1
|
Gruca-Stryjak K, Maciak K, Winiewska-Szajewska M, Jurkiewicz A, Gora M, Kacprzak MM, Drgas O, Bialek-Proscinska A, Sobczynska-Tomaszewska A, Pluta KD, Jamsheer A, Markwitz W, Poznanski J, Burzynska B. A novel NEK1 variant disturbs the interaction between the C-terminal fragment of NEK1 and the VDAC1 channel, causing lethal short-rib polydactyly syndrome. Bone 2025; 195:117471. [PMID: 40147672 DOI: 10.1016/j.bone.2025.117471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/07/2025] [Accepted: 03/20/2025] [Indexed: 03/29/2025]
Abstract
The NIMA-related kinase 1 (NEK1) gene belongs to the Never in Mitosis Gene A (NIMA) kinase family, a group whose members play essential roles in cell cycle regulation, specifically in cell division and ciliogenesis. Mutations in the NEK1 gene have been implicated in several diseases, including short-rib polydactyly syndrome (SRPS). SRPS is a bone growth disorder characterized by severe congenital anomalies. Here, we describe a family with a lethal form of SRPS due to a novel intronic variant in the NEK1 gene. Basing on whole-exome sequencing of fetuses with SRPS we identified a homozygous variant of the NEK1 gene at position c.3584-10T>A as the causative mutation. Bioinformatic methods and minigene splicing assays were then used to assess the harmfulness and functional impact of the variant. We found that the identified mutation leads to the synthesis of the NEK1 protein lacking 90C-terminal residues following the last coiled-coil region. Additional experiments were performed to identify proteins that interact with the C-terminal fragment of NEK1 absent in the mutated protein. We suggest that the interaction between the C-terminal fragment of NEK1 and the VDAC1 channel is essential for the VDAC1 phosphorylation, the absence of which is likely to affect ciliogenesis.
Collapse
Affiliation(s)
- Karolina Gruca-Stryjak
- Department of Perinatology, Poznan University of Medical Sciences, Poznan, Poland; Centers of Medical Genetics GENESIS, Poznan, Poland
| | - Karolina Maciak
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | | | - Aneta Jurkiewicz
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Monika Gora
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | | | | | | | | | - Krzysztof D Pluta
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Warsaw, Poland
| | - Aleksander Jamsheer
- Department of Medical Genetics, Poznan University of Medical Sciences, Poznan, Poland
| | - Wieslaw Markwitz
- Department of Perinatology, Poznan University of Medical Sciences, Poznan, Poland
| | - Jaroslaw Poznanski
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Beata Burzynska
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland.
| |
Collapse
|
2
|
Li M, Chen Z, Guo Z, Wang Y, Chai Y, Li W, Ou G. Alpha-tubulin tails regulate axoneme differentiation. Proc Natl Acad Sci U S A 2025; 122:e2414731122. [PMID: 40198703 DOI: 10.1073/pnas.2414731122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 02/19/2025] [Indexed: 04/10/2025] Open
Abstract
The tubulin tail is a key element for microtubule (MT) functionality, but the functional redundancy of tubulin genes complicates the genetic determination of their physiological functions. Here, we removed the C-terminal tail of five alpha- and four beta-tubulin genes in the C. elegans genome. Sensory cilia typically exhibit an axoneme that longitudinally differentiates into a middle segment with doublet MTs and a distal segment with singlet MTs. However, the excision of the alpha-tubulin tail, but not the beta-tubulin tail, resulted in the ectopic formation of doublet MTs in the distal segments. Molecular dynamics simulations suggest that the alpha-tubulin tail could prevent the B-tubule from docking on the surface of A-tubule. Using recombinant tubulins, we demonstrated that removing the alpha-tubulin tail efficiently promoted doublet MTs formation in vitro. These results reveal the vital and unique contributions of tubulin tails to the structural integrity and accuracy of axoneme MT organization.
Collapse
Affiliation(s)
- Ming Li
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
- Beijing Frontier Research Center for Biological Structure, Tsinghua University, Beijing 100084, China
- McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China
- State Key Laboratory of Membrane Biology, Tsinghua University, Beijing 100084, China
- School of Life Sciences and Ministry of Education Key Laboratory for Protein Science, Tsinghua University, Beijing 100084, China
| | - Zhe Chen
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
- Beijing Frontier Research Center for Biological Structure, Tsinghua University, Beijing 100084, China
- McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China
- State Key Laboratory of Membrane Biology, Tsinghua University, Beijing 100084, China
- School of Life Sciences and Ministry of Education Key Laboratory for Protein Science, Tsinghua University, Beijing 100084, China
| | - Zhengyang Guo
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
- Beijing Frontier Research Center for Biological Structure, Tsinghua University, Beijing 100084, China
- McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China
- State Key Laboratory of Membrane Biology, Tsinghua University, Beijing 100084, China
- School of Life Sciences and Ministry of Education Key Laboratory for Protein Science, Tsinghua University, Beijing 100084, China
| | - Yang Wang
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
- Beijing Frontier Research Center for Biological Structure, Tsinghua University, Beijing 100084, China
- McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China
- State Key Laboratory of Membrane Biology, Tsinghua University, Beijing 100084, China
- School of Life Sciences and Ministry of Education Key Laboratory for Protein Science, Tsinghua University, Beijing 100084, China
| | - Yongping Chai
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
- Beijing Frontier Research Center for Biological Structure, Tsinghua University, Beijing 100084, China
- McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China
- State Key Laboratory of Membrane Biology, Tsinghua University, Beijing 100084, China
- School of Life Sciences and Ministry of Education Key Laboratory for Protein Science, Tsinghua University, Beijing 100084, China
| | - Wei Li
- School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Guangshuo Ou
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China
- Beijing Frontier Research Center for Biological Structure, Tsinghua University, Beijing 100084, China
- McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China
- State Key Laboratory of Membrane Biology, Tsinghua University, Beijing 100084, China
- School of Life Sciences and Ministry of Education Key Laboratory for Protein Science, Tsinghua University, Beijing 100084, China
| |
Collapse
|
3
|
Li B, He YY, Yao WX, Jin DD, Luo HN, Li MY, Wu Y, Yang ZM. Primary cilia prevent activation of the cGAS-STING pathway during mouse decidualization. Commun Biol 2025; 8:607. [PMID: 40229503 DOI: 10.1038/s42003-025-08030-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 04/02/2025] [Indexed: 04/16/2025] Open
Abstract
Primary cilia are antenna-like organelles that sense extracellular signals and function as signaling hubs essential for vertebrate development and homeostasis. Decidualization is crucial for pregnancy establishment and maintenance in both humans and mice. While primary cilia are present in endometrial stromal cells, their role in pregnancy remains unknown. Here, we identify TMEM67, a key component of the ciliary transition zone, as a critical regulator of mouse decidualization. Loss of primary cilia triggers RhoA-MLC2-dependent actomyosin contraction, which transmits mechanical forces to the nuclear lamina, leading to micronuclei formation. Within these micronuclei, double-stranded DNA (dsDNA) can directly bind to cyclic GMP-AMP synthase (cGAS) in situ, initiating downstream signaling. This activation of the cGAS-STING pathway reduces CCL6 production and impairs decidualization. Furthermore, pharmacological inhibition of actin polymerization or RhoA-ROCK signaling alleviates mechanical forces surrounding stromal cells, restores ciliogenesis, maintains nuclear integrity, suppresses the cGAS-STING pathway activation, and ultimately rescues decidualization. Our findings reveal a previously unrecognized mechanism by which primary cilia regulate the actin cytoskeleton to maintain nuclear integrity and prevent DNA leakage. This safeguards against aberrant activation of the cGAS-STING pathway, which would otherwise trigger detrimental immune signaling and impair decidualization.
Collapse
Affiliation(s)
- Bo Li
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountain Region, College of Animal Science, Guizhou University, Guiyang, 550025, China
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Yu-Ying He
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Wen-Xu Yao
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountain Region, College of Animal Science, Guizhou University, Guiyang, 550025, China
| | - Dan-Dan Jin
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountain Region, College of Animal Science, Guizhou University, Guiyang, 550025, China
| | - Hui-Na Luo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Meng-Yuan Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Ying Wu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Zeng-Ming Yang
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountain Region, College of Animal Science, Guizhou University, Guiyang, 550025, China.
| |
Collapse
|
4
|
Roig J. NEK8, a NIMA-family protein kinase at the core of the ciliary INV complex. Cell Commun Signal 2025; 23:170. [PMID: 40189576 PMCID: PMC11974183 DOI: 10.1186/s12964-025-02143-w] [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: 12/10/2024] [Accepted: 03/07/2025] [Indexed: 04/09/2025] Open
Abstract
Here we describe the current knowledge about the ciliary kinase NEK8, highlighting what we know and what we don't know about its regulation, substrates and potential functions. We also review the literature about the pathological consequences of different NEK8 variants in patients of nephronophthisis, renal-hepatic-pancreatic dysplasia and autosomal dominant polycystic kidney disease, three different types of ciliopathies. NEK8 belongs to the NIMA family of serine/threonine protein kinases. Like its closest relative, NEK9, it contains a protein kinase and an RCC1 domain, but lacks the C-terminal region that is key for NEK9's regulation as a G2/M kinase. Importantly, NEK8 localizes to cilia as part of a multimeric protein complex that assembles in a fibrillar fashion at the proximal half of this signaling organelle, defining what is known as the INV compartment. NEK8 and its INV compartment partners inversin, ANKS6 and NPHP3 are necessary for left-right determination and the correct development of different organs such as the kidney, the heart and the liver. But the kinase substrates, regulatory mechanism and activating cues and thus the molecular basis of NEK8 important physiological roles remain elusive. We present the current findings regarding NEK8 and also highlight what we miss in order to progress towards the understanding of the kinase and the function of the INV complex at the cilia.
Collapse
Affiliation(s)
- Joan Roig
- Department of Cells and Tissues, Cell Cycle and Signaling Research Group, Molecular Biology Institute of Barcelona (IBMB-CSIC), Baldiri I Reixac 10-12, Barcelona, 08028, Spain.
| |
Collapse
|
5
|
Mitra A, Gioukakis E, Mul W, Peterman EJG. Delivery of intraflagellar transport proteins to the ciliary base and assembly into trains. SCIENCE ADVANCES 2025; 11:eadr1716. [PMID: 40184459 PMCID: PMC11970479 DOI: 10.1126/sciadv.adr1716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 03/03/2025] [Indexed: 04/06/2025]
Abstract
Anterograde intraflagellar transport (IFT) trains, composed of IFT-B, IFT-A, and BBSome subcomplexes, are responsible for transporting ciliary proteins into the cilium. How IFT subcomplexes reach the ciliary base and assemble into IFT trains is poorly understood. Here, we perform quantitative single-molecule imaging in Caenorhabditis elegans chemosensory cilia to uncover how IFT subcomplexes arrive at the base, organize in IFT trains, and enter the cilium. We find that BBSomes reach the base via diffusion where they either associate with assembling IFT trains or with the membrane surrounding the base. In contrast, IFT-B and IFT-A reach the base via directed transport most likely on vesicles that stop at distinct locations near the base. Individual subcomplexes detach from the vesicles into a diffusive pool and associate to assembling trains. Our results show that IFT-B is first incorporated into IFT trains, followed by IFT-A, and finally BBSomes, indicating that the assembly of IFT trains is a highly regulated, step-wise process.
Collapse
Affiliation(s)
| | - Evangelos Gioukakis
- Department of Physics and Astronomy and LaserLaB, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Wouter Mul
- Department of Physics and Astronomy and LaserLaB, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Erwin J. G. Peterman
- Department of Physics and Astronomy and LaserLaB, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
6
|
Nikonorova IA, desRanleau E, Jacobs KC, Saul J, Walsh JD, Wang J, Barr MM. Polycystins recruit cargo to distinct ciliary extracellular vesicle subtypes in C. elegans. Nat Commun 2025; 16:2899. [PMID: 40180912 PMCID: PMC11968823 DOI: 10.1038/s41467-025-57512-3] [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: 04/13/2024] [Accepted: 02/19/2025] [Indexed: 04/05/2025] Open
Abstract
Therapeutic use of tiny extracellular vesicles (EVs) requires understanding cargo loading mechanisms. Here, we use a modular proximity labeling approach to identify the cargo of ciliary EVs associated with the transient receptor potential channel polycystin-2 PKD-2 of C. elegans. Polycystins are conserved ciliary proteins and cargo of EVs; dysfunction causes polycystic kidney disease in humans and mating deficits in C. elegans. We discover that polycystins localize with specific cargo on ciliary EVs: polycystin-associated channel-like protein PACL-1, dorsal and ventral polycystin-associated membrane C-type lectins PAMLs, and conserved tumor necrosis factor receptor-associated factor (TRAF) TRF-1 and TRF-2. Loading of these components to EVs relies on polycystin-1 LOV-1. Our modular EV-TurboID approach can be applied in both cell- and tissue-specific manners to define the composition of distinct EV subtypes, addressing a major challenge of the EV field.
Collapse
Affiliation(s)
- Inna A Nikonorova
- Department of Genetics and Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ, USA.
| | - Elizabeth desRanleau
- Department of Genetics and Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Katherine C Jacobs
- Department of Genetics and Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Josh Saul
- Department of Genetics and Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Jonathon D Walsh
- Department of Genetics and Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Juan Wang
- Department of Genetics and Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Maureen M Barr
- Department of Genetics and Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ, USA.
| |
Collapse
|
7
|
Son J, Park J, Jeong JW, Lee SH, Kim JE. SIRT2 inhibition attenuates myofibroblast transition through autophagy-mediated ciliogenesis in renal epithelial cells. Int J Biochem Cell Biol 2025; 181:106754. [PMID: 39988243 DOI: 10.1016/j.biocel.2025.106754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 02/09/2025] [Accepted: 02/16/2025] [Indexed: 02/25/2025]
Abstract
Myofibroblast transition plays a crucial role in both fibrotic diseases and wound healing. Although SIRT2 regulates fibrosis, its mechanisms of action remain poorly understood. This study aimed to investigate the effects of SIRT2 inhibition on myofibroblast transition in human renal cells under quiescent conditions. HK-2 kidney proximal tubular epithelial cells were starved of serum, resulting in the formation of primary cilia. Transforming growth factor-β (TGF-β) stimulation reduced both the number of ciliated cells and ciliary length. The ciliary defects resulted from a failure in autophagy termination, leading to the accumulation of OFD1, a negative regulator of ciliogenesis, at centriolar satellites. This phenomenon was correlated with the upregulation of fibrosis-related proteins. To elucidate the role of SIRT2 in the autophagy-ciliogenesis-fibrosis axis, cells were treated with AGK2, a specific inhibitor of SIRT2. AGK2 treatment promoted the formation of both autophagosomes and autolysosomes and facilitated OFD1 degradation at the centriolar satellites, resulting in the lengthening of primary cilia. Restoration of primary cilia by AGK2 was associated with the suppression of myofibroblast transition. In conclusion, SIRT2 inhibition attenuates TGF-β-induced fibrosis by promoting autophagy-mediated ciliogenesis. This study highlights SIRT2 as a potential therapeutic target for fibrotic diseases.
Collapse
Affiliation(s)
- Juyoung Son
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jaejung Park
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Joo-Won Jeong
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Seung Hyeun Lee
- Department of Precision Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Ja-Eun Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; Department of Precision Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; Department of Pharmacology, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea.
| |
Collapse
|
8
|
Shahdadnejad K, Yazdanparast R. The influence of IMPDH activity on ciliogenesis and adipogenesis of 3T3-L1 cells while undergoing differentiation. Biochim Biophys Acta Mol Cell Biol Lipids 2025; 1870:159603. [PMID: 39961477 DOI: 10.1016/j.bbalip.2025.159603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 01/18/2025] [Accepted: 02/13/2025] [Indexed: 02/23/2025]
Abstract
The functional roles of primary cilia and inosine 5'-monophosphate dehydrogenase (IMPDH) are among the hot topics in today's adipogenesis research. Considering the reported interaction between IMPDH and ADP Ribosylation Factor-Like GTPase 13B (ARL13B), as a key ciliary protein, our study focused on this interaction during the ciliogenesis process while 3T3-L1 pre-adipocytes undergoing differentiation to lipid-accumulating adipocytes. Our results indicated that, in the early days of differentiation, when cilium length is long, IMPDH expression is high and its interaction with ARL13B is low. Conversely, in the last days of differentiation, the cilia length and IMPDH expression reduced while, the IMPDH/ARL13B interaction remains high relative to the initial days. In either of these two situations, IMPDH was not documented within the cilia. The extent of the interaction between IMPDH and ARL13B might account for the lack of co-localization of IMPDH and ARL13B within cilia during the process of differentiation. Although, inhibiting IMPDH in the early days of differentiation did not have a significant effect on cilia length, it did reduce adipogenesis by limiting mitotic clonal expansion through arresting cells in the G1/G0 phase. These findings provide the ground for further research to investigate the relationship between the IMPDH/ARL13B interaction and cilia length, which decline in obesity.
Collapse
Affiliation(s)
| | - Razieh Yazdanparast
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran.
| |
Collapse
|
9
|
Ding D, Tian R, Yang X, Ren Z, Jing ZC, Wu XT, Sun LW. The impact of ciliary length on the mechanical response of osteocytes to fluid shear stress. Nitric Oxide 2025; 155:1-11. [PMID: 39734011 DOI: 10.1016/j.niox.2024.12.003] [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: 11/19/2024] [Revised: 12/15/2024] [Accepted: 12/27/2024] [Indexed: 12/31/2024]
Abstract
BACKGROUND Osteocytes are crucial for detecting mechanical stimuli and translating them into biochemical responses within the bone. The primary cilium, a cellular 'antenna,' plays a vital role in this process. However, there is a lack of direct correlation between cilium length changes and osteocyte mechanosensitivity changes. This study aims to reveal the relationship between ciliary length and nitric oxide (NO) release in osteocytes to show how primary cilia may be involved in reducing osteocyte mechanosensitivity caused by microgravity. MATERIALS AND METHODS We used the MLO-Y4 cell line and primary osteoblasts to adjust the ciliary length using chloral hydrate (CH) for shortening and lithium ions (Li+) for elongation. We then examined the impact of varied ciliary lengths on osteocyte response to fluid shear stress, focusing on the PC1/PC2-Ca2+-NO signaling pathway. Co-culture systems assessed downstream effects on osteoblast function, including collagen secretion and mineralization. RESULTS We observed a significant correlation between ciliary length and osteocyte mechanosensitivity, with longer primary cilia enhancing Ca2+ influx and NO release in response to fluid shear stress. However, contrary to expectations, calmodulin (CaM) expression did not increase with ciliary length, suggesting alternative pathways, such as PKC or Akt/PKB, may modulate p-eNOS activity. Co-cultured osteoblasts showed altered osteogenic functions regulated by osteocyte-derived signals influenced by primary cilia length. CONCLUSION Our findings clarify the role of primary cilia length in modulating osteocyte mechanosensitivity and their influence on osteoblast function, highlighting a complex regulatory network that may not solely rely on CaM for NO release. These insights contribute to a deeper understanding of bone mechanotransduction and could have implications for developing therapeutic targets for osteocyte-related disorders.
Collapse
Affiliation(s)
- Dong Ding
- National Infrastructures for Translational Medicine, Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China; Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology, National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Ran Tian
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology, National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Xiao Yang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology, National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Zhe Ren
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology, National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China; Hospital of Beihang University, Beijing, 100191, China
| | - Zhi-Cheng Jing
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital Guangdong Academy of Medical Sciences, Southern Medical University. Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xin-Tong Wu
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology, National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China.
| | - Lian-Wen Sun
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology, National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China.
| |
Collapse
|
10
|
Pant K, Richard S, Peixoto E, Baral S, Yang R, Ren Y, Masyuk TV, LaRusso NF, Gradilone SA. Cholangiocyte ciliary defects induce sustained epidermal growth factor receptor signaling. Hepatology 2025; 81:1132-1145. [PMID: 39186465 DOI: 10.1097/hep.0000000000001055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 07/29/2024] [Indexed: 08/28/2024]
Abstract
BACKGROUND AND AIMS The primary cilium, an organelle that protrudes from cell surfaces, is essential for sensing extracellular signals. With disturbed cellular communication and chronic liver pathologies, this organelle's dysfunctions have been linked to disorders, including polycystic liver disease and cholangiocarcinoma. The goal of this study was to elucidate the relationship between primary cilia and the crucial regulator of cellular proliferation, the epidermal growth factor receptor (EGFR) signaling pathway, which has been associated with various clinical conditions. APPROACH AND RESULTS The study identified aberrant EGFR signaling pathways in cholangiocytes lacking functional primary cilia using liver-specific intraflagellar transport 88 knockout mice, a Pkhd1 mutant rat model, and human cell lines that did not have functional cilia. Cilia-deficient cholangiocytes showed persistent EGFR activation because of impaired receptor degradation, in contrast to their normal counterparts, where EGFR localization to the cilia promotes appropriate signaling. Using histone deacetylase 6 inhibitors to restore primary cilia accelerates EGFR degradation, thereby reducing maladaptive signaling. Importantly, experimental intervention with the histone deacetylase 6 inhibitor tubastatin A in an orthotopic rat model moved EGFR to cilia and reduced ERK phosphorylation. Concurrent administration of EGFR and histone deacetylase 6 inhibitors in cholangiocarcinoma and polycystic liver disease cells demonstrated synergistic antiproliferative effects, which were associated with the restoration of functioning primary cilia. CONCLUSIONS This study's findings shed light on ciliary function and robust EGFR signaling with slower receptor turnover. We could use therapies that restore the function of primary cilia to treat EGFR-driven diseases in polycystic liver disease and cholangiocarcinoma.
Collapse
Affiliation(s)
- Kishor Pant
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
| | - Seth Richard
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
| | | | - Subheksha Baral
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
| | - Rendong Yang
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Yanan Ren
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Tatyana V Masyuk
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, USA
| | - Nicholas F LaRusso
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, USA
| | - Sergio A Gradilone
- The Hormel Institute, University of Minnesota, Austin, Minnesota, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
11
|
Mao S, Song R, Jin S, Pang S, Jovanovic A, Zimmerman A, Li P, Wu X, Wendland MF, Lin K, Chen WC, Choksi SP, Chen G, Holtzman MJ, Reiter JF, Wan Y, Xuan Z, Xiang YK, Xu CS, Upadhyayula S, Hess HF, He L. Multicilia dynamically transduce Sonic Hedgehog signaling to regulate choroid plexus functions. Cell Rep 2025; 44:115383. [PMID: 40057957 DOI: 10.1016/j.celrep.2025.115383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 11/17/2024] [Accepted: 02/11/2025] [Indexed: 03/29/2025] Open
Abstract
The choroid plexus is a major site for cerebrospinal fluid (CSF) production, characterized by a multiciliated epithelial monolayer that regulates CSF production. We demonstrate that defective choroid plexus ciliogenesis or intraflagellar transport yields neonatal hydrocephalus, at least in part due to increased water channel Aqp1 and ion transporter Atp1a2 expression. We demonstrate choroid plexus multicilia as sensory cilia, transducing both canonical and non-canonical Sonic Hedgehog (Shh) signaling. Interestingly, it is the non-canonical Shh signaling that represses Aqp1 and Atp1a2 expression by the Smoothened (Smo)/Gαi/cyclic AMP (cAMP) pathway. Choroid plexus multicilia exhibit unique ciliary ultrastructure, carrying features of both primary and motile cilia. Unlike most cilia that elongate during maturation, choroid plexus ciliary length decreases during development, causing a decline of Shh signaling intensity in the developing choroid plexus, a derepression of Aqp1 and Atp1a2, and, ultimately, increased CSF production. Hence, the developmental dynamics of choroid plexus multicilia dampens the Shh signaling intensity to promote CSF production.
Collapse
Affiliation(s)
- Suifang Mao
- Division of Cellular and Developmental Biology, MCB Department, University of California, Berkeley, Berkeley, CA 94705, USA
| | - Rui Song
- Division of Cellular and Developmental Biology, MCB Department, University of California, Berkeley, Berkeley, CA 94705, USA
| | - Shibo Jin
- Division of Cellular and Developmental Biology, MCB Department, University of California, Berkeley, Berkeley, CA 94705, USA
| | - Song Pang
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Aleksandra Jovanovic
- Division of Cellular and Developmental Biology, MCB Department, University of California, Berkeley, Berkeley, CA 94705, USA
| | - Adam Zimmerman
- Division of Cellular and Developmental Biology, MCB Department, University of California, Berkeley, Berkeley, CA 94705, USA
| | - Peng Li
- Division of Cellular and Developmental Biology, MCB Department, University of California, Berkeley, Berkeley, CA 94705, USA
| | - Xinying Wu
- Division of Cellular and Developmental Biology, MCB Department, University of California, Berkeley, Berkeley, CA 94705, USA
| | - Michael F Wendland
- Division of Cellular and Developmental Biology, MCB Department, University of California, Berkeley, Berkeley, CA 94705, USA
| | - Kerry Lin
- Division of Cellular and Developmental Biology, MCB Department, University of California, Berkeley, Berkeley, CA 94705, USA
| | - Wei-Chi Chen
- Division of Cellular and Developmental Biology, MCB Department, University of California, Berkeley, Berkeley, CA 94705, USA
| | - Semil P Choksi
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Gang Chen
- Chongqing Key Laboratory of Cytomics, Chongqing 400038, China
| | - Michael J Holtzman
- Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Jeremy F Reiter
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94143, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Ying Wan
- Chongqing Key Laboratory of Cytomics, Chongqing 400038, China
| | - Zhenyu Xuan
- Department of Molecular and Cell Biology, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Yang K Xiang
- Department of Pharmacology, University of California at Davis, Davis, CA 95616, USA; VA Northern California Health Care System, Mather, CA 95655, USA
| | - C Shan Xu
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA; Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Srigokul Upadhyayula
- Division of Cellular and Developmental Biology, MCB Department, University of California, Berkeley, Berkeley, CA 94705, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA.
| | - Harald F Hess
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA.
| | - Lin He
- Division of Cellular and Developmental Biology, MCB Department, University of California, Berkeley, Berkeley, CA 94705, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA.
| |
Collapse
|
12
|
Bear R, Wei C, Caspary T. In vivo genetic labeling of primary cilia in developing astrocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.19.644181. [PMID: 40166229 PMCID: PMC11957109 DOI: 10.1101/2025.03.19.644181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Astrocyte cilia are largely understudied due to the lack of available tools. Astrocyte research advanced with the establishment of Aldh1l1-Cre ERT2 , an inducible Cre line that specifically targets the astrocyte lineage. Here, we develop and compare genetic models that label astrocyte cilia in the developing prefrontal cortex (PFC) using Aldh1l1-Cre ERT2 and Cre-dependent cilia reporters. We evaluate these models by testing different tamoxifen-induction protocols and quantifying the percentage of astrocytes labeled with the cilia reporters. We show that tamoxifen dosage impacts the expression of cilia reporters in astrocytes. We validate the maximum cilia-labeling efficiency of tamoxifen using constitutively-expressed cilia reporters. The data reveal that only a subset of SOX9- positive astrocytes in the PFC possess cilia throughout development. Our work highlights the utility of Cre-Lox systems to target specific cell types and the importance of carefully validating genetic models.
Collapse
Affiliation(s)
- Rachel Bear
- Department of Human Genetics, Emory University School of Medicine, 615 Michael Street Suite 301, Atlanta GA 30322, United States
- Emory Graduate Program in Neuroscience
| | - Claire Wei
- Department of Human Genetics, Emory University School of Medicine, 615 Michael Street Suite 301, Atlanta GA 30322, United States
| | - Tamara Caspary
- Department of Human Genetics, Emory University School of Medicine, 615 Michael Street Suite 301, Atlanta GA 30322, United States
| |
Collapse
|
13
|
Bae JE, Jang S, Kim JB, Park NY, Jo DS, Hyung H, Kim P, Kim MS, Ryu HY, Lee HS, Lee DS, Baes M, Ryoo ZY, Cho DH. HSD17B4 deficiency causes dysregulation of primary cilia and is alleviated by acetyl-CoA. Nat Commun 2025; 16:2663. [PMID: 40102401 PMCID: PMC11920078 DOI: 10.1038/s41467-025-57793-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 03/02/2025] [Indexed: 03/20/2025] Open
Abstract
Primary cilia are dynamic sensory organelles orchestrating key signaling pathways, and disruption of primary ciliogenesis is implicated in a spectrum of genetic disorders. The peroxisomal bifunctional enzyme HSD17B4 is pivotal for peroxisomal β-oxidation and acetyl-CoA synthesis, and its deficiency profoundly impairs peroxisomal metabolism. While patients with HSD17B4 deficiency exhibit ciliopathy-like symptoms due to dysfunctional primary cilia, the molecular connection between HSD17B4 and ciliopathy remains poorly understood. Here, we demonstrate that HSD17B4 deficiency impairs primary ciliogenesis and alters cilia-mediated signaling, suggesting a potential link between peroxisomal metabolism and ciliary function. Notably, elevation of acetyl-CoA rescues ciliary defects via HDAC6-mediated ciliogenesis in HSD17B4-deficient cells. Strikingly, acetate administration restores motor function, enhances primary cilia formation, and preserves the Purkinje layer in Hsd17B4-knockout mice. These findings provide insights into the functional link between HSD17B4 and primary cilia, highlighting acetyl-CoA as a potential therapeutic target for HSD17B4 deficiency and ciliopathy.
Collapse
Affiliation(s)
- Ji-Eun Bae
- Organelle Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Soyoung Jang
- Organelle Institute, Kyungpook National University, Daegu, Republic of Korea
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
| | - Joon Bum Kim
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
| | - Na Yeon Park
- Organelle Institute, Kyungpook National University, Daegu, Republic of Korea
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
| | - Doo Sin Jo
- ORGASIS Corp, Suwon, Gyeonggi-do, Republic of Korea
| | - Hyejin Hyung
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
| | - Pansoo Kim
- ORGASIS Corp, Suwon, Gyeonggi-do, Republic of Korea
| | - Min-Seon Kim
- Division of Endocrinology and Metabolism, Asan Medical Center, Seoul, Republic of Korea
| | - Hong-Yeoul Ryu
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
- KNU G-LAMP Project Group, KNU Institute of Basic Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Hyun-Shik Lee
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
- KNU G-LAMP Project Group, KNU Institute of Basic Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Dong-Seok Lee
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
| | - Myriam Baes
- Laboratory of Cell Metabolism, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Zae Young Ryoo
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea.
| | - Dong-Hyung Cho
- Organelle Institute, Kyungpook National University, Daegu, Republic of Korea.
- School of Life Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea.
- ORGASIS Corp, Suwon, Gyeonggi-do, Republic of Korea.
| |
Collapse
|
14
|
Lee E, Chou L, Chen Z, Wong BJF. Optical Imaging of Cilia in the Head and Neck. J Clin Med 2025; 14:2059. [PMID: 40142867 PMCID: PMC11943365 DOI: 10.3390/jcm14062059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 03/05/2025] [Accepted: 03/07/2025] [Indexed: 03/28/2025] Open
Abstract
Background/Objectives: Cilia are hair-like organelles with various mechanosensory and chemosensory functions. In particular, motile cilia generate directional fluid flow important for multiple processes. Motile ciliopathies have serious clinical implications, including developmental and respiratory disorders. Evaluating the most suitable imaging methods for studying ciliary structure and function has great clinical significance. Methods: Here, we provide an overview of ciliary function, imaging modalities, and applications in ciliopathic diseases. Results: Optical imaging has become a crucial tool for studying ciliary structure and function, providing high-resolution, non-invasive imaging capabilities that are valuable for in vivo applications. Optical coherence tomography (OCT) is well suited for the visualization of ciliary anatomy and quantitative studies of microfluidic flow. Conclusions: A deeper understanding of ciliary biology can lead to novel approaches in diagnosing, treating, and monitoring ciliopathies, contributing to more effective and individualized care.
Collapse
Affiliation(s)
- Elizabeth Lee
- Beckman Laser Institute, University of California, Irvine, CA 92697, USA; (E.L.); (L.C.); (Z.C.)
- Department of Otolaryngology-Head and Neck Surgery, University of California, Irvine, CA 92612, USA
| | - Lidek Chou
- Beckman Laser Institute, University of California, Irvine, CA 92697, USA; (E.L.); (L.C.); (Z.C.)
| | - Zhongping Chen
- Beckman Laser Institute, University of California, Irvine, CA 92697, USA; (E.L.); (L.C.); (Z.C.)
- Department of Biomedical Engineering, University of California, Irvine, CA 92612, USA
| | - Brian J. F. Wong
- Beckman Laser Institute, University of California, Irvine, CA 92697, USA; (E.L.); (L.C.); (Z.C.)
- Department of Otolaryngology-Head and Neck Surgery, University of California, Irvine, CA 92612, USA
- Department of Biomedical Engineering, University of California, Irvine, CA 92612, USA
| |
Collapse
|
15
|
Valls A, Ruiz-Roldán C, Immanuel J, Alonso-Martín S, Gallardo E, Fernández-Torrón R, Bonilla M, Lersundi A, Hernández-Laín A, Domínguez-González C, Vílchez JJ, Iruzubieta P, López de Munain A, Sáenz A. The Role of Integrin β1D Mislocalization in the Pathophysiology of Calpain 3-Related Limb-Girdle Muscular Dystrophy. Cells 2025; 14:446. [PMID: 40136695 PMCID: PMC11941428 DOI: 10.3390/cells14060446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 03/06/2025] [Accepted: 03/10/2025] [Indexed: 03/27/2025] Open
Abstract
Limb-girdle muscular dystrophy R1 (LGMDR1) is characterized by progressive proximal muscle weakness due to mutations in the CAPN3 gene. Little is known about CAPN3's function in muscle, but its loss results in aberrant sarcomere formation. Human muscle structure was analyzed in this study, with observations including integrin β1D isoform (ITGβ1D) mislocalization, a lack of Talin-1 (TLN1) in the sarcolemma and the irregular expression of focal adhesion kinase (FAK) in LGMDR1 muscles, suggesting a lack of integrin activation with an altered sarcolemma, extracellular matrix (ECM) assembly and signaling pathway deregulation, which may cause frailty in LGMDR1 muscle fibers. Additionally, altered nuclear morphology, centrosome distribution and microtubule organization have been found in muscle cells derived from LGMDR1 patients.
Collapse
Affiliation(s)
- Andrea Valls
- Neuromuscular Diseases Group, Neurosciences Area, Biogipuzkoa Health Research Institute, 20014 San Sebastian, Spain
- Center for Biomedical Network Research on Neurodegenerative Diseases (CIBERNED), Spanish Ministry of Science & Innovation, Carlos III Health Institute, 28029 Madrid, Spain
| | - Cristina Ruiz-Roldán
- Neuromuscular Diseases Group, Neurosciences Area, Biogipuzkoa Health Research Institute, 20014 San Sebastian, Spain
| | - Jenita Immanuel
- Neuromuscular Diseases Group, Neurosciences Area, Biogipuzkoa Health Research Institute, 20014 San Sebastian, Spain
- Center for Biomedical Network Research on Neurodegenerative Diseases (CIBERNED), Spanish Ministry of Science & Innovation, Carlos III Health Institute, 28029 Madrid, Spain
| | - Sonia Alonso-Martín
- Center for Biomedical Network Research on Neurodegenerative Diseases (CIBERNED), Spanish Ministry of Science & Innovation, Carlos III Health Institute, 28029 Madrid, Spain
- Stem Cells and Aging Group, Bioengineering Area, Biogipuzkoa Health Research Institute, 20014 San Sebastian, Spain
| | - Eduard Gallardo
- Neuromuscular Diseases Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain
- Institut de Recerca Sant Pau, IR-SantPau, 08041 Barcelona, Spain
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Spanish Ministry of Science & Innovation, Carlos III Health Institute, 28029 Madrid, Spain
| | - Roberto Fernández-Torrón
- Neuromuscular Diseases Group, Neurosciences Area, Biogipuzkoa Health Research Institute, 20014 San Sebastian, Spain
- Center for Biomedical Network Research on Neurodegenerative Diseases (CIBERNED), Spanish Ministry of Science & Innovation, Carlos III Health Institute, 28029 Madrid, Spain
- Department of Neurology, Hospital Universitario Donostia, Osakidetza, 20014 San Sebastian, Spain
| | - Mario Bonilla
- Stem Cells and Aging Group, Bioengineering Area, Biogipuzkoa Health Research Institute, 20014 San Sebastian, Spain
- Department of Traumatology, Donostialdea Integrated Health Organisation, Osakidetza, 20014 San Sebastian, Spain
| | - Ana Lersundi
- Department of Traumatology, Donostialdea Integrated Health Organisation, Osakidetza, 20014 San Sebastian, Spain
- Department of Surgery, University of the Basque Country UPV/EHU, 20014 San Sebastian, Spain
| | - Aurelio Hernández-Laín
- Department of Neuropathology, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
- Department of Pathology, Faculty of Medicine, Complutense University of Madrid (UCM), 28040 Madrid, Spain
| | - Cristina Domínguez-González
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Spanish Ministry of Science & Innovation, Carlos III Health Institute, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
- Neuromuscular Unit, Department of Neurology, Hospital 12 de Octubre, 28041 Madrid, Spain
| | - Juan Jesús Vílchez
- Center for Biomedical Network Research on Rare Diseases (CIBERER), Spanish Ministry of Science & Innovation, Carlos III Health Institute, 28029 Madrid, Spain
- Neuromuscular and Ataxias Research Group, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
- Neuromuscular Diseases Unit, Neurology Department, Hospital Universitari I Politècnic La Fe, 46026 Valencia, Spain
| | - Pablo Iruzubieta
- Center for Biomedical Network Research on Neurodegenerative Diseases (CIBERNED), Spanish Ministry of Science & Innovation, Carlos III Health Institute, 28029 Madrid, Spain
- Neurogenetics, RNA Biology and Therapies Group, Neurosciences Area, Biogipuzkoa Health Research Institute, 20014 San Sebastian, Spain
- Department of Neurology and Neurosurgery, Montreal Neurological Hospital and Institute, McGill University, Montreal, QC H3A 2B4, Canada
| | - Adolfo López de Munain
- Neuromuscular Diseases Group, Neurosciences Area, Biogipuzkoa Health Research Institute, 20014 San Sebastian, Spain
- Center for Biomedical Network Research on Neurodegenerative Diseases (CIBERNED), Spanish Ministry of Science & Innovation, Carlos III Health Institute, 28029 Madrid, Spain
- Department of Neurology, Hospital Universitario Donostia, Osakidetza, 20014 San Sebastian, Spain
- Department of Neurosciences, University of the Basque Country UPV-EHU, 20014 San Sebastian, Spain
- Faculty of Medicine, University of Deusto, 48007 Bilbao, Spain
| | - Amets Sáenz
- Neuromuscular Diseases Group, Neurosciences Area, Biogipuzkoa Health Research Institute, 20014 San Sebastian, Spain
- Center for Biomedical Network Research on Neurodegenerative Diseases (CIBERNED), Spanish Ministry of Science & Innovation, Carlos III Health Institute, 28029 Madrid, Spain
| |
Collapse
|
16
|
Ku PI, Sreeja JS, Chadha A, Williams DS, Engelke MF, Subramanian R. Collaborative role of two distinct cilium-specific cytoskeletal systems in driving Hedgehog-responsive transcription factor trafficking. SCIENCE ADVANCES 2025; 11:eadt5439. [PMID: 40073114 PMCID: PMC11900865 DOI: 10.1126/sciadv.adt5439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 02/05/2025] [Indexed: 03/14/2025]
Abstract
In vertebrate Hedgehog (Hh) signaling, the precise output of the final effectors, GLI (glioma-associated oncogene) transcription factors, depends on the primary cilium. Upon pathway initiation, generating the precise levels of the activator form of GLI depends on its concentration at the cilium tip. The mechanisms underlying this critical step in Hh signaling are unclear. We developed an assay to visualize GLI2, the primary GLI activator isoform, at single-particle resolution within the cilium. We found that GLI2 is a cargo of intraflagellar transport (IFT) machinery. Anterograde-biased IFT loading of GLI2 in a restricted time window following pathway activation results in the tip accumulation of GLI2. Unexpectedly, we found that the conserved Hh regulator KIF7, a nonmotile kinesin, is important for the temporal control of IFT-mediated GLI2 transport and retention of GLI2 at the cilium tip. Our findings underscore a design principle where a cilia-specific cytoskeletal transport system and an Hh pathway-specific cytoskeletal protein collaboratively regulate GLI2 trafficking for Hh signaling.
Collapse
Affiliation(s)
- Pei-I Ku
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Jamuna S. Sreeja
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Abhishek Chadha
- Departments of Ophthalmology and Neurobiology, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - David S. Williams
- Departments of Ophthalmology and Neurobiology, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
| | - Martin F. Engelke
- Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, TN, USA
| | - Radhika Subramanian
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
17
|
Beyer T, Diwan GD, Leonhard T, Dahlke K, Klose F, Stehle IF, Seda M, Bolz S, Woerz F, Russell RB, Jenkins D, Ueffing M, Boldt K. Ciliopathy-Associated Missense Mutations in IFT140 are Tolerated by the Inherent Resilience of the IFT Machinery. Mol Cell Proteomics 2025; 24:100916. [PMID: 39880085 PMCID: PMC11907452 DOI: 10.1016/j.mcpro.2025.100916] [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: 04/25/2024] [Revised: 11/26/2024] [Accepted: 01/23/2025] [Indexed: 01/31/2025] Open
Abstract
Genotype-phenotype correlations of rare diseases are complicated by low patient number, high phenotype variability, and compound heterozygosity. Mutations may cause instability of single proteins, and affect protein complex formation or overall robustness of a specific process in a given cell. Ciliopathies offer an interesting case for studying genotype-phenotype correlations as they have a spectrum of severity and include diverse phenotypes depending on different mutations in the same protein. For instance, mutations in the intraflagellar transport protein IFT140 cause a vast spectrum of ciliopathies ranging from isolated retinal dystrophy to severe skeletal abnormalities and multi-organ diseases such as Mainzer-Saldino and Jeune syndrome. Here, the quantitative effects of 23 missense mutations in IFT140, which forms part of the crucial IFT-A complex of the ciliary machinery, were analyzed using affinity purification coupled with mass spectrometry (AP-MS). A subset of 10 mutations led to a significant and domain-specific reduction in IFT140-IFT-A complex interaction indicating complex formation issues and potentially hampering its molecular function. Knockout of IFT140 led to loss of cilia, as shown before. However, phenotypically only mild effects concerning cilia assembly were observed for two out of four tested IFT140 missense mutations. Therefore, our results demonstrate the utility of AP-MS in discerning pathogenic MMs from polymorphisms, and we postulate that reduced function is tolerated by the evolutionarily highly conserved IFT-A system.
Collapse
Affiliation(s)
- Tina Beyer
- Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Germany.
| | - Gaurav D Diwan
- BioQuant, University of Heidelberg, Heidelberg, Germany; Biochemistry Center (BZH), Heidelberg University, Heidelberg, Germany
| | - Tobias Leonhard
- Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Katrin Dahlke
- Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Franziska Klose
- Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Isabel F Stehle
- Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Marian Seda
- UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Sylvia Bolz
- Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Franziska Woerz
- Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Robert B Russell
- BioQuant, University of Heidelberg, Heidelberg, Germany; Biochemistry Center (BZH), Heidelberg University, Heidelberg, Germany
| | - Dagan Jenkins
- UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom.
| | - Marius Ueffing
- Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Karsten Boldt
- Institute for Ophthalmic Research, Center for Ophthalmology, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
18
|
Yang Y, Xue Z, Lai J, Zhang J, Pang C, Zhong J, Kuang Z, Zou B, Liu Y, Sun L. Kibra knockdown inhibits the aberrant Hippo pathway, suppresses renal cyst formation and ameliorates renal fibrosis in nphp1 KO mice. Clin Transl Med 2025; 15:e70245. [PMID: 39995111 PMCID: PMC11850762 DOI: 10.1002/ctm2.70245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 01/11/2025] [Accepted: 02/13/2025] [Indexed: 02/26/2025] Open
Abstract
INTRODUCTION Nephronophthisis (NPH) is an autosomal recessive interstitial cystic kidney disease, which is the most common genetic cause of end-stage renal disease (ESRD) in childhood. The Hippo pathway is regulated by the cilium and has been suggested to be linked to NPH. The aim of the study was to investigate the involvement of Hippo pathway in the pathogenesis of nphp1 defect-associated NPH (NPH1). METHOD Nphp1 knockout (nphp1KO) Madin-Darby Canine Kidney (MDCK) cells and nphp1KO C57BL/6J mice were generated via CRISPR gene editing strategy. The siRNAs targeting Kibra, MST1 and LATS1 were designed. An AAV9 vector was designed for Kibra knockdown. The expression and phosphorylation of core Hippo pathway molecules were evaluated. Pathological renal changes were evaluated via light microscopy respectively with haematoxylin-eosin and Masson staining. RESULTS In nphp1KO MDCK cells, nphp1KO mice and NPH1 patients' kidneys, Kibra, p-MST1/2, p-LATS and p-YAP exhibited a notable increase in levels, with an even greater elevation observed in renal cyst cells, indicating the Hippo pathway activated in these nphp1-deficient contexts. Nphp1 re-expression reversed the Hippo pathway activation in cells, indicating that the Hippo pathway activation is related to nphp1 deficiency in vitro. Meanwhile, in vitro, MST1 knockdown downregulated LATS1 and YAP phosphorylation, LATS1 knockdown downregulated YAP phosphorylation, suggesting the activation of the canonical Hippo pathway in nphp1-deficient contexts. Knockdown of the upstream regulator Kibra inhibited the Hippo pathway activation in both nphp1KO MDCK cells and mice. Following Kibra knockdown, the organisation of nphp1KO MDCK cells became more compact, the intensity of the actin fibres increased. Besides, decreased renal fibrosis and cyst formation were observed in nphp1KO mice. CONCLUSIONS The canonical Hippo pathway is aberrantly activated in nphp1-deficient conditions. Kibra may serve as a crucial upstream regulator of nphp1 deficiency-related Hippo pathway activation. Kibra upregulation and activation of the Hippo pathway are involved in the pathogenesis of NPH1. KEY POINTS Canonical Hippo pathway activated in nphp1-deficient disease models and patients. Kibra was a key upstream molecule in regulating the activation of canonical Hippo pathway in nphp1-deficient disease models and patients and closely related to renal cyst formation and fibrosis in nphp1KO mice.
Collapse
Affiliation(s)
- Yichen Yang
- Department of PediatricsNanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Zhihe Xue
- Department of PediatricsNanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Jiayong Lai
- Department of PediatricsNanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Jinglan Zhang
- Department of PediatricsNanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Changmiao Pang
- Department of PediatricsNanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Jinglin Zhong
- Department of PediatricsShenzhen Maternity and Child Healthcare HospitalShenzhenChina
| | - Zhanpeng Kuang
- Department of PediatricsNanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Baojuan Zou
- Department of PediatricsNanfang Hospital, Southern Medical UniversityGuangzhouChina
| | - Yaqing Liu
- Department of PediatricsThe First Affiliated Hospital, Gannan Medical UniversityGanzhouChina
| | - Liangzhong Sun
- Department of PediatricsNanfang Hospital, Southern Medical UniversityGuangzhouChina
| |
Collapse
|
19
|
Laporte D, Sagot I. Microtubule Reorganization and Quiescence: an Intertwined Relationship. Physiology (Bethesda) 2025; 40:0. [PMID: 39378102 DOI: 10.1152/physiol.00036.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/01/2024] [Accepted: 10/02/2024] [Indexed: 10/10/2024] Open
Abstract
Quiescence is operationally defined as a reversible proliferation arrest. This cellular state is central to both organism development and homeostasis, and its dysregulation causes many pathologies. The quiescent state encompasses very diverse cellular situations depending on the cell type and its environment. Further, quiescent cell properties evolve with time, a process that is thought to be the origin of aging in multicellular organisms. Microtubules are found in all eukaryotes and are essential for cell proliferation as they support chromosome segregation and intracellular trafficking. Upon proliferation cessation and quiescence establishment, the microtubule cytoskeleton was shown to undergo significant remodeling. The purpose of this review is to examine the literature in search of evidence to determine whether the observed microtubule reorganizations are merely a consequence of quiescence establishment or if they somehow participate in this cell fate decision.
Collapse
Affiliation(s)
- Damien Laporte
- Centre National de la Recherche ScientifiqueUniversité de Bordeaux-Institut de Biochimie et Génétique Cellulaires, UMR5095, Bordeaux, France
| | - Isabelle Sagot
- Centre National de la Recherche ScientifiqueUniversité de Bordeaux-Institut de Biochimie et Génétique Cellulaires, UMR5095, Bordeaux, France
| |
Collapse
|
20
|
Yamaguchi H, Meyer MD, Barrell WB, Faisal M, Berdeaux R, Liu KJ, Komatsu Y. The primary cilia: Orchestrating cranial neural crest cell development. Differentiation 2025; 142:100818. [PMID: 39500655 PMCID: PMC11911094 DOI: 10.1016/j.diff.2024.100818] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/28/2024] [Accepted: 10/28/2024] [Indexed: 03/17/2025]
Abstract
Primary cilia (hereafter "cilia") are microtubule-based antenna-like organelles projecting from the surface of vertebrate cells. Cilia can serve as cellular antennae controlling cell growth and differentiation. Absent or dysfunctional cilia frequently lead to craniofacial anomalies known as craniofacial ciliopathies. However, the detailed pathological mechanisms of craniofacial ciliopathies remain unclear. This perspective discusses our current understanding of the role of cilia in cranial neural crest cells. We also describe potential mechanisms of ciliogenesis in cranial neural crest cells, which may contribute to unraveling the complex pathogenesis of craniofacial ciliopathies.
Collapse
Affiliation(s)
- Hiroyuki Yamaguchi
- Department of Pediatrics, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Matthew D Meyer
- Shared Equipment Authority, Rice University, Houston, TX, USA
| | - William B Barrell
- Centre for Craniofacial and Regenerative Biology, King's College London, SE1 9RT, London, UK
| | - Maryam Faisal
- Department of Pediatrics, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, USA; Department of Bioengineering, Rice University George R. Brown School of Engineering, 77005, Houston, TX, USA
| | - Rebecca Berdeaux
- Department of Integrative Biology and Pharmacology, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, USA; CellChorus INC, Houston, TX, USA
| | - Karen J Liu
- Centre for Craniofacial and Regenerative Biology, King's College London, SE1 9RT, London, UK
| | - Yoshihiro Komatsu
- Department of Pediatrics, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, USA; Graduate Program in Genetics & Epigenetics, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, 77030, Houston, TX, USA.
| |
Collapse
|
21
|
Shim MS, Sim EJ, Betsch K, Desikan V, Su CC, Pastor-Valverde D, Sun Y, Liton PB. Class I PI3Ks activate stretch-induced autophagy in trabecular meshwork cells. Cell Mol Life Sci 2025; 82:82. [PMID: 39985671 PMCID: PMC11846827 DOI: 10.1007/s00018-025-05615-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 01/13/2025] [Accepted: 02/04/2025] [Indexed: 02/24/2025]
Abstract
Elevated intraocular pressure (IOP) is the primary risk factor for glaucoma, a leading cause of irreversible blindness worldwide. IOP homeostasis is maintained through a balance between aqueous humor production and its drainage through the trabecular meshwork (TM)/Schlemm's Canal (SC) outflow pathway. Prior studies by our laboratory reported a key role of mechanical forces and primary cilia (PC)-dependent stretch-induced autophagy in IOP homeostasis. However, the precise mechanism regulating this process remains elusive. In this study, we investigated the upstream signaling pathway orchestrating autophagy activation during cyclic mechanical stretch (CMS) in primary cultured human TM cells, using biochemical and cell biological analyses. Our results indicate that TM cells express catalytic subunits of class IA PI3Ks (PIK3CA, B, and D), and that inhibition of class IA isoforms, but not class II and III, significantly prevent CMS-induced autophagy. Importantly, PIK3CA was found to localize in the PC. Furthermore, we identified a coordinated action of Class IA PI3Ks along INPP4A/B, a 4' inositol phosphatase, responsible for the formation of PI(3,4)P2 and PI(3)P and stretch-induced autophagy in TM cells. These findings contribute to a deeper understanding of the molecular mechanisms underlying IOP homeostasis.
Collapse
Affiliation(s)
- Myoung Sup Shim
- Department of Ophthalmology, Duke Eye Center, Duke University, AERI Bldg, Office 4004, Erwin, Rd. Box 3802, Durham, NC, 27713, USA
| | - Ethan J Sim
- Department of Ophthalmology, Duke Eye Center, Duke University, AERI Bldg, Office 4004, Erwin, Rd. Box 3802, Durham, NC, 27713, USA
| | - Kevin Betsch
- Department of Ophthalmology, Duke Eye Center, Duke University, AERI Bldg, Office 4004, Erwin, Rd. Box 3802, Durham, NC, 27713, USA
| | - Vaibhav Desikan
- Department of Ophthalmology, Duke Eye Center, Duke University, AERI Bldg, Office 4004, Erwin, Rd. Box 3802, Durham, NC, 27713, USA
| | - Chien-Chia Su
- Department of Ophthalmology, Duke Eye Center, Duke University, AERI Bldg, Office 4004, Erwin, Rd. Box 3802, Durham, NC, 27713, USA
| | - Diego Pastor-Valverde
- Department of Ophthalmology, Duke Eye Center, Duke University, AERI Bldg, Office 4004, Erwin, Rd. Box 3802, Durham, NC, 27713, USA
| | - Yang Sun
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Paloma B Liton
- Department of Ophthalmology, Duke Eye Center, Duke University, AERI Bldg, Office 4004, Erwin, Rd. Box 3802, Durham, NC, 27713, USA.
| |
Collapse
|
22
|
Norppa AJ, Shcherbii MV, Frilander MJ. Connecting genotype and phenotype in minor spliceosome diseases. RNA (NEW YORK, N.Y.) 2025; 31:284-299. [PMID: 39761998 PMCID: PMC11874965 DOI: 10.1261/rna.080337.124] [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: 02/21/2025]
Abstract
Minor spliceosome is responsible for recognizing and excising a specific subset of divergent introns during the pre-mRNA splicing process. Mutations in the unique snRNA and protein components of the minor spliceosome are increasingly being associated with a variety of germline and somatic human disorders, collectively termed as minor spliceosomopathies. Understanding the mechanistic basis of these diseases has been challenging due to limited functional information on many minor spliceosome components. However, recently published cryo-electron microscopy (cryo-EM) structures of various minor spliceosome assembly intermediates have marked a significant advancement in elucidating the roles of these components during splicing. These structural breakthroughs have not only enhanced our comprehension of the minor spliceosome's functionality but also shed light on how disease-associated mutations disrupt its functions. Consequently, research focus is now shifting toward investigating how these splicing defects translate into broader pathological processes within gene expression pathways. Here we outline the current structural and functional knowledge of the minor spliceosome, explore the mechanistic consequences of its mutations, and discuss emerging challenges in connecting molecular dysfunctions to clinical phenotypes.
Collapse
Affiliation(s)
- Antto J Norppa
- Institute of Biotechnology, 000014 University of Helsinki, Finland
| | | | | |
Collapse
|
23
|
Kim B, Weon B, Kim E, Park S, Jin W, Shin N, Oh YK, Lim CS, Lee JP, Kwon O, Lee J. Clinical implications of proximal tubular multicilia in glomerular diseases. Heliyon 2025; 11:e42416. [PMID: 39991237 PMCID: PMC11847256 DOI: 10.1016/j.heliyon.2025.e42416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 01/02/2025] [Accepted: 01/30/2025] [Indexed: 02/25/2025] Open
Abstract
Introduction Multiciliated cells (MCCs) have been identified in the proximal tubules of patients with kidney disease; however, their clinical significance is unknown. We aimed to investigate whether MCCs are associated with clinical outcomes in patients with glomerular diseases. Methods Between August 2012 and April 2021, 134 patients (including 126 with glomerular disease patients and 8 controls) who were hospitalized at Seoul National University Boramae Medical Center and Seoul National University Hospital were included in this study. The ratio of MCCs to total proximal tubular cells was calculated using immunohistochemistry. The relationship between the MCC ratio and kidney disease-related clinical features was then analyzed. Results MCCs were exclusively detected in patients with glomerular diseases (68.3 %), not those in the control group. Patients with diabetic kidney disease (88.9 %) or antineutrophil cytoplasmic antibody (ANCA)-associated glomerulonephritis (GN, 86.4 %) had higher MCC ratios. MCC-positivity and MCC ratios were significantly associated with increased age and proteinuria, and a decreased estimated glomerular filtration rate (eGFR). Patients with higher MCC ratios had a significantly higher risk of end-stage kidney disease (ESKD) and composite outcomes with death. In multivariable analysis, MCC ratios were significantly correlated with an increased risk of ESKD (hazard ratio [HR], 1.413; 95 % confidence interval [CI], 1.012-1.972) and composite outcome (HR, 1.401; 95 % CI, 1.028-1.909). Conclusion Higher MCC ratios were correlated with poorer prognosis; therefore, quantification of MCCs in the proximal tubules can serve as a valuable prognostic marker in clinical practice.
Collapse
Affiliation(s)
- Bohye Kim
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, South Korea
- Genomic Medicine Institute, Medical Research Center, Seoul National University, Seoul, South Korea
| | - Boram Weon
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, South Korea
| | - Evonne Kim
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, South Korea
- Department of Biomedical Sciences, BK21 FOUR Biomedical Science Program, Seoul National University College of Medicine, Seoul, South Korea
| | - Sohee Park
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, South Korea
- Department of Biomedical Sciences, BK21 FOUR Biomedical Science Program, Seoul National University College of Medicine, Seoul, South Korea
| | - Wencheng Jin
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, South Korea
- Department of Internal Medicine, College of Medicine, Seoul National University, Seoul, South Korea
| | - Nayeon Shin
- Department of Internal Medicine, College of Medicine, Seoul National University, Seoul, South Korea
| | - Yun Kyu Oh
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, South Korea
- Department of Internal Medicine, College of Medicine, Seoul National University, Seoul, South Korea
| | - Chun Soo Lim
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, South Korea
- Department of Internal Medicine, College of Medicine, Seoul National University, Seoul, South Korea
| | - Jung Pyo Lee
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, South Korea
- Department of Internal Medicine, College of Medicine, Seoul National University, Seoul, South Korea
| | - Obin Kwon
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, South Korea
- Genomic Medicine Institute, Medical Research Center, Seoul National University, Seoul, South Korea
- Department of Biomedical Sciences, BK21 FOUR Biomedical Science Program, Seoul National University College of Medicine, Seoul, South Korea
- Sensory Organ Research Institute, Medical Research Center, Seoul National University, Seoul, South Korea
| | - Jeonghwan Lee
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, South Korea
- Department of Internal Medicine, College of Medicine, Seoul National University, Seoul, South Korea
| |
Collapse
|
24
|
Benk Vysloužil D, Bernatík O, Lánská E, Renzová T, Binó L, Lacigová A, Drahošová T, Lánský Z, Čajánek L. Tau-tubulin kinase 2 restrains microtubule-depolymerizer KIF2A to support primary cilia growth. Cell Commun Signal 2025; 23:73. [PMID: 39930500 PMCID: PMC11809056 DOI: 10.1186/s12964-025-02072-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 01/29/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Primary cilia facilitate cellular signalling and play critical roles in development, homeostasis, and disease. Their assembly is under the control of Tau-Tubulin Kinase 2 (TTBK2), a key enzyme mutated in patients with spinocerebellar ataxia. Recent work has implicated TTBK2 in the regulation of cilia maintenance and function, but the underlying molecular mechanisms are not understood. METHODS To dissect the role of TTBK2 during cilia growth and maintenance in human cells, we examined disease-related TTBK2 truncations. We used biochemical approaches, proteomics, genetic engineering, and advanced microscopy techniques to unveil molecular events triggered by TTBK2. RESULTS We demonstrate that truncated TTBK2 protein moieties, unable to localize to the mother centriole, create unique semi-permissive conditions for cilia assembly, under which cilia begin to form but fail to elongate. Subsequently, we link the defects in cilia growth to aberrant turnover of a microtubule-depolymerizing kinesin KIF2A, which we find restrained by TTBK2 phosphorylation. CONCLUSIONS Together, our data imply that the regulation of KIF2A by TTBK2 represents an important mechanism governing cilia elongation and maintenance. Further, the requirement for concentrating TTBK2 activity to the mother centriole to initiate ciliogenesis can be under specific conditions bypassed, revealing TTBK2 recruitment-independent functions of its key partner, CEP164.
Collapse
Affiliation(s)
- David Benk Vysloužil
- Laboratory of Cilia and Centrosome Biology, Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 3, Brno, 62500, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, Brno, 62500, Czech Republic
| | - Ondřej Bernatík
- Laboratory of Cilia and Centrosome Biology, Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 3, Brno, 62500, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, Brno, 62500, Czech Republic
| | - Eva Lánská
- Institute of Biotechnology, Czech Academy of Sciences, BIOCEV, Průmyslová 595, Vestec, Prague, 252 50, Czech Republic
- Department of Cell Biology, Faculty of Science, Charles University, Viničná 7, Prague, 12800, Czech Republic
| | - Tereza Renzová
- Laboratory of Cilia and Centrosome Biology, Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 3, Brno, 62500, Czech Republic
| | - Lucia Binó
- Laboratory of Cilia and Centrosome Biology, Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 3, Brno, 62500, Czech Republic
| | - Andrea Lacigová
- Laboratory of Cilia and Centrosome Biology, Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 3, Brno, 62500, Czech Republic
| | - Tereza Drahošová
- Laboratory of Cilia and Centrosome Biology, Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 3, Brno, 62500, Czech Republic
| | - Zdeněk Lánský
- Institute of Biotechnology, Czech Academy of Sciences, BIOCEV, Průmyslová 595, Vestec, Prague, 252 50, Czech Republic
| | - Lukáš Čajánek
- Laboratory of Cilia and Centrosome Biology, Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Kamenice 3, Brno, 62500, Czech Republic.
- Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, Brno, 62500, Czech Republic.
| |
Collapse
|
25
|
Chen J, Li Q, Liu X, Lin F, Jing Y, Yang J, Zhao L. Potential biomarkers and immune infiltration linking endometriosis with recurrent pregnancy loss based on bioinformatics and machine learning. Front Mol Biosci 2025; 12:1529507. [PMID: 39963268 PMCID: PMC11830612 DOI: 10.3389/fmolb.2025.1529507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 01/20/2025] [Indexed: 02/20/2025] Open
Abstract
Objective Endometriosis (EMs) is a chronic inflammatory disease characterized by the presence of endometrial tissue in the non-uterine cavity, resulting in dysmenorrhea, pelvic pain, and infertility. Epidemiologic data have suggested the correlation between EMs and recurrent pregnancy loss (RPL), but the pathological mechanism is unclear. This study aims to investigate the potential biomarkers and immune infiltration in EMs and RPL, providing a basis for early detection and treatment of the two diseases. Methods Two RPL and six EMs transcriptomic datasets from the Gene Expression Omnibus (GEO) database were used for differential analysis via limma package, followed by weighted gene co-expression network analysis (WGCNA) for key modules screening. Protein-protein interaction (PPI) network and two machine learning algorithms were applied to identify the common core genes in both diseases. The diagnostic capabilities of the core genes were assessed by receiver operating characteristic (ROC) curves. Moreover, immune cell infiltration was estimated using CIBERSORTx, and the Cancer Genome Atlas (TCGA) database was employed to elucidate the role of key genes in endometrial carcinoma (EC). Results 26 common differentially expressed genes (DEGs) were screened in both diseases, three of which were identified as common core genes (MAN2A1, PAPSS1, RIBC2) through the combination of WGCNA, PPI network, and machine learning-based feature selection. The area under the curve (AUC) values generated by the ROC indicates excellent diagnostic powers in both EMs and RPL. The key genes were found to be significantly associated with the infiltration of several immune cells. Interestingly, MAN2A1 and RIBC2 may play a predominant role in the development and prognostic stratification of EC. Conclusion We identified three key genes linking EMs and RPL, emphasizing the heterogeneity of immune infiltration in the occurrence of both diseases. These findings may provide new mechanistic insights or therapeutic targets for further research of EMs and RPL.
Collapse
Affiliation(s)
- Jianhui Chen
- Prenatal Diagnosis Center, Center of Reproductive Medicine, Suining Central Hospital, Suining, Sichuan, China
| | - Qun Li
- Department of Radiology, Suining Central Hospital, Suining, Sichuan, China
| | - Xiaofang Liu
- Prenatal Diagnosis Center, Center of Reproductive Medicine, Suining Central Hospital, Suining, Sichuan, China
| | - Fang Lin
- Prenatal Diagnosis Center, Center of Reproductive Medicine, Suining Central Hospital, Suining, Sichuan, China
| | - Yaling Jing
- Prenatal Diagnosis Center, Center of Reproductive Medicine, Suining Central Hospital, Suining, Sichuan, China
| | - Jiayan Yang
- Prenatal Diagnosis Center, Center of Reproductive Medicine, Suining Central Hospital, Suining, Sichuan, China
| | - Lianfang Zhao
- Prenatal Diagnosis Center, Center of Reproductive Medicine, Suining Central Hospital, Suining, Sichuan, China
| |
Collapse
|
26
|
He K, Sun X, Chen C, Luc S, Robichaud JH, Zhang Y, Huang Y, Ji B, Ku PI, Subramanian R, Ling K, Hu J. Non-canonical CDK6 activity promotes cilia disassembly by suppressing axoneme polyglutamylation. J Cell Biol 2025; 224:e202405170. [PMID: 39636239 PMCID: PMC11619382 DOI: 10.1083/jcb.202405170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 10/02/2024] [Accepted: 11/01/2024] [Indexed: 12/07/2024] Open
Abstract
Tubulin polyglutamylation is a posttranslational modification that occurs primarily along the axoneme of cilia. Defective axoneme polyglutamylation impairs cilia function and has been correlated with ciliopathies, including Joubert Syndrome (JBTS). However, the precise mechanisms regulating proper axoneme polyglutamylation remain vague. Here, we show that cyclin-dependent kinase 6 (CDK6), but not its paralog CDK4, localizes to the cilia base and suppresses axoneme polyglutamylation by phosphorylating RAB11 family interacting protein 5 (FIP5) at site S641, a critical regulator of cilia import of glutamylases. S641 phosphorylation disrupts the ciliary recruitment of FIP5 and its association with RAB11, thereby reducing the ciliary import of glutamylases. Encouragingly, the FDA-approved CDK4/6 inhibitor Abemaciclib can effectively restore cilia function in JBTS cells with defective glutamylation. In summary, our study elucidates the regulatory mechanisms governing axoneme polyglutamylation and suggests that developing CDK6-specific inhibitors could be a promising therapeutic strategy to enhance cilia function in ciliopathy patients.
Collapse
Affiliation(s)
- Kai He
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Xiaobo Sun
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Chuan Chen
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - San Luc
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Jielu Hao Robichaud
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Yingyi Zhang
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Yan Huang
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Biyun Ji
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Pei-I Ku
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Radhika Subramanian
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Kun Ling
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Jinghua Hu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
- Mayo Clinic Robert M. and Billie Kelley Pirnie Translational Polycystic Kidney Disease Center, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
27
|
Takahashi Y, Ishida Y, Yoshida S, Shin HW, Katoh Y, Nakayama K. Counterregulatory roles of GLI2 and GLI3 in osteogenic differentiation via Gli1 expression. J Cell Sci 2025; 138:jcs263556. [PMID: 39801296 DOI: 10.1242/jcs.263556] [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: 09/20/2024] [Accepted: 12/31/2024] [Indexed: 02/11/2025] Open
Abstract
The GLI1, GLI2 and GLI3 transcription factors mediate Hedgehog (Hh) signaling, which is crucial for bone development. During intramembranous ossification, mesenchymal stem cells (MSCs) are directly differentiated into osteoblasts. Under basal and Hh pathway-stimulated conditions, primary cilia play essential roles in proteolytic processing of GLI3 to its repressor form (GLI3R) and in activation of GLI2. Although previous studies in mice have suggested that Gli1 expression depends on GLI2 and GLI3, coordinated roles of GLI1, GLI2 and GLI3 in osteogenic differentiation are not fully understood at the cellular level. From the MSC line C3H10T1/2, we established Gli2-knockout (KO) and Gli3-KO cells, as well as constitutively GLI3R-producing (cGLI3R) cells, and expressed GLI1, GLI2 and GLI3 constructs in these cell lines. The results demonstrate at the cellular level that GLI2 and GLI3R counterregulate osteogenic differentiation via activation and repression of Gli1 expression, respectively; GLI3R, which results from GLI3 processing requiring protein kinase A-mediated phosphorylation, downregulates expression of Gli2 as well as Gli1; and GLI1 upregulates expression of Gli1 itself and Gli2, constituting a GLI1-GLI2 positive feedback loop.
Collapse
Affiliation(s)
- Yuto Takahashi
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Yamato Ishida
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Saishu Yoshida
- Department of Biochemistry, The Jikei University School of Medicine, Tokyo 105-8461, Japan
| | - Hye-Won Shin
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Yohei Katoh
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Kazuhisa Nakayama
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| |
Collapse
|
28
|
Ojeda-Naharros I, Das T, Castro RA, Bazan JF, Vaisse C, Nachury MV. Tonic ubiquitination of the central body weight regulator melanocortin receptor 4 (MC4R) promotes its constitutive exit from cilia. PLoS Biol 2025; 23:e3003025. [PMID: 39899600 PMCID: PMC11825094 DOI: 10.1371/journal.pbio.3003025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 02/13/2025] [Accepted: 01/17/2025] [Indexed: 02/05/2025] Open
Abstract
The G protein-coupled receptor (GPCR) melanocortin receptor 4 (MC4R) is an essential regulator of body weight homeostasis. MC4R is unusual among GPCRs in that its activity is regulated by 2 opposing physiological ligands, the agonist ⍺-MSH and the antagonist/inverse agonist AgRP. Paradoxically, while MC4R localizes and functions at the cilium of hypothalamic neurons, the ciliary levels of MC4R are very low under unrestricted feeding conditions. Here, we find that the constitutive activity of MC4R is responsible for the continuous depletion of MC4R from cilia and that inhibition of MC4R's activity via AgRP leads to a robust accumulation of MC4R in cilia. Ciliary targeting of MC4R is mediated by its partner MRAP2 and the constitutive exit of MC4R from cilia relies on the sensor of activation β-arrestin, on ubiquitination, and on the BBSome ciliary trafficking complex. Thus, while MC4R exits cilia via conventional mechanisms, it only accumulates in cilia when its activity is suppressed by AgRP.
Collapse
Affiliation(s)
- Irene Ojeda-Naharros
- Department of Ophthalmology, University of California San Francisco, California, United States of America
- Cardiovascular Research Institute, University of California San Francisco, California, United States of America
| | - Tirthasree Das
- Department of Ophthalmology, University of California San Francisco, California, United States of America
- Cardiovascular Research Institute, University of California San Francisco, California, United States of America
| | - Ralph A. Castro
- Department of Ophthalmology, University of California San Francisco, California, United States of America
- Cardiovascular Research Institute, University of California San Francisco, California, United States of America
| | - J. Fernando Bazan
- Unit for Structural Biology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- ħ bioconsulting llc, Stillwater, Minnesota, United States of America
| | - Christian Vaisse
- Diabetes Center, University of California San Francisco; San Francisco, California, United States of America
| | - Maxence V. Nachury
- Department of Ophthalmology, University of California San Francisco, California, United States of America
- Cardiovascular Research Institute, University of California San Francisco, California, United States of America
| |
Collapse
|
29
|
Fan A, Zhang Y, Li Y, Meng W, Wu F, Pan W, Ma Z, Chen W. Primary Cilia Formation Mediated by Hsa_Circ_0005185/OTUB1/RAB8A Complex Inhibits Prostate Cancer Progression by Suppressing Hedgehog Signaling Pathway. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411675. [PMID: 39785769 PMCID: PMC11848605 DOI: 10.1002/advs.202411675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 12/26/2024] [Indexed: 01/12/2025]
Abstract
Prostate cancer (PCa) is one of the most common malignancies for male individuals globally. Androgen deprivation therapy (ADT) initially demonstrated significant efficacy in treating PCa; however, most cases of PCa eventually progress to castration-resistant prostate cancer (CRPC), which becomes increasingly challenging to manage. Notably, the loss or disruption of primary cilia in PCa cells may play a critical role in the progression of the disease, and there are no reports on the role of circular RNAs in ciliogenesis. Thus, this warrants further investigation.In this study, key circular RNAs linked to prostate cancer progression, and enzalutamide resistance is identified. Specifically, it is found that hsa_circ_0005185 interacts with OTUB1 and RAB8A, serving as a molecular scaffold. Hsa_circ_0005185 mediates the binding of the deubiquitinase OTUB1 to RAB8A, resulting in the deubiquitination of RAB8A. Consequently, the stable expression of RAB8A promotes the regeneration of primary cilia and enhances the production of GLI3R, an inhibitory factor in the Hedgehog signaling pathway, thereby suppressing AR activity and slowing the progression of CRPC.
Collapse
Affiliation(s)
- Aoyu Fan
- Department of UrologyZhongshan HospitalFudan UniversityShanghai200030China
| | - Yunyan Zhang
- Department of UrologyZhongshan HospitalFudan UniversityShanghai200030China
| | - Yunpeng Li
- Department of UrologyZhongshan HospitalFudan UniversityShanghai200030China
| | - Wei Meng
- Lab for Noncoding RNA and CancerSchool of Life SciencesShanghai UniversityShanghai200444China
| | - Fan Wu
- Lab for Noncoding RNA and CancerSchool of Life SciencesShanghai UniversityShanghai200444China
| | - Wei Pan
- Lab for Noncoding RNA and CancerSchool of Life SciencesShanghai UniversityShanghai200444China
| | - Zhongliang Ma
- Lab for Noncoding RNA and CancerSchool of Life SciencesShanghai UniversityShanghai200444China
| | - Wei Chen
- Department of UrologyZhongshan HospitalFudan UniversityShanghai200030China
| |
Collapse
|
30
|
He L, Xu H, Liu M, Tan Y, Huang S, Yin X, Luo X, Chung HY, Gao M, Li Y, Ding W, Zhou H, Huang Y. The ignored structure in female fertility: cilia in the fallopian tubes. Reprod Biomed Online 2025; 50:104346. [PMID: 39740369 DOI: 10.1016/j.rbmo.2024.104346] [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: 02/18/2024] [Revised: 06/12/2024] [Accepted: 06/18/2024] [Indexed: 01/02/2025]
Abstract
Cilia in the fallopian tubes (CFT) play an important role in female infertility, but have not been explored comprehensively. This review reveals the detection techniques for CFT function and morphology, and the related analysis of female infertility and other gynaecological disorders. CFT differentiate from progenitor cells, and develop into primary cilia and motile cilia. Primary cilia coordinate multiple signalling pathways, and motile cilia produce laminar flow through bidirectional intraflagellar transport, which drives the movement of oocytes and gametes. Several methods for quantitative detection and protein analysis have been used to explore the factors contributing to the decrease in ciliary beat frequency (CBF), and the cellular mechanism of ciliary cell death and shedding. In both primary and secondary ciliary disorders associated with reproductive diseases, abnormal alterations in ciliary quantity, ciliary structure, CBF and ciliary signalling pathways result in abnormal tubal laminar flow, and diminished oocyte retrieval and transport capabilities.
Collapse
Affiliation(s)
- Liuqing He
- Department of Gynaecology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Haofei Xu
- Department of Gynaecology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Min Liu
- Department of Gynaecology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ying Tan
- Department of Gynaecology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shiyu Huang
- Department of Gynaecology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaoxiao Yin
- Department of Gynaecology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xinyu Luo
- Department of Gynaecology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hui Yee Chung
- Department of Gynaecology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ming Gao
- Department of Gynaecology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yujie Li
- Department of Gynaecology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Weijun Ding
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hang Zhou
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yefang Huang
- Department of Gynaecology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
31
|
Mao S, Song R, Jin S, Pang S, Jovanovic A, Zimmerman A, Li P, Wu X, Wendland MF, Lin K, Chen WC, Choksi SP, Chen G, Holtzman MJ, Reiter JF, Wan Y, Xuan Z, Xiang YK, Xu CS, Upadhyayula S, Hess HF, He L. Multicilia dynamically transduce Shh signaling to regulate choroid plexus functions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.21.633415. [PMID: 39896593 PMCID: PMC11785054 DOI: 10.1101/2025.01.21.633415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Choroid plexus is a major site for cerebrospinal fluid (CSF) production, characterized by a multiciliated epithelial monolayer that regulates CSF production. We demonstrate that defective choroid plexus ciliogenesis or Intraflagellar transport yields neonatal hydrocephalus, at least in part, due to increased water channel Aqp1 and ion transporter Atp1a2 expression. We demonstrate choroid plexus multicilia as sensory cilia, transducing both canonical and non-canonical Shh signaling. Interestingly, it is the non-canonical Shh signaling that represses Aqp1 and Atp1a2 expression by Smo/Gαi/cAMP pathway. Choroid plexus multicilia exhibit unique ciliary ultrastructure, carrying features of both primary and motile cilia. Unlike most cilia that elongate during maturation, choroid plexus ciliary length decreases during development, causing a decline of Shh signaling intensity in developing choroid plexus, a derepression of Aqp1 and Atp1a2, and ultimately, an increased CSF production. Hence, developmental dynamics of choroid plexus multicilia dampens the Shh signaling intensity to promote CSF production.
Collapse
|
32
|
Chao YY, Lin RC, Su PJ, Wang CA, Tu TY, Hou YC, Tsai YT, Peng IC, Tsai SJ, Shan YS, Wang CY. Melanophilin-induced primary cilia promote pancreatic cancer metastasis. Cell Death Dis 2025; 16:22. [PMID: 39820281 PMCID: PMC11739566 DOI: 10.1038/s41419-025-07344-2] [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: 05/15/2024] [Revised: 12/11/2024] [Accepted: 01/08/2025] [Indexed: 01/30/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most malignant tumors because of its high metastatic ability. The glutamine (Gln)-deficient microenvironment contributes to PDAC metastasis; however, the underlying molecular mechanisms remain unclear. Here, we demonstrated that melanophilin (MLPH) promotes PDAC metastasis by inducing the regrowth of primary cilia. Using RNA sequencing, we found that MLPH was upregulated in Gln-deficient conditions. MLPH facilitated PDAC metastasis in vitro and in vivo. Clinically, high MLPH expression is positively correlated with metastasis and poor PDAC prognosis. MLPH localized to the centrosome and facilitated the regrowth of primary cilia. The primary ciliogenesis upregulated phospholipase C γ-1 (PLCG1) to promote PDAC metastasis. Interestingly, PLCG1 was localized to the primary cilia, and depletion of PLCG1 alleviated primary ciliogenesis, suggesting a feedforward role for PLCG1 in mediating primary ciliogenesis. Thus, our study revealed a novel function of the MLPH-primary cilia-PLCG1 axis in facilitating PDAC metastasis under Gln deficiency both in vitro and in vivo.
Collapse
Affiliation(s)
- Yu-Ying Chao
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ruei-Ci Lin
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ping-Jui Su
- Division of General Surgery, Department of Surgery, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Chu-An Wang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ting-Yuan Tu
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Ya-Chin Hou
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Tzui Tsai
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - I-Chen Peng
- Department of Life Sciences, National Cheng Kung University, Tainan, Taiwan
| | - Shaw-Jenq Tsai
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yan-Shen Shan
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Surgery, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chia-Yih Wang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
33
|
Qin H, Liang T, Zhang C, Wu J, Sheng X. The bidirectional relationship between cilia and PCP signaling pathway core protein Vangl2. Sci Prog 2025; 108:368504241311964. [PMID: 39819247 PMCID: PMC11748379 DOI: 10.1177/00368504241311964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Vangl2, a core component of the PCP signaling pathway, serves as a scaffold protein on the cell membrane, playing a crucial role in organizing protein complexes. Cilia, dynamic structures on the cell surface, carry out a wide range of functions. Research has highlighted a bidirectional regulatory interaction between Vangl2 and cilia, underscoring their interconnected roles in cellular processes. This relationship is demonstrated by the localization of Vangl2 at the base and proximal regions of cilia, where it plays essential roles in ciliary positioning, asymmetric distribution, and ciliogenesis. In contrast, the absence of cilia can disrupt Vangl2-mediated signal transduction processes. This review offers a narrative review of recent research on Vangl2's function in cilia and examines the regulatory effects of cilia on Vangl2-mediated signaling.
Collapse
Affiliation(s)
- Huanyong Qin
- Department of Biochemistry and Molecular Biology, School of Preclinical Medicine, Zunyi Medical University, Zunyi, Guizhou, China
| | - Ting Liang
- Department of Biochemistry and Molecular Biology, School of Preclinical Medicine, Zunyi Medical University, Zunyi, Guizhou, China
| | - Chuanfen Zhang
- Department of Biochemistry and Molecular Biology, School of Preclinical Medicine, Zunyi Medical University, Zunyi, Guizhou, China
| | - Junlin Wu
- Department of Biochemistry and Molecular Biology, School of Preclinical Medicine, Zunyi Medical University, Zunyi, Guizhou, China
| | - Xin Sheng
- Department of Biochemistry and Molecular Biology, School of Preclinical Medicine, Zunyi Medical University, Zunyi, Guizhou, China
| |
Collapse
|
34
|
Zhang X, Cao Y, Wang M, Li Y, Yin H, Ni H, Yang S, Yu F, Yang J, Peng L, Hu M, Li D, Liu D. Primary Cilia Regulate the Homeostasis and Regeneration of the Stem Cell Niche in the Tooth. J Cell Physiol 2025; 240:e31517. [PMID: 39734274 DOI: 10.1002/jcp.31517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 12/10/2024] [Accepted: 12/14/2024] [Indexed: 12/31/2024]
Abstract
Primary cilia, functioning as crucial hubs for signal sensing and transduction, are integral to the development and maintenance of homeostasis across various organs. However, their roles in tooth homeostasis and repair remain inadequately understood. In this study, we reveal an indispensable role for primary cilia in regulating the homeostasis and regeneration of teeth, primarily through the regulation of cell proliferation. Using cilium-deficient mice, we demonstrate that disruption of ciliary homeostasis leads to abnormal tooth morphology, stunted growth and notably impaired tooth repair. RNA sequencing reveals a dysregulation in genes associated with various biological processes such as cell proliferation, differentiation, and cycle regulation. Furthermore, we show that cilium-deficient mice display reduced cell proliferation. Our findings highlight a critical function for primary cilia in the regulation of tooth homeostasis and regeneration and have important implications for the development of tooth regeneration therapies.
Collapse
Affiliation(s)
- Xinming Zhang
- Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, Tianjin Medical University School of Stomatology, Tianjin Medical University, Tianjin, China
| | - Yuxin Cao
- Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, Tianjin Medical University School of Stomatology, Tianjin Medical University, Tianjin, China
| | - Mengge Wang
- Haihe Laboratory of Cell Ecosystem, Tianjin Medical University, Tianjin, China
| | - Yujia Li
- Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, Tianjin Medical University School of Stomatology, Tianjin Medical University, Tianjin, China
| | - Hanxiao Yin
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Hua Ni
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Song Yang
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Fan Yu
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Jia Yang
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Lisu Peng
- Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, Tianjin Medical University School of Stomatology, Tianjin Medical University, Tianjin, China
| | - Meilin Hu
- Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, Tianjin Medical University School of Stomatology, Tianjin Medical University, Tianjin, China
| | - Dengwen Li
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Dayong Liu
- Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, Tianjin Medical University School of Stomatology, Tianjin Medical University, Tianjin, China
- School and Hospital of Stomatology, Hebei Medical University & Hebei Key Laboratory of Stomatology & Hebei Clinical Research Center for Oral Diseases Shijiazhuang, Shijiazhuang, China
| |
Collapse
|
35
|
Zhu D, Pan Y, Yang Y, Wang S. Regulation of the Cilia as a Potential Treatment for Senescence and Tumors: A Review. J Cell Physiol 2025; 240:e31499. [PMID: 39660388 DOI: 10.1002/jcp.31499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 11/15/2024] [Accepted: 11/20/2024] [Indexed: 12/12/2024]
Abstract
Millions of people worldwide die from malignant tumors every year, and the current clinical treatment is still based on radiotherapy and chemotherapy. Immunotherapy-adjuvant chemotherapy is widely applied, yet resistance to various factors persists in the management of advanced malignancies. Recently researchers have gradually discovered that the integrity of primary cilia is closely related to many diseases. The phenotypic changes in primary cilia are found in some cases of progeria, tumorigenesis, and drug resistance. Primary cilia seem to mediate signaling during these diseases. Hedgehog inhibitors have emerged in recent years to treat tumors by controlling signaling proteins on primary cilia. There is evidence for the use of anti-tumor drugs to treat senescence-related disease. Considering the close relationship between aging and obesity, as well as the obesity is the phenotype of many ciliopathies. Therefore, we speculate that some anti-tumor or anti-aging drugs can treat ciliopathies. Additionally, there is evidence suggesting that anti-aging drugs for tumor treatment, in which the process may be mediated by cilia. This review elucidates for the first time that cilia may be involved in the regulation of senescence, metabolic, tumorigenesis, and tumor resistance and hypothesizes that cilia can be regulated to treat these diseases in the future.
Collapse
Affiliation(s)
- Danping Zhu
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yuqin Pan
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yong Yang
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Shukui Wang
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Collaborative Innovation Center on Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
36
|
Niehrs C, Da Silva F, Seidl C. Cilia as Wnt signaling organelles. Trends Cell Biol 2025; 35:24-32. [PMID: 38697898 DOI: 10.1016/j.tcb.2024.04.001] [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: 02/08/2024] [Revised: 04/03/2024] [Accepted: 04/08/2024] [Indexed: 05/05/2024]
Abstract
Cilia and Wnt signaling have a complex relationship, wherein Wnt regulates cilia and, conversely, cilia may affect Wnt signaling. Recently, it was shown that Wnt receptors are present in flagella, primary cilia, and multicilia, where they transmit an intraciliary signal that is independent of β-catenin. Intraciliary Wnt signaling promotes ciliogenesis, affecting male fertility, adipogenesis, and mucociliary clearance. Wnt also stimulates the beating of motile cilia, highlighting that these nanomotors, too, are chemosensory. Intraciliary Wnt signaling employs a Wnt-protein phosphatase 1 (PP1) signaling axis, involving the canonical Wnt pathway's inhibition of glycogen synthase kinase 3 (GSK3) to repress PP1 activity. Collectively, these findings support that cilia are Wnt signaling organelles, with implications for ciliopathies and cancer.
Collapse
Affiliation(s)
- Christof Niehrs
- Division of Molecular Embryology, DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany; Institute of Molecular Biology (IMB), 55128 Mainz, Germany.
| | - Fabio Da Silva
- Division of Molecular Embryology, DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany
| | - Carina Seidl
- Division of Molecular Embryology, DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany
| |
Collapse
|
37
|
Palicharla VR, Badgandi HB, Hwang SH, Legué E, Liem KF, Mukhopadhyay S. A defined tubby domain β-barrel surface region of TULP3 mediates ciliary trafficking of diverse cargoes. Mol Biol Cell 2025; 36:ar1. [PMID: 39565681 PMCID: PMC11742108 DOI: 10.1091/mbc.e24-09-0426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 11/05/2024] [Accepted: 11/12/2024] [Indexed: 11/22/2024] Open
Abstract
The primary cilium is a paradigmatic subcellular compartment at the nexus of numerous cellular and morphogenetic pathways. The tubby family protein TULP3 acts as an adapter of the intraflagellar transport complex A in transporting integral membrane and membrane-associated lipidated proteins into cilia. However, the mechanisms by which TULP3 coordinates ciliary transport of diverse cargoes is not well understood. Here, we provide molecular insights into TULP3-mediated ciliary cargo recognition. We screened for critical TULP3 residues by proximity biotinylation-mass spectrometry, structural analysis, and testing TULP3 variants in human patients with hepatorenal fibrocystic disease and spina bifida. The TULP3 residues we identified 1) were located on one side of the β-barrel of the tubby domain away from the phosphoinositide binding site, 2) mediated ciliary trafficking of lipidated and transmembrane cargoes, and 3) determined proximity with these cargoes in vivo without affecting ciliary localization, phosphoinositide binding or hydrodynamic properties of TULP3. Overall, these findings implicate a specific region of one of the surfaces of the TULP3 β-barrel in ciliary trafficking of diverse cargoes. This region overlooks the β-strands 8-12 of the β-barrel and is away from the membrane anchoring phosphoinositide binding site. Targeting the TULP3-cargo interactions could provide therapeutics in ciliary trafficking diseases.
Collapse
Affiliation(s)
- Vivek Reddy Palicharla
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Hemant B. Badgandi
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Sun-Hee Hwang
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Emilie Legué
- Vertebrate Developmental Biology Program, Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06520
| | - Karel F. Liem
- Vertebrate Developmental Biology Program, Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06520
| | - Saikat Mukhopadhyay
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| |
Collapse
|
38
|
Karuntu JS, Almushattat H, Nguyen XTA, Plomp AS, Wanders RJA, Hoyng CB, van Schooneveld MJ, Schalij-Delfos NE, Brands MM, Leroy BP, van Karnebeek CDM, Bergen AA, van Genderen MM, Boon CJF. Syndromic Retinitis Pigmentosa. Prog Retin Eye Res 2024:101324. [PMID: 39733931 DOI: 10.1016/j.preteyeres.2024.101324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 12/31/2024]
Abstract
Retinitis pigmentosa (RP) is a progressive inherited retinal dystrophy, characterized by the degeneration of photoreceptors, presenting as a rod-cone dystrophy. Approximately 20-30% of patients with RP also exhibit extra-ocular manifestations in the context of a syndrome. This manuscript discusses the broad spectrum of syndromes associated with RP, pathogenic mechanisms, clinical manifestations, differential diagnoses, clinical management approaches, and future perspectives. Given the diverse clinical and genetic landscape of syndromic RP, the diagnosis may be challenging. However, an accurate and timely diagnosis is essential for optimal clinical management, prognostication, and potential treatment. Broadly, the syndromes associated with RP can be categorized into ciliopathies, inherited metabolic disorders, mitochondrial disorders, and miscellaneous syndromes. Among the ciliopathies associated with RP, Usher syndrome and Bardet-Biedl syndrome are the most well-known. Less common ciliopathies include Cohen syndrome, Joubert syndrome, cranioectodermal dysplasia, asphyxiating thoracic dystrophy, Mainzer-Saldino syndrome, and RHYNS syndrome. Several inherited metabolic disorders can present with RP including Zellweger spectrum disorders, adult Refsum disease, α-methylacyl-CoA racemase deficiency, certain mucopolysaccharidoses, ataxia with vitamin E deficiency, abetalipoproteinemia, several neuronal ceroid lipofuscinoses, mevalonic aciduria, PKAN/HARP syndrome, PHARC syndrome, and methylmalonic acidaemia with homocystinuria type cobalamin (cbl) C disease. Due to the mitochondria's essential role in supplying continuous energy to the retina, disruption of mitochondrial function can lead to RP, as seen in Kearns-Sayre syndrome, NARP syndrome, primary coenzyme Q10 deficiency, SSBP1-associated disease, and long chain 3-hydroxyacyl-CoA dehydrogenase deficiency. Lastly, Cockayne syndrome and PERCHING syndrome can present with RP, but they do not fit the abovementioned hierarchy and are thus categorized as 'Miscellaneous'. Several first-in-human clinical trials are underway or in preparation for some of these syndromic forms of RP.
Collapse
Affiliation(s)
- Jessica S Karuntu
- Department of Ophthalmology, Leiden University Medical Center, Leiden, the Netherlands
| | - Hind Almushattat
- Department of Ophthalmology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Xuan-Thanh-An Nguyen
- Department of Ophthalmology, Leiden University Medical Center, Leiden, the Netherlands
| | - Astrid S Plomp
- Department of Human Genetics, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Reproduction & Development Institute, Amsterdam, the Netherlands
| | - Ronald J A Wanders
- Department of Paediatrics, Division of Metabolic Diseases, Amsterdam UMC location University of Amsterdam, Emma Children's Hospital, Amsterdam, The Netherlands; Laboratory Genetic Metabolic Diseases, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, the Netherlands
| | - Carel B Hoyng
- Department of Ophthalmology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Mary J van Schooneveld
- Department of Ophthalmology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | | | - Marion M Brands
- Amsterdam Reproduction & Development Institute, Amsterdam, the Netherlands; Department of Paediatrics, Division of Metabolic Diseases, Amsterdam UMC location University of Amsterdam, Emma Children's Hospital, Amsterdam, The Netherlands; Amsterdam Gastroenterology Endocrinology Metabolism, Inborn errors of metabolism, Amsterdam, The Netherlands
| | - Bart P Leroy
- Department of Ophthalmology & Center for Medical Genetics, Ghent University, Ghent, Belgium; Department of Head & Skin, Ghent University, Ghent, Belgium
| | - Clara D M van Karnebeek
- Department of Paediatrics, Division of Metabolic Diseases, Amsterdam UMC location University of Amsterdam, Emma Children's Hospital, Amsterdam, The Netherlands; Emma Center for Personalized Medicine, Departments of Pediatrics and Human Genetics, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Arthur A Bergen
- Department of Human Genetics, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands; Emma Center for Personalized Medicine, Departments of Pediatrics and Human Genetics, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Maria M van Genderen
- Department of Ophthalmology, University Medical Center Utrecht, Utrecht, the Netherlands; Diagnostic Center for Complex Visual Disorders, Zeist, the Netherlands
| | - Camiel J F Boon
- Department of Ophthalmology, Leiden University Medical Center, Leiden, the Netherlands; Department of Ophthalmology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
39
|
Constable S, Ott CM, Lemire AL, White K, Xun Y, Lim A, Lippincott-Schwartz J, Mukhopadhyay S. Permanent cilia loss during cerebellar granule cell neurogenesis involves withdrawal of cilia maintenance and centriole capping. Proc Natl Acad Sci U S A 2024; 121:e2408083121. [PMID: 39705308 DOI: 10.1073/pnas.2408083121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 11/09/2024] [Indexed: 12/22/2024] Open
Abstract
Brain neurons utilize the primary cilium as a privileged compartment to detect and respond to extracellular ligands such as Sonic hedgehog (SHH). However, cilia in cerebellar granule cell (GC) neurons disassemble during differentiation through ultrastructurally unique intermediates, a process we refer to as cilia deconstruction. In addition, mature neurons do not reciliate despite having docked centrioles. Here, we identify molecular changes that accompany cilia deconstruction and centriole docking in GC neurons. We used single cell transcriptomic and immunocytological analyses to compare the transcript levels and subcellular localization of proteins between progenitor, differentiating, and mature GCs. Differentiating GCs lacked transcripts for key activators of premitotic cilia resorption, indicating that cilia disassembly in differentiating cells is distinct from premitotic cilia resorption. Instead, during differentiation, transcripts of many genes required for cilia maintenance-specifically those encoding components of intraflagellar transport, pericentrosomal material, and centriolar satellites-decreased. The abundance of several corresponding proteins in and around cilia and centrosomes also decreased. These changes coincided with downregulation of SHH signaling prior to differentiation, even in a mutant with excessive SHH activation. Finally, mother centrioles in maturing granule neurons recruited the cap complex protein, CEP97. These data suggest that a global, developmentally programmed decrease in cilium maintenance in differentiating GCs mediates cilia deconstruction, while capping of docked mother centrioles prevents cilia regrowth and dysregulated SHH signaling. Our study provides mechanistic insights expanding our understanding of permanent cilia loss in multiple tissue-specific contexts.
Collapse
Affiliation(s)
- Sandii Constable
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Carolyn M Ott
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147
| | - Andrew L Lemire
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147
| | - Kevin White
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Yu Xun
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Amin Lim
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | | | - Saikat Mukhopadhyay
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| |
Collapse
|
40
|
Yamazaki S, Fujii T, Chiba S, Shin HW, Nakayama K, Katoh Y. TXNDC15, an ER-localized thioredoxin-like transmembrane protein, contributes to ciliary transition zone integrity. J Cell Sci 2024; 137:jcs262123. [PMID: 39679447 DOI: 10.1242/jcs.262123] [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: 03/18/2024] [Accepted: 11/11/2024] [Indexed: 12/17/2024] Open
Abstract
Primary cilia have specific proteins on their membrane to fulfill their sensory functions. Preservation of the specific protein composition of cilia relies on the barrier function of the transition zone (TZ) located at the ciliary base. Defects in cilia and the TZ cause ciliopathies, which have diverse clinical manifestations, including Meckel syndrome (MKS). Many of the proteins mutated in individuals with MKS are known to constitute the MKS module of the TZ. Although TXNDC15 (also known as MKS14) is a thioredoxin-related transmembrane protein that is localized mainly in the endoplasmic reticulum (ER) and is mutated in individuals with MKS, its role at the TZ or within cilia has not been characterized. Here, we show that TXNDC15-knockout cells have defects in MKS module assembly and in ciliary membrane protein localization. These defects in TXNDC15-knockout cells were not rescued by exogenous expression of any of the TXNDC15 constructs with MKS variations in the thioredoxin domain. Furthermore, TXNDC15 with mutations of two cysteine residues within the thioredoxin domain failed to rescue defects in TXNDC15-knockout cells, suggesting that TXNDC15 controls the TZ integrity from outside the TZ via its thioredoxin domain.
Collapse
Affiliation(s)
- Shingo Yamazaki
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Taiju Fujii
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Shuhei Chiba
- Laboratory of Molecular and Cellular Biology, Tohoku University, Aobayama, Sendai, Miyagi 980-8578, Japan
| | - Hye-Won Shin
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Kazuhisa Nakayama
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Yohei Katoh
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
41
|
Monteillet L, Perrot G, Evrard F, Miliano A, Silva M, Leblond A, Nguyen C, Terzi F, Mithieux G, Rajas F. Impaired Glucose Metabolism, Primary Cilium Defects, and Kidney Cystogenesis in Glycogen Storage Disease Type Ia. J Am Soc Nephrol 2024; 35:1639-1654. [PMID: 39141438 PMCID: PMC11617483 DOI: 10.1681/asn.0000000000000452] [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: 01/07/2024] [Accepted: 07/31/2024] [Indexed: 08/16/2024] Open
Abstract
Key Points Metabolism adaptations due to glucose-6 phosphate accumulation in glycogen storage disease type Ia kidneys, toward a Warburg-like metabolism, promoted cell proliferation. Metabolic perturbations directly affected primary cilium structure and cystogenesis in glycogen storage disease type Ia kidneys. Background Glycogen storage disease type Ia (GSDIa) is a rare metabolic disorder caused by mutations in the catalytic subunit of glucose-6 phosphatase (G6PC1). This leads to severe hypoglycemia, and most young patients with GSDIa develop CKD. The kidney pathology is characterized by the development of cysts, which typically occur at an advanced stage of CKD. Methods To elucidate the molecular mechanisms responsible for cyst formation, we characterized renal metabolism, molecular pathways involved in cell proliferation, and primary cilium integrity using mice in which G6pc1 was specifically deleted in the kidney from an in utero stage. Results GSDIa mice exhibited kidney fibrosis, high inflammation, and cyst formation, leading to kidney dysfunction. In addition, the loss of G6PC1 led to the ectopic accumulation of glycogen and lipids in the kidneys and a metabolic shift toward a Warburg-like metabolism. This metabolic adaptation was due to an excess of glucose-6 phosphate, which supports cell proliferation, driven by the mitogen-activated protein kinase/extracellular signal–regulated kinases and protein kinase B/mammalian target of rapamycin pathways. Treatment of GSDIa mice with rapamycin, a target of the mammalian target of rapamycin pathway, reduced cell proliferation and kidney damage. Our results also identified lipocalin 2 as a contributor to renal inflammation and an early biomarker of CKD progression in GSDIa mice. Its inactivation partially prevented kidney lesions in GSDIa. Importantly, primary cilium defects were observed in the kidneys of GSDIa mice. Conclusions Metabolic adaptations because of glucose-6 phosphate accumulation in GSDIa renal tubules, toward a Warburg-like metabolism, promoted cell proliferation and cyst formation in a similar manner to that observed in various cystic kidney diseases. This was associated with downregulation of primary cilium gene expression and, consequently, altered cilium morphology.
Collapse
Affiliation(s)
- Laure Monteillet
- Université Claude Bernard Lyon 1, INSERM, UMR_S1213, NUDICE, Villeurbanne, France
| | - Gwendoline Perrot
- Université Claude Bernard Lyon 1, INSERM, UMR_S1213, NUDICE, Villeurbanne, France
| | - Félicie Evrard
- Université Claude Bernard Lyon 1, INSERM, UMR_S1213, NUDICE, Villeurbanne, France
| | - Alexane Miliano
- Université Claude Bernard Lyon 1, INSERM, UMR_S1213, NUDICE, Villeurbanne, France
| | - Marine Silva
- Université Claude Bernard Lyon 1, INSERM, UMR_S1213, NUDICE, Villeurbanne, France
| | - Alicia Leblond
- Université Claude Bernard Lyon 1, INSERM, UMR_S1213, NUDICE, Villeurbanne, France
| | - Clément Nguyen
- Université de Paris Cité, INSERM U1151, CNRS UMR 8253, Institut Necker Enfants Malades, Département “Croissance et Signalisation,” Paris, France
| | - Fabiola Terzi
- Université de Paris Cité, INSERM U1151, CNRS UMR 8253, Institut Necker Enfants Malades, Département “Croissance et Signalisation,” Paris, France
| | - Gilles Mithieux
- Université Claude Bernard Lyon 1, INSERM, UMR_S1213, NUDICE, Villeurbanne, France
| | - Fabienne Rajas
- Université Claude Bernard Lyon 1, INSERM, UMR_S1213, NUDICE, Villeurbanne, France
| |
Collapse
|
42
|
del Rosario-Gilabert D, Valenzuela-Miralles A, Esquiva G. Advances in mechanotransduction and sonobiology: effects of audible acoustic waves and low-vibration stimulations on mammalian cells. Biophys Rev 2024; 16:783-812. [PMID: 39830129 PMCID: PMC11735818 DOI: 10.1007/s12551-024-01242-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 09/25/2024] [Indexed: 01/22/2025] Open
Abstract
In recent decades, research on mechanotransduction has advanced considerably, focusing on the effects of audible acoustic waves (AAWs) and low-vibration stimulation (LVS), which has propelled the field of sonobiology forward. Taken together, the current evidence demonstrates the influence of these biosignals on key cellular processes, such as growth, differentiation and migration in mammalian cells, emphasizing the determining role of specific physical parameters during stimulation, such as frequency, sound pressure level/amplitude and exposure time. These mechanical waves interact with various cellular elements, including ion channels, primary cilia, cell-cell adhesion receptors, cell-matrix and extracellular matrix proteins, and focal adhesion complexes. These components connect with the cytoskeletal fibre network, enabling the transmission of mechanical stimuli towards the nucleus. The nucleus, in turn, linked to the cytoskeleton via the linkers of the nucleoskeleton and cytoskeleton complex, acts as a mechanosensitive centre, not only responding to changes in cytoskeletal stiffness and nuclear tension but also regulating gene expression through the transcriptional co-activator YAP/TAZ and interactions between chromatin and the nuclear envelope. This intricate chain of mechanisms highlights the potential of sonobiology in various fields, including dentistry, regenerative medicine, tissue engineering and cancer research. However, progress in these fields requires the establishment of standardized measurement methodologies and biocompatible experimental setups to ensure the reproducibility of results.
Collapse
Affiliation(s)
- D. del Rosario-Gilabert
- Department of Optics, Pharmacology and Anatomy, University of Alicante, San Vicente del Raspeig, Spain
- Department of Physics, Systems Engineering and Signal Theory, University of Alicante, San Vicente del Raspeig, Spain
- Department of Computer Technology, University of Alicante, San Vicente del Raspeig, Spain
- Institute for Advanced Neuroscience of Barcelona (INAB), Barcelona, Spain
| | - A. Valenzuela-Miralles
- Department of Optics, Pharmacology and Anatomy, University of Alicante, San Vicente del Raspeig, Spain
| | - G. Esquiva
- Department of Optics, Pharmacology and Anatomy, University of Alicante, San Vicente del Raspeig, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain
| |
Collapse
|
43
|
Sun J, Yuan H, Yu Y, Li A, Zhao Z, Tang Y, Zheng F. Immunomodulatory potential of primary cilia in the skin. Front Immunol 2024; 15:1456875. [PMID: 39676858 PMCID: PMC11638010 DOI: 10.3389/fimmu.2024.1456875] [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: 06/29/2024] [Accepted: 11/15/2024] [Indexed: 12/17/2024] Open
Abstract
Primary cilia (PC) are essential signaling hubs for proper epithelial formation and the maintenance of skin homeostasis. Found on most cells in the human body, including skin cells, PC facilitate signal transduction that allows ciliated cells to interact with the immune system via multiple pathways, helping to maintain immune system homeostasis. PC can be altered by various microenvironmental stimuli to develop corresponding regulatory functions. Both PC and ciliary signaling pathways have been shown to be involved in the immune processes of various skin lesions. However, the mechanisms by which PC regulate cellular functions and maintain immune homeostasis in tissues are highly complex, and our understanding of them in the skin remains limited. In this paper, we discuss key ciliary signaling pathways and ciliated cells in the skin, with a focus on their immunomodulatory functions. We have compiled evidence from various cells, tissues and disease models to help explore the potential immunomodulatory effects of PC in the skin and their molecular mechanisms.
Collapse
Affiliation(s)
- Jingwei Sun
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Huimin Yuan
- Department of Dermatology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Yanru Yu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Aorou Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Zihe Zhao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yang Tang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Fengjie Zheng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
44
|
Procházková K, Uhlík J. Influence of Hypoxia on the Airway Epithelium. Physiol Res 2024; 73:S557. [PMID: 39589303 PMCID: PMC11627265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 06/26/2024] [Indexed: 11/27/2024] Open
Abstract
The necessity of oxygen for metabolic processes means that hypoxia can lead to serious cell and tissue damage. On the other hand, in some situations, hypoxia occurs under physiological conditions and serves as an important regulation factor. The airway epithelium is specific in that it gains oxygen not only from the blood supply but also directly from the luminal air. Many respiratory diseases are associated with airway obstruction or excessive mucus production thus leading to luminal hypoxia. The main goal of this review is to point out how the airway epithelium reacts to hypoxic conditions. Cells detect low oxygen levels using molecular mechanisms involving hypoxia-inducible factors (HIFs). In addition, the cells of the airway epithelium appear to overexpress HIFs in hypoxic conditions. HIFs then regulate many aspects of epithelial cell functions. The effects of hypoxia include secretory cell stimulation and hyperplasia, epithelial barrier changes, and ciliogenesis impairment. All the changes can impair mucociliary clearance, exacerbate infection, and promote inflammation leading to damage of airway epithelium and subsequent airway wall remodeling. The modulation of hypoxia regulatory mechanisms may be one of the strategies for the treatment of obstructive respiratory diseases or diseases with mucus hyperproduction. Keywords: Secretory cells, Motile cilia, Epithelial barrier, Oxygenation, Obstructive respiratory diseases.
Collapse
Affiliation(s)
- K Procházková
- Department of Histology and Embryology, 2nd Faculty of Medicine, Charles University, Prague, Czech Republic.
| | | |
Collapse
|
45
|
Wloga D, Joachimiak E, Osinka A, Ahmadi S, Majhi S. Motile Cilia in Female and Male Reproductive Tracts and Fertility. Cells 2024; 13:1974. [PMID: 39682722 PMCID: PMC11639810 DOI: 10.3390/cells13231974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Motile cilia are evolutionarily conserved organelles. In humans, multiciliated cells (MCCs), assembling several hundred motile cilia on their apical surface, are components of the monolayer epithelia lining lower and upper airways, brain ventricles, and parts of the reproductive tracts, the fallopian tube and uterus in females, and efferent ductules in males. The coordinated beating of cilia generates a force that enables a shift of the tubular fluid, particles, or cells along the surface of the ciliated epithelia. Uncoordinated or altered cilia motion or cilia immotility may result in subfertility or even infertility. Here, we summarize the current knowledge regarding the localization and function of MCCs in the human reproductive tracts, discuss how cilia and cilia beating-generated fluid flow directly and indirectly contribute to the processes in these organs, and how lack or improper functioning of cilia influence human fertility.
Collapse
Affiliation(s)
- Dorota Wloga
- Laboratory of Cytoskeleton and Cilia Biology, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland; (E.J.); (A.O.); (S.A.); (S.M.)
| | | | | | | | | |
Collapse
|
46
|
Hu Y, Peng L, Zhuo X, Yang C, Zhang Y. Hedgehog Signaling Pathway in Fibrosis and Targeted Therapies. Biomolecules 2024; 14:1485. [PMID: 39766192 PMCID: PMC11727624 DOI: 10.3390/biom14121485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/17/2024] [Accepted: 11/20/2024] [Indexed: 01/12/2025] Open
Abstract
Hedgehog (Hh) signaling is a well-established developmental pathway; it is crucial for early embryogenesis, cell differentiation, and damage-driven regeneration. It is being increasingly recognized that dysregulated Hh signaling is also involved in fibrotic diseases, which are characterized by excessive extracellular matrix deposition that compromises tissue architecture and function. As in-depth insights into the mechanisms of Hh signaling are obtained, its complex involvement in fibrosis is gradually being illuminated. Notably, some Hh-targeted inhibitors are currently under exploration in preclinical and clinical trials as a means to prevent fibrosis progression. In this review, we provide a concise overview of the biological mechanisms involved in Hh signaling. We summarize the latest advances in our understanding of the roles of Hh signaling in fibrogenesis across the liver, kidneys, airways, and lungs, as well as other tissues and organs, with an emphasis on both the shared features and, more critically, the distinct functional variations observed across these tissues and organs. We thus highlight the context dependence of Hh signaling, as well as discuss the current status and the challenges of Hh-targeted therapies for fibrosis.
Collapse
Affiliation(s)
- Yuchen Hu
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu 610041, China; (Y.H.); (L.P.); (X.Z.)
- Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Linrui Peng
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu 610041, China; (Y.H.); (L.P.); (X.Z.)
- Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xinyu Zhuo
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu 610041, China; (Y.H.); (L.P.); (X.Z.)
- Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chan Yang
- Division of Endocrinology and Metabolism, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu 610041, China;
| | - Yuwei Zhang
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu 610041, China; (Y.H.); (L.P.); (X.Z.)
- Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
47
|
Barbelanne M, Lu Y, Kumar K, Zhang X, Li C, Park K, Warner A, Xu XZS, Shaham S, Leroux MR. C. elegans PPEF-type phosphatase (Retinal degeneration C ortholog) functions in diverse classes of cilia to regulate nematode behaviors. Sci Rep 2024; 14:28347. [PMID: 39550471 PMCID: PMC11569196 DOI: 10.1038/s41598-024-79057-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 11/06/2024] [Indexed: 11/18/2024] Open
Abstract
Primary (non-motile) cilia represent structurally and functionally diverse organelles whose roles as specialized cellular antenna are central to animal cell signaling pathways, sensory physiology and development. An ever-growing number of ciliary proteins, including those found in vertebrate photoreceptors, have been uncovered and linked to human disorders termed ciliopathies. Here, we demonstrate that an evolutionarily-conserved PPEF-family serine-threonine phosphatase, not functionally linked to cilia in any organism but associated with rhabdomeric (non-ciliary) photoreceptor degeneration in the Drosophila rdgC (retinal degeneration C) mutant, is a bona fide ciliary protein in C. elegans. The nematode protein, PEF-1, depends on transition zone proteins, which make up a 'ciliary gate' in the proximal-most region of the cilium, for its compartmentalization within cilia. Animals lacking PEF-1 protein function display structural defects to several types of cilia, including potential degeneration of microtubules. They also exhibit anomalies to cilium-dependent behaviors, including impaired responses to chemical, temperature, light, and noxious CO2 stimuli. Lastly, we demonstrate that PEF-1 function depends on conserved myristoylation and palmitoylation signals. Collectively, our findings broaden the role of PPEF proteins to include cilia, and suggest that the poorly-characterized mammalian PPEF1 and PPEF2 orthologs may also have ciliary functions and thus represent ciliopathy candidates.
Collapse
Affiliation(s)
- Marine Barbelanne
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
- Centre for Cell Biology, Development, and Disease, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
| | - Yun Lu
- Laboratory of Developmental Genetics, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| | - Keerthana Kumar
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
- Centre for Cell Biology, Development, and Disease, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
| | - Xinxing Zhang
- Life Sciences Institute, Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Chunmei Li
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
- Centre for Cell Biology, Development, and Disease, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
| | - Kwangjin Park
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
- Centre for Cell Biology, Development, and Disease, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
| | - Adam Warner
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
- Centre for Cell Biology, Development, and Disease, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
| | - X Z Shawn Xu
- Life Sciences Institute, Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Shai Shaham
- Laboratory of Developmental Genetics, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| | - Michel R Leroux
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada.
- Centre for Cell Biology, Development, and Disease, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada.
| |
Collapse
|
48
|
Iwaya C, Suzuki A, Iwata J. Loss of Sc5d results in micrognathia due to a failure in osteoblast differentiation. J Adv Res 2024; 65:153-165. [PMID: 38086515 PMCID: PMC11519736 DOI: 10.1016/j.jare.2023.12.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 11/30/2023] [Accepted: 12/09/2023] [Indexed: 01/01/2024] Open
Abstract
INTRODUCTION Mutations in genes related to cholesterol metabolism, or maternal diet and health status, affect craniofacial bone formation. However, the precise role of intracellular cholesterol metabolism in craniofacial bone development remains unclear. OBJECTIVE The aim of this study is to determine how cholesterol metabolism aberrations affect craniofacial bone development. METHODS Mice with a deficiency in Sc5d, which encodes an enzyme involved in cholesterol synthesis, were analyzed with histology, micro computed tomography (microCT), and cellular and molecular biological methods. RESULTS Sc5d null mice exhibited mandible hypoplasia resulting from defects in osteoblast differentiation. The activation of the hedgehog and WNT/β-catenin signaling pathways, which induce expression of osteogenic genes Col1a1 and Spp1, was compromised in the mandible of Sc5d null mice due to a failure in the formation of the primary cilium, a cell surface structure that senses extracellular cues. Treatments with an inducer of hedgehog or WNT/β-catenin signaling or with simvastatin, a drug that restores abnormal cholesterol production, partially rescued the defects in osteoblast differentiation seen in Sc5d mutant cells. CONCLUSION Our results indicate that loss of Sc5d results in mandibular hypoplasia through defective primary cilia-mediated hedgehog and WNT/β-catenin signaling pathways.
Collapse
Affiliation(s)
- Chihiro Iwaya
- Department of Diagnostic & Biomedical Sciences, The University of Texas Health Science Center at Houston, School of Dentistry, Houston, TX 77054, USA; Center for Craniofacial Research, The University of Texas Health Science Center at Houston, School of Dentistry, Houston, TX 77054, USA
| | - Akiko Suzuki
- Department of Diagnostic & Biomedical Sciences, The University of Texas Health Science Center at Houston, School of Dentistry, Houston, TX 77054, USA; Center for Craniofacial Research, The University of Texas Health Science Center at Houston, School of Dentistry, Houston, TX 77054, USA
| | - Junichi Iwata
- Department of Diagnostic & Biomedical Sciences, The University of Texas Health Science Center at Houston, School of Dentistry, Houston, TX 77054, USA; Center for Craniofacial Research, The University of Texas Health Science Center at Houston, School of Dentistry, Houston, TX 77054, USA; MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA.
| |
Collapse
|
49
|
Maddirevula S, Shagrani M, Ji AR, Horne CR, Young SN, Mather LJ, Alqahtani M, McKerlie C, Wood G, Potter PK, Abdulwahab F, AlSheddi T, van der Woerd WL, van Gassen KLI, AlBogami D, Kumar K, Muhammad Akhtar AS, Binomar H, Almanea H, Faqeih E, Fuchs SA, Scott JW, Murphy JM, Alkuraya FS. Large-scale genomic investigation of pediatric cholestasis reveals a novel hepatorenal ciliopathy caused by PSKH1 mutations. Genet Med 2024; 26:101231. [PMID: 39132680 DOI: 10.1016/j.gim.2024.101231] [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: 02/10/2024] [Revised: 08/03/2024] [Accepted: 08/06/2024] [Indexed: 08/13/2024] Open
Abstract
PURPOSE Pediatric cholestasis is the phenotypic expression of clinically and genetically heterogeneous disorders of bile acid synthesis and flow. Although a growing number of monogenic causes of pediatric cholestasis have been identified, the majority of cases remain undiagnosed molecularly. METHODS In a cohort of 299 pediatric participants (279 families) with intrahepatic cholestasis, we performed exome sequencing as a first-tier diagnostic test. RESULTS A likely causal variant was identified in 135 families (48.56%). These comprise 135 families that harbor variants spanning 37 genes with established or tentative links to cholestasis. In addition, we propose a novel candidate gene (PSKH1) (HGNC:9529) in 4 families. PSKH1 was particularly compelling because of strong linkage in 3 consanguineous families who shared a novel hepatorenal ciliopathy phenotype. Two of the 4 families shared a founder homozygous variant, whereas the third and fourth had different homozygous variants in PSKH1. PSKH1 encodes a putative protein serine kinase of unknown function. Patient fibroblasts displayed abnormal cilia that are long and show abnormal transport. A homozygous Pskh1 mutant mouse faithfully recapitulated the human phenotype and displayed abnormally long cilia. The phenotype could be rationalized by the loss of catalytic activity observed for each recombinant PSKH1 variant using in vitro kinase assays. CONCLUSION Our results support the use of genomics in the workup of pediatric cholestasis and reveal PSKH1-related hepatorenal ciliopathy as a novel candidate monogenic form.
Collapse
Affiliation(s)
- Sateesh Maddirevula
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Mohammad Shagrani
- Pediatric Transplant Gastro & Hepatology, Organ Transplant Centre of Excellence, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia; College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Ae-Ri Ji
- Translational Medicine Research Program, The Hospital for Sick Children, Toronto, ON, Canada; The Centre for Phenogenomics, Toronto, ON, Canada
| | - Christopher R Horne
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia; Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Victoria, Australia
| | - Samuel N Young
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Lucy J Mather
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Mashael Alqahtani
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Colin McKerlie
- Translational Medicine Research Program, The Hospital for Sick Children, Toronto, ON, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Geoffrey Wood
- Department of Pathobiology, University of Guelph, Guelph, ON, Canada
| | - Paul K Potter
- Department of Biomedical Sciences, Faculty of Health and Life Sciences, Oxford Brookes University, Oxford, United Kingdom
| | - Firdous Abdulwahab
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Tarfa AlSheddi
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Wendy L van der Woerd
- Department of Pediatric Gastroenterology, Hepatology and Nutrition, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Koen L I van Gassen
- Department of Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Dalal AlBogami
- Pediatric Transplant Gastro & Hepatology, Organ Transplant Centre of Excellence, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia; College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Kishwer Kumar
- Pediatric Transplant Gastro & Hepatology, Organ Transplant Centre of Excellence, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia; College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Ali Syed Muhammad Akhtar
- Pediatric Transplant Gastro & Hepatology, Organ Transplant Centre of Excellence, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia; College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Hiba Binomar
- Pediatric Transplant Gastro & Hepatology, Organ Transplant Centre of Excellence, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia; College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Hadeel Almanea
- Department of Pathology and Laboratory Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Eissa Faqeih
- Section of Medical Genetics, Department of Pediatric Subspecialties, Children Specialized Hospital, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Sabine A Fuchs
- Department of Pediatric Gastroenterology, Hepatology and Nutrition, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
| | - John W Scott
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Victoria, Australia; The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia; St Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
| | - James M Murphy
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia; Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Victoria, Australia
| | - Fowzan S Alkuraya
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia.
| |
Collapse
|
50
|
Baird DA, Mubeen H, Doganli C, Miltenburg JB, Thomsen OK, Ali Z, Naveed T, Rehman AU, Baig SM, Christensen ST, Farooq M, Larsen LA. Rare homozygous cilia gene variants identified in consanguineous congenital heart disease patients. Hum Genet 2024; 143:1323-1339. [PMID: 39347817 PMCID: PMC11522069 DOI: 10.1007/s00439-024-02703-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 09/09/2024] [Indexed: 10/01/2024]
Abstract
Congenital heart defects (CHD) appear in almost one percent of live births. Asian countries have the highest birth prevalence of CHD in the world. Recessive genotypes may represent a CHD risk factor in Asian populations with a high degree of consanguineous marriages. Genetic analysis of consanguineous families may represent a relatively unexplored source for investigating CHD etiology. To obtain insight into the contribution of recessive genotypes in CHD we analysed a cohort of forty-nine Pakistani CHD probands, originating from consanguineous unions. The majority (82%) of patient's malformations were septal defects. We identified protein altering, rare homozygous variants (RHVs) in the patient's coding genome by whole exome sequencing. The patients had a median of seven damaging RHVs each, and our analysis revealed a total of 758 RHVs in 693 different genes. By prioritizing these genes based on variant severity, loss-of-function intolerance and specific expression in the developing heart, we identified a set of 23 candidate disease genes. These candidate genes were significantly enriched for genes known to cause heart defects in recessive mouse models (P < 2.4e-06). In addition, we found a significant enrichment of cilia genes in both the initial set of 693 genes (P < 5.4e-04) and the 23 candidate disease genes (P < 5.2e-04). Functional investigation of ADCY6 in cell- and zebrafish-models verified its role in heart development. Our results confirm a significant role for cilia genes in recessive forms of CHD and suggest important functions of cilia genes in cardiac septation.
Collapse
Affiliation(s)
- Daniel A Baird
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Hira Mubeen
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Allama Iqbal Road, Faisalabad, 38000, Pakistan
- Department of Biotechnology, University of Central Punjab, Lahore, Pakistan
| | - Canan Doganli
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | - Jasmijn B Miltenburg
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
| | | | - Zafar Ali
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark
- Centre for Biotechnology and Microbiology, University of Swat, Swat, Pakistan
| | - Tahir Naveed
- Rawalpindi Institute of Cardiology, Rawalpindi, Pakistan
| | | | - Shahid Mahmood Baig
- Faculty of Life Sciences, Health Services Academy, Islamabad, Pakistan
- Department of Biological and Biomedical Sciences, Aga Khan University, Karachi, Pakistan
| | | | - Muhammad Farooq
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Allama Iqbal Road, Faisalabad, 38000, Pakistan.
| | - Lars Allan Larsen
- Department of Cellular and Molecular Medicine, University of Copenhagen, Blegdamsvej 3, 2200, Copenhagen, Denmark.
| |
Collapse
|