1
|
Song T, Han X, Yin H, Zhao J, Ma M, Wen X, Liu C, Yue Y, Zhao H, Zhou J, Yang Y, Ran J, Liu M. HDAC6 deacetylates ENKD1 to regulate mitotic spindle behavior and corneal epithelial homeostasis. EMBO Rep 2025; 26:2597-2621. [PMID: 40155750 PMCID: PMC12116779 DOI: 10.1038/s44319-025-00438-0] [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: 07/24/2024] [Revised: 02/11/2025] [Accepted: 03/06/2025] [Indexed: 04/01/2025] Open
Abstract
Corneal diseases can cause severe visual impairment and even blindness, which have been linked to the interruption of corneal epithelial homeostasis. However, the underlying molecular mechanisms are largely unknown. In this study, by comparing the transcriptomes of keratoconus, bacterial keratitis, viral keratitis, and healthy corneas, we found a steady upregulation of histone deacetylase 6 (HDAC6) in corneal diseases. Consistently, a significant increase in HDAC6 was observed in mouse corneas with bacterial keratitis. Overexpression of HDAC6 in mice results in a significant thickening of the corneal epithelium. Mechanistic studies reveal that HDAC6 overexpression disrupts mitotic spindle orientation and positioning in corneal epithelial cells. Our data further show that HDAC6 deacetylates enkurin domain-containing protein 1 (ENKD1) at lysine 98 and thereby impedes its interaction with γ-tubulin, restraining the centrosomal localization of ENKD1 and its proper function in regulating mitotic spindle behavior. These findings uncover a pivotal role for HDAC6-mediated deacetylation of ENKD1 in the control of corneal epithelial homeostasis, providing potential therapeutic targets for treating corneal diseases.
Collapse
Affiliation(s)
- Ting Song
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, 250014, Jinan, China
| | - Xueqing Han
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, 250014, Jinan, China
| | - Hanxiao Yin
- Department of Genetics and Cell Biology, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, 300071, Tianjin, China
| | - Junkui Zhao
- Department of Genetics and Cell Biology, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, 300071, Tianjin, China
| | - Mingming Ma
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, 250014, Jinan, China
| | - Xiaonuan Wen
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, 250014, Jinan, China
| | - Chunli Liu
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, 250014, Jinan, China
| | - Yiyang Yue
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, 250014, Jinan, China
| | - Huijie Zhao
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, 250014, Jinan, China
| | - Jun Zhou
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, 250014, Jinan, China
- Department of Genetics and Cell Biology, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, 300071, Tianjin, China
| | - Yang Yang
- Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, China.
| | - Jie Ran
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, 250014, Jinan, China.
| | - Min Liu
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, 250014, Jinan, China.
- Laboratory of Tissue Homeostasis, Haihe Laboratory of Cell Ecosystem, 300462, Tianjin, China.
| |
Collapse
|
2
|
Li X, Li J, Zhou L, Li Y, Wu X, Chen J. MiR-760 protects against lipopolysaccharide-induced septic acute kidney injury by targeting ENKD1. Clin Exp Nephrol 2025:10.1007/s10157-025-02690-z. [PMID: 40304823 DOI: 10.1007/s10157-025-02690-z] [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: 12/09/2024] [Accepted: 04/22/2025] [Indexed: 05/02/2025]
Abstract
BACKGROUND Sepsis, a systemic inflammatory disease, often leads to severe complications, including septic acute kidney injury (AKI). Despite its prevalence, effective treatments for septic AKI remain elusive. MicroRNAs (miRNAs) are critical regulators of gene expression and play essential roles in sepsis pathogenesis. This study investigates the function and regulatory pathway of miR-760 in septic AKI. METHODS Septic AKI models were developed using lipopolysaccharide (LPS)-treated mice and HK-2 renal tubular epithelial cells. Renal pathology was assessed via H&E staining, while renal function was assessed through the measurement of blood urea nitrogen (BUN) and creatinine levels. Apoptosis, inflammation, and oxidative stress markers were analyzed using functional assays, ELISA, and flow cytometry. Bioinformatics analysis, RT-qPCR, and dual luciferase reporter assays were used to identify and validate miR-760's target, enkurin domain containing 1 (ENKD1). RESULTS Database analysis and experimental validation revealed markedly decreased level of miR-760-3p levels in LPS-induced septic AKI mice model. MiR-760-3p overexpression in vivo ameliorated renal damage, improved kidney function, and reduced tubular apoptosis, inflammation, and oxidative stress. In vitro, miR-760 mimics enhanced cell proliferation while inhibiting apoptosis, inflammation, and oxidative stress in LPS-stimulated HK-2 cells. Mechanistically, ENKD1 was identified as a direct target of miR-760. ENKD1 overexpression negated the protective effects of miR-760, exacerbating renal injury, apoptosis, and inflammatory responses. CONCLUSION This study demonstrates that miR-760 alleviates septic AKI by targeting ENKD1, reducing apoptosis, inflammation, and oxidative stress. The miR-760/ENKD1 axis offers a promising therapeutic avenue to address septic AKI.
Collapse
Affiliation(s)
- Xinxin Li
- Department of Emergency Intensive Care Medicine and Emergency Medicine, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, No. 166, Yulong West Road, Tinghu District, Yancheng, 224000, Jiangsu, China
| | - Jianchao Li
- Department of Emergency Intensive Care Medicine and Emergency Medicine, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, No. 166, Yulong West Road, Tinghu District, Yancheng, 224000, Jiangsu, China
| | - Liangliang Zhou
- Department of Emergency Intensive Care Medicine and Emergency Medicine, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, No. 166, Yulong West Road, Tinghu District, Yancheng, 224000, Jiangsu, China
| | - Yingbin Li
- Department of Emergency Intensive Care Medicine and Emergency Medicine, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, No. 166, Yulong West Road, Tinghu District, Yancheng, 224000, Jiangsu, China
| | - Xufeng Wu
- Department of Emergency Intensive Care Medicine and Emergency Medicine, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, No. 166, Yulong West Road, Tinghu District, Yancheng, 224000, Jiangsu, China
| | - Jianjun Chen
- Department of Emergency Intensive Care Medicine and Emergency Medicine, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, No. 166, Yulong West Road, Tinghu District, Yancheng, 224000, Jiangsu, China.
| |
Collapse
|
3
|
Zhang T, Wang Y, Nie X, Chen X, Jin Y, Sun L, Yang R, Wang J, Xu W, Song T, Xie W, Chen X, Li C, Zhou J, Wu S, Li Y, Li T. ENKD1 modulates innate immune responses through enhanced geranylgeranyl pyrophosphate synthase activity. Cell Rep 2025; 44:115397. [PMID: 40048432 DOI: 10.1016/j.celrep.2025.115397] [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: 08/27/2024] [Revised: 12/22/2024] [Accepted: 02/14/2025] [Indexed: 03/29/2025] Open
Abstract
Inflammation is a crucial element of immune responses, with pivotal roles in host defenses against pathogens. Comprehensive understanding of the molecular mechanisms underlying inflammation is imperative for developing effective strategies to combat infectious diseases. Here, we conducted a screening analysis and identified enkurin domain-containing protein 1 (ENKD1) as a promising regulator of inflammation. We observed that ENKD1 expression was significantly reduced on activation of multiple Toll-like receptor (TLR) molecules. Deletion of ENKD1 resulted in enhanced innate immune system activation and exacerbation of septic inflammation. Mechanistically, ENKD1 interacted with geranylgeranyl diphosphate synthase 1 (GGPS1) and modulated its enzymatic activity, thereby influencing geranylgeranyl diphosphate production. This interaction ultimately led to Ras-related C3 botulinum toxin substrate 1 (RAC1) inactivation and suppression of pro-inflammatory signaling pathways. Our findings establish ENKD1 as a critical regulator of innate immune cell activation, underscoring its significant role in septic inflammation.
Collapse
Affiliation(s)
- Tianyu Zhang
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250358, China
| | - Yixuan Wang
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250358, China
| | - Xiaotong Nie
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250358, China
| | - Xiangrong Chen
- Key Laboratory for Applied Technology of Sophisticated Analytical Instruments, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Science), Jinan, Shandong, China
| | - Yueyi Jin
- Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou 215123, China
| | - Lulu Sun
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250358, China
| | - Ruqian Yang
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250358, China
| | - Jie Wang
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250358, China
| | - Wenqing Xu
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250358, China
| | - Ting Song
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250358, China
| | - Wei Xie
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Xiangfeng Chen
- Key Laboratory for Applied Technology of Sophisticated Analytical Instruments, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Science), Jinan, Shandong, China
| | - Chaojun Li
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine and School of Medicine, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Jun Zhou
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250358, China; State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Cell Ecosystem, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Sijin Wu
- Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou 215123, China.
| | - Yan Li
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250358, China.
| | - Tianliang Li
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250358, China.
| |
Collapse
|
4
|
Lossius-Cott C, Annoh A, Bens M, Nietzsche S, Hoffmann B, Figge MT, Rauner M, Hofbauer LC, Müller JP. Oncogenic FLT3 internal tandem duplications (ITD) and CD45/PTPRC control osteoclast functions and bone microarchitecture. JBMR Plus 2025; 9:ziae173. [PMID: 39906260 PMCID: PMC11788565 DOI: 10.1093/jbmrpl/ziae173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 12/16/2024] [Accepted: 01/29/2025] [Indexed: 02/06/2025] Open
Abstract
Activating internal tandem duplications (ITD) in the juxtamembrane domain of receptor tyrosine kinase FLT3 occur frequently in patients with acute myeloid leukemia (AML). Constitutive active FLT3-ITD mutations induce aberrant signaling and promote leukemic cell transformation. Inactivation of the attenuating receptor protein tyrosine phosphatase CD45 (PTPRC) in FLT3-ITD mice resulted in the development of a severe hematopoietic phenotype with characteristics of AML. In addition, abnormal bone structures and ectopic bone formation were observed in these mice, suggesting a previously unknown role of FLT3 to control bone development and remodeling. While Ptprc knockout and Flt3-ITD mutant mice showed a largely normal bone microarchitecture, micro-CT analysis of femurs from Flt3-ITD Ptprc knockout mice revealed trabecularization of the cortical bone. This resulted in increased trabecular bone volume at the metaphysis, while the cortical bone at the diaphysis was thinner and less dense. In the metaphysis, severely reduced osteoclast and osteoblast numbers were observed. Reduced capacity of ex vivo differentiation of CD11b-positive bone marrow stem cells to mature osteoclast was accompanied by their abnormal morphology and reduced size. Transcriptome analysis revealed reduced expression of osteoclastogenic genes. Unexpectedly, cumulative resorption activity of osteoclasts was increased. Size and structure of resorption pits of differentiated osteoclasts remained similar to those observed in osteoclast cultures derived from control animals. Enhanced proliferation of cells in osteoclast cultures derived from FLT3-ITD-expressing mice was mediated by increased expression of STAT5 target genes. Transcriptome analysis of differentiated osteoclasts showed dysregulated signaling pathways influencing their differentiation as well as the coupling of bone resorption and formation. Taken together, inactivation of attenuating CD45 in mice expressing oncogenic FLT3-ITD resulted in marked abnormalities of the osteo-hematopoietic niche, which can be explained by aberrant STAT5 activation.
Collapse
Affiliation(s)
- Carolin Lossius-Cott
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), Jena University Hospital, 07745 Jena, Germany
| | - Akua Annoh
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), Jena University Hospital, 07745 Jena, Germany
- Medical Clinic 3, Division of Nephrology, University Dresden, 01307 Dresden, Germany
| | - Martin Bens
- Core Facility Next Generation Sequencing, Leibniz Institute on Aging—Fritz Lipmann Institute, 07745 Jena, Germany
| | - Sandor Nietzsche
- Center for Electron Microscopy, Jena University Hospital, 07743 Jena, Germany
| | - Bianca Hoffmann
- Applied Systems Biology, Leibniz Institute for Natural Products, Research and Infection Biology, Hans Knoell Institute, 07745 Jena, Germany
| | - Marc Thilo Figge
- Applied Systems Biology, Leibniz Institute for Natural Products, Research and Infection Biology, Hans Knoell Institute, 07745 Jena, Germany
- Faculty of Biological Sciences, Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Martina Rauner
- Department of Medicine III and Center for Healthy Aging, University Dresden, 01307 Dresden, Germany
| | - Lorenz C Hofbauer
- Department of Medicine III and Center for Healthy Aging, University Dresden, 01307 Dresden, Germany
| | - Jörg P Müller
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), Jena University Hospital, 07745 Jena, Germany
| |
Collapse
|
5
|
Yang S, Yu F, Yang M, Ni H, Bu W, Yin H, Yang J, Wang W, Zhai D, Wu X, Ma N, Li T, Hao H, Ran J, Song T, Li D, Yoshida S, Lu Q, Yang Y, Zhou J, Liu M. CYLD Maintains Retinal Homeostasis by Deubiquitinating ENKD1 and Promoting the Phagocytosis of Photoreceptor Outer Segments. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404067. [PMID: 39373352 PMCID: PMC11615780 DOI: 10.1002/advs.202404067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 09/13/2024] [Indexed: 10/08/2024]
Abstract
Phagocytosis of shed photoreceptor outer segments by the retinal pigment epithelium (RPE) is essential for retinal homeostasis. Dysregulation of the phagocytotic process is associated with irreversible retinal degenerative diseases. However, the molecular mechanisms underlying the phagocytic activity of RPE cells remain elusive. In an effort to uncover proteins orchestrating retinal function, the cylindromatosis (CYLD) deubiquitinase is identified as a critical regulator of photoreceptor outer segment phagocytosis. CYLD-deficient mice exhibit abnormal retinal structure and function. Mechanistically, CYLD interacts with enkurin domain containing protein 1 (ENKD1) and deubiquitinates ENKD1 at lysine residues K141 and K242. Deubiquitinated ENKD1 interacts with Ezrin, a membrane-cytoskeleton linker, and stimulates the microvillar localization of Ezrin, which is essential for the phagocytic activity of RPE cells. These findings thus reveal a crucial role for the CYLD-ENKD1-Ezrin axis in regulating retinal homeostasis and may have important implications for the prevention and treatment of retinal degenerative diseases.
Collapse
Affiliation(s)
- Song Yang
- Department of Genetics and Cell BiologyCollege of Life SciencesState Key Laboratory of Medicinal Chemical BiologyHaihe Laboratory of Cell EcosystemNankai UniversityTianjin300071China
- School of Health and Life SciencesQingdao Central HospitalUniversity of Health and Rehabilitation SciencesQingdao266113China
| | - Fan Yu
- Department of Genetics and Cell BiologyCollege of Life SciencesState Key Laboratory of Medicinal Chemical BiologyHaihe Laboratory of Cell EcosystemNankai UniversityTianjin300071China
- School of Health and Life SciencesQingdao Central HospitalUniversity of Health and Rehabilitation SciencesQingdao266113China
| | - Mulin Yang
- Department of Genetics and Cell BiologyCollege of Life SciencesState Key Laboratory of Medicinal Chemical BiologyHaihe Laboratory of Cell EcosystemNankai UniversityTianjin300071China
| | - Hua Ni
- Department of Genetics and Cell BiologyCollege of Life SciencesState Key Laboratory of Medicinal Chemical BiologyHaihe Laboratory of Cell EcosystemNankai UniversityTianjin300071China
| | - Weiwen Bu
- Department of Genetics and Cell BiologyCollege of Life SciencesState Key Laboratory of Medicinal Chemical BiologyHaihe Laboratory of Cell EcosystemNankai UniversityTianjin300071China
| | - Hanxiao Yin
- Department of Genetics and Cell BiologyCollege of Life SciencesState Key Laboratory of Medicinal Chemical BiologyHaihe Laboratory of Cell EcosystemNankai UniversityTianjin300071China
| | - Jia Yang
- Department of Genetics and Cell BiologyCollege of Life SciencesState Key Laboratory of Medicinal Chemical BiologyHaihe Laboratory of Cell EcosystemNankai UniversityTianjin300071China
| | - Weishu Wang
- Department of Genetics and Cell BiologyCollege of Life SciencesState Key Laboratory of Medicinal Chemical BiologyHaihe Laboratory of Cell EcosystemNankai UniversityTianjin300071China
| | - Denghui Zhai
- Department of Genetics and Cell BiologyCollege of Life SciencesState Key Laboratory of Medicinal Chemical BiologyHaihe Laboratory of Cell EcosystemNankai UniversityTianjin300071China
| | - Xuemei Wu
- Department of Genetics and Cell BiologyCollege of Life SciencesState Key Laboratory of Medicinal Chemical BiologyHaihe Laboratory of Cell EcosystemNankai UniversityTianjin300071China
| | - Nan Ma
- Department of Genetics and Cell BiologyCollege of Life SciencesState Key Laboratory of Medicinal Chemical BiologyHaihe Laboratory of Cell EcosystemNankai UniversityTianjin300071China
| | - Te Li
- Department of Genetics and Cell BiologyCollege of Life SciencesState Key Laboratory of Medicinal Chemical BiologyHaihe Laboratory of Cell EcosystemNankai UniversityTianjin300071China
| | - Huijie Hao
- Department of Genetics and Cell BiologyCollege of Life SciencesState Key Laboratory of Medicinal Chemical BiologyHaihe Laboratory of Cell EcosystemNankai UniversityTianjin300071China
| | - Jie Ran
- Center for Cell Structure and FunctionShandong Provincial Key Laboratory of Animal Resistance BiologyCollege of Life SciencesShandong Normal UniversityJinan250014China
| | - Ting Song
- Center for Cell Structure and FunctionShandong Provincial Key Laboratory of Animal Resistance BiologyCollege of Life SciencesShandong Normal UniversityJinan250014China
| | - Dengwen Li
- Department of Genetics and Cell BiologyCollege of Life SciencesState Key Laboratory of Medicinal Chemical BiologyHaihe Laboratory of Cell EcosystemNankai UniversityTianjin300071China
| | - Sei Yoshida
- Department of Genetics and Cell BiologyCollege of Life SciencesState Key Laboratory of Medicinal Chemical BiologyHaihe Laboratory of Cell EcosystemNankai UniversityTianjin300071China
| | - Quanlong Lu
- Department of Genetics and Cell BiologyCollege of Life SciencesState Key Laboratory of Medicinal Chemical BiologyHaihe Laboratory of Cell EcosystemNankai UniversityTianjin300071China
| | - Yunfan Yang
- Department of Cell BiologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinan250012China
| | - Jun Zhou
- Department of Genetics and Cell BiologyCollege of Life SciencesState Key Laboratory of Medicinal Chemical BiologyHaihe Laboratory of Cell EcosystemNankai UniversityTianjin300071China
- Center for Cell Structure and FunctionShandong Provincial Key Laboratory of Animal Resistance BiologyCollege of Life SciencesShandong Normal UniversityJinan250014China
| | - Min Liu
- Laboratory of Tissue HomeostasisHaihe Laboratory of Cell EcosystemTianjin300462China
| |
Collapse
|
6
|
Sun S, Yang Y, Zhou J, Liu P. Liquid-liquid phase separation of microtubule-binding proteins in the regulation of spindle assembly. Cell Prolif 2024; 57:e13649. [PMID: 38736355 PMCID: PMC11471393 DOI: 10.1111/cpr.13649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 03/19/2024] [Accepted: 04/15/2024] [Indexed: 05/14/2024] Open
Abstract
Cell division is a highly regulated process essential for the accurate segregation of chromosomes. Central to this process is the assembly of a bipolar mitotic spindle, a highly dynamic microtubule (MT)-based structure responsible for chromosome movement. The nucleation and dynamics of MTs are intricately regulated by MT-binding proteins. Over the recent years, various MT-binding proteins have been reported to undergo liquid-liquid phase separation, forming either single- or multi-component condensates on MTs. Herein, we provide a comprehensive summary of the phase separation characteristics of these proteins. We underscore their critical roles in MT nucleation, spindle assembly and kinetochore-MT attachment during the cell division process. Furthermore, we discuss the current challenges and various remaining unsolved problems, highlights the ongoing research efforts aimed at a deeper understanding of the role of the phase separation process during spindle assembly and orientation. Our review aims to contribute to the collective knowledge in this area and stimulate further investigations that will enhance our comprehension of the intricate mechanisms governing cell division.
Collapse
Affiliation(s)
- Shuang Sun
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, College of Life SciencesShandong Normal UniversityJinanChina
| | - Yang Yang
- Translational Medicine CenterThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Jun Zhou
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, College of Life SciencesShandong Normal UniversityJinanChina
- State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Cell Ecosystem, College of Life SciencesNankai UniversityTianjinChina
| | - Peiwei Liu
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, College of Life SciencesShandong Normal UniversityJinanChina
| |
Collapse
|
7
|
Belay S, Belay G, Nigussie H, Ahbara AM, Tijjani A, Dessie T, Tarekegn GM, Jian-Lin H, Mor S, Woldekiros HS, Dobney K, Lebrasseur O, Hanotte O, Mwacharo JM. Anthropogenic events and responses to environmental stress are shaping the genomes of Ethiopian indigenous goats. Sci Rep 2024; 14:14908. [PMID: 38942813 PMCID: PMC11213886 DOI: 10.1038/s41598-024-65303-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: 04/12/2023] [Accepted: 06/19/2024] [Indexed: 06/30/2024] Open
Abstract
Anthropological and biophysical processes have shaped livestock genomes over Millenia and can explain their current geographic distribution and genetic divergence. We analyzed 57 Ethiopian indigenous domestic goat genomes alongside 67 equivalents of east, west, and north-west African, European, South Asian, Middle East, and wild Bezoar goats. Cluster, ADMIXTURE (K = 4) and phylogenetic analysis revealed four genetic groups comprising African, European, South Asian, and wild Bezoar goats. The Middle Eastern goats had an admixed genome of these four genetic groups. At K = 5, the West African Dwarf and Moroccan goats were separated from East African goats demonstrating a likely historical legacy of goat arrival and dispersal into Africa via the coastal Mediterranean Sea and the Horn of Africa. FST, XP-EHH, and Hp analysis revealed signatures of selection in Ethiopian goats overlaying genes for thermo-sensitivity, oxidative stress response, high-altitude hypoxic adaptation, reproductive fitness, pathogen defence, immunity, pigmentation, DNA repair, modulation of renal function and integrated fluid and electrolyte homeostasis. Notable examples include TRPV1 (a nociception gene); PTPMT1 (a critical hypoxia survival gene); RETREG (a regulator of reticulophagy during starvation), and WNK4 (a molecular switch for osmoregulation). These results suggest that human-mediated translocations and adaptation to contrasting environments are shaping indigenous African goat genomes.
Collapse
Affiliation(s)
- Shumuye Belay
- Tigray Agricultural Research Institute, Mekelle, Ethiopia.
- Department of Microbial, Cellular and Molecular Biology, Addis Ababa University, Addis Ababa, Ethiopia.
- International Livestock Research Institute (ILRI), Addis Ababa, Ethiopia.
| | - Gurja Belay
- Department of Microbial, Cellular and Molecular Biology, Addis Ababa University, Addis Ababa, Ethiopia
| | - Helen Nigussie
- Department of Microbial, Cellular and Molecular Biology, Addis Ababa University, Addis Ababa, Ethiopia
| | - Abulgasim M Ahbara
- Animal and Veterinary Sciences, Scotland's Rural College (SRUC), Roslin Institute Building, Midlothian, UK
- Department of Zoology, Misurata University, Misurata, Libya
| | - Abdulfatai Tijjani
- The Jackson Laboratory, Bar Harbor, ME, 04609, USA
- International Livestock Research Institute (ILRI), Addis Ababa, Ethiopia
| | - Tadelle Dessie
- International Livestock Research Institute (ILRI), Addis Ababa, Ethiopia
| | - Getinet M Tarekegn
- Animal and Veterinary Sciences, Scotland's Rural College (SRUC), Roslin Institute Building, Midlothian, UK
- Institute of Biotechnology (IoB), Addis Ababa University, Addis Ababa, Ethiopia
| | - Han Jian-Lin
- CAAS-ILRI Joint Laboratory on Livestock and Forage Genetic Resources, Beijing, China
- Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - Siobhan Mor
- International Livestock Research Institute (ILRI), Addis Ababa, Ethiopia
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Helina S Woldekiros
- Department of Anthropology, Washington University in St. Louis, St. Louis, USA
| | - Keith Dobney
- Department of Archaeology, Classics and Egyptology, University of Liverpool, Liverpool, UK
- University of Sydney, Sydney, Australia
| | - Ophelie Lebrasseur
- Palaeogenomics and Bioarchaeology Research Network, School of Archaeology, University of Oxford, Oxford, UK
| | - Olivier Hanotte
- International Livestock Research Institute (ILRI), Addis Ababa, Ethiopia
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Joram M Mwacharo
- Animal and Veterinary Sciences, Scotland's Rural College (SRUC), Roslin Institute Building, Midlothian, UK.
- Small Ruminant Genomics, International Centre for Agricultural Research in the Dry Areas (ICARDA), Addis Ababa, Ethiopia.
| |
Collapse
|
8
|
Zhang R, Xie S, Ran J, Li T. Restraining the power of Proteolysis Targeting Chimeras in the cage: A necessary and important refinement for therapeutic safety. J Cell Physiol 2024; 239:e31255. [PMID: 38501341 DOI: 10.1002/jcp.31255] [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: 01/22/2024] [Revised: 03/02/2024] [Accepted: 03/06/2024] [Indexed: 03/20/2024]
Abstract
Proteolysis Targeting Chimeras (PROTACs) represent a significant advancement in therapeutic drug development by leveraging the ubiquitin-proteasome system to enable targeted protein degradation, particularly impacting oncology. This review delves into the various types of PROTACs, such as peptide-based, nucleic acid-based, and small molecule PROTACs, each addressing distinct challenges in protein degradation. It also discusses innovative strategies like bridged PROTACs and conditional switch-activated PROTACs, offering precise targeting of previously "undruggable" proteins. The potential of PROTACs extends beyond oncology, with ongoing research and technological advancements needed to maximize their therapeutic potential. Future progress in this field relies on interdisciplinary collaboration and the integration of advanced computational tools to open new treatment avenues across various diseases.
Collapse
Affiliation(s)
- Renshuai Zhang
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Center for Cell Structure and Function, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Songbo Xie
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Center for Cell Structure and Function, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Jie Ran
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Center for Cell Structure and Function, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Te Li
- Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Medicinal Chemical Biology, Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, China
| |
Collapse
|
9
|
Zhai D, Li L, Chen C, Wang X, Liu R, Shan Y. INPP5E Regulates the Distribution of Phospholipids on Cilia in RPE1 Cells. J Clin Lab Anal 2024; 38:e25031. [PMID: 38514901 PMCID: PMC11033345 DOI: 10.1002/jcla.25031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 02/28/2024] [Accepted: 03/06/2024] [Indexed: 03/23/2024] Open
Abstract
BACKGROUND Primary cilia are static microtubule-based structures protruding from the cell surface and present on most vertebrate cells. The appropriate localization of phospholipids is essential for cilia formation and stability. INPP5E is a cilia-localized inositol 5-phosphatase; its deletion alters the phosphoinositide composition in the ciliary membrane, disrupting ciliary function. METHODS The EGFP-2xP4MSidM, PHPLCδ1-EGFP, and SMO-tRFP plasmids were constructed by the Gateway system to establish a stable RPE1 cell line. The INPP5E KO RPE1 cell line was constructed with the CRISPR/Cas9 system. The localization of INPP5E and the distribution of PI(4,5)P2 and PI4P were examined by immunofluorescence microscopy. The fluorescence intensity co-localized with cilia was quantified by ImageJ. RESULTS In RPE1 cells, PI4P is localized at the ciliary membrane, whereas PI(4,5)P2 is localized at the base of cilia. Knocking down or knocking out INPP5E alters this distribution, resulting in the distribution of PI(4,5)P2 along the ciliary membrane and the disappearance of PI4P from the cilia. Meanwhile, PI(4,5)P2 is located in the ciliary membrane labeled by SMO-tRFP. CONCLUSIONS INPP5E regulates the distribution of phosphoinositide on cilia. PI(4,5)P2 localizes at the ciliary membrane labeled with SMO-tRFP, indicating that ciliary pocket membrane contains PI(4,5)P2, and phosphoinositide composition in early membrane structures may differ from that in mature ciliary membrane.
Collapse
Affiliation(s)
- Denghui Zhai
- State Key Laboratory of Medicinal Chemical Biology, College of Life SciencesNankai UniversityTianjinChina
| | - Lamei Li
- State Key Laboratory of Medicinal Chemical Biology, College of Life SciencesNankai UniversityTianjinChina
| | - Cheng Chen
- State Key Laboratory of Medicinal Chemical Biology, College of Life SciencesNankai UniversityTianjinChina
| | - Xue Wang
- State Key Laboratory of Medicinal Chemical Biology, College of Life SciencesNankai UniversityTianjinChina
| | - Ruming Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Life SciencesNankai UniversityTianjinChina
| | - Ying Shan
- State Key Laboratory of Medicinal Chemical Biology, College of Life SciencesNankai UniversityTianjinChina
| |
Collapse
|
10
|
Bu W, Di J, Zhao J, Liu R, Wu Y, Ran J, Li T. Dynein Light Intermediate Chains Exhibit Different Arginine Methylation Patterns. J Clin Lab Anal 2024; 38:e25030. [PMID: 38525916 PMCID: PMC11033342 DOI: 10.1002/jcla.25030] [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: 01/29/2024] [Revised: 02/28/2024] [Accepted: 03/04/2024] [Indexed: 03/26/2024] Open
Abstract
BACKGROUND The motor protein dynein is integral to retrograde transport along microtubules and interacts with numerous cargoes through the recruitment of cargo-specific adaptor proteins. This interaction is mediated by dynein light intermediate chain subunits LIC1 (DYNC1LI1) and LIC2 (DYNC1LI2), which govern the adaptor binding and are present in distinct dynein complexes with overlapping and unique functions. METHODS Using bioinformatics, we analyzed the C-terminal domains (CTDs) of LIC1 and LIC2, revealing similar structural features but diverse post-translational modifications (PTMs). The methylation status of LIC2 and the proteins involved in this modification were examined through immunoprecipitation and immunoblotting analyses. The specific methylation sites on LIC2 were identified through a site-directed mutagenesis analysis, contributing to a deeper understanding of the regulatory mechanisms of the dynein complex. RESULTS We found that LIC2 is specifically methylated at the arginine 397 residue, a reaction that is catalyzed by protein arginine methyltransferase 1 (PRMT1). CONCLUSIONS The distinct PTMs of the LIC subunits offer a versatile mechanism for dynein to transport diverse cargoes efficiently. Understanding how these PTMs influence the functions of LIC2, and how they differ from LIC1, is crucial for elucidating the role of dynein-related transport pathways in a range of diseases. The discovery of the arginine 397 methylation site on LIC2 enhances our insight into the regulatory PTMs of dynein functions.
Collapse
Affiliation(s)
- Weiwen Bu
- Department of Genetics and Cell Biology, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Medicinal Chemical Biology, College of Life SciencesNankai UniversityTianjinChina
| | - Jie Di
- Department of Genetics and Cell Biology, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Medicinal Chemical Biology, College of Life SciencesNankai UniversityTianjinChina
| | - Junkui Zhao
- Department of Genetics and Cell Biology, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Medicinal Chemical Biology, College of Life SciencesNankai UniversityTianjinChina
| | - Ruming Liu
- Department of Genetics and Cell Biology, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Medicinal Chemical Biology, College of Life SciencesNankai UniversityTianjinChina
| | - Yue Wu
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life SciencesShandong Normal UniversityJinanChina
| | - Jie Ran
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life SciencesShandong Normal UniversityJinanChina
| | - Te Li
- Department of Genetics and Cell Biology, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Medicinal Chemical Biology, College of Life SciencesNankai UniversityTianjinChina
| |
Collapse
|
11
|
Song T, He N, Hao Z, Yang Y. Upregulation of ENKD1 disrupts cellular homeostasis to promote lymphoma development. J Cell Physiol 2023; 238:1308-1323. [PMID: 36960713 DOI: 10.1002/jcp.31012] [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: 02/03/2023] [Revised: 03/09/2023] [Accepted: 03/14/2023] [Indexed: 03/25/2023]
Abstract
Diffuse large B cell lymphoma (DLBCL) is a common and aggressive form of B cell lymphoma. Approximately 40% of DLBCL patients are incurable despite modern therapeutic approaches. To explore the molecular mechanisms driving the growth and progression of DLBCL, we analyzed genes with differential expression in DLBCL using the Gene Expression Profiling Interactive Analysis database. Enkurin domain-containing protein 1 (ENKD1), a centrosomal protein-encoding gene, was found to be highly expressed in DLBCL samples compared with normal samples. The phylogenetic analysis revealed that ENKD1 is evolutionarily conserved. Depletion of ENKD1 in cultured DLBCL cells induced apoptosis, suppressed cell proliferation, and blocked cell cycle progression in the G2/M phase. Moreover, ENKD1 expression positively correlates with the expression levels of a number of cellular homeostatic regulators, including Sperm-associated antigen 5, a gene encoding an important mitotic regulator. These findings thus demonstrate a critical function for ENKD1 in regulating the cellular homeostasis and suggest a potential value of targeting ENKD1 for the treatment of DLBCL.
Collapse
Affiliation(s)
- Ting Song
- Department of Cell Biology, School of Basic Medical Sciences, Cheeloo Medical College, Shandong University, Jinan, China
| | - Na He
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
| | - Ziqian Hao
- College of Artificial Intelligence and Big Data for Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Yunfan Yang
- Department of Cell Biology, School of Basic Medical Sciences, Cheeloo Medical College, Shandong University, Jinan, China
| |
Collapse
|
12
|
Zhou P, Yang G, Xie W. Organization of cortical microtubules in differentiated cells. J Cell Physiol 2023; 238:1141-1147. [PMID: 36960617 DOI: 10.1002/jcp.31011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/08/2023] [Accepted: 03/14/2023] [Indexed: 03/25/2023]
Abstract
The microtubule cytoskeleton plays a critical role in a variety of cellular activities, and its structures and functions have been extensively studied. However, little is known about cell differentiation-related microtubule remodeling, its regulatory mechanisms, and its physiological functions. Recent studies have shown that microtubule-binding proteins as well as cell junctions, such as desmosomes and adherens junctions, are involved in the remodeling of microtubules in response to cell differentiation. In addition, the microtubule-organizing activity and structural integrity of centrosomes undergo dramatic changes during cell differentiation to promote microtubule remodeling. Here we summarize recent advances revealing the dynamic changes in microtubule organization and functions during cell differentiation. We also highlight the molecular mechanisms underlying microtubule modeling in differentiated cells, focusing on the key roles played by microtubule-binding proteins, cell junctions, and centrosomes.
Collapse
Affiliation(s)
- Ping Zhou
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Guiwen Yang
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Wei Xie
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| |
Collapse
|
13
|
Yin H, Hu M, Li D. Regulation of epidermal stratification and development by basal keratinocytes. J Cell Physiol 2023; 238:742-748. [PMID: 36815398 DOI: 10.1002/jcp.30978] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 01/23/2023] [Accepted: 02/07/2023] [Indexed: 02/24/2023]
Abstract
The epidermis is a stratified squamous epithelium distributed in the outermost layer of the skin and is intimately involved in the formation of a physical barrier to pathogens. Basal keratinocytes possess the properties of stem cells and play an essential role in epidermal development and skin damage recovery. Therefore, understanding the molecular mechanism of how basal keratinocytes participate in epidermal development and stratification is vital for preventing and treating skin lesions. During epidermal morphogenesis, the symmetric division of basal keratinocytes contributes to the extension of skin tissues, while their asymmetric division and migration facilitate epidermal stratification. In this review, we summarize the process of epidermal stratification and illustrate the molecular mechanisms underlying epidermal morphogenesis. Furthermore, we discuss the coordination of multiple signaling pathways and transcription factors in epidermal stratification, together with the roles of cell polarity and cell dynamics during the process.
Collapse
Affiliation(s)
- Hanxiao Yin
- State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Cell Ecosystem, Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Mingzheng Hu
- State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Cell Ecosystem, Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Dengwen Li
- State Key Laboratory of Medicinal Chemical Biology, Haihe Laboratory of Cell Ecosystem, Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, China
| |
Collapse
|
14
|
Ge R, Cao M, Chen M, Liu M, Xie S. Cytoskeletal networks in primary cilia: Current knowledge and perspectives. J Cell Physiol 2022; 237:3975-3983. [PMID: 36000703 DOI: 10.1002/jcp.30865] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 07/08/2022] [Accepted: 08/11/2022] [Indexed: 11/12/2022]
Abstract
Primary cilia, microtubule-based protrusions present on the surface of most mammalian cells, function as sensory organelles that monitor extracellular signals and transduce them into intracellular biochemical responses. There is renewed research interest in primary cilia due to their essential roles in development, tissue homeostasis, and human diseases. Primary cilia dysfunction causes a large spectrum of human diseases, collectively known as ciliopathies. Despite significant advances in our understanding of primary cilia, there are still no effective agents for treating ciliopathies. Primary ciliogenesis is a highly ordered process involving membrane trafficking, basal body maturation, vesicle docking and fusion, transition zone assembly, and axoneme extension, in which actin and microtubule networks play critical and multiple roles. Actin and microtubule network architecture, isotropy, and dynamics are tightly controlled by cytoskeleton-associated proteins, a growing number of which are now recognized as responsible for cilium formation and maintenance. Here we summarize the roles of actin and microtubules and their associated proteins in primary ciliogenesis and maintenance. In doing so, we highlight that targeting cytoskeleton-associated proteins may be a promising therapeutic strategy for the treatment of ciliopathies.
Collapse
Affiliation(s)
- Ruixin Ge
- Department of Cell Biology, College of Life Sciences, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, Shandong Normal University, Jinan, China
| | - Minghui Cao
- Department of Cell Biology, College of Life Sciences, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, Shandong Normal University, Jinan, China
| | - Miao Chen
- Department of Bioscience, School of Life Sciences and Medicine, Shandong University of Technology, Zibo, China
| | - Min Liu
- Department of Cell Biology, College of Life Sciences, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, Shandong Normal University, Jinan, China
| | - Songbo Xie
- Department of Cell Biology, College of Life Sciences, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, Shandong Normal University, Jinan, China.,Department of Bioscience, School of Life Sciences and Medicine, Shandong University of Technology, Zibo, China
| |
Collapse
|