1
|
Li M, Böke E, Yang J. Centrosome-assisted assembly of the Balbiani body. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.11.637656. [PMID: 39990491 PMCID: PMC11844453 DOI: 10.1101/2025.02.11.637656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
The Balbiani body (Bb), which was discovered about 170 years ago, is a membraneless organelle in the oocyte in most species. In organisms like Xenopus and Zebrafish, Bb accumulates mitochondria, endoplasmic reticulum (ER), and germline determinants and regulates the proper localization of germline determinants. The Bb forms around the centrosome in the oocyte during early oogenesis. The mechanism behind its assembly has gained attention only very recently. Here, we report that overexpression of the germ plasm matrix protein Xvelo leads to the formation of a 'Bb-like' structure in somatic cells. The 'Bb-like' structure assembles around the centrosome and selectively recruits mitochondria, ER, and germline determinants. Taking advantage of this system, we investigated the roles of centrosome components on the assembly of Xvelo. Our results reveal that multiple components of the centrosome, including Sas6, Cenexin, and DZIP1, interact with Xvelo and promote its assembly, with Sas6 exhibiting the most prominent activity. Importantly, knocking down Sas6, Cenexin, and DZIP1 individually or in combination resulted in reduced Xvelo aggregates. Taken together, our work suggests that the centrosome may function as a nucleation center to promote the initiation of Xvelo assembly, resulting in the formation of the Bb around the centrosome.
Collapse
|
2
|
Zhang Y, Liu YJ, Mei J, Yang ZX, Qian XP, Huang W. An Analysis Regarding the Association Between DAZ Interacting Zinc Finger Protein 1 (DZIP1) and Colorectal Cancer (CRC). Mol Biotechnol 2025; 67:527-547. [PMID: 38334905 DOI: 10.1007/s12033-024-01065-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 12/21/2023] [Indexed: 02/10/2024]
Abstract
Colorectal cancer (CRC) is the third most common malignant disease worldwide, and its incidence is increasing, but the molecular mechanisms of this disease are highly heterogeneous and still far from being fully understood. Increasing evidence suggests that fibrosis mediated by abnormal activation of fibroblasts based in the microenvironment is associated with a poor prognosis. However, the function and pathogenic mechanisms of fibroblasts in CRC remain unclear. Here, combining scrna-seq and clinical specimen data, DAZ Interacting Protein 1 (DZIP1) was found to be expressed on fibroblasts and cancer cells and positively correlated with stromal deposition. Importantly, pseudotime-series analysis showed that DZIP1 levels were up-regulated in malignant transformation of fibroblasts and experimentally confirmed that DZIP1 modulates activation of fibroblasts and promotes epithelial-mesenchymal transition (EMT) in tumor cells. Further studies showed that DZIP1 expressed by tumor cells also has a driving effect on EMT and contributes to the recruitment of more fibroblasts. A similar phenomenon was observed in xenografted nude mice. And it was confirmed in xenograft mice that downregulation of DZIP1 expression significantly delayed tumor formation and reduced tumor size in CRC cells. Taken together, our findings suggested that DZIP1 was a regulator of the CRC mesenchymal phenotype. The revelation of targeting DZIP1 provides a new avenue for CRC therapy.
Collapse
Affiliation(s)
- Yu Zhang
- Comprehensive Cancer Center, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, 210008, Jiangsu, China
- Department of Medical Oncology, Affiliated Jinling Hospital, Medical School Nanjing University, Nanjing, 210029, Jiangsu, China
- Department of Oncology, Nanjing Tianyinshan Hospital, Nanjing, 211199, Jiangsu, China
| | - Yuan-Jie Liu
- Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China
| | - Jia Mei
- Department of Pathology, Affiliated Jinling Hospital, Medical School Nanjing University, Nanjing, 210029, Jiangsu, China
| | - Zhao-Xu Yang
- Department of Medical Oncology, Affiliated Jinling Hospital, Medical School Nanjing University, Nanjing, 210029, Jiangsu, China
| | - Xiao-Ping Qian
- Comprehensive Cancer Center, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, 210008, Jiangsu, China.
- Comprehensive Cancer Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Clinical Cancer Institute of Nanjing University, Nanjing, 210008, Jiangsu, China.
| | - Wei Huang
- Department of Oncology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Hanzhong Road No.155, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
3
|
Chattannavar G, Ger M, Balasubramanian J, Mandal S, Jalali S, Takkar B, Pisuchpen P, de Guimaraes TAC, Capasso JE, Kumar Padhy S, Levin AV. Bardet-Biedl syndrome with chorioretinal coloboma: a case series and review of literature. Ophthalmic Genet 2024; 45:616-622. [PMID: 39402987 DOI: 10.1080/13816810.2024.2411257] [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: 09/09/2024] [Accepted: 09/26/2024] [Indexed: 10/19/2024]
Abstract
INTRODUCTION Bardet-Biedl Syndrome (BBS) is a ciliopathy causing developmental defects and progressive retinal dystrophy, whereas choroidal coloboma is a developmental defect causing structural deficiency in the posterior retina. Both are rarely reported together. METHODS Here, we describe the phenotype and genotype of three unrelated patients with co-occurrence of Bardet-Biedl Syndrome and chorioretinal coloboma and review the pertinent literature. RESULTS We describe three unrelated patients, with variable clinical features of Bardet Biedl syndrome. None had family history of BBS or coloboma. Each carried biallelic variants in BBS1, BBS9 and TTC8 gene, respectively. Two had unilateral chorioretinal coloboma, while one had bilateral chorioretinal coloboma. DISCUSSION Although there may be other explanatory factors yet to be revealed, our data suggests that chorioretinal coloboma may be associated with BBS. The Hedgehog (Hh) signaling pathway, an intercellular communicator for development of the eye, is dependent on the primary cilia and plays a crucial role in the closure of the optic fissure. Both disorders therefore involve disruption of primary cilia function which may explain their association.
Collapse
Affiliation(s)
- Goura Chattannavar
- Flaum Eye Institute, Ocular Genetics, Golisano Children's Hospital, Rochester, New York, USA
- Jasti V. Ramanamma Children's Eye Care Centre, Child Sight Institute, L. V. Prasad Eye Institute, Hyderabad, India
| | - Marina Ger
- Anant Bajaj Retina Institute, L. V. Prasad eye Institute, Visakhapatnam, India
| | | | - Sohini Mandal
- Department of Ophthalmology, All India Institute of Medical Sciences, New Delhi, India
| | - Subhadra Jalali
- Jasti V. Ramanamma Children's Eye Care Centre, Child Sight Institute, L. V. Prasad Eye Institute, Hyderabad, India
- Srimati Kannuri Santhamma Centre for Vitreoretinal Diseases, Anant Bajaj Retina Institute, L. V. Prasad eye Institute, Hyderabad, India
| | - Brijesh Takkar
- Srimati Kannuri Santhamma Centre for Vitreoretinal Diseases, Anant Bajaj Retina Institute, L. V. Prasad eye Institute, Hyderabad, India
| | - Phattrawan Pisuchpen
- Department of Ophthalmology, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
- Division of Academic Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Thales A C de Guimaraes
- Institute of Ophthalmology, University College London, London, UK
- Department of Ophthalmology, Moorfields Eye Hospital NHS Foundation Trust, London, UK
| | - Jenina E Capasso
- Flaum Eye Institute, Ocular Genetics, Golisano Children's Hospital, Rochester, New York, USA
| | - Srikanta Kumar Padhy
- Vitreo-Retina, Anant Bajaj Retina Institute, L. V. Prasad Eye Institute, Bhubaneswar, India
| | - Alex V Levin
- Flaum Eye Institute, Ocular Genetics, Golisano Children's Hospital, Rochester, New York, USA
| |
Collapse
|
4
|
Turgeon A, Fu J, Divyanshi, Ma M, Jin Z, Hwang H, Li M, Qiao H, Mei W, Yang J. Dzip1 is dynamically expressed in the vertebrate germline and regulates the development of Xenopus primordial germ cells. Dev Biol 2024; 514:28-36. [PMID: 38880277 PMCID: PMC11934228 DOI: 10.1016/j.ydbio.2024.06.003] [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: 04/19/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 06/18/2024]
Abstract
Primordial germ cells (PGCs) are the precursors of sperms and oocytes. Proper development of PGCs is crucial for the survival of the species. In many organisms, factors responsible for PGC development are synthesized during early oogenesis and assembled into the germ plasm. During early embryonic development, germ plasm is inherited by a few cells, leading to the formation of PGCs. While germline development has been extensively studied, how components of the germ plasm regulate PGC development is not fully understood. Here, we report that Dzip1 is dynamically expressed in vertebrate germline and is a novel component of the germ plasm in Xenopus and zebrafish. Knockdown of Dzip1 impairs PGC development in Xenopus embryos. At the molecular level, Dzip1 physically interacts with Dazl, an evolutionarily conserved RNA-binding protein that plays a multifaced role during germline development. We further showed that the sequence between amino acid residues 282 and 550 of Dzip1 is responsible for binding to Dazl. Disruption of the binding between Dzip1 and Dazl leads to defective PGC development. Taken together, our results presented here demonstrate that Dzip1 is dynamically expressed in the vertebrate germline and plays a novel function during Xenopus PGC development.
Collapse
Affiliation(s)
- Aurora Turgeon
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Jia Fu
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Divyanshi
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Meng Ma
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Zhigang Jin
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Hyojeong Hwang
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Department of Biochemistry and Biophysics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Meining Li
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Huanyu Qiao
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Wenyan Mei
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Jing Yang
- Department of Comparative Biosciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
5
|
Pyatnitskiy MA, Poverennaya EV. Transcript-Level Biomarkers of Early Lung Carcinogenesis in Bronchial Lesions. Cancers (Basel) 2024; 16:2260. [PMID: 38927965 PMCID: PMC11202239 DOI: 10.3390/cancers16122260] [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: 05/10/2024] [Revised: 06/09/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Premalignant lesions within the bronchial epithelium signify the initial phases of squamous cell lung carcinoma, posing challenges for detection via conventional methods. Instead of focusing solely on gene expression, in this study, we explore transcriptomic alterations linked to lesion progression, with an emphasis on protein-coding transcripts. We reanalyzed a publicly available RNA-Seq dataset on airway epithelial cells from 82 smokers with and without premalignant lesions. Transcript and gene abundance were quantified using kallisto, while differential expression and transcript usage analysis was performed utilizing sleuth and RATs packages. Functional characterization involved overrepresentation analysis via clusterProfiler, weighted coexpression network analysis (WGCNA), and network analysis via Enrichr-KG. We detected 5906 differentially expressed transcripts and 4626 genes, exhibiting significant enrichment within pathways associated with oxidative phosphorylation and mitochondrial function. Remarkably, transcript-level WGCNA revealed a single module correlated with dysplasia status, notably enriched in cilium-related biological processes. Notable hub transcripts included RABL2B (ENST00000395590), DNAH1 (ENST00000420323), EFHC1 (ENST00000635996), and VWA3A (ENST00000563389) along with transcription factors such as FOXJ1 and ZNF474 as potential regulators. Our findings underscore the value of transcript-level analysis in uncovering novel insights into premalignant bronchial lesion biology, including identification of potential biomarkers associated with early lung carcinogenesis.
Collapse
Affiliation(s)
- Mikhail A. Pyatnitskiy
- Institute of Biomedical Chemistry, Moscow 119121, Russia;
- National Research University Higher School of Economics, Moscow 101000, Russia
| | | |
Collapse
|
6
|
Chen H, Wu Z, Yan Z, Chen C, Zhang Y, Wang Q, Gao Y, Ling K, Hu J, Wei Q. The ARPKD Protein DZIP1L Regulates Ciliary Protein Entry by Modulating the Architecture and Function of Ciliary Transition Fibers. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308820. [PMID: 38634253 PMCID: PMC11200010 DOI: 10.1002/advs.202308820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 03/13/2024] [Indexed: 04/19/2024]
Abstract
Serving as the cell's sensory antennae, primary cilia are linked to numerous human genetic diseases when they malfunction. DZIP1L, identified as one of the genetic causes of human autosomal recessive polycystic kidney disease (ARPKD), is an evolutionarily conserved ciliary basal body protein. Although it has been reported that DZIP1L is involved in the ciliary entry of PKD proteins, the underlying mechanism remains elusive. Here, an uncharacterized role of DZIP1L is reported in modulating the architecture and function of transition fibers (TFs), striking ciliary base structures essential for selective cilia gating. Using C. elegans as a model, C01G5.7 (hereafter termed DZIP-1) is identified as the sole homolog of DZIP1L, which specifically localizes to TFs. While DZIP-1 or ANKR-26 (the ortholog of ANKRD26) deficiency shows subtle impact on TFs, co-depletion of DZIP-1 and ANKR-26 disrupts TF assembly and cilia gating for soluble and membrane proteins, including the ortholog of ADPKD protein polycystin-2. Notably, the synergistic role for DZIP1L and ANKRD26 in the formation and function of TFs is highly conserved in mammalian cilia. Hence, the findings illuminate an evolutionarily conserved role of DZIP1L in TFs architecture and function, highlighting TFs as a vital part of the ciliary gate implicated in ciliopathies ARPKD.
Collapse
Affiliation(s)
- Huicheng Chen
- CAS Key Laboratory of Insect Developmental and Evolutionary BiologyCAS Center for Excellence in Molecular Plant SciencesChinese Academy of SciencesShanghai200032China
- University of Chinese Academy of SciencesBeijing100039China
- Center for Energy Metabolism and ReproductionInstitute of Biomedicine and BiotechnologyShenzhen Institutes of Advanced TechnologyChinese Academy of Sciences (CAS)Shenzhen518055China
| | - Zhimao Wu
- Center for Energy Metabolism and ReproductionInstitute of Biomedicine and BiotechnologyShenzhen Institutes of Advanced TechnologyChinese Academy of Sciences (CAS)Shenzhen518055China
| | - Ziwei Yan
- CAS Key Laboratory of Insect Developmental and Evolutionary BiologyCAS Center for Excellence in Molecular Plant SciencesChinese Academy of SciencesShanghai200032China
- University of Chinese Academy of SciencesBeijing100039China
| | - Chuan Chen
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMN55905USA
| | - Yingying Zhang
- Center for Energy Metabolism and ReproductionInstitute of Biomedicine and BiotechnologyShenzhen Institutes of Advanced TechnologyChinese Academy of Sciences (CAS)Shenzhen518055China
| | - Qiaoling Wang
- Institute of Medicine and Pharmaceutical SciencesZhengzhou UniversityZhengzhou430000China
| | - Yuqing Gao
- Center for Energy Metabolism and ReproductionInstitute of Biomedicine and BiotechnologyShenzhen Institutes of Advanced TechnologyChinese Academy of Sciences (CAS)Shenzhen518055China
| | - Kun Ling
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMN55905USA
| | - Jinghua Hu
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMN55905USA
| | - Qing Wei
- Center for Energy Metabolism and ReproductionInstitute of Biomedicine and BiotechnologyShenzhen Institutes of Advanced TechnologyChinese Academy of Sciences (CAS)Shenzhen518055China
- School of Synthetic BiologyShanxi Key Laboratory of Nucleic Acid BiopesticidesShanxi UniversityTaiyuan030006China
| |
Collapse
|
7
|
Turgeon A, Fu J, Divyanshi, Ma M, Jin Z, Hwang H, Li M, Qiao H, Mei W, Yang J. Dzip1 is dynamically expressed in the vertebrate germline and regulates the development of Xenopus primordial germ cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.20.590349. [PMID: 38712275 PMCID: PMC11071414 DOI: 10.1101/2024.04.20.590349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Primordial germ cells (PGCs) are the precursors of sperms and oocytes. Proper development of PGCs is crucial for the survival of the species. In many organisms, factors responsible for PGC development are synthesized during early oogenesis and assembled into the germ plasm. During early embryonic development, germ plasm is inherited by a few cells, leading to the formation of PGCs. While germline development has been extensively studied, how components of the germ plasm regulate PGC development is not fully understood. Here, we report that Dzip1 is dynamically expressed in vertebrate germline and is a novel component of the germ plasm in Xenopus and zebrafish. Knockdown of Dzip1 impairs PGC development in Xenopus embryos. At the molecular level, Dzip1 physically interacts with Dazl, an evolutionarily conserved RNA-binding protein that plays a multifaced role during germline development. We further showed that the sequence between amino acid residues 282 and 550 of Dzip1 is responsible for binding to Dazl. Disruption of the binding between Dzip1 and Dazl leads to defective PGC development. Taken together, our results presented here demonstrate that Dzip1 is dynamically expressed in the vertebrate germline and plays a novel function during Xenopus PGC development.
Collapse
|
8
|
Shaikh Qureshi WM, Hentges KE. Functions of cilia in cardiac development and disease. Ann Hum Genet 2024; 88:4-26. [PMID: 37872827 PMCID: PMC10952336 DOI: 10.1111/ahg.12534] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/08/2023] [Accepted: 10/02/2023] [Indexed: 10/25/2023]
Abstract
Errors in embryonic cardiac development are a leading cause of congenital heart defects (CHDs), including morphological abnormalities of the heart that are often detected after birth. In the past few decades, an emerging role for cilia in the pathogenesis of CHD has been identified, but this topic still largely remains an unexplored area. Mouse forward genetic screens and whole exome sequencing analysis of CHD patients have identified enrichment for de novo mutations in ciliary genes or non-ciliary genes, which regulate cilia-related pathways, linking cilia function to aberrant cardiac development. Key events in cardiac morphogenesis, including left-right asymmetric development of the heart, are dependent upon cilia function. Cilia dysfunction during left-right axis formation contributes to CHD as evidenced by the substantial proportion of heterotaxy patients displaying complex CHD. Cilia-transduced signaling also regulates later events during heart development such as cardiac valve formation, outflow tract septation, ventricle development, and atrioventricular septa formation. In this review, we summarize the role of motile and non-motile (primary cilia) in cardiac asymmetry establishment and later events during heart development.
Collapse
Affiliation(s)
- Wasay Mohiuddin Shaikh Qureshi
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine, and Health, Manchester Academic Health Science CentreUniversity of ManchesterManchesterUK
| | - Kathryn E. Hentges
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine, and Health, Manchester Academic Health Science CentreUniversity of ManchesterManchesterUK
| |
Collapse
|
9
|
Yin Y, Liu Y, Wang Y, Li J, Liang S, Zhang W, Ma Z, Liu S, Zou X. DZIP1 expressed in fibroblasts and tumor cells may affect immunosuppression and metastatic potential in gastric cancer. Int Immunopharmacol 2023; 117:109886. [PMID: 36805200 DOI: 10.1016/j.intimp.2023.109886] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 02/09/2023] [Accepted: 02/10/2023] [Indexed: 02/19/2023]
Abstract
The tumor microenvironment (TME) contains complex components, of which the most well-known one is the tumor-associated fibroblast (CAF) that participates in the development and progression of tumors. A high abundance of CAFs implies that tumor stroma is also abundant and often predicts a poor prognosis, especially in terms of immunotherapeutic resistance. In this study, DAZ interacting zinc finger protein 1 (DZIP1) was identified to be upregulated in CAFs and malignant epithelial cells based on single-cell sequencing. Furthermore, results from The Cancer Genome Atlas database showed that this gene was highly positively associated with the mesenchymal phenotype in gastric cancer (GC). In addition, molecular experiments verified that DZIP1 directly promoted the proliferation of CAFs and enhanced the epithelial-mesenchymal transition (EMT) of GC cells to drive angiogenesis. Also, the upregulated DZIP1 in GC cells was found to directly promote invasion and metastasis. Finally, multiplex immunofluorescence and immunohistochemistry showed that DZIP1 was correlated with the immunosuppressive microenvironment of GC and resulted in a poor response to immunotherapy. Overall, our findings suggest that DZIP1 is expressed in both tumor parenchyma and mesenchyme and that it is involved in shaping the immunosuppressive microenvironment and inducing EMT by participating in tumor-stromal signaling crosstalk.
Collapse
Affiliation(s)
- Yi Yin
- Affiliated Hospital of Nanjing the University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210023, Jiangsu Province, China; No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Yuanjie Liu
- Affiliated Hospital of Nanjing the University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210023, Jiangsu Province, China; No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Yunya Wang
- Affiliated Hospital of Nanjing the University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210023, Jiangsu Province, China; No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Jiepin Li
- Affiliated Hospital of Nanjing the University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210023, Jiangsu Province, China; No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China; Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang 215600, Jiangsu Province, China
| | - Shuo Liang
- Affiliated Hospital of Nanjing the University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210023, Jiangsu Province, China; No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Wei Zhang
- Affiliated Hospital of Nanjing the University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210023, Jiangsu Province, China
| | - Zhibin Ma
- Hunan Aifang Biological Company Limited, Changsha 410000, Hunan Province, China
| | - Shenlin Liu
- Affiliated Hospital of Nanjing the University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210023, Jiangsu Province, China; No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China.
| | - Xi Zou
- Affiliated Hospital of Nanjing the University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210023, Jiangsu Province, China; No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu Province, China; Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumor, Nanjing 210023, Jiangsu Province, China.
| |
Collapse
|
10
|
Platova S, Poliushkevich L, Kulakova M, Nesterenko M, Starunov V, Novikova E. Gotta Go Slow: Two Evolutionarily Distinct Annelids Retain a Common Hedgehog Pathway Composition, Outlining Its Pan-Bilaterian Core. Int J Mol Sci 2022; 23:ijms232214312. [PMID: 36430788 PMCID: PMC9695228 DOI: 10.3390/ijms232214312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/11/2022] [Accepted: 11/13/2022] [Indexed: 11/19/2022] Open
Abstract
Hedgehog signaling is one of the key regulators of morphogenesis, cell differentiation, and regeneration. While the Hh pathway is present in all bilaterians, it has mainly been studied in model animals such as Drosophila and vertebrates. Despite the conservatism of its core components, mechanisms of signal transduction and additional components vary in Ecdysozoa and Deuterostomia. Vertebrates have multiple copies of the pathway members, which complicates signaling implementation, whereas model ecdysozoans appear to have lost some components due to fast evolution rates. To shed light on the ancestral state of Hh signaling, models from the third clade, Spiralia, are needed. In our research, we analyzed the transcriptomes of two spiralian animals, errantial annelid Platynereis dumerilii (Nereididae) and sedentarian annelid Pygospio elegans (Spionidae). We found that both annelids express almost all Hh pathway components present in Drosophila and mouse. We performed a phylogenetic analysis of the core pathway components and built multiple sequence alignments of the additional key members. Our results imply that the Hh pathway compositions of both annelids share more similarities with vertebrates than with the fruit fly. Possessing an almost complete set of single-copy Hh pathway members, lophotrochozoan signaling composition may reflect the ancestral features of all three bilaterian branches.
Collapse
Affiliation(s)
- Sofia Platova
- Faculty of Biology, St. Petersburg State University, Saint Petersburg 199034, Russia
- Zoological Institute RAS, Saint Petersburg 199034, Russia
| | | | - Milana Kulakova
- Faculty of Biology, St. Petersburg State University, Saint Petersburg 199034, Russia
- Zoological Institute RAS, Saint Petersburg 199034, Russia
- Correspondence: (M.K.); (E.N.)
| | | | - Viktor Starunov
- Faculty of Biology, St. Petersburg State University, Saint Petersburg 199034, Russia
- Zoological Institute RAS, Saint Petersburg 199034, Russia
| | - Elena Novikova
- Faculty of Biology, St. Petersburg State University, Saint Petersburg 199034, Russia
- Zoological Institute RAS, Saint Petersburg 199034, Russia
- Correspondence: (M.K.); (E.N.)
| |
Collapse
|
11
|
Hedgehog Morphogens Act as Growth Factors Critical to Pre- and Postnatal Cardiac Development and Maturation: How Primary Cilia Mediate Their Signal Transduction. Cells 2022; 11:cells11121879. [PMID: 35741008 PMCID: PMC9221318 DOI: 10.3390/cells11121879] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/27/2022] [Accepted: 05/30/2022] [Indexed: 02/06/2023] Open
Abstract
Primary cilia are crucial for normal cardiac organogenesis via the formation of cyto-architectural, anatomical, and physiological boundaries in the developing heart and outflow tract. These tiny, plasma membrane-bound organelles function in a sensory-integrative capacity, interpreting both the intra- and extra-cellular environments and directing changes in gene expression responses to promote, prevent, and modify cellular proliferation and differentiation. One distinct feature of this organelle is its involvement in the propagation of a variety of signaling cascades, most notably, the Hedgehog cascade. Three ligands, Sonic, Indian, and Desert hedgehog, function as growth factors that are most commonly dependent on the presence of intact primary cilia, where the Hedgehog receptors Patched-1 and Smoothened localize directly within or at the base of the ciliary axoneme. Hedgehog signaling functions to mediate many cell behaviors that are critical for normal embryonic tissue/organ development. However, inappropriate activation and/or upregulation of Hedgehog signaling in postnatal and adult tissue is known to initiate oncogenesis, as well as the pathogenesis of other diseases. The focus of this review is to provide an overview describing the role of Hedgehog signaling and its dependence upon the primary cilium in the cell types that are most essential for mammalian heart development. We outline the breadth of developmental defects and the consequential pathologies resulting from inappropriate changes to Hedgehog signaling, as it pertains to congenital heart disease and general cardiac pathophysiology.
Collapse
|
12
|
Nandamuri SP, Lusk S, Kwan KM. Loss of zebrafish dzip1 results in inappropriate recruitment of periocular mesenchyme to the optic fissure and ocular coloboma. PLoS One 2022; 17:e0265327. [PMID: 35286359 PMCID: PMC8920261 DOI: 10.1371/journal.pone.0265327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/28/2022] [Indexed: 01/13/2023] Open
Abstract
Cilia are essential for the development and function of many different tissues. Although cilia machinery is crucial in the eye for photoreceptor development and function, a role for cilia in early eye development and morphogenesis is still somewhat unclear: many zebrafish cilia mutants retain cilia at early stages due to maternal deposition of cilia components. An eye phenotype has been described in the mouse Arl13 mutant, however, zebrafish arl13b is maternally deposited, and an early role for cilia proteins has not been tested in zebrafish eye development. Here we use the zebrafish dzip1 mutant, which exhibits a loss of cilia throughout stages of early eye development, to examine eye development and morphogenesis. We find that in dzip1 mutants, initial formation of the optic cup proceeds normally, however, the optic fissure subsequently fails to close and embryos develop the structural eye malformation ocular coloboma. Further, neural crest cells, which are implicated in optic fissure closure, do not populate the optic fissure correctly, suggesting that their inappropriate localization may be the underlying cause of coloboma. Overall, our results indicate a role for dzip1 in proper neural crest localization in the optic fissure and optic fissure closure.
Collapse
Affiliation(s)
- Sri Pratima Nandamuri
- Department of Human Genetics, University of Utah, Salt Lake City, UT, United States of America
| | - Sarah Lusk
- Department of Human Genetics, University of Utah, Salt Lake City, UT, United States of America
| | - Kristen M. Kwan
- Department of Human Genetics, University of Utah, Salt Lake City, UT, United States of America
| |
Collapse
|
13
|
Guo L, Beck T, Fulmer D, Ramos‐Ortiz S, Glover J, Wang C, Moore K, Gensemer C, Morningstar J, Moore R, Schott J, Le Tourneau T, Koren N, Norris RA. DZIP1 regulates mammalian cardiac valve development through a Cby1-β-catenin mechanism. Dev Dyn 2021; 250:1432-1449. [PMID: 33811421 PMCID: PMC8518365 DOI: 10.1002/dvdy.342] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 03/03/2021] [Accepted: 03/26/2021] [Indexed: 11/21/2022] Open
Abstract
Background Mitral valve prolapse (MVP) is a common and progressive cardiovascular disease with developmental origins. How developmental errors contribute to disease pathogenesis are not well understood. Results A multimeric complex was identified that consists of the MVP gene Dzip1, Cby1, and β‐catenin. Co‐expression during valve development revealed overlap at the basal body of the primary cilia. Biochemical studies revealed a DZIP1 peptide required for stabilization of the complex and suppression of β‐catenin activities. Decoy peptides generated against this interaction motif altered nuclear vs cytosolic levels of β‐catenin with effects on transcriptional activity. A mutation within this domain was identified in a family with inherited non‐syndromic MVP. This novel mutation and our previously identified DZIP1S24R variant resulted in reduced DZIP1 and CBY1 stability and increased β‐catenin activities. The β‐catenin target gene, MMP2 was up‐regulated in the Dzip1S14R/+ valves and correlated with loss of collagenous ECM matrix and myxomatous phenotype. Conclusion Dzip1 functions to restrain β‐catenin signaling through a CBY1 linker during cardiac development. Loss of these interactions results in increased nuclear β‐catenin/Lef1 and excess MMP2 production, which correlates with developmental and postnatal changes in ECM and generation of a myxomatous phenotype.
Collapse
Affiliation(s)
- Lilong Guo
- Department of Regenerative Medicine and Cell BiologyMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Tyler Beck
- Department of Regenerative Medicine and Cell BiologyMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Diana Fulmer
- Department of Regenerative Medicine and Cell BiologyMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Sandra Ramos‐Ortiz
- Department of Regenerative Medicine and Cell BiologyMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Janiece Glover
- Department of Regenerative Medicine and Cell BiologyMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Christina Wang
- Department of Regenerative Medicine and Cell BiologyMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Kelsey Moore
- Department of Regenerative Medicine and Cell BiologyMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Cortney Gensemer
- Department of Regenerative Medicine and Cell BiologyMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Jordan Morningstar
- Department of Regenerative Medicine and Cell BiologyMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Reece Moore
- Department of Regenerative Medicine and Cell BiologyMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | | | | | - Natalie Koren
- Department of Regenerative Medicine and Cell BiologyMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Russell A. Norris
- Department of Regenerative Medicine and Cell BiologyMedical University of South CarolinaCharlestonSouth CarolinaUSA
| |
Collapse
|
14
|
Role of DZIP1-CBY-FAM92 transition zone complex in the basal body to membrane attachment and ciliary budding. Biochem Soc Trans 2021; 48:1067-1075. [PMID: 32491167 DOI: 10.1042/bst20191007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 04/20/2020] [Accepted: 05/07/2020] [Indexed: 02/07/2023]
Abstract
Cilia play important signaling or motile functions in various organisms. In Human, cilia dysfunctions are responsible for a wide range of diseases, called ciliopathies. Cilia assembly is a tightly controlled process, which starts with the conversion of the centriole into a basal body, leading to the formation of the ciliary bud that protrudes inside a ciliary vesicle and/or ultimately at the cell surface. Ciliary bud formation is associated with the assembly of the transition zone (TZ), a complex architecture of proteins of the ciliary base which plays critical functions in gating proteins in and out of the ciliary compartment. Many proteins are involved in the assembly of the TZ, which shows structural and functional variations in different cell types or organisms. In this review, we discuss how a particular complex, composed of members of the DZIP1, CBY and FAM92 families of proteins, is required for the initial stages of cilia assembly leading to ciliary bud formation and how their functional hierarchy contributes to TZ assembly. Moreover, we summarize how evidences in Drosophila reveal functional differences of the DZIP1-CBY-FAM92 complex in the different ciliated tissues of this organism. Whereas it is essential for proper TZ assembly in the two types of ciliated tissues, it is involved in stable anchoring of basal bodies to the plasma membrane in male germ cells. Overall, the DZIP1-CBY-FAM92 complex reveals a molecular assembly pathway required for the initial stages of ciliary bud formation and that is conserved from Drosophila to Human.
Collapse
|
15
|
Wu Z, Pang N, Zhang Y, Chen H, Peng Y, Fu J, Wei Q. CEP290 is essential for the initiation of ciliary transition zone assembly. PLoS Biol 2020; 18:e3001034. [PMID: 33370260 PMCID: PMC7793253 DOI: 10.1371/journal.pbio.3001034] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 01/08/2021] [Accepted: 12/16/2020] [Indexed: 11/24/2022] Open
Abstract
Cilia play critical roles during embryonic development and adult homeostasis. Dysfunction of cilia leads to various human genetic diseases, including many caused by defects in transition zones (TZs), the "gates" of cilia. The evolutionarily conserved TZ component centrosomal protein 290 (CEP290) is the most frequently mutated human ciliopathy gene, but its roles in ciliogenesis are not completely understood. Here, we report that CEP290 plays an essential role in the initiation of TZ assembly in Drosophila. Mechanistically, the N-terminus of CEP290 directly recruits DAZ interacting zinc finger protein 1 (DZIP1), which then recruits Chibby (CBY) and Rab8 to promote early ciliary membrane formation. Complete deletion of CEP290 blocks ciliogenesis at the initiation stage of TZ assembly, which can be mimicked by DZIP1 deletion mutants. Remarkably, expression of the N-terminus of CEP290 alone restores the TZ localization of DZIP1 and subsequently ameliorates the defects in TZ assembly initiation in cep290 mutants. Our results link CEP290 to DZIP1-CBY/Rab8 module and uncover a previously uncharacterized important function of CEP290 in the coordination of early ciliary membrane formation and TZ assembly.
Collapse
Affiliation(s)
- Zhimao Wu
- Chinese Academy of Sciences Key Laboratory of Insect Developmental and Evolutionary Biology, Chinese Academy of Sciences Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
- Center for Energy Metabolism and Reproduction, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Nan Pang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Yingying Zhang
- Chinese Academy of Sciences Key Laboratory of Insect Developmental and Evolutionary Biology, Chinese Academy of Sciences Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
- Center for Energy Metabolism and Reproduction, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Huicheng Chen
- Chinese Academy of Sciences Key Laboratory of Insect Developmental and Evolutionary Biology, Chinese Academy of Sciences Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
- Center for Energy Metabolism and Reproduction, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Ying Peng
- Institute of Medicine and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jingyan Fu
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Qing Wei
- Center for Energy Metabolism and Reproduction, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
16
|
Kobayashi T, Tanaka K, Mashima Y, Shoda A, Tokuda M, Itoh H. CEP164 Deficiency Causes Hyperproliferation of Pancreatic Cancer Cells. Front Cell Dev Biol 2020; 8:587691. [PMID: 33251215 PMCID: PMC7674857 DOI: 10.3389/fcell.2020.587691] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 10/13/2020] [Indexed: 12/21/2022] Open
Abstract
Primary cilia are hair-like projections that protrude from most mammalian cells and mediate various extracellular signaling pathways. Pancreatic ductal adenocarcinoma (PDAC) cells are known to lose their primary cilia, but the relevance of this phenomenon remains unclear. In this study, we generated PDAC-originated Panc1 cells devoid of primary cilia by mutating a centriolar protein, centrosomal protein 164 (CEP164), which is required for ciliogenesis. CEP164 depletion enhanced the clonogenicity of Panc1 cells, along with chemically induced elimination of primary cilia, suggesting that a lack of these organelles promotes PDAC cells proliferation. In addition, the loss of CEP164 altered the cell cycle progression irrespective of absence of primary cilia. We found that CEP164 was co-localized with the GLI2 transcription factor at the mother centriole and controlled its activation, thus inducing Cyclin D-CDK6 expression. Furthermore, CEP164-mutated Panc1 cells were significantly tolerant to KRAS depletion-dependent growth inhibition. This study suggests that CEP164 deficiency is advantageous for PDAC cells proliferation due to not only lack of ciliation but also cilia-independent GLI2-Cyclin D/CDK6 activation, and that CEP164 is a potential therapeutic target for PDAC.
Collapse
Affiliation(s)
- Tetsuo Kobayashi
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma, Japan
| | - Kosuke Tanaka
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma, Japan
| | - Yu Mashima
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma, Japan
| | - Ayano Shoda
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma, Japan
| | - Mio Tokuda
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma, Japan
| | - Hiroshi Itoh
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma, Japan
| |
Collapse
|
17
|
Avidor-Reiss T, Carr A, Fishman EL. The sperm centrioles. Mol Cell Endocrinol 2020; 518:110987. [PMID: 32810575 PMCID: PMC7606549 DOI: 10.1016/j.mce.2020.110987] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 08/07/2020] [Accepted: 08/10/2020] [Indexed: 12/13/2022]
Abstract
Centrioles are eukaryotic subcellular structures that produce and regulate massive cytoskeleton superstructures. They form centrosomes and cilia, regulate new centriole formation, anchor cilia to the cell, and regulate cilia function. These basic centriolar functions are executed in sperm cells during their amplification from spermatogonial stem cells during their differentiation to spermatozoa, and finally, after fertilization, when the sperm fuses with the egg. However, sperm centrioles exhibit many unique characteristics not commonly observed in other cell types, including structural remodeling, centriole-flagellum transition zone migration, and cell membrane association during meiosis. Here, we discuss five roles of sperm centrioles: orchestrating early spermatogenic cell divisions, forming the spermatozoon flagella, linking the spermatozoon head and tail, controlling sperm tail beating, and organizing the cytoskeleton of the zygote post-fertilization. We present the historic discovery of the centriole as a sperm factor that initiates embryogenesis, and recent genetic studies in humans and other mammals evaluating the current evidence for the five functions of sperm centrioles. We also examine information connecting the various sperm centriole functions to distinct clinical phenotypes. The emerging picture is that centrioles are essential sperm components with remarkable functional diversity and specialization that will require extensive and in-depth future studies.
Collapse
Affiliation(s)
- Tomer Avidor-Reiss
- Department of Biological Sciences, College of Natural Sciences and Mathematics, University of Toledo, Toledo, OH, USA; Department of Urology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, USA.
| | - Alexa Carr
- Department of Biological Sciences, College of Natural Sciences and Mathematics, University of Toledo, Toledo, OH, USA
| | | |
Collapse
|
18
|
Toomer KA, Yu M, Fulmer D, Guo L, Moore KS, Moore R, Drayton KD, Glover J, Peterson N, Ramos-Ortiz S, Drohan A, Catching BJ, Stairley R, Wessels A, Lipschutz JH, Delling FN, Jeunemaitre X, Dina C, Collins RL, Brand H, Talkowski ME, Del Monte F, Mukherjee R, Awgulewitsch A, Body S, Hardiman G, Hazard ES, da Silveira WA, Wang B, Leyne M, Durst R, Markwald RR, Le Scouarnec S, Hagege A, Le Tourneau T, Kohl P, Rog-Zielinska EA, Ellinor PT, Levine RA, Milan DJ, Schott JJ, Bouatia-Naji N, Slaugenhaupt SA, Norris RA. Primary cilia defects causing mitral valve prolapse. Sci Transl Med 2020; 11:11/493/eaax0290. [PMID: 31118289 PMCID: PMC7331025 DOI: 10.1126/scitranslmed.aax0290] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 04/25/2019] [Indexed: 12/15/2022]
Abstract
Mitral valve prolapse (MVP) affects 1 in 40 people and is the most common indication for mitral valve surgery. MVP can cause arrhythmias, heart failure, and sudden cardiac death, and to date, the causes of this disease are poorly understood. We now demonstrate that defects in primary cilia genes and their regulated pathways can cause MVP in familial and sporadic nonsyndromic MVP cases. Our expression studies and genetic ablation experiments confirmed a role for primary cilia in regulating ECM deposition during cardiac development. Loss of primary cilia during development resulted in progressive myxomatous degeneration and profound mitral valve pathology in the adult setting. Analysis of a large family with inherited, autosomal dominant nonsyndromic MVP identified a deleterious missense mutation in a cilia gene, DZIP1 A mouse model harboring this variant confirmed the pathogenicity of this mutation and revealed impaired ciliogenesis during development, which progressed to adult myxomatous valve disease and functional MVP. Relevance of primary cilia in common forms of MVP was tested using pathway enrichment in a large population of patients with MVP and controls from previously generated genome-wide association studies (GWAS), which confirmed the involvement of primary cilia genes in MVP. Together, our studies establish a developmental basis for MVP through altered cilia-dependent regulation of ECM and suggest that defects in primary cilia genes can be causative to disease phenotype in some patients with MVP.
Collapse
Affiliation(s)
- Katelynn A Toomer
- Cardiovascular Developmental Biology Center, Department of Regenerative Medicine and Cell Biology, College of Medicine, Children's Research Institute, Medical University of South Carolina, 171 Ashley Avenue, Charleston, SC 29425, USA
| | - Mengyao Yu
- INSERM, UMR-970, Paris Cardiovascular Research Center, 75015 Paris, France.,Paris Descartes University, Sorbonne Paris Cité, Faculty of Medicine, 75006 Paris, France
| | - Diana Fulmer
- Cardiovascular Developmental Biology Center, Department of Regenerative Medicine and Cell Biology, College of Medicine, Children's Research Institute, Medical University of South Carolina, 171 Ashley Avenue, Charleston, SC 29425, USA
| | - Lilong Guo
- Cardiovascular Developmental Biology Center, Department of Regenerative Medicine and Cell Biology, College of Medicine, Children's Research Institute, Medical University of South Carolina, 171 Ashley Avenue, Charleston, SC 29425, USA
| | - Kelsey S Moore
- Cardiovascular Developmental Biology Center, Department of Regenerative Medicine and Cell Biology, College of Medicine, Children's Research Institute, Medical University of South Carolina, 171 Ashley Avenue, Charleston, SC 29425, USA
| | - Reece Moore
- Cardiovascular Developmental Biology Center, Department of Regenerative Medicine and Cell Biology, College of Medicine, Children's Research Institute, Medical University of South Carolina, 171 Ashley Avenue, Charleston, SC 29425, USA
| | - Ka'la D Drayton
- Cardiovascular Developmental Biology Center, Department of Regenerative Medicine and Cell Biology, College of Medicine, Children's Research Institute, Medical University of South Carolina, 171 Ashley Avenue, Charleston, SC 29425, USA
| | - Janiece Glover
- Cardiovascular Developmental Biology Center, Department of Regenerative Medicine and Cell Biology, College of Medicine, Children's Research Institute, Medical University of South Carolina, 171 Ashley Avenue, Charleston, SC 29425, USA
| | - Neal Peterson
- Cardiovascular Developmental Biology Center, Department of Regenerative Medicine and Cell Biology, College of Medicine, Children's Research Institute, Medical University of South Carolina, 171 Ashley Avenue, Charleston, SC 29425, USA
| | - Sandra Ramos-Ortiz
- Cardiovascular Developmental Biology Center, Department of Regenerative Medicine and Cell Biology, College of Medicine, Children's Research Institute, Medical University of South Carolina, 171 Ashley Avenue, Charleston, SC 29425, USA
| | - Alex Drohan
- Cardiovascular Developmental Biology Center, Department of Regenerative Medicine and Cell Biology, College of Medicine, Children's Research Institute, Medical University of South Carolina, 171 Ashley Avenue, Charleston, SC 29425, USA
| | - Breiona J Catching
- Cardiovascular Developmental Biology Center, Department of Regenerative Medicine and Cell Biology, College of Medicine, Children's Research Institute, Medical University of South Carolina, 171 Ashley Avenue, Charleston, SC 29425, USA
| | - Rebecca Stairley
- Cardiovascular Developmental Biology Center, Department of Regenerative Medicine and Cell Biology, College of Medicine, Children's Research Institute, Medical University of South Carolina, 171 Ashley Avenue, Charleston, SC 29425, USA
| | - Andy Wessels
- Cardiovascular Developmental Biology Center, Department of Regenerative Medicine and Cell Biology, College of Medicine, Children's Research Institute, Medical University of South Carolina, 171 Ashley Avenue, Charleston, SC 29425, USA
| | - Joshua H Lipschutz
- Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA.,Department of Medicine, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29401, USA
| | - Francesca N Delling
- Department of Medicine, Division of Cardiology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Xavier Jeunemaitre
- INSERM, UMR-970, Paris Cardiovascular Research Center, 75015 Paris, France.,Paris Descartes University, Sorbonne Paris Cité, Faculty of Medicine, 75006 Paris, France.,Assistance Publique-Hôpitaux de Paris, Département de Génétique, Hôpital Européen Georges Pompidou, 75015 Paris, France
| | - Christian Dina
- INSERM, CNRS, Univ Nantes, L'Institut du Thorax, Nantes 44093, France.,CHU Nantes, L'Institut du Thorax, Service de Cardiologie, Nantes 44093, France
| | - Ryan L Collins
- Center for Genomic Medicine, Department of Neurology, Massachusetts General Hospital Research Institute, Harvard Medical School, 185 Cambridge St., Boston, MA 02114, USA
| | - Harrison Brand
- Center for Genomic Medicine, Department of Neurology, Massachusetts General Hospital Research Institute, Harvard Medical School, 185 Cambridge St., Boston, MA 02114, USA
| | - Michael E Talkowski
- Center for Genomic Medicine, Department of Neurology, Massachusetts General Hospital Research Institute, Harvard Medical School, 185 Cambridge St., Boston, MA 02114, USA
| | - Federica Del Monte
- Gazes Cardiac Research Institute, Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Rupak Mukherjee
- Gazes Cardiac Research Institute, Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Alexander Awgulewitsch
- Cardiovascular Developmental Biology Center, Department of Regenerative Medicine and Cell Biology, College of Medicine, Children's Research Institute, Medical University of South Carolina, 171 Ashley Avenue, Charleston, SC 29425, USA
| | - Simon Body
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Gary Hardiman
- Center for Genomic Medicine, Medical University of South Carolina, 135 Cannon Street, Suite 303 MSC 835, Charleston, SC 29425, USA.,Faculty of Medicine, Health and Life Sciences School of Biological Sciences, Institute for Global Food Security (IGFS), Queen's University Belfast, Belfast, Northern Ireland, BT7 1NN, UK
| | - E Starr Hazard
- Center for Genomic Medicine, Medical University of South Carolina, 135 Cannon Street, Suite 303 MSC 835, Charleston, SC 29425, USA
| | - Willian A da Silveira
- Center for Genomic Medicine, Medical University of South Carolina, 135 Cannon Street, Suite 303 MSC 835, Charleston, SC 29425, USA
| | - Baolin Wang
- Department of Genetic Medicine, Weill Medical College of Cornell University, New York, NY 10065, USA
| | - Maire Leyne
- Center for Genomic Medicine, Department of Neurology, Massachusetts General Hospital Research Institute, Harvard Medical School, 185 Cambridge St., Boston, MA 02114, USA
| | - Ronen Durst
- Cardiology Division, Hadassah Hebrew University Medical Center, POB 12000, Jerusalem, Israel
| | - Roger R Markwald
- Cardiovascular Developmental Biology Center, Department of Regenerative Medicine and Cell Biology, College of Medicine, Children's Research Institute, Medical University of South Carolina, 171 Ashley Avenue, Charleston, SC 29425, USA
| | | | - Albert Hagege
- INSERM, UMR-970, Paris Cardiovascular Research Center, 75015 Paris, France.,Paris Descartes University, Sorbonne Paris Cité, Faculty of Medicine, 75006 Paris, France.,Assistance Publique-Hôpitaux de Paris, Department of Cardiology, Hôpital Européen Georges Pompidou, 75015 Paris, France
| | - Thierry Le Tourneau
- INSERM, CNRS, Univ Nantes, L'Institut du Thorax, Nantes 44093, France.,CHU Nantes, L'Institut du Thorax, Service de Cardiologie, Nantes 44093, France
| | - Peter Kohl
- University Heart Center Freiburg, Bad Krozingen and Faculty of Medicine of the Albert-Ludwigs University Freiburg, Institute for Experimental Cardiovascular Medicine, Elsässerstr 2Q, 79110 Freiburg, Germany
| | - Eva A Rog-Zielinska
- University Heart Center Freiburg, Bad Krozingen and Faculty of Medicine of the Albert-Ludwigs University Freiburg, Institute for Experimental Cardiovascular Medicine, Elsässerstr 2Q, 79110 Freiburg, Germany
| | - Patrick T Ellinor
- Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital Research Institute, Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA
| | - Robert A Levine
- Cardiac Ultrasound Laboratory, Cardiology Division, Massachusetts General Hospital Research Institute, Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA
| | - David J Milan
- Cardiovascular Research Center, Cardiology Division, Massachusetts General Hospital Research Institute, Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA.,Leducq Foundation, 265 Franklin Street, Suite 1902, Boston, MA, 02110, USA
| | - Jean-Jacques Schott
- INSERM, CNRS, Univ Nantes, L'Institut du Thorax, Nantes 44093, France.,CHU Nantes, L'Institut du Thorax, Service de Cardiologie, Nantes 44093, France
| | - Nabila Bouatia-Naji
- INSERM, UMR-970, Paris Cardiovascular Research Center, 75015 Paris, France.,Paris Descartes University, Sorbonne Paris Cité, Faculty of Medicine, 75006 Paris, France
| | - Susan A Slaugenhaupt
- Center for Genomic Medicine, Department of Neurology, Massachusetts General Hospital Research Institute, Harvard Medical School, 185 Cambridge St., Boston, MA 02114, USA
| | - Russell A Norris
- Cardiovascular Developmental Biology Center, Department of Regenerative Medicine and Cell Biology, College of Medicine, Children's Research Institute, Medical University of South Carolina, 171 Ashley Avenue, Charleston, SC 29425, USA.
| |
Collapse
|
19
|
Gabriel GC, Young CB, Lo CW. Role of cilia in the pathogenesis of congenital heart disease. Semin Cell Dev Biol 2020; 110:2-10. [PMID: 32418658 DOI: 10.1016/j.semcdb.2020.04.017] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/22/2020] [Accepted: 04/25/2020] [Indexed: 12/13/2022]
Abstract
An essential role for cilia in the pathogenesis of congenital heart disease (CHD) has emerged from findings of a large-scale mouse forward genetic screen. High throughput screening with fetal ultrasound imaging followed by whole exome sequencing analysis recovered a preponderance of cilia related genes and cilia transduced cell signaling genes among mutations identified to cause CHD. The perturbation of left-right patterning in CHD pathogenesis is suggested by the association of CHD with heterotaxy, but also by the finding of the co-occurrence of laterality defects with CHD in birth defect registries. Many of the cilia and cilia cell signaling genes recovered were found to be related to Hedgehog signaling. Studies in mice showed cilia transduced hedgehog signaling coordinates left-right patterning with heart looping and differentiation of the heart tube. Cilia transduced Shh signaling also regulates later events in heart development, including outflow tract septation and formation of the atrioventricular septum. More recent work has shown mutations in cilia related genes may also contribute to valve disease that largely manifest in adult life. Overall, these and other findings show cilia play an important role in CHD and also in more common valve diseases.
Collapse
Affiliation(s)
- George C Gabriel
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201, United States
| | - Cullen B Young
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201, United States
| | - Cecilia W Lo
- Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15201, United States.
| |
Collapse
|
20
|
Fulmer D, Toomer KA, Glover J, Guo L, Moore K, Moore R, Stairley R, Gensemer C, Abrol S, Rumph MK, Emetu F, Lipschutz JH, McDowell C, Bian J, Wang C, Beck T, Wessels A, Renault MA, Norris RA. Desert hedgehog-primary cilia cross talk shapes mitral valve tissue by organizing smooth muscle actin. Dev Biol 2020; 463:26-38. [PMID: 32151560 DOI: 10.1016/j.ydbio.2020.03.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 02/27/2020] [Accepted: 03/02/2020] [Indexed: 01/01/2023]
Abstract
Non-syndromic mitral valve prolapse (MVP) is the most common heart valve disease affecting 2.4% of the population. Recent studies have identified genetic defects in primary cilia as causative to MVP, although the mechanism of their action is currently unknown. Using a series of gene inactivation approaches, we define a paracrine mechanism by which endocardially-expressed Desert Hedgehog (DHH) activates primary cilia signaling on neighboring valve interstitial cells. High-resolution imaging and functional assays show that DHH de-represses smoothened at the primary cilia, resulting in kinase activation of RAC1 through the RAC1-GEF, TIAM1. Activation of this non-canonical hedgehog pathway stimulates α-smooth actin organization and ECM remodeling. Genetic or pharmacological perturbation of this pathway results in enlarged valves that progress to a myxomatous phenotype, similar to valves seen in MVP patients. These data identify a potential molecular origin for MVP as well as establish a paracrine DHH-primary cilium cross-talk mechanism that is likely applicable across developmental tissue types.
Collapse
Affiliation(s)
- Diana Fulmer
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Katelynn A Toomer
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA; Department of Genetic Medicine, John Hopkins, Baltimore, MD, USA
| | - Janiece Glover
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Lilong Guo
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Kelsey Moore
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Reece Moore
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Rebecca Stairley
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Cortney Gensemer
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Sameer Abrol
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Mary Kate Rumph
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Faith Emetu
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Joshua H Lipschutz
- Department of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Colin McDowell
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Justin Bian
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Christina Wang
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Tyler Beck
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Andy Wessels
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | | | - Russell A Norris
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA; Department of Medicine, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
21
|
Lv M, Liu W, Chi W, Ni X, Wang J, Cheng H, Li WY, Yang S, Wu H, Zhang J, Gao Y, Liu C, Li C, Yang C, Tan Q, Tang D, Zhang J, Song B, Chen YJ, Li Q, Zhong Y, Zhang Z, Saiyin H, Jin L, Xu Y, Zhou P, Wei Z, Zhang C, He X, Zhang F, Cao Y. Homozygous mutations in DZIP1 can induce asthenoteratospermia with severe MMAF. J Med Genet 2020; 57:445-453. [PMID: 32051257 PMCID: PMC7361034 DOI: 10.1136/jmedgenet-2019-106479] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 12/02/2019] [Accepted: 12/21/2019] [Indexed: 02/01/2023]
Abstract
BACKGROUND Asthenoteratospermia, one of the most common causes for male infertility, often presents with defective sperm heads and/or flagella. Multiple morphological abnormalities of the sperm flagella (MMAF) is one of the common clinical manifestations of asthenoteratospermia. Variants in several genes including DNAH1, CEP135, CATSPER2 and SUN5 are involved in the genetic pathogenesis of asthenoteratospermia. However, more than half of the asthenoteratospermia cases cannot be explained by the known pathogenic genes. METHODS AND RESULTS Two asthenoteratospermia-affected men with severe MMAF (absent flagella in >90% spermatozoa) from consanguineous families were subjected to whole-exome sequencing. The first proband had a homozygous missense mutation c.188G>A (p.Arg63Gln) of DZIP1 and the second proband had a homozygous stop-gain mutation c.690T>G (p.Tyr230*). Both of the mutations were neither detected in the human population genome data (1000 Genomes Project, Exome Aggregation Consortium) nor in our own data of a cohort of 875 Han Chinese control populations. DZIP1 encodes a DAZ (a protein deleted in azoospermia) interacting protein, which was associated with centrosomes in mammalian cells. Immunofluorescence staining of the centriolar protein Centrin1 indicated that the spermatozoa of the proband presented with abnormal centrosomes, including no concentrated centriolar dot or more than two centriolar dots. HEK293T cells transfected with two DZIP1-mutated constructs showed reduced DZIP1 level or truncated DZIP1. The Dzip1-knockout mice, generated by the CRSIPR-Cas9, revealed consistent phenotypes of severe MMAF. CONCLUSION Our study strongly suggests that homozygous DZIP1 mutations can induce asthenoteratospermia with severe MMAF. The deficiency of DZIP1 induces sperm centrioles dysfunction and causes the absence of flagella.
Collapse
Affiliation(s)
- Mingrong Lv
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, China.,Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, China
| | - Wangjie Liu
- Obstetrics and Gynecology Hospital, NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), State Key Laboratory of Genetic Engineering at School of Life Sciences, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China.,State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Wangfei Chi
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing, China
| | - Xiaoqing Ni
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, China.,Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, China
| | - Jiajia Wang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, China.,Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, China
| | - Huiru Cheng
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, Hefei, China.,Anhui Provincial Engineering Technology Research Center for Biopreservation and Artificial Organs, Hefei, China
| | - Wei-Yu Li
- Obstetrics and Gynecology Hospital, NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), State Key Laboratory of Genetic Engineering at School of Life Sciences, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China.,State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Shenmin Yang
- Center for Reproduction and Genetics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Huan Wu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, Hefei, China.,Anhui Provincial Engineering Technology Research Center for Biopreservation and Artificial Organs, Hefei, China
| | - Junqiang Zhang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, Hefei, China.,Anhui Provincial Engineering Technology Research Center for Biopreservation and Artificial Organs, Hefei, China
| | - Yang Gao
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, China.,Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, China
| | - Chunyu Liu
- Obstetrics and Gynecology Hospital, NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), State Key Laboratory of Genetic Engineering at School of Life Sciences, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China.,State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Caihua Li
- Genesky Biotechnologies Inc, Shanghai, Shanghai, China
| | - Chenyu Yang
- Center of Cryo-Electron Microscopy, Zhejiang University, Hangzhou, China
| | - Qing Tan
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, Hefei, China.,Anhui Provincial Engineering Technology Research Center for Biopreservation and Artificial Organs, Hefei, China
| | - Dongdong Tang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, Hefei, China.,Anhui Provincial Engineering Technology Research Center for Biopreservation and Artificial Organs, Hefei, China
| | - Jingjing Zhang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, Hefei, China.,Anhui Provincial Engineering Technology Research Center for Biopreservation and Artificial Organs, Hefei, China
| | - Bing Song
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, Hefei, China.,Anhui Provincial Engineering Technology Research Center for Biopreservation and Artificial Organs, Hefei, China
| | - Yu-Jie Chen
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, Hefei, China.,Anhui Provincial Engineering Technology Research Center for Biopreservation and Artificial Organs, Hefei, China
| | - Qiang Li
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, Hefei, China.,Anhui Provincial Engineering Technology Research Center for Biopreservation and Artificial Organs, Hefei, China
| | - Yading Zhong
- Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhihua Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China
| | - Hexige Saiyin
- Obstetrics and Gynecology Hospital, NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), State Key Laboratory of Genetic Engineering at School of Life Sciences, Fudan University, Shanghai, China
| | - Li Jin
- Obstetrics and Gynecology Hospital, NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), State Key Laboratory of Genetic Engineering at School of Life Sciences, Fudan University, Shanghai, China
| | - Yuping Xu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, Hefei, China.,Anhui Provincial Engineering Technology Research Center for Biopreservation and Artificial Organs, Hefei, China
| | - Ping Zhou
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, Hefei, China.,Anhui Provincial Engineering Technology Research Center for Biopreservation and Artificial Organs, Hefei, China
| | - Zhaolian Wei
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Anhui Province Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, Hefei, China.,Anhui Provincial Engineering Technology Research Center for Biopreservation and Artificial Organs, Hefei, China
| | - Chuanmao Zhang
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, College of Life Sciences, Peking University, Beijing, China
| | - Xiaojin He
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China .,NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, China.,Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, China
| | - Feng Zhang
- Obstetrics and Gynecology Hospital, NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), State Key Laboratory of Genetic Engineering at School of Life Sciences, Fudan University, Shanghai, China .,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China.,State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Yunxia Cao
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China .,NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, China.,Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, China
| |
Collapse
|
22
|
Lapart JA, Gottardo M, Cortier E, Duteyrat JL, Augière C, Mangé A, Jerber J, Solassol J, Gopalakrishnan J, Thomas J, Durand B. Dzip1 and Fam92 form a ciliary transition zone complex with cell type specific roles in Drosophila. eLife 2019; 8:49307. [PMID: 31821146 PMCID: PMC6904220 DOI: 10.7554/elife.49307] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 11/12/2019] [Indexed: 12/12/2022] Open
Abstract
Cilia and flagella are conserved eukaryotic organelles essential for cellular signaling and motility. Cilia dysfunctions cause life-threatening ciliopathies, many of which are due to defects in the transition zone (TZ), a complex structure of the ciliary base. Therefore, understanding TZ assembly, which relies on ordered interactions of multiprotein modules, is of critical importance. Here, we show that Drosophila Dzip1 and Fam92 form a functional module which constrains the conserved core TZ protein, Cep290, to the ciliary base. We identify cell type specific roles of this functional module in two different tissues. While it is required for TZ assembly in all Drosophila ciliated cells, it also regulates basal-body growth and docking to the plasma membrane during spermatogenesis. We therefore demonstrate a novel regulatory role for Dzip1 and Fam92 in mediating membrane/basal-body interactions and show that these interactions exhibit cell type specific functions in basal-body maturation and TZ organization. Many animal cells have hair-like structures called cilia on their surface, which help them to sense and interact with their surroundings. The cilia are supported by protein filaments and must assemble correctly because faulty cilia can lead to several life-threatening diseases. Problems in an area at the base of the cilia, known as the ‘transition zone’, account for the most severe forms of these diseases in humans. The transition zone is responsible for selecting which proteins are allowed in and out of the cilia. The transition zone itself is made up of many proteins that work together to determine the cilia composition. But not all of these proteins are known, and it is unclear how those that are known affect cilia structure. One protein found in transition zones of several animals, including fruit flies and mice, is called Cby. Lapart et al. set out to understand which other proteins interact with Cby in fruit flies to better understand what this protein does in the transition zone. A series of experiments showed that Cby interacts with two proteins called Dzip1 and Fam92 to regulate the assembly of transition zones. Together these three proteins constrain a core component of the transition zone, a fourth protein called Cep290, to the base of the cilia. Fruit flies only have cilia on cells in their sensory organs and testes and, in both types of tissue, cilia could only form properly when Dzip1 and Fam92 were present. Lapart et al. also showed that, in the fruit fly testes, Dzip1 and Fam92 helped to anchor the newly forming cilia to the cell surface. This anchoring role was particularly important for the fruit flies’ sperm to grow their characteristic whip-like tails, which are a specialized type of cilia that allow sperm cells to move. Overall, the findings show how some transition zone proteins work together and that they can have different effects in different tissues. Understanding the mechanisms behind healthy cilia assembly will likely be key to tackling cilia-related diseases.
Collapse
Affiliation(s)
- Jean-André Lapart
- Institut NeuroMyoGène, CNRS UMR 5310, INSERM U1217, Université Claude Bernard Lyon 1, Lyon, France
| | - Marco Gottardo
- Institute of Human Genetics, Universitätsklinikum Düsseldorf, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| | - Elisabeth Cortier
- Institut NeuroMyoGène, CNRS UMR 5310, INSERM U1217, Université Claude Bernard Lyon 1, Lyon, France
| | - Jean-Luc Duteyrat
- Institut NeuroMyoGène, CNRS UMR 5310, INSERM U1217, Université Claude Bernard Lyon 1, Lyon, France
| | - Céline Augière
- Institut NeuroMyoGène, CNRS UMR 5310, INSERM U1217, Université Claude Bernard Lyon 1, Lyon, France
| | - Alain Mangé
- IRCM, INSERM, Université de Montpellier, ICM, Montpellier, France
| | - Julie Jerber
- Institut NeuroMyoGène, CNRS UMR 5310, INSERM U1217, Université Claude Bernard Lyon 1, Lyon, France
| | - Jérôme Solassol
- IRCM, INSERM, Université de Montpellier, ICM, Montpellier, France
| | - Jay Gopalakrishnan
- Institute of Human Genetics, Universitätsklinikum Düsseldorf, Heinrich-Heine-Universität Düsseldorf, Düsseldorf, Germany
| | - Joëlle Thomas
- Institut NeuroMyoGène, CNRS UMR 5310, INSERM U1217, Université Claude Bernard Lyon 1, Lyon, France
| | - Bénédicte Durand
- Institut NeuroMyoGène, CNRS UMR 5310, INSERM U1217, Université Claude Bernard Lyon 1, Lyon, France
| |
Collapse
|
23
|
Yan W, Deng Y, Zhang Y, Luo J, Lu D, Wan Q, Mao L, Chen Y. DZIP1 Promotes Proliferation, Migration, and Invasion of Oral Squamous Carcinoma Through the GLI1/3 Pathway. Transl Oncol 2019; 12:1504-1515. [PMID: 31450126 PMCID: PMC6717062 DOI: 10.1016/j.tranon.2019.07.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 06/29/2019] [Accepted: 07/08/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND DZIP1 is an oncogene involved in the progression and stemness of carcinoma through the Wnt/β-catenin pathway, and the potential mechanism of DZIP1 in oral squamous cancer remains unknown. The aim of this study was to uncover the effect and mechanism of DZIP1 in the progression of oral squamous carcinoma. METHODS TCGA database scanning was applied to verify dysregulated genes in oral squamous carcinoma. quantitative real-time polymerase chain reaction, immunohistochemistry, and Western blotting assays were used to detect the expression of DZIP1 in tissues and cell lines. We established stable DZIP1-overexpressing and DZIP1 knockdown cell lines. We investigated the biological function and the underlying mechanism of DZIP1 through a series of experiments. RESULTS DZIP1 was one of the genes discovered by the scanning strategy to be upregulated in cancer tissue and negatively correlated with the overall survival (OS) of patients. DZIP1 promotes proliferation, migration, and invasion in an oral squamous carcinoma cell line through EMT in a GLI1/3-dependent manner. CONCLUSIONS DZIP1 promotes the proliferation, migration, and invasion of oral squamous carcinoma through the GLI1/3 pathway.
Collapse
Affiliation(s)
- Wangxiang Yan
- Department of Stomatology, First Affiliated Hospital of Sun Yat-Sen University, China.
| | - Yun Deng
- Department of Stomatology, First Affiliated Hospital of Sun Yat-Sen University, China.
| | - Yuhang Zhang
- Department of Stomatology, First Affiliated Hospital of Sun Yat-Sen University, China.
| | - Jing Luo
- Department of Stomatology, General Hospital of Southern Theatre Command, China.
| | - Dunlang Lu
- Department of Stomatology, First Affiliated Hospital of Sun Yat-Sen University, China.
| | - Quan Wan
- Department of Oral and Maxillofacial Surgery, Sun Yat-Sen Memorial Hospital, China.
| | - Lijuan Mao
- Department of Radiology, First Affiliated Hospital of Sun Yat-Sen University, China.
| | - Yu Chen
- Department of Stomatology, First Affiliated Hospital of Sun Yat-Sen University, China.
| |
Collapse
|
24
|
Xu S, Liu Y, Meng Q, Wang B. Rab34 small GTPase is required for Hedgehog signaling and an early step of ciliary vesicle formation in mouse. J Cell Sci 2018; 131:jcs.213710. [PMID: 30301781 DOI: 10.1242/jcs.213710] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 09/21/2018] [Indexed: 12/30/2022] Open
Abstract
The primary cilium is a microtubule-based organelle that protrudes from the cell surface and plays essential roles in embryonic development. Ciliogenesis begins with the successive fusion of preciliary vesicles to form ciliary vesicles, which then dock onto the distal end of the mother centriole. Rab proteins have been linked to cilia formation in cultured cells, but not yet in vivo In the present study, we demonstrate that endocytic recycling protein Rab34 localizes to cilia, and that its mutation results in significant decrease of ciliogenesis in both cultured cells and mice. Rab34 is required for the successive fusion of preciliary vesicles to generate ciliary vesicles and for the migration of the mother centriole from perinuclear region to plasma membrane. We also show that Rab34 mutant mice exhibit polydactyly, and cleft-lip and -palate. These phenotypes are consistent with observations that nonciliated Rab34 mutant cells fail to respond to Hedgehog signaling and that processing of full-length Gli3 to its C-terminally truncated form is reduced in Rab34 mutant embryos. Therefore, Rab34 is required for an early step of ciliary vesicle formation and Hh signaling in vivo This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Shouying Xu
- Department of Genetic Medicine, Weill Medical College of Cornell University, 1300 York Avenue, W404, New York, NY 10065, USA.,Institute of Biological Sciences and Biotechnology, Donghua University, Shanghai 201620, China
| | - Yang Liu
- Department of Genetic Medicine, Weill Medical College of Cornell University, 1300 York Avenue, W404, New York, NY 10065, USA.,Department of Veterinary Public Health, College of Veterinary Medicine, Jilin University, Jilin 130000, China
| | - Qing Meng
- Institute of Biological Sciences and Biotechnology, Donghua University, Shanghai 201620, China
| | - Baolin Wang
- Department of Genetic Medicine, Weill Medical College of Cornell University, 1300 York Avenue, W404, New York, NY 10065, USA .,Department of Cell and Developmental Biology, Weill Medical College of Cornell University, 1300 York Avenue, W404, New York, NY 10065, USA
| |
Collapse
|
25
|
Wang C, Li J, Takemaru KI, Jiang X, Xu G, Wang B. Centrosomal protein Dzip1l binds Cby, promotes ciliary bud formation, and acts redundantly with Bromi to regulate ciliogenesis in the mouse. Development 2018; 145:dev.164236. [PMID: 29487109 DOI: 10.1242/dev.164236] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 02/16/2018] [Indexed: 12/14/2022]
Abstract
The primary cilium is a microtubule-based organelle required for Hedgehog (Hh) signaling and consists of a basal body, a ciliary axoneme and a compartment between the first two structures, called the transition zone (TZ). The TZ serves as a gatekeeper to control protein composition in cilia, but less is known about its role in ciliary bud formation. Here, we show that centrosomal protein Dzip1l is required for Hh signaling between Smoothened and Sufu. Dzip1l colocalizes with basal body appendage proteins and Rpgrip1l, a TZ protein. Loss of Dzip1l results in reduced ciliogenesis and dysmorphic cilia in vivo Dzip1l interacts with, and acts upstream of, Cby, an appendage protein, in ciliogenesis. Dzip1l also has overlapping functions with Bromi (Tbc1d32) in ciliogenesis, cilia morphogenesis and neural tube patterning. Loss of Dzip1l arrests ciliogenesis at the stage of ciliary bud formation from the TZ. Consistent with this, Dzip1l mutant cells fail to remove the capping protein Cp110 (Ccp110) from the distal end of mother centrioles and to recruit Rpgrip1l to the TZ. Therefore, Dzip1l promotes ciliary bud formation and is required for the integrity of the TZ.
Collapse
Affiliation(s)
- Chengbing Wang
- Department of Genetic Medicine, Weill Medical College of Cornell University, 1300 York Avenue, W404, New York, NY 10065, USA
| | - Jia Li
- Department of Genetic Medicine, Weill Medical College of Cornell University, 1300 York Avenue, W404, New York, NY 10065, USA
| | - Ken-Ichi Takemaru
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY 11794, USA
| | - Xiaogang Jiang
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and College of Pharmaceutical Sciences, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, Jiangsu 215123, China
| | - Guoqiang Xu
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and College of Pharmaceutical Sciences, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, Jiangsu 215123, China
| | - Baolin Wang
- Department of Genetic Medicine, Weill Medical College of Cornell University, 1300 York Avenue, W404, New York, NY 10065, USA .,Department of Cell and Developmental Biology, Weill Medical College of Cornell University, 1300 York Avenue, W404, New York, NY 10065, USA
| |
Collapse
|
26
|
Wu C, Li J, Peterson A, Tao K, Wang B. Loss of dynein-2 intermediate chain Wdr34 results in defects in retrograde ciliary protein trafficking and Hedgehog signaling in the mouse. Hum Mol Genet 2017; 26:2386-2397. [PMID: 28379358 DOI: 10.1093/hmg/ddx127] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 03/28/2017] [Indexed: 11/14/2022] Open
Abstract
The Wdr34 gene encodes an intermediate chain of cytoplasmic dynein 2, the motor for retrograde intraflagellar transport (IFT) in primary cilia. Although mutations in human WDR34 have recently been reported, the association of WDR34 function with Hedgehog (Hh) signaling has not been established, and actual cilia defects in the WDR34 mutant cells have also not been completely characterized. In the present study, we show that Wdr34 mutant mice die in midgestation and exhibit open brain and polydactyly phenotypes. Several Hh-dependent ventral neural cell types are not specified in the mutant neural tube. The expression of the direct Hh targets, Gli1 and Patched 1, is inhibited, while the expression of limb patterning genes that are normally inhibited by the Gli3 repressor is anteriorly expanded in mutant limbs. Comparison of cilia phenotype and function among wild type, Dnchc2 (dynein 2 heavy chain), and Wdr34 mutant cells demonstrates that cilia in both Dnchc2 and Wdr34 mutant cells are stumpy. Several ciliary proteins examined abnormally accumulate in the cilia of both mutant cells. Consistent with its function, overexpressed Wdr34 is occasionally localized to cilia, and Wdr34 is required for the ciliary localization of dynein 2 light intermediate chain Lic3. More interestingly, we show that both Dnchc2 and Wdr34 act between Smo and Gli2/Gli3 in the Hh pathway. Therefore, like Dnchc2, Wdr34 is required for ciliogenesis, retrograde ciliary protein trafficking, and the regulation of Gli2/Gli3 activators and repressors. Furthermore, both Wdr34 and Dnchc2 promote microtubule growth, a novel dynein 2 function in a non-cilia structure.
Collapse
Affiliation(s)
- Chuanqing Wu
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.,Department of Genetic Medicine, Weill Medical College of Cornell University, W404, New York, NY 10065,USA
| | - Jia Li
- Department of Genetic Medicine, Weill Medical College of Cornell University, W404, New York, NY 10065,USA.,Institute of Biological Sciences and Biotechnology, Donghua University, Shanghai 201620, China
| | - Andrew Peterson
- Department of Molecular Biology, Genentech, South San Francisco, CA 94080, USA
| | - Kaixiong Tao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Baolin Wang
- Department of Genetic Medicine, Weill Medical College of Cornell University, W404, New York, NY 10065,USA.,Department of Cell and Developmental Biology, Weill Medical College of Cornell University, W404, New York, NY 10065, USA
| |
Collapse
|
27
|
Li J, Wang C, Wu C, Cao T, Xu G, Meng Q, Wang B. PKA-mediated Gli2 and Gli3 phosphorylation is inhibited by Hedgehog signaling in cilia and reduced in Talpid3 mutant. Dev Biol 2017; 429:147-157. [PMID: 28673820 DOI: 10.1016/j.ydbio.2017.06.035] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 06/28/2017] [Accepted: 06/29/2017] [Indexed: 10/19/2022]
Abstract
Hedgehog (Hh) signaling is thought to occur in primary cilia, but the molecular basis of Gli2 and Gli3 activation by Hh signaling in cilia is unknown. Similarly, how ciliary gene mutations result in reduced Gli3 processing that generates a repressor is also not clear. Here we show that Hh signaling inhibits Gli2 and Gli3 phosphorylation by protein kinase A (PKA) in cilia. The cilia related gene Talpid3 (Ta3) mutation results in the reduced processing and phosphorylation of Gli2 and Gli3. Interestingly, Ta3 interacts and colocalizes with PKA regulatory subunit PKARIIβ at centrioles in the cell. The centriolar localization and PKA binding regions are located in the N- and C-terminal regions of Ta3, respectively. PKARIIβ fails to localize at centrioles in some Ta3 mutant cells. Therefore, our study provides the direct evidence that Gli2 and Gli3 are dephosphorylated and activated in cilia and that impaired Gli2 and Gli3 processing in Ta3 mutant is at least in part due to a decrease in Gli2 and Gli3 phosphorylation.
Collapse
Affiliation(s)
- Jia Li
- Institute of Biological Sciences and Biotechnology, Donghua University, Shanghai 201620, China; Department of Genetic Medicine, Weill Medical College of Cornell University, 1300 York Avenue, W404, New York, NY 10065, USA
| | - Chengbing Wang
- Department of Genetic Medicine, Weill Medical College of Cornell University, 1300 York Avenue, W404, New York, NY 10065, USA
| | - Chuanqing Wu
- Department of Genetic Medicine, Weill Medical College of Cornell University, 1300 York Avenue, W404, New York, NY 10065, USA; Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Ting Cao
- Department of Genetic Medicine, Weill Medical College of Cornell University, 1300 York Avenue, W404, New York, NY 10065, USA; College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Guoqiang Xu
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and College of Pharmaceutical Sciences, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, Jiangsu, China
| | - Qing Meng
- Institute of Biological Sciences and Biotechnology, Donghua University, Shanghai 201620, China.
| | - Baolin Wang
- Department of Genetic Medicine, Weill Medical College of Cornell University, 1300 York Avenue, W404, New York, NY 10065, USA; Department of Cell and Developmental Biology, Weill Medical College of Cornell University, 1300 York Avenue, W404, New York, NY 10065, USA.
| |
Collapse
|
28
|
Lu H, Galeano MCR, Ott E, Kaeslin G, Kausalya PJ, Kramer C, Ortiz-Brüchle N, Hilger N, Metzis V, Hiersche M, Tay SY, Tunningley R, Vij S, Courtney AD, Whittle B, Wühl E, Vester U, Hartleben B, Neuber S, Frank V, Little MH, Epting D, Papathanasiou P, Perkins AC, Wright GD, Hunziker W, Gee HY, Otto EA, Zerres K, Hildebrandt F, Roy S, Wicking C, Bergmann C. Mutations in DZIP1L, which encodes a ciliary-transition-zone protein, cause autosomal recessive polycystic kidney disease. Nat Genet 2017; 49:1025-1034. [PMID: 28530676 DOI: 10.1038/ng.3871] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 04/24/2017] [Indexed: 12/21/2022]
Abstract
Autosomal recessive polycystic kidney disease (ARPKD), usually considered to be a genetically homogeneous disease caused by mutations in PKHD1, has been associated with ciliary dysfunction. Here, we describe mutations in DZIP1L, which encodes DAZ interacting protein 1-like, in patients with ARPKD. We further validated these findings through loss-of-function studies in mice and zebrafish. DZIP1L localizes to centrioles and to the distal ends of basal bodies, and interacts with septin2, a protein implicated in maintenance of the periciliary diffusion barrier at the ciliary transition zone. In agreement with a defect in the diffusion barrier, we found that the ciliary-membrane translocation of the PKD proteins polycystin-1 and polycystin-2 is compromised in DZIP1L-mutant cells. Together, these data provide what is, to our knowledge, the first conclusive evidence that ARPKD is not a homogeneous disorder and further establish DZIP1L as a second gene involved in ARPKD pathogenesis.
Collapse
Affiliation(s)
- Hao Lu
- Institute of Molecular and Cell Biology, Singapore
| | - Maria C Rondón Galeano
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Elisabeth Ott
- Department of Medicine IV, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Geraldine Kaeslin
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | | | - Carina Kramer
- Department of Medicine IV, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | | - Nadescha Hilger
- Institute of Human Genetics, RWTH Aachen University, Aachen, Germany
| | - Vicki Metzis
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Milan Hiersche
- Center for Human Genetics, Bioscientia, Ingelheim, Germany
| | | | - Robert Tunningley
- John Curtin School of Medical Research, Australian National University, Acton, Australian Capital Territory, Australia
| | - Shubha Vij
- Institute of Molecular and Cell Biology, Singapore
| | - Andrew D Courtney
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Belinda Whittle
- John Curtin School of Medical Research, Australian National University, Acton, Australian Capital Territory, Australia
| | - Elke Wühl
- Division of Pediatric Nephrology, University Children's Hospital Center for Child and Adolescent Medicine, Heidelberg University Hospital, Heidelberg, Germany
| | - Udo Vester
- Department of Pediatric Nephrology, University Children's Hospital Essen, Essen, Germany
| | - Björn Hartleben
- Institute of Pathology, MHH University Medical School Hannover, Hannover, Germany
| | - Steffen Neuber
- Center for Human Genetics, Bioscientia, Ingelheim, Germany
| | - Valeska Frank
- Center for Human Genetics, Bioscientia, Ingelheim, Germany
| | - Melissa H Little
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Daniel Epting
- Department of Medicine IV, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Peter Papathanasiou
- John Curtin School of Medical Research, Australian National University, Acton, Australian Capital Territory, Australia
| | - Andrew C Perkins
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia.,Mater Research Institute, Faculty of Medicine and Biomedical Sciences, The University of Queensland, Woolloongabba, Queensland, Australia
| | | | - Walter Hunziker
- Institute of Molecular and Cell Biology, Singapore.,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Singapore Eye Research Institute, Singapore
| | - Heon Yung Gee
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA.,Department of Pharmacology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Edgar A Otto
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan, USA
| | - Klaus Zerres
- Institute of Human Genetics, RWTH Aachen University, Aachen, Germany
| | - Friedhelm Hildebrandt
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Sudipto Roy
- Institute of Molecular and Cell Biology, Singapore.,Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Department of Biological Sciences, National University of Singapore, Singapore
| | - Carol Wicking
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Carsten Bergmann
- Department of Medicine IV, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Institute of Human Genetics, RWTH Aachen University, Aachen, Germany.,Center for Human Genetics, Bioscientia, Ingelheim, Germany
| |
Collapse
|
29
|
Bangs F, Anderson KV. Primary Cilia and Mammalian Hedgehog Signaling. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a028175. [PMID: 27881449 DOI: 10.1101/cshperspect.a028175] [Citation(s) in RCA: 448] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
It has been a decade since it was discovered that primary cilia have an essential role in Hedgehog (Hh) signaling in mammals. This discovery came from screens in the mouse that identified a set of genes that are required for both normal Hh signaling and for the formation of primary cilia. Since then, dozens of mouse mutations have been identified that disrupt cilia in a variety of ways and have complex effects on Hedgehog signaling. Here, we summarize the genetic and developmental studies used to deduce how Hedgehog signal transduction is linked to cilia and the complex effects that perturbation of cilia structure can have on Hh signaling. We conclude by describing the current status of our understanding of the cell-type-specific regulation of ciliogenesis and how that determines the ability of cells to respond to Hedgehog ligands.
Collapse
Affiliation(s)
- Fiona Bangs
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| | - Kathryn V Anderson
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| |
Collapse
|
30
|
Zhang B, Wang G, Xu X, Yang S, Zhuang T, Wang G, Ren H, Cheng SY, Jiang Q, Zhang C. DAZ-interacting Protein 1 (Dzip1) Phosphorylation by Polo-like Kinase 1 (Plk1) Regulates the Centriolar Satellite Localization of the BBSome Protein during the Cell Cycle. J Biol Chem 2017; 292:1351-1360. [PMID: 27979967 PMCID: PMC5270478 DOI: 10.1074/jbc.m116.765438] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 12/13/2016] [Indexed: 12/29/2022] Open
Abstract
The function of the primary cilia, which is assembled in most vertebrate cells, is achieved by transport in and out of kinds of signaling receptors. The BBSome protein complex could recognize and target membrane proteins to the cilia, but how the BBSome itself is transported into the cilia is poorly understood. Here we demonstrate that the centrosome protein Dzip1 mediates the assembly of the BBSome-Dzip1-PCM1 complex in the centriolar satellites (CS) at the G0 phase for ciliary translocation of the BBSome. Phosphorylation of Dzip1 at Ser-210 by Plk1 (polo-like kinase 1) during the G2 phase promotes disassembly of this complex, resulting in removal of Dzip1 and the BBSome from the CS. Inhibiting the kinase activity of Plk1 maintains the CS localization of the BBSome and Dzip1 at the G2 phase. Collectively, our findings reveal the cell cycle-dependent regulation of BBSome transport to the CS and highlight a potential mechanism that the BBSome-mediated signaling pathways are accordingly regulated during the cell cycle.
Collapse
Affiliation(s)
- Boyan Zhang
- From the Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education and the State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing 100871 and
| | - Gang Wang
- From the Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education and the State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing 100871 and
| | - Xiaowei Xu
- From the Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education and the State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing 100871 and
| | - Sisi Yang
- From the Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education and the State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing 100871 and
| | - Tenghan Zhuang
- From the Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education and the State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing 100871 and
| | - Guopeng Wang
- From the Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education and the State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing 100871 and
| | - He Ren
- From the Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education and the State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing 100871 and
| | - Steven Y Cheng
- the Department of Developmental Genetics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing 210029, China
| | - Qing Jiang
- From the Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education and the State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing 100871 and
| | - Chuanmao Zhang
- From the Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education and the State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing 100871 and
| |
Collapse
|
31
|
BAR Domain-Containing FAM92 Proteins Interact with Chibby1 To Facilitate Ciliogenesis. Mol Cell Biol 2016; 36:2668-2680. [PMID: 27528616 DOI: 10.1128/mcb.00160-16] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 08/05/2016] [Indexed: 02/03/2023] Open
Abstract
Chibby1 (Cby1) is a small, conserved coiled-coil protein that localizes to centrioles/basal bodies and plays a crucial role in the formation and function of cilia. During early stages of ciliogenesis, Cby1 is required for the efficient recruitment of small vesicles at the distal end of centrioles to facilitate basal body docking to the plasma membrane. Here, we identified family with sequence similarity 92, member A (FAM92A) and FAM92B, which harbor predicted lipid-binding BAR domains, as novel Cby1-interacting partners using tandem affinity purification and mass spectrometry. We found that in cultured cell lines, FAM92A colocalizes with Cby1 at the centrioles/basal bodies of primary cilia, while FAM92B is undetectable. In airway multiciliated cells, both FAM92A and -92B colocalize with Cby1 at the base of cilia. Notably, the centriolar localization of FAM92A and -92B depends largely on Cby1. Knockdown of FAM92A in RPE1 cells impairs ciliogenesis. Consistent with the membrane-remodeling properties of BAR domains, FAM92A and -92B in cooperation with Cby1 induce deformed membrane-like structures containing the small GTPase Rab8 in cultured cells. Our results therefore suggest that FAM92 proteins interact with Cby1 to promote ciliogenesis via regulation of membrane-remodeling processes.
Collapse
|
32
|
Wei Q, Zhang Y, Schouteden C, Zhang Y, Zhang Q, Dong J, Wonesch V, Ling K, Dammermann A, Hu J. The hydrolethalus syndrome protein HYLS-1 regulates formation of the ciliary gate. Nat Commun 2016; 7:12437. [PMID: 27534274 PMCID: PMC4992140 DOI: 10.1038/ncomms12437] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 06/30/2016] [Indexed: 12/18/2022] Open
Abstract
Transition fibres (TFs), together with the transition zone (TZ), are basal ciliary structures thought to be crucial for cilium biogenesis and function by acting as a ciliary gate to regulate selective protein entry and exit. Here we demonstrate that the centriolar and basal body protein HYLS-1, the C. elegans orthologue of hydrolethalus syndrome protein 1, is required for TF formation, TZ organization and ciliary gating. Loss of HYLS-1 compromises the docking and entry of intraflagellar transport (IFT) particles, ciliary gating for both membrane and soluble proteins, and axoneme assembly. Additional depletion of the TF component DYF-19 in hyls-1 mutants further exacerbates TZ anomalies and completely abrogates ciliogenesis. Our data support an important role for HYLS-1 and TFs in establishment of the ciliary gate and underline the importance of selective protein entry for cilia assembly.
Collapse
Affiliation(s)
- Qing Wei
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota 55905, USA.,Key Laboratory of Insect Developmental and Evolutionary Biology, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| | - Yingyi Zhang
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota 55905, USA
| | - Clementine Schouteden
- Max F. Perutz Laboratories, Vienna Biocenter (VBC), University of Vienna, A-1030 Vienna, Austria
| | - Yuxia Zhang
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota 55905, USA
| | - Qing Zhang
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota 55905, USA
| | - Jinhong Dong
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota 55905, USA
| | - Veronika Wonesch
- Max F. Perutz Laboratories, Vienna Biocenter (VBC), University of Vienna, A-1030 Vienna, Austria
| | - Kun Ling
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota 55905, USA
| | - Alexander Dammermann
- Max F. Perutz Laboratories, Vienna Biocenter (VBC), University of Vienna, A-1030 Vienna, Austria
| | - Jinghua Hu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota 55905, USA.,Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota 55905, USA.,Mayo Translational PKD Center, Mayo Clinic, Rochester, Minnesota 55905, USA
| |
Collapse
|
33
|
Emechebe U, Kumar P P, Rozenberg JM, Moore B, Firment A, Mirshahi T, Moon AM. T-box3 is a ciliary protein and regulates stability of the Gli3 transcription factor to control digit number. eLife 2016; 5. [PMID: 27046536 PMCID: PMC4829432 DOI: 10.7554/elife.07897] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 03/05/2016] [Indexed: 12/17/2022] Open
Abstract
Crucial roles for T-box3 in development are evident by severe limb malformations and other birth defects caused by T-box3 mutations in humans. Mechanisms whereby T-box3 regulates limb development are poorly understood. We discovered requirements for T-box at multiple stages of mouse limb development and distinct molecular functions in different tissue compartments. Early loss of T-box3 disrupts limb initiation, causing limb defects that phenocopy Sonic Hedgehog (Shh) mutants. Later ablation of T-box3 in posterior limb mesenchyme causes digit loss. In contrast, loss of anterior T-box3 results in preaxial polydactyly, as seen with dysfunction of primary cilia or Gli3-repressor. Remarkably, T-box3 is present in primary cilia where it colocalizes with Gli3. T-box3 interacts with Kif7 and is required for normal stoichiometry and function of a Kif7/Sufu complex that regulates Gli3 stability and processing. Thus, T-box3 controls digit number upstream of Shh-dependent (posterior mesenchyme) and Shh-independent, cilium-based (anterior mesenchyme) Hedgehog pathway function. DOI:http://dx.doi.org/10.7554/eLife.07897.001 Mutations in the gene that encodes a protein called T-box3 cause serious birth defects, including deformities of the hands and feet, via poorly understood mechanisms. Several other proteins are also important for ensuring that limbs develop correctly. These include the Sonic Hedgehog protein, which controls a signaling pathway that determines whether a protein called Gli3 is converted into its “repressor” form. The hair-like structures called primary cilia that sit on the surface of animal cells also contain Gli3, and processes within these structures control the production of the Gli3-repressor. Emechebe, Kumar et al. have now studied genetically engineered mice in which the production of the T-box3 protein was stopped at different stages of mouse development. This revealed that turning off T-box3 production early in development causes many parts of the limb not to form. This type of defect appears to be the same as that seen in mice that lack the Sonic Hedgehog protein. If the production of T-box3 is turned off later in mouse development in the rear portion of the developing limb, the limb starts to develop but doesn’t develop enough rear toes. When T-box3 production is turned off in the front portion of the developing limbs, mice are born with too many front toes. This latter problem mimics the effects seen in mice that are unable to produce Gli3-repressor or that have defective primary cilia. Further investigation unexpectedly revealed that T-box3 is found in primary cilia and localizes to the same regions of the cilia as the Gli3-repressor. Furthermore, T-box3 also interacts with a protein complex that controls the stability of Gli3 and processes it into the Gli3-repressor form. In the future, it will be important to determine how T-box3 controls the stability of Gli3 and whether that process occurs in the primary cilia or in other parts of the cell where T-box3 and Gli3 coexist, such as the nucleus. This could help us understand how T-box3 and Sonic Hedgehog signaling contribute to other aspects of development and to certain types of cancer. DOI:http://dx.doi.org/10.7554/eLife.07897.002
Collapse
Affiliation(s)
- Uchenna Emechebe
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, United States
| | - Pavan Kumar P
- Weis Center for Research, Geisinger Clinic, Danville, United States
| | | | - Bryn Moore
- Weis Center for Research, Geisinger Clinic, Danville, United States
| | - Ashley Firment
- Weis Center for Research, Geisinger Clinic, Danville, United States
| | - Tooraj Mirshahi
- Weis Center for Research, Geisinger Clinic, Danville, United States
| | - Anne M Moon
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, United States.,Weis Center for Research, Geisinger Clinic, Danville, United States.,Department of Human Genetics, University of Utah, Salt Lake City, United States.,Department of Pediatrics, University of Utah, Salt Lake City, United States
| |
Collapse
|
34
|
Wei Q, Ling K, Hu J. The essential roles of transition fibers in the context of cilia. Curr Opin Cell Biol 2015; 35:98-105. [PMID: 25988548 DOI: 10.1016/j.ceb.2015.04.015] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 04/18/2015] [Accepted: 04/23/2015] [Indexed: 12/18/2022]
Abstract
Once thought of as a vestigial organelle, the primary cilium is now recognized as a signaling hub for key cellular pathways in vertebrate development. The recent renaissance in cilia studies significantly improved our understanding of how cilia form and function, but little is known about how ciliogenesis is initiated and how ciliary proteins enter cilia. These important ciliary events require transition fibers (TFs) that are positioned at the ciliary base as symmetric nine-bladed propeller fibrous structures. Up until recently, TFs have been the most underappreciated ciliary structures due to limited knowledge about their molecular composition and function. Here, we highlight recent advances in our understanding of TF composition and the indispensable roles of TFs in regulating the initiation of ciliogenesis and the selective import of ciliary proteins.
Collapse
Affiliation(s)
- Qing Wei
- Department of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA; Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Kun Ling
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Jinghua Hu
- Department of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA; Mayo Translational PKD Center, Rochester, MN, USA; Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
35
|
Zhang B, Zhang T, Wang G, Wang G, Chi W, Jiang Q, Zhang C. GSK3β-Dzip1-Rab8 cascade regulates ciliogenesis after mitosis. PLoS Biol 2015; 13:e1002129. [PMID: 25860027 PMCID: PMC4393111 DOI: 10.1371/journal.pbio.1002129] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 03/13/2015] [Indexed: 11/17/2022] Open
Abstract
The primary cilium, which disassembles before mitotic entry and reassembles after mitosis, organizes many signal transduction pathways that are crucial for cell life and individual development. However, how ciliogenesis is regulated during the cell cycle remains largely unknown. Here we show that GSK3β, Dzip1, and Rab8 co-regulate ciliogenesis by promoting the assembly of the ciliary membrane after mitosis. Immunofluorescence and super-resolution microscopy showed that Dzip1 was localized to the periciliary diffusion barrier and enriched at the mother centriole. Knockdown of Dzip1 by short hairpin RNAs led to failed ciliary localization of Rab8, and Rab8 accumulation at the basal body. Dzip1 preferentially bound to Rab8GDP and promoted its dissociation from its inhibitor GDI2 at the pericentriolar region, as demonstrated by sucrose gradient centrifugation of purified basal bodies, immunoprecipitation, and acceptor-bleaching fluorescence resonance energy transfer assays. By means of in vitro phosphorylation, in vivo gel shift, phospho-peptide identification by mass spectrometry, and GST pulldown assays, we demonstrated that Dzip1 was phosphorylated by GSK3β at S520 in G0 phase, which increased its binding to GDI2 to promote the release of Rab8GDP at the cilium base. Moreover, ciliogenesis was inhibited by overexpression of the GSK3β-nonphosphorylatable Dzip1 mutant or by disabling of GSK3β by specific inhibitors or knockout of GSK3β in cells. Collectively, our data reveal a unique cascade consisting of GSK3β, Dzip1, and Rab8 that regulates ciliogenesis after mitosis.
Collapse
Affiliation(s)
- Boyan Zhang
- The Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education and the State Key Laboratory of Biomembrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing, China
| | - Tingting Zhang
- The Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education and the State Key Laboratory of Biomembrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing, China
| | - Guopeng Wang
- The Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education and the State Key Laboratory of Biomembrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing, China
| | - Gang Wang
- The Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education and the State Key Laboratory of Biomembrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing, China
| | - Wangfei Chi
- The Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education and the State Key Laboratory of Biomembrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing, China
| | - Qing Jiang
- The Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education and the State Key Laboratory of Biomembrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing, China
| | - Chuanmao Zhang
- The Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education and the State Key Laboratory of Biomembrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing, China
| |
Collapse
|
36
|
Di Gioia SA, Farinelli P, Letteboer SJF, Arsenijevic Y, Sharon D, Roepman R, Rivolta C. Interactome analysis reveals that FAM161A, deficient in recessive retinitis pigmentosa, is a component of the Golgi-centrosomal network. Hum Mol Genet 2015; 24:3359-71. [PMID: 25749990 DOI: 10.1093/hmg/ddv085] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Accepted: 03/04/2015] [Indexed: 11/13/2022] Open
Abstract
Defects in FAM161A, a protein of unknown function localized at the cilium of retinal photoreceptor cells, cause retinitis pigmentosa, a form of hereditary blindness. By using different fragments of this protein as baits to screen cDNA libraries of human and bovine retinas, we defined a yeast two-hybrid-based FAM161A interactome, identifying 53 bona fide partners. In addition to statistically significant enrichment in ciliary proteins, as expected, this interactome revealed a substantial bias towards proteins from the Golgi apparatus, the centrosome and the microtubule network. Validation of interaction with key partners by co-immunoprecipitation and proximity ligation assay confirmed that FAM161A is a member of the recently recognized Golgi-centrosomal interactome, a network of proteins interconnecting Golgi maintenance, intracellular transport and centrosome organization. Notable FAM161A interactors included AKAP9, FIP3, GOLGA3, KIFC3, KLC2, PDE4DIP, NIN and TRIP11. Furthermore, analysis of FAM161A localization during the cell cycle revealed that this protein followed the centrosome during all stages of mitosis, likely reflecting a specific compartmentalization related to its role at the ciliary basal body during the G0 phase. Altogether, these findings suggest that FAM161A's activities are probably not limited to ciliary tasks but also extend to more general cellular functions, highlighting possible novel mechanisms for the molecular pathology of retinal disease.
Collapse
Affiliation(s)
| | - Pietro Farinelli
- Department of Medical Genetics, University of Lausanne, Lausanne, Switzerland
| | - Stef J F Letteboer
- Department of Human Genetics and Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands and
| | - Yvan Arsenijevic
- Unit of Gene Therapy and Stem Cell Biology, Jules-Gonin Eye Hospital, University of Lausanne, Lausanne, Switzerland
| | - Dror Sharon
- Department of Ophthalmology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Ronald Roepman
- Department of Human Genetics and Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands and
| | - Carlo Rivolta
- Department of Medical Genetics, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
37
|
Arnold CR, Lamont RE, Walker JT, Spice PJ, Chan CK, Ho CY, Childs SJ. Comparative analysis of genes regulated by Dzip1/iguana and hedgehog in zebrafish. Dev Dyn 2015; 244:211-23. [PMID: 25476803 DOI: 10.1002/dvdy.24237] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 11/04/2014] [Accepted: 11/30/2014] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND The zebrafish genetic mutant iguana (igu) has defects in the ciliary basal body protein Dzip1, causing improper cilia formation. Dzip1 also interacts with the downstream transcriptional activators of Hedgehog (Hh), the Gli proteins, and Hh signaling is disrupted in igu mutants. Hh governs a wide range of developmental processes, including stabilizing developing blood vessels to prevent hemorrhage. Using igu mutant embryos and embryos treated with the Hh pathway antagonist cyclopamine, we conducted a microarray to determine genes involved in Hh signaling mediating vascular stability. RESULTS We identified 40 genes with significantly altered expression in both igu mutants and cyclopamine-treated embryos. For a subset of these, we used in situ hybridization to determine localization during embryonic development and confirm the expression changes seen on the array. CONCLUSIONS Through comparing gene expression changes in a genetic model of vascular instability with a chemical inhibition of Hh signaling, we identified a set of 40 differentially expressed genes with potential roles in vascular stabilization.
Collapse
Affiliation(s)
- Corey R Arnold
- Department of Biochemistry and Molecular Biology and Alberta Children's Hospital Research Institute, University of Calgary, Canada
| | | | | | | | | | | | | |
Collapse
|
38
|
A function for the Joubert syndrome protein Arl13b in ciliary membrane extension and ciliary length regulation. Dev Biol 2014; 397:225-36. [PMID: 25448689 DOI: 10.1016/j.ydbio.2014.11.009] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 10/31/2014] [Accepted: 11/11/2014] [Indexed: 11/20/2022]
Abstract
Cilia perform a variety of functions in a number of developmental and physiological contexts, and are implicated in the pathogenesis of a wide spectrum of human disorders. While the ciliary axoneme is assembled by intraflagellar transport, how ciliary membrane length is regulated is not completely understood. Here, we show that zebrafish embryos as well as mammalian cells overexpressing the ciliary membrane protein Arl13b, an ARF family small GTPase that is essential for ciliary differentiation, showed pronounced increase in ciliary length. Intriguingly, this increase in cilia length occurred as a function of the amounts of overexpressed Arl13b. While the motility of Arl13b overexpressing excessively long motile cilia was obviously disrupted, surprisingly, the abnormally long immotile primary cilia seemed to retain their signaling capacity. arl13b is induced by FoxJ1 and Rfx, and these ciliogenic transcription factors are unable to promote ciliary length increase when Arl13b activity is inhibited. Conversely, overexpression of Arl13b was sufficient to restore ciliary length in zebrafish embryos deficient in FoxJ1 function. We show that Arl13b increases cilia length by inducing protrusion of the ciliary membrane, which is then followed by the extension of the axonemal microtubules. Using mutant versions of Arl13b, one of which has been shown to be causative of the ciliopathy Joubert syndrome, we establish that the GTPase activity of the protein is essential for ciliary membrane extension. Taken together, our findings identify Arl13b as an important effector of ciliary membrane biogenesis and ciliary length regulation, and provide insights into possible mechanisms of dysfunction of the protein in Joubert syndrome.
Collapse
|
39
|
Wu C, Yang M, Li J, Wang C, Cao T, Tao K, Wang B. Talpid3-binding centrosomal protein Cep120 is required for centriole duplication and proliferation of cerebellar granule neuron progenitors. PLoS One 2014; 9:e107943. [PMID: 25251415 PMCID: PMC4176001 DOI: 10.1371/journal.pone.0107943] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 08/18/2014] [Indexed: 02/06/2023] Open
Abstract
Granule neuron progenitors (GNPs) are the most abundant neuronal type in the cerebellum. GNP proliferation and thus cerebellar development require Sonic hedgehog (Shh) secreted from Purkinje cells. Shh signaling occurs in primary cilia originating from the mother centriole. Centrioles replicate only once during a typical cell cycle and are responsible for mitotic spindle assembly and organization. Recent studies have linked cilia function to cerebellar morphogenesis, but the role of centriole duplication in cerebellar development is not known. Here we show that centrosomal protein Cep120 is asymmetrically localized to the daughter centriole through its interaction with Talpid3 (Ta3), another centrosomal protein. Cep120 null mutant mice die in early gestation with abnormal heart looping. Inactivation of Cep120 in the central nervous system leads to both hydrocephalus, due to the loss of cilia on ependymal cells, and severe cerebellar hypoplasia, due to the failed proliferation of GNPs. The mutant GNPs lack Hedgehog pathway activity. Cell biological studies show that the loss of Cep120 results in failed centriole duplication and consequently ciliogenesis, which together underlie Cep120 mutant cerebellar hypoplasia. Thus, our study for the first time links a centrosomal protein necessary for centriole duplication to cerebellar morphogenesis.
Collapse
Affiliation(s)
- Chuanqing Wu
- Department of General Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Department of Genetic Medicine, Weill Medical College of Cornell University, New York, New York, United States of America
| | - Mei Yang
- Department of Genetic Medicine, Weill Medical College of Cornell University, New York, New York, United States of America
- Department of Human Anatomy, Institute of Neuroscience, Chongqing Medical University, Chongqing, China
| | - Juan Li
- Department of Genetic Medicine, Weill Medical College of Cornell University, New York, New York, United States of America
- Institute of Developmental Immunology, College of Life Science, Shandong University, Jinan, Shandong, China
| | - Chengbing Wang
- Department of Genetic Medicine, Weill Medical College of Cornell University, New York, New York, United States of America
| | - Ting Cao
- Department of Genetic Medicine, Weill Medical College of Cornell University, New York, New York, United States of America
- Institute of Life Science, Nanjing University, Nanjing, Jiangsu, China
| | - Kaixiong Tao
- Department of General Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Baolin Wang
- Department of Genetic Medicine, Weill Medical College of Cornell University, New York, New York, United States of America
- Department of Cell Biology and Development, Weill Medical College of Cornell University, New York, New York, United States of America
| |
Collapse
|
40
|
Shigunov P, Sotelo-Silveira J, Stimamiglio MA, Kuligovski C, Irigoín F, Badano JL, Munroe D, Correa A, Dallagiovanna B. Ribonomic analysis of human DZIP1 reveals its involvement in ribonucleoprotein complexes and stress granules. BMC Mol Biol 2014; 15:12. [PMID: 24993635 PMCID: PMC4091656 DOI: 10.1186/1471-2199-15-12] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Accepted: 06/19/2014] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND DZIP1 (DAZ-interacting protein 1) has been described as a component of the Hh signaling pathway with a putative regulatory role in ciliogenesis. DZIP1 interacts with DAZ RNA binding proteins in embryonic stem cells and human germ cells suggesting a role in mRNA regulation. RESULTS We investigated DZIP1 function in HeLa cells and its involvement in ribonucleoprotein complexes. DZIP1 was predominantly located in granules in the cytoplasm. Under oxidative stress conditions, DZIP1 re-localized to stress granules. DZIP appears to be important for the formation of stress granules during the stress response. We used immunoprecipitation assays with antibodies against DZIP1 and microarray hybridization to identify mRNAs associated with DZIP1. The genetic networks formed by the DZIP1-associated mRNAs were involved in cell cycle and gene expression regulation. DZIP1 is involved in the Hedgehog signaling pathway. We used cyclopamine, a specific inhibitor of this pathway, to analyze the expression of DZIP1 and its associated mRNAs. The abundance of DZIP1-associated mRNAs increased with treatment; however, the silencing or overexpression of DZIP1 in HeLa cells had no effect on the accumulation of the associated mRNAs. Polysomal profile analysis by sucrose gradient centrifugation demonstrated the presence of DZIP1 in the polysomal fraction. CONCLUSIONS Our results suggest that DZIP1 is part of an RNP complex that occupies various subcellular locations. The diversity of the mRNAs associated with DZIP1 suggests that this protein is a component of different RNPs associated with translating polysomes and with RNA granules.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Bruno Dallagiovanna
- Stem Cells Basic Biology Laboratory, Instituto Carlos Chagas, FIOCRUZ, Algacyr Munhoz Mader 3775, Curitiba 81350-010, Brazil.
| |
Collapse
|
41
|
Collins MM, Ryan AK. Are there conserved roles for the extracellular matrix, cilia, and junctional complexes in left-right patterning? Genesis 2014; 52:488-502. [PMID: 24668924 DOI: 10.1002/dvg.22774] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Accepted: 03/19/2014] [Indexed: 01/11/2023]
Abstract
Many different types of molecules have essential roles in patterning the left-right axis and directing asymmetric morphogenesis. In particular, the relationship between signaling molecules and transcription factors has been explored extensively. Another group of proteins implicated in left-right patterning are components of the extracellular matrix, apical junctions, and cilia. These structural molecules have the potential to participate in the conversion of morphogenetic cues from the extracellular environment into morphogenetic patterning via their interactions with the actin cytoskeleton. Although it has been relatively easy to temporally position these proteins within the hierarchy of the left-right patterning pathway, it has been more difficult to define how they mechanistically fit into these pathways. Consequently, our understanding of how these factors impart patterning information to influence the establishment of the left-right axis remains limited. In this review, we will discuss those structural molecules that have been implicated in early phases of left-right axis development.
Collapse
Affiliation(s)
- Michelle M Collins
- Department of Human Genetics, McGill University, Montréal, Québec, Canada
| | | |
Collapse
|
42
|
C2cd3 is critical for centriolar distal appendage assembly and ciliary vesicle docking in mammals. Proc Natl Acad Sci U S A 2014; 111:2164-9. [PMID: 24469809 DOI: 10.1073/pnas.1318737111] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The primary cilium plays critical roles in vertebrate development and physiology, but the mechanisms underlying its biogenesis remain poorly understood. We investigated the molecular function of C2 calcium-dependent domain containing 3 (C2cd3), an essential regulator of primary cilium biogenesis. We show that C2cd3 is localized to the centriolar satellites in a microtubule- and Pcm1-dependent manner; however, C2cd3 is dispensable for centriolar satellite integrity. C2cd3 is also localized to the distal ends of both mother and daughter centrioles and is required for the recruitment of five centriolar distal appendage proteins: Sclt1, Ccdc41, Cep89, Fbf1, and Cep164. Furthermore, loss of C2cd3 results in failure in the recruitment of Ttbk2 to the ciliary basal body as well as the removal of Cp110 from the ciliary basal body, two critical steps in initiating ciliogenesis. C2cd3 is also required for recruiting the intraflagellar transport proteins Ift88 and Ift52 to the mother centriole. Consistent with a role in distal appendage assembly, C2cd3 is essential for ciliary vesicle docking to the mother centriole. Our results suggest that C2cd3 regulates cilium biogenesis by promoting the assembly of centriolar distal appendages critical for docking ciliary vesicles and recruiting other essential ciliogenic proteins.
Collapse
|
43
|
Schwend T, Jin Z, Jiang K, Mitchell BJ, Jia J, Yang J. Stabilization of speckle-type POZ protein (Spop) by Daz interacting protein 1 (Dzip1) is essential for Gli turnover and the proper output of Hedgehog signaling. J Biol Chem 2013; 288:32809-32820. [PMID: 24072710 DOI: 10.1074/jbc.m113.512962] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The Hedgehog (Hh) pathway is essential for embryonic development and adult tissue homeostasis. The Gli/Cubitus interruptus (Ci) family of transcription factors acts at the downstream end of the pathway to mediate Hh signaling. Both Hh-dependent and -independent Gli regulatory mechanisms are important for the output of Hh signaling. Daz interacting protein 1 (Dzip1) has bipartite positive and negative functions in the Hh pathway. The positive Hh regulatory function appears to be attributed to a requirement for Dzip1 during ciliogenesis. The mechanism by which Dzip1 inhibits Hh signaling, however, remains largely unclear. We recently found that Dzip1 is required for Gli turnover, which may account for its inhibitory function in Hh signaling. Here, we report that Dzip1 regulates Gli/Ci turnover by preventing degradation of speckle-type POZ protein (Spop), a protein that promotes proteasome-dependent turnover of Gli proteins. We provide evidence that Dzip1 regulates the stability of Spop independent of its function in ciliogenesis. Partial knockdown of Dzip1 to levels insufficient for perturbing ciliogenesis, sensitized Xenopus embryos to Hh signaling, leading to phenotypes that resemble activation of Hh signaling. Importantly, overexpression of Spop was able to restore proper Gli protein turnover and rescue phenotypes in Dzip1-depleted embryos. Consistently, depletion of Dzip1 in Drosophila S2 cells destabilized Hh-induced BTB protein (HIB), the Drosophila homolog of Spop, and increased the level of Ci. Thus, Dzip1-dependent stabilization of Spop/HIB is evolutionarily conserved and essential for proper regulation of Gli/Ci proteins in the Hh pathway.
Collapse
Affiliation(s)
- Tyler Schwend
- From the Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois 61802
| | - Zhigang Jin
- From the Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois 61802
| | - Kai Jiang
- Markey Cancer Center, Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky 40536-0509
| | - Brian J Mitchell
- Department of Cell and Molecular Biology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois 60611
| | - Jianhang Jia
- Markey Cancer Center, Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky 40536-0509
| | - Jing Yang
- From the Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois 61802,.
| |
Collapse
|