue the BBS-like phenotypes of IFT27-knockout cells. Furthermore, the BBS variants of IFT74 rescued the ciliogenesis defect of IFT74-knockout cells, but the rescued cells demonstrated BBS-like abnormal phenotypes. Taken together, we conclude that the impaired interaction between IFT74-IFT81 and IFT25-IFT27 causes the BBS-associated ciliary defects.
Collapse
Affiliation(s)
- Zhuang Zhou
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Hantian Qiu
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Roiner-Francisco Castro-Araya
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Ryota Takei
- 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
| | - Yohei Katoh
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| |
Collapse
|
39
|
Gupta S, Ozimek-Kulik JE, Phillips JK. Nephronophthisis-Pathobiology and Molecular Pathogenesis of a Rare Kidney Genetic Disease. Genes (Basel) 2021; 12:genes12111762. [PMID: 34828368 PMCID: PMC8623546 DOI: 10.3390/genes12111762] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/26/2021] [Accepted: 10/27/2021] [Indexed: 12/17/2022] Open
Abstract
The exponential rise in our understanding of the aetiology and pathophysiology of genetic cystic kidney diseases can be attributed to the identification of cystogenic genes over the last three decades. The foundation of this was laid by positional cloning strategies which gradually shifted towards next-generation sequencing (NGS) based screenings. This shift has enabled the discovery of novel cystogenic genes at an accelerated pace unlike ever before and, most notably, the past decade has seen the largest increase in identification of the genes which cause nephronophthisis (NPHP). NPHP is a monogenic autosomal recessive cystic kidney disease caused by mutations in a diverse clade of over 26 identified genes and is the most common genetic cause of renal failure in children. NPHP gene types present with some common pathophysiological features alongside a diverse range of extra-renal phenotypes associated with specific syndromic presentations. This review provides a timely update on our knowledge of this disease, including epidemiology, pathophysiology, anatomical and molecular features. We delve into the diversity of the NPHP causing genes and discuss known molecular mechanisms and biochemical pathways that may have possible points of intersection with polycystic kidney disease (the most studied renal cystic pathology). We delineate the pathologies arising from extra-renal complications and co-morbidities and their impact on quality of life. Finally, we discuss the current diagnostic and therapeutic modalities available for disease management, outlining possible avenues of research to improve the prognosis for NPHP patients.
Collapse
Affiliation(s)
- Shabarni Gupta
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia; (J.E.O.-K.); (J.K.P.)
- Correspondence:
| | - Justyna E. Ozimek-Kulik
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia; (J.E.O.-K.); (J.K.P.)
- School of Women’s and Children’s Health, University of New South Wales, Sydney, NSW 2031, Australia
- Department of Paediatric Nephrology, Sydney Children’s Hospital Network, Children’s Hospital at Westmead, Sydney, NSW 2145, Australia
| | - Jacqueline Kathleen Phillips
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW 2109, Australia; (J.E.O.-K.); (J.K.P.)
| |
Collapse
|
40
|
Quidwai T, Wang J, Hall EA, Petriman NA, Leng W, Kiesel P, Wells JN, Murphy LC, Keighren MA, Marsh JA, Lorentzen E, Pigino G, Mill P. A WDR35-dependent coat protein complex transports ciliary membrane cargo vesicles to cilia. eLife 2021; 10:e69786. [PMID: 34734804 PMCID: PMC8754431 DOI: 10.7554/elife.69786] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 11/04/2021] [Indexed: 11/13/2022] Open
Abstract
Intraflagellar transport (IFT) is a highly conserved mechanism for motor-driven transport of cargo within cilia, but how this cargo is selectively transported to cilia is unclear. WDR35/IFT121 is a component of the IFT-A complex best known for its role in ciliary retrograde transport. In the absence of WDR35, small mutant cilia form but fail to enrich in diverse classes of ciliary membrane proteins. In Wdr35 mouse mutants, the non-core IFT-A components are degraded and core components accumulate at the ciliary base. We reveal deep sequence homology of WDR35 and other IFT-A subunits to α and ß' COPI coatomer subunits and demonstrate an accumulation of 'coat-less' vesicles that fail to fuse with Wdr35 mutant cilia. We determine that recombinant non-core IFT-As can bind directly to lipids and provide the first in situ evidence of a novel coat function for WDR35, likely with other IFT-A proteins, in delivering ciliary membrane cargo necessary for cilia elongation.
Collapse
Affiliation(s)
- Tooba Quidwai
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| | - Jiaolong Wang
- Department of Molecular Biology and Genetics, Aarhus UniversityAarhusDenmark
| | - Emma A Hall
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| | - Narcis A Petriman
- Department of Molecular Biology and Genetics, Aarhus UniversityAarhusDenmark
| | - Weihua Leng
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
| | - Petra Kiesel
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
| | - Jonathan N Wells
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| | - Laura C Murphy
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| | - Margaret A Keighren
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| | - Joseph A Marsh
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| | - Esben Lorentzen
- Department of Molecular Biology and Genetics, Aarhus UniversityAarhusDenmark
| | - Gaia Pigino
- Max Planck Institute of Molecular Cell Biology and GeneticsDresdenGermany
- Human TechnopoleMilanItaly
| | - Pleasantine Mill
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of EdinburghEdinburghUnited Kingdom
| |
Collapse
|
41
|
Abraham SP, Nita A, Krejci P, Bosakova M. Cilia kinases in skeletal development and homeostasis. Dev Dyn 2021; 251:577-608. [PMID: 34582081 DOI: 10.1002/dvdy.426] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/22/2021] [Accepted: 09/22/2021] [Indexed: 11/08/2022] Open
Abstract
Primary cilia are dynamic compartments that regulate multiple aspects of cellular signaling. The production, maintenance, and function of cilia involve more than 1000 genes in mammals, and their mutations disrupt the ciliary signaling which manifests in a plethora of pathological conditions-the ciliopathies. Skeletal ciliopathies are genetic disorders affecting the development and homeostasis of the skeleton, and encompass a broad spectrum of pathologies ranging from isolated polydactyly to lethal syndromic dysplasias. The recent advances in forward genetics allowed for the identification of novel regulators of skeletogenesis, and revealed a growing list of ciliary proteins that are critical for signaling pathways implicated in bone physiology. Among these, a group of protein kinases involved in cilia assembly, maintenance, signaling, and disassembly has emerged. In this review, we summarize the functions of cilia kinases in skeletal development and disease, and discuss the available and upcoming treatment options.
Collapse
Affiliation(s)
- Sara P Abraham
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Alexandru Nita
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Pavel Krejci
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic.,Institute of Animal Physiology and Genetics of the CAS, Brno, Czech Republic.,International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Michaela Bosakova
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic.,Institute of Animal Physiology and Genetics of the CAS, Brno, Czech Republic.,International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| |
Collapse
|
42
|
Cai E, Zhang J, Ge X. Control of the Hedgehog pathway by compartmentalized PKA in the primary cilium. SCIENCE CHINA-LIFE SCIENCES 2021; 65:500-514. [PMID: 34505970 DOI: 10.1007/s11427-021-1975-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 07/01/2021] [Indexed: 01/20/2023]
Abstract
The Hedgehog (Hh) signaling is one of the essential signaling pathways during embryogenesis and in adults. Hh signal transduction relies on primary cilium, a specialized cell surface organelle viewed as the hub of cell signaling. Protein kinase A (PKA) has been recognized as a potent negative regulator of the Hh pathway, raising the question of how such a ubiquitous kinase specifically regulates one signaling pathway. We reviewed recent genetic, molecular and biochemical studies that have advanced our mechanistic understanding of PKA's role in Hh signaling in vertebrates, focusing on the compartmentalized PKA at the centrosome and in the primary cilium. We outlined the recently developed genetic and optical tools that can be harvested to study PKA activities during the course of Hh signal transduction.
Collapse
Affiliation(s)
- Eva Cai
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, CA, 95340, USA
| | - Jingyi Zhang
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, CA, 95340, USA
| | - Xuecai Ge
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, CA, 95340, USA.
| |
Collapse
|
43
|
Pakvasa M, Tucker AB, Shen T, He TC, Reid RR. The Pleiotropic Intricacies of Hedgehog Signaling: From Craniofacial Patterning to Carcinogenesis. FACE (THOUSAND OAKS, CALIF.) 2021; 2:260-274. [PMID: 35812774 PMCID: PMC9268505 DOI: 10.1177/27325016211024326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Hedgehog signaling was discovered more than 40 years ago in experiments demonstrating that it is a fundamental mediator of limb development. Since that time, it has been shown to be important in development, homeostasis, and disease. The hedgehog pathway proceeds through a pathway highly conserved throughout animals beginning with the extracellular diffusion of hedgehog ligands, proceeding through an intracellular signaling cascade, and ending with the activation of specific target genes. A vast amount of research has been done elucidating hedgehog signaling mechanisms and regulation. This research has found a complex system of genetics and signaling that helps determine how organisms develop and function. This review provides an overview of what is known about hedgehog genetics and signaling, followed by an in-depth discussion of the role of hedgehog signaling in craniofacial development and carcinogenesis.
Collapse
Affiliation(s)
- Mikhail Pakvasa
- Pritzker School of Medicine, University of Chicago, Chicago, IL USA
- Molecular Oncology Lab, Department of Orthopedic Surgery & Rehabilitation Medicine,University of Chicago Medicine, Chicago, IL
| | - Andrew B. Tucker
- Pritzker School of Medicine, University of Chicago, Chicago, IL USA
- Molecular Oncology Lab, Department of Orthopedic Surgery & Rehabilitation Medicine,University of Chicago Medicine, Chicago, IL
| | - Timothy Shen
- Pritzker School of Medicine, University of Chicago, Chicago, IL USA
| | - Tong-Chuan He
- Molecular Oncology Lab, Department of Orthopedic Surgery & Rehabilitation Medicine,University of Chicago Medicine, Chicago, IL
| | - Russell R. Reid
- Molecular Oncology Lab, Department of Orthopedic Surgery & Rehabilitation Medicine,University of Chicago Medicine, Chicago, IL
- Section of Plastic and Reconstructive Surgery, University of Chicago Medicine, Chicago, IL
| |
Collapse
|
44
|
Chlamydomonas LZTFL1 mediates phototaxis via controlling BBSome recruitment to the basal body and its reassembly at the ciliary tip. Proc Natl Acad Sci U S A 2021; 118:2101590118. [PMID: 34446551 DOI: 10.1073/pnas.2101590118] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Many G protein-coupled receptors and other signaling proteins localize to the ciliary membrane for regulating diverse cellular processes. The BBSome composed of multiple Bardet-Biedl syndrome (BBS) proteins is an intraflagellar transport (IFT) cargo adaptor essential for sorting signaling proteins in and/or out of cilia via IFT. Leucine zipper transcription factor-like 1 (LZTFL1) protein mediates ciliary signaling by controlling BBSome ciliary content, reflecting how LZTFL1 mutations could cause BBS. However, the mechanistic mechanism underlying this process remains elusive thus far. Here, we show that LZTFL1 maintains BBSome ciliary dynamics by finely controlling BBSome recruitment to the basal body and its reassembly at the ciliary tip simultaneously in Chlamydomonas reinhardtii LZTFL1 directs BBSome recruitment to the basal body via promoting basal body targeting of Arf-like 6 GTPase BBS3, thus deciding the BBSome amount available for loading onto anterograde IFT trains for entering cilia. Meanwhile, LZTFL1 stabilizes the IFT25/27 component of the IFT-B1 subcomplex in the cell body so as to control its presence and amount at the basal body for entering cilia. Since IFT25/27 promotes BBSome reassembly at the ciliary tip for loading onto retrograde IFT trains, LZTFL1 thus also directs BBSome removal out of cilia. Therefore, LZTFL1 dysfunction deprives the BBSome of ciliary presence and generates Chlamydomonas cells defective in phototaxis. In summary, our data propose that LZTFL1 maintains BBSome dynamics in cilia by such a dual-mode system, providing insights into how LZTFL1 mediates ciliary signaling through maintaining BBSome ciliary dynamics and the pathogenetic mechanism of the BBS disorder as well.
Collapse
|
45
|
Lv B, Stuck MW, Desai PB, Cabrera OA, Pazour GJ. E3 ubiquitin ligase Wwp1 regulates ciliary dynamics of the Hedgehog receptor Smoothened. J Cell Biol 2021; 220:212435. [PMID: 34161574 PMCID: PMC8236919 DOI: 10.1083/jcb.202010177] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 05/01/2021] [Accepted: 06/01/2021] [Indexed: 12/26/2022] Open
Abstract
The Hedgehog pathway, critical to vertebrate development, is organized in primary cilia. Activation of signaling causes the Hedgehog receptor Ptch1 to exit cilia, allowing a second receptor, Smo, to accumulate in cilia and activate the downstream steps of the pathway. Mechanisms regulating the dynamics of these receptors are unknown, but the ubiquitination of Smo regulates its interaction with the intraflagellar transport system to control ciliary levels. A focused screen of ubiquitin-related genes identified nine required for maintaining low ciliary Smo at the basal state. These included cytoplasmic E3s (Arih2, Mgrn1, and Maea), a ciliary localized E3 (Wwp1), a ciliary localized E2 (Ube2l3), a deubiquitinase (Bap1), and three adaptors (Kctd5, Skp1a, and Skp2). The ciliary E3, Wwp1, binds Ptch1 and localizes to cilia at the basal state. Activation of signaling removes both Ptch1 and Wwp1 from cilia, thus providing an elegant mechanism for Ptch1 to regulate ciliary Smo levels.
Collapse
Affiliation(s)
- Bo Lv
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Michael W Stuck
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Paurav B Desai
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Oscar A Cabrera
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Gregory J Pazour
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA
| |
Collapse
|
46
|
Wang W, Jack BM, Wang HH, Kavanaugh MA, Maser RL, Tran PV. Intraflagellar Transport Proteins as Regulators of Primary Cilia Length. Front Cell Dev Biol 2021; 9:661350. [PMID: 34095126 PMCID: PMC8170031 DOI: 10.3389/fcell.2021.661350] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 04/06/2021] [Indexed: 12/21/2022] Open
Abstract
Primary cilia are small, antenna-like organelles that detect and transduce chemical and mechanical cues in the extracellular environment, regulating cell behavior and, in turn, tissue development and homeostasis. Primary cilia are assembled via intraflagellar transport (IFT), which traffics protein cargo bidirectionally along a microtubular axoneme. Ranging from 1 to 10 μm long, these organelles typically reach a characteristic length dependent on cell type, likely for optimum fulfillment of their specific roles. The importance of an optimal cilia length is underscored by the findings that perturbation of cilia length can be observed in a number of cilia-related diseases. Thus, elucidating mechanisms of cilia length regulation is important for understanding the pathobiology of ciliary diseases. Since cilia assembly/disassembly regulate cilia length, we review the roles of IFT in processes that affect cilia assembly/disassembly, including ciliary transport of structural and membrane proteins, ectocytosis, and tubulin posttranslational modification. Additionally, since the environment of a cell influences cilia length, we also review the various stimuli encountered by renal epithelia in healthy and diseased states that alter cilia length and IFT.
Collapse
Affiliation(s)
- Wei Wang
- Department of Anatomy and Cell Biology, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, United States
| | - Brittany M Jack
- Department of Anatomy and Cell Biology, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, United States
| | - Henry H Wang
- Department of Anatomy and Cell Biology, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, United States
| | - Matthew A Kavanaugh
- Department of Anatomy and Cell Biology, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, United States
| | - Robin L Maser
- Department of Clinical Laboratory Sciences, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, United States
| | - Pamela V Tran
- Department of Anatomy and Cell Biology, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
47
|
Stuck MW, Chong WM, Liao JC, Pazour GJ. Rab34 is necessary for early stages of intracellular ciliogenesis. Curr Biol 2021; 31:2887-2894.e4. [PMID: 33989524 DOI: 10.1016/j.cub.2021.04.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 03/17/2021] [Accepted: 04/09/2021] [Indexed: 01/04/2023]
Abstract
Primary cilia are sensory organelles present on most vertebrate cells and are critical for development and health. Ciliary dysfunction is associated with a large class of human pathologies collectively known as ciliopathies. These include cystic kidneys, blindness, obesity, skeletal malformations, and other organ anomalies. Using a proximity biotinylation with Ift27 as bait, we identified the small guanosine triphosphatase (GTPase) Rab34 as a ciliary protein. Rab34 localizes to the centrosomes near the mother centriole, the axoneme of developed cilia, and highly dynamic tubule structures in the centrosomal region. Rab34 is required for cilia formation in fibroblasts, where we find that Rab34 loss blocks ciliogenesis at an early step of ciliary vesicle formation. In inner medullary collecting duct (IMCD3) epithelial cells, the requirement is more complex, with Rab34 needed in cells grown at low density but becoming less important as cell density increases. Ciliogenesis can proceed by an internal pathway where cilia form in the cytoplasm before being displayed on the ciliary surface or cilia can assemble by an external pathway where the centriole docks on the plasma membrane before ciliary assembly. Fibroblasts are thought to use the internal pathway, although IMCD3 cells are thought to use the external pathway. However, we find that IMCD3 cells can use the internal assembly pathway and significant numbers of internally assembling cilia are observed in low-density cells. Together, our work indicates that Rab34 is required for internal assembly of cilia, but not for cilia built on the cell surface.
Collapse
Affiliation(s)
- Michael W Stuck
- Program in Molecular Medicine, University of Massachusetts Medical School, Biotech II, Suite 213, 373 Plantation Street, Worcester, MA 01605, USA
| | - Weng Man Chong
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei 10617, Taiwan
| | - Jung-Chi Liao
- Institute of Atomic and Molecular Sciences, Academia Sinica, Taipei 10617, Taiwan
| | - Gregory J Pazour
- Program in Molecular Medicine, University of Massachusetts Medical School, Biotech II, Suite 213, 373 Plantation Street, Worcester, MA 01605, USA.
| |
Collapse
|
48
|
Alfadhel M, Umair M, Almuzzaini B, Asiri A, Al Tuwaijri A, Alhamoudi K, Alyafee Y, Al-Owain M. Identification of the TTC26 Splice Variant in a Novel Complex Ciliopathy Syndrome with Biliary, Renal, Neurological, and Skeletal Manifestations. Mol Syndromol 2021; 12:133-140. [PMID: 34177428 DOI: 10.1159/000513829] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 12/16/2020] [Indexed: 12/27/2022] Open
Abstract
Ciliopathies constitute heterogeneous disorders that result from mutations in ciliary proteins. These proteins play an important role in the development of organs, physiology, and signaling pathways, and sequence variations in the genes encoding these proteins are associated with multisystem disorders. In this study, we describe a severe ciliopathy disorder that segregates in an autosomal recessive manner in a nonconsanguineous Saudi family. The proband exhibited features such as cholestasis, cystic dilatation of intrahepatic biliary ducts, diabetes insipidus, dysmorphic facial features, optic atrophy, pituitary hypoplasia, hydrocephalus, aqueductal stenosis, hyperextensible knee joints, bilateral knee dislocation, polydactyly, and syndactyly. Whole-genome sequencing and Sanger sequencing revealed a homozygous splice site variant (c.4-1G>C; NM_024926.3) in the tetratricopeptide repeat domain 26 (TTC26) gene located in chromosome 7q34, which cosegregated perfectly with the disease phenotype. qRT-PCR revealed a substantial decrease in the expression of the TTC26 gene as compared to the normal control, suggesting the pathogenicity of the identified variant. This report further strengthens the evidence that homozygous variants in the TTC26 gene cause severe ciliopathies with diverse phenotypes. We named this newly characterized condition as BRENS syndrome, which stands for biliary, renal, neurological, and skeletal features.
Collapse
Affiliation(s)
- Majid Alfadhel
- Medical Genomics Research Department, King Abdullah International Medical Research Center, Ministry of National Guard Health Affairs (MNGH), Riyadh, Saudi Arabia.,King Saud Bin Abdulaziz University for Health Sciences, MNGH, Riyadh, Saudi Arabia.,Division of Genetics, Department of Pediatrics, King Abdullah Specialized Children's Hospital, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | - Muhammad Umair
- Medical Genomics Research Department, King Abdullah International Medical Research Center, Ministry of National Guard Health Affairs (MNGH), Riyadh, Saudi Arabia.,King Saud Bin Abdulaziz University for Health Sciences, MNGH, Riyadh, Saudi Arabia
| | - Bader Almuzzaini
- Medical Genomics Research Department, King Abdullah International Medical Research Center, Ministry of National Guard Health Affairs (MNGH), Riyadh, Saudi Arabia.,King Saud Bin Abdulaziz University for Health Sciences, MNGH, Riyadh, Saudi Arabia
| | - Abdulaziz Asiri
- Medical Genomics Research Department, King Abdullah International Medical Research Center, Ministry of National Guard Health Affairs (MNGH), Riyadh, Saudi Arabia.,Faculty of Applied Medical Sciences, University of Bisha, Bisha, Saudi Arabia
| | - Abeer Al Tuwaijri
- Medical Genomics Research Department, King Abdullah International Medical Research Center, Ministry of National Guard Health Affairs (MNGH), Riyadh, Saudi Arabia.,King Saud Bin Abdulaziz University for Health Sciences, MNGH, Riyadh, Saudi Arabia
| | - Khaloud Alhamoudi
- Medical Genomics Research Department, King Abdullah International Medical Research Center, Ministry of National Guard Health Affairs (MNGH), Riyadh, Saudi Arabia.,King Saud Bin Abdulaziz University for Health Sciences, MNGH, Riyadh, Saudi Arabia
| | - Yusra Alyafee
- Medical Genomics Research Department, King Abdullah International Medical Research Center, Ministry of National Guard Health Affairs (MNGH), Riyadh, Saudi Arabia.,King Saud Bin Abdulaziz University for Health Sciences, MNGH, Riyadh, Saudi Arabia
| | - Mohammed Al-Owain
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| |
Collapse
|
49
|
Desai PB, Stuck MW, Lv B, Pazour GJ. Ubiquitin links smoothened to intraflagellar transport to regulate Hedgehog signaling. J Cell Biol 2021; 219:151798. [PMID: 32435793 PMCID: PMC7337509 DOI: 10.1083/jcb.201912104] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 03/17/2020] [Accepted: 04/19/2020] [Indexed: 12/17/2022] Open
Abstract
In the absence of Hedgehog ligand, patched-1 (Ptch1) localizes to cilia and prevents ciliary accumulation and activation of smoothened (Smo). Upon ligand binding, Ptch1 is removed from cilia, and Smo is derepressed and accumulates in cilia where it activates signaling. The mechanisms regulating these dynamic movements are not well understood, but defects in intraflagellar transport components, including Ift27 and the BBSome, cause Smo to accumulate in cilia without pathway activation. We find that in the absence of ligand-induced pathway activation, Smo is ubiquitinated and removed from cilia, and this process is dependent on Ift27 and BBSome components. Activation of Hedgehog signaling decreases Smo ubiquitination and ciliary removal, resulting in its accumulation. Blocking ubiquitination of Smo by an E1 ligase inhibitor or by mutating two lysine residues in intracellular loop three causes Smo to aberrantly accumulate in cilia without pathway activation. These data provide a mechanism to control Smo's ciliary level during Hedgehog signaling by regulating the ubiquitination state of the receptor.
Collapse
Affiliation(s)
- Paurav B Desai
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Michael W Stuck
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Bo Lv
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Gregory J Pazour
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA
| |
Collapse
|
50
|
Allard BA, Wang W, Pottorf TS, Mumtaz H, Jack BM, Wang HH, Silva LM, Jacobs DT, Wang J, Bumann EE, Tran PV. Thm2 interacts with paralog, Thm1, and sensitizes to Hedgehog signaling in postnatal skeletogenesis. Cell Mol Life Sci 2021; 78:3743-3762. [PMID: 33683377 DOI: 10.1007/s00018-021-03806-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 02/06/2021] [Accepted: 02/27/2021] [Indexed: 11/25/2022]
Abstract
Mutations in the intraflagellar transport-A (IFT-A) gene, THM1, have been identified in skeletal ciliopathies. Here, we report a genetic interaction between Thm1, and its paralog, Thm2, in postnatal skeletogenesis. THM2 localizes to primary cilia, but Thm2 deficiency does not affect ciliogenesis and Thm2-null mice survive into adulthood. However, by postnatal day 14, Thm2-/-; Thm1aln/+ mice exhibit small stature and small mandible. Radiography and microcomputed tomography reveal Thm2-/-; Thm1aln/+ tibia are less opaque and have reduced cortical and trabecular bone mineral density. In the mutant tibial growth plate, the proliferation zone is expanded and the hypertrophic zone is diminished, indicating impaired chondrocyte differentiation. Additionally, mutant growth plate chondrocytes show increased Hedgehog signaling. Yet deletion of one allele of Gli2, a major transcriptional activator of the Hedgehog pathway, exacerbated the Thm2-/-; Thm1aln/+ small phenotype, and further revealed that Thm2-/-; Gli2+/- mice have small stature. In Thm2-/-; Thm1aln/+ primary osteoblasts, a Hedgehog signaling defect was not detected, but bone nodule formation was markedly impaired. This indicates a signaling pathway is altered, and we propose that this pathway may potentially interact with Gli2. Together, our data reveal that loss of Thm2 with one allele of Thm1, Gli2, or both, present new IFT mouse models of osteochondrodysplasia. Our data also suggest Thm2 as a modifier of Hedgehog signaling in postnatal skeletal development.
Collapse
Affiliation(s)
- Bailey A Allard
- Department of Anatomy and Cell Biology, Jared Grantham Kidney Institute, University of Kansas Medical Center, 3901 Rainbow Blvd., MS #3038, Kansas City, KS, 66160, USA
| | - Wei Wang
- Department of Anatomy and Cell Biology, Jared Grantham Kidney Institute, University of Kansas Medical Center, 3901 Rainbow Blvd., MS #3038, Kansas City, KS, 66160, USA
| | - Tana S Pottorf
- Department of Anatomy and Cell Biology, Jared Grantham Kidney Institute, University of Kansas Medical Center, 3901 Rainbow Blvd., MS #3038, Kansas City, KS, 66160, USA
| | - Hammad Mumtaz
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Brittany M Jack
- Department of Anatomy and Cell Biology, Jared Grantham Kidney Institute, University of Kansas Medical Center, 3901 Rainbow Blvd., MS #3038, Kansas City, KS, 66160, USA
| | - Henry H Wang
- Department of Anatomy and Cell Biology, Jared Grantham Kidney Institute, University of Kansas Medical Center, 3901 Rainbow Blvd., MS #3038, Kansas City, KS, 66160, USA
| | - Luciane M Silva
- Department of Anatomy and Cell Biology, Jared Grantham Kidney Institute, University of Kansas Medical Center, 3901 Rainbow Blvd., MS #3038, Kansas City, KS, 66160, USA
| | - Damon T Jacobs
- Department of Anatomy and Cell Biology, Jared Grantham Kidney Institute, University of Kansas Medical Center, 3901 Rainbow Blvd., MS #3038, Kansas City, KS, 66160, USA
| | - Jinxi Wang
- Department of Orthopedic Surgery, Medical Center, University of Kansas, Kansas City, KS, 66160, USA
| | - Erin E Bumann
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Pamela V Tran
- Department of Anatomy and Cell Biology, Jared Grantham Kidney Institute, University of Kansas Medical Center, 3901 Rainbow Blvd., MS #3038, Kansas City, KS, 66160, USA.
| |
Collapse
|