1
|
Guo Y, He X, Liu J, Tan Y, Zhang C, Chen S, Zhang S. The relationship between HYDIN and fallopian tubal cilia loss in patients with epithelial ovarian cancer. Front Oncol 2025; 14:1495753. [PMID: 39850822 PMCID: PMC11754247 DOI: 10.3389/fonc.2024.1495753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 12/09/2024] [Indexed: 01/25/2025] Open
Abstract
Introduction Primary cilia play an important role in the development of cancer by regulating signaling pathways. Several studies have demonstrated that women with BRCA mutations have, on average, 50% fewer ciliated cells compared with general women. However, the role of tubal cilia loss in the development of epithelial ovarian cancer (EOC) remains unclear. Few specific studies have been found in linking HYDIN, a ciliary defect associated gene that encodes HYDIN axonemal central pair apparatus protein, which is involved in the transduction of Hedgehog (Hh) signal and is considered a cancer associated antigen, to ovarian cancer. Therefore, our study aimed to investigate the correlation between HYDIN gene mutations and tubal cilia loss in EOC. Methods A whole exome sequencing (WES), immunohistochemistry (IHC), western blot, and reverse transcription quantitative (RT q) PCR were performed in 80 patients with EOC and 50 cases of non ovarian cancer to detect the mutations and expression of tubal ciliary marker, ciliary morphology, and abnormal rate. Results We found that the incidence of tubal cilia loss was higher in EOC group with decreased expression of HYDIN compared with the control group (P<0.05). Discussion This study suggests that tubal ciliary loss is evident in epithelial fallopian tube carcinoma, and ciliary cells may be involved in the occurrence and development of EOC, and cilia-related gene HYDIN is expected to be a tumor marker for epithelial ovarian cancer.
Collapse
Affiliation(s)
- Yuanli Guo
- Department of Pathology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Xinxin He
- Department of Pathology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Junfeng Liu
- Department of Pathology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | | | | | - Shan Chen
- Department of Gynecology, The Six Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Sheng Zhang
- Department of Pathology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| |
Collapse
|
2
|
Yavuz Saricay L, Baldwin G, Moulton EA, Gonzalez E, Rajabi F, Hunter DG, Fulton AB. Refractive errors in patients with Bardet Biedl syndrome. Ophthalmic Genet 2024; 45:435-440. [PMID: 38953718 DOI: 10.1080/13816810.2024.2357296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 05/04/2024] [Accepted: 05/08/2024] [Indexed: 07/04/2024]
Abstract
PURPOSE Bardet-Biedl Syndrome (BBS) is a rare autosomal recessive ciliopathy. Within corneal development, primary cilia serve a critical role. We sought to investigate the association of BBS with corneal astigmatism among a cohort of patients with BBS. METHODS This was a cross-sectional, retrospective study performed at a pediatric ophthalmology department of a tertiary hospital. The study enrolled 45 patients with genetically confirmed Bardet-Biedl syndrome, encompassing a total of 90 eyes observed from February 2011 to August 2021. Spherical and cylindrical refractive errors and keratometry outcome measures, including diopter (D) values at the flattest and steepest axes, were recorded. Corneal astigmatism of greater than 3D is considered extreme corneal astigmatism based on previously published data. RESULTS Among 45 patients (M:26; F:19), the mean age was 16.4 ± 8.2 years, and the mean best-corrected visual acuity was 20/60. The most common molecular diagnosis was BBS1, seen in 24 of 45 (53.3%). Among all the patients, the mean spherical refractive error was -2.9 ± 3.8D. The mean cylindrical refractive error was 2.6 ± 1.5D. The mean keratometry values at the flattest axis was 43.5 ± 5.3D (39.4-75.0) and at the steepest axis was 47.2 ± 7.3D(41.5-84.0). Among all the patients with BBS, the mean corneal astigmatism was 3.7 ± 1.0D(0.5-7.1), which is considered extreme. CONCLUSION A cohort of individuals with BBS demonstrated high corneal astigmatism. These results suggest an association between corneal astigmatism and primary ciliary dysfunction and may assist in clinical management and future therapeutic targets among BBS and other corneal disorders.
Collapse
Affiliation(s)
- Leyla Yavuz Saricay
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Eric A Moulton
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Anesthesia, Critical Care and Pain Medicine, Boston Children's Hospital, Harvard Medical School, Brain and Eye Pain Imaging Lab, Pain and Affective Neuroscience Center, Boston, Massachusetts, USA
| | - Efren Gonzalez
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Farah Rajabi
- Department of Pediatrics, Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - David G Hunter
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Anne B Fulton
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
3
|
Zhong BH, Dong M. The implication of ciliary signaling pathways for epithelial-mesenchymal transition. Mol Cell Biochem 2024; 479:1535-1543. [PMID: 37490178 PMCID: PMC11224103 DOI: 10.1007/s11010-023-04817-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 07/15/2023] [Indexed: 07/26/2023]
Abstract
Epithelial-to-mesenchymal transition (EMT), which plays an essential role in development, tissue repair and fibrosis, and cancer progression, is a reversible cellular program that converts epithelial cells to mesenchymal cell states characterized by motility-invasive properties. The mostly signaling pathways that initiated and controlled the EMT program are regulated by a solitary, non-motile organelle named primary cilium. Acting as a signaling nexus, primary cilium dynamically concentrates signaling molecules to respond to extracellular cues. Recent research has provided direct evidence of connection between EMT and primary ciliogenesis in multiple contexts, but the mechanistic understanding of this relationship is complicated and still undergoing. In this review, we describe the current knowledge about the ciliary signaling pathways involved in EMT and list the direct evidence that shows the link between them, trying to figure out the intricate relationship between EMT and primary ciliogenesis, which may aid the future development of primary cilium as a novel therapeutic approach targeted to EMT.
Collapse
Affiliation(s)
- Bang-Hua Zhong
- Department of Gastrointestinal Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Ming Dong
- Department of Gastrointestinal Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
4
|
Ni H, Li L, Hu D, Yang M, Wang D, Ma H, Bu W, Yang J, Zhu LE, Zhai D, Song T, Yang S, Lu Q, Li D, Ran J, Liu M. Dynamic changes of endothelial and stromal cilia are required for the maintenance of corneal homeostasis. J Cell Physiol 2024; 239:e31215. [PMID: 38308657 DOI: 10.1002/jcp.31215] [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: 11/07/2023] [Revised: 01/21/2024] [Accepted: 01/24/2024] [Indexed: 02/05/2024]
Abstract
Primary cilia are distributed extensively within the corneal epithelium and endothelium. However, the presence of cilia in the corneal stroma and the dynamic changes and roles of endothelial and stromal cilia in corneal homeostasis remain largely unknown. Here, we present compelling evidence for the presence of primary cilia in the corneal stroma, both in vivo and in vitro. We also demonstrate dynamic changes of both endothelial and stromal cilia during corneal development. In addition, our data show that cryoinjury triggers dramatic cilium formation in the corneal endothelium and stroma. Furthermore, depletion of cilia in mutant mice lacking intraflagellar transport protein 88 compromises the corneal endothelial capacity to establish the effective tissue barrier, leading to an upregulation of α-smooth muscle actin within the corneal stroma in response to cryoinjury. These observations underscore the essential involvement of corneal endothelial and stromal cilia in maintaining corneal homeostasis and provide an innovative strategy for the treatment of corneal injuries and diseases.
Collapse
Affiliation(s)
- Hua Ni
- Department of Genetics and Cell Biology, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
- College of Life and Geographic Sciences, Kashi University, Kashi, China
| | - Lamei Li
- Department of Genetics and Cell Biology, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Die Hu
- Department of Genetics and Cell Biology, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Mulin Yang
- Department of Genetics and Cell Biology, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Difei Wang
- Department of Genetics and Cell Biology, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Hongbo Ma
- Department of Genetics and Cell Biology, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Weiwen Bu
- Department of Genetics and Cell Biology, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Jia Yang
- Department of Genetics and Cell Biology, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Li-E Zhu
- Department of Genetics and Cell Biology, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Denghui Zhai
- Department of Genetics and Cell Biology, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Ting Song
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Song Yang
- Department of Genetics and Cell Biology, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Quanlong Lu
- Department of Genetics and Cell Biology, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Dengwen Li
- Department of Genetics and Cell Biology, Haihe Laboratory of Cell Ecosystem, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Jie Ran
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Min Liu
- Laboratory of Tissue Homeostasis, Haihe Laboratory of Cell Ecosystem, Tianjin, China
| |
Collapse
|
5
|
Tian Z, Li X, Yu X, Yan S, Sun J, Ma W, Zhu X, Tang Y. The role of primary cilia in thyroid diseases. Front Endocrinol (Lausanne) 2024; 14:1306550. [PMID: 38260150 PMCID: PMC10801159 DOI: 10.3389/fendo.2023.1306550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 12/05/2023] [Indexed: 01/24/2024] Open
Abstract
Primary cilia (PC) are non-motile and microtube-based organelles protruding from the surface of almost all thyroid follicle cells. They maintain homeostasis in thyrocytes and loss of PC can result in diverse thyroid diseases. The dysfunction of structure and function of PC are found in many patients with common thyroid diseases. The alterations are associated with the cause, development, and recovery of the diseases and are regulated by PC-mediated signals. Restoring normal PC structure and function in thyrocytes is a promising therapeutic strategy to treat thyroid diseases. This review explores the function of PC in normal thyroid glands. It summarizes the pathology caused by PC alterations in thyroid cancer (TC), autoimmune thyroid diseases (AITD), hypothyroidism, and thyroid nodules (TN) to provide comprehensive references for further study.
Collapse
Affiliation(s)
- Zijiao Tian
- College of Traditional Chinese Medicine of Beijing University of Chinese Medicine, Beijing, China
| | - Xinlin Li
- College of Traditional Chinese Medicine of Beijing University of Chinese Medicine, Beijing, China
| | - Xue Yu
- College of Traditional Chinese Medicine of Beijing University of Chinese Medicine, Beijing, China
| | - Shuxin Yan
- College of Traditional Chinese Medicine of Beijing University of Chinese Medicine, Beijing, China
| | - Jingwei Sun
- College of Traditional Chinese Medicine of Beijing University of Chinese Medicine, Beijing, China
| | - Wenxin Ma
- College of Traditional Chinese Medicine of Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoyun Zhu
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yang Tang
- College of Traditional Chinese Medicine of Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
6
|
Guan YT, Zhang C, Zhang HY, Wei WL, Yue W, Zhao W, Zhang DH. Primary cilia: Structure, dynamics, and roles in cancer cells and tumor microenvironment. J Cell Physiol 2023; 238:1788-1807. [PMID: 37565630 DOI: 10.1002/jcp.31092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/24/2023] [Accepted: 07/13/2023] [Indexed: 08/12/2023]
Abstract
Despite the initiation of tumor arises from tumorigenic transformation signaling in cancer cells, cancer cell survival, invasion, and metastasis also require a dynamic and reciprocal association with extracellular signaling from tumor microenvironment (TME). Primary cilia are the antenna-like structure that mediate signaling sensation and transduction in different tissues and cells. Recent studies have started to uncover that the heterogeneous ciliation in cancer cells and cells from the TME in tumor growth impels asymmetric paracellular signaling in the TME, indicating the essential functions of primary cilia in homeostasis maintenance of both cancer cells and the TME. In this review, we discussed recent advances in the structure and assembly of primary cilia, and the role of primary cilia in tumor and TME formation, as well as the therapeutic potentials that target ciliary dynamics and signaling from the cells in different tumors and the TME.
Collapse
Affiliation(s)
- Yi-Ting Guan
- Zhanjiang Institute of Clinical Medicine, Central People's Hospital of Zhanjiang, Guangdong Medical University Zhanjiang Central Hospital, Zhanjiang, P. R. China
| | - Chong Zhang
- Zhanjiang Institute of Clinical Medicine, Central People's Hospital of Zhanjiang, Guangdong Medical University Zhanjiang Central Hospital, Zhanjiang, P. R. China
| | - Hong-Yong Zhang
- Zhanjiang Institute of Clinical Medicine, Central People's Hospital of Zhanjiang, Guangdong Medical University Zhanjiang Central Hospital, Zhanjiang, P. R. China
| | - Wen-Lu Wei
- Zhanjiang Institute of Clinical Medicine, Central People's Hospital of Zhanjiang, Guangdong Medical University Zhanjiang Central Hospital, Zhanjiang, P. R. China
| | - Wei Yue
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Wei Zhao
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, P. R. China
- Department of Posthodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, P. R. China
| | - Dong-Hui Zhang
- Zhanjiang Institute of Clinical Medicine, Central People's Hospital of Zhanjiang, Guangdong Medical University Zhanjiang Central Hospital, Zhanjiang, P. R. China
| |
Collapse
|
7
|
Girardet L, Cyr DG, Belleannée C. Arl13b controls basal cell stemness properties and Hedgehog signaling in the mouse epididymis. Cell Mol Life Sci 2022; 79:556. [PMID: 36261680 PMCID: PMC11803030 DOI: 10.1007/s00018-022-04570-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 09/09/2022] [Accepted: 09/22/2022] [Indexed: 11/30/2022]
Abstract
Epithelial cells orchestrate a series of intercellular signaling events in response to tissue damage. While the epididymis is composed of a pseudostratified epithelium that controls the acquisition of male fertility, the maintenance of its integrity in the context of tissue damage or inflammation remains largely unknown. Basal cells of the epididymis contain a primary cilium, an organelle that controls cellular differentiation in response to Hedgehog signaling cues. Hypothesizing its contribution to epithelial homeostasis, we knocked out the ciliary component ARL13B in keratin 5-positive basal cells. In this model, the reduced size of basal cell primary cilia was associated with impaired Hedgehog signaling and the loss of KRT5, KRT14, and P63 basal cell markers. When subjected to tissue injury, the epididymal epithelium from knock-out mice displayed imbalanced rates of cell proliferation/apoptosis and failed to properly regenerate in vivo. This response was associated with changes in the transcriptomic landscape related to immune response, cell differentiation, cell adhesion, and triggered severe hypoplasia of the epithelium. Together our results indicate that the ciliary GTPase, ARL13B, participates in the transduction of the Hedgehog signaling pathway to maintain basal cell stemness needed for tissue regeneration. These findings provide new insights into the role of basal cell primary cilia as safeguards of pseudostratified epithelia.
Collapse
Affiliation(s)
- Laura Girardet
- Faculty of Medicine, Department of Obstetrics, Gynecology and Reproduction, Université Laval, CHU de Québec Research Center (CHUL), Quebec City, QC, Canada
| | - Daniel G Cyr
- Faculty of Medicine, Department of Obstetrics, Gynecology and Reproduction, Université Laval, CHU de Québec Research Center (CHUL), Quebec City, QC, Canada
- Laboratory for Reproductive Toxicology, INRS-Centre Armand-Frappier Santé Biotechnologie, Université du Québec, Laval, QC, Canada
| | - Clémence Belleannée
- Faculty of Medicine, Department of Obstetrics, Gynecology and Reproduction, Université Laval, CHU de Québec Research Center (CHUL), Quebec City, QC, Canada.
| |
Collapse
|
8
|
Hibino E, Ichiyama Y, Tsukamura A, Senju Y, Morimune T, Ohji M, Maruo Y, Nishimura M, Mori M. Bex1 is essential for ciliogenesis and harbours biomolecular condensate-forming capacity. BMC Biol 2022; 20:42. [PMID: 35144600 PMCID: PMC8830175 DOI: 10.1186/s12915-022-01246-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 02/02/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Primary cilia are sensory organelles crucial for organ development. The pivotal structure of the primary cilia is a microtubule that is generated via tubulin polymerization reaction that occurs in the basal body. It remains to be elucidated how molecules with distinct physicochemical properties contribute to the formation of the primary cilia. RESULTS Here we show that brain expressed X-linked 1 (Bex1) plays an essential role in tubulin polymerization and primary cilia formation. The Bex1 protein shows the physicochemical property of being an intrinsically disordered protein (IDP). Bex1 shows cell density-dependent accumulation as a condensate either in nucleoli at a low cell density or at the apical cell surface at a high cell density. The apical Bex1 localizes to the basal body. Bex1 knockout mice present ciliopathy phenotypes and exhibit ciliary defects in the retina and striatum. Bex1 recombinant protein shows binding capacity to guanosine triphosphate (GTP) and forms the condensate that facilitates tubulin polymerization in the reconstituted system. CONCLUSIONS Our data reveals that Bex1 plays an essential role for the primary cilia formation through providing the reaction field for the tubulin polymerization.
Collapse
Affiliation(s)
- Emi Hibino
- Molecular Neuroscience Research Centre (MNRC), Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, Shiga, 520-2192, Japan.,Graduate School of Pharmaceutical Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8601, Japan
| | - Yusuke Ichiyama
- Department of Ophthalmology, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, Shiga, 520-2192, Japan
| | - Atsushi Tsukamura
- Molecular Neuroscience Research Centre (MNRC), Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, Shiga, 520-2192, Japan.,Department of Paediatrics, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, Shiga, 520-2192, Japan.,Department of Vascular Physiology, National Cerebral and Cardiovascular Centre Research Institute, 6-1 Kishibe-Shimmachi, Suita, Osaka, 564-8565, Japan
| | - Yosuke Senju
- Research Institute for Interdisciplinary Science, Okayama University, 3-1-1 Tsushima-naka, Kita-ku, Okayama, 700-8530, Japan
| | - Takao Morimune
- Molecular Neuroscience Research Centre (MNRC), Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, Shiga, 520-2192, Japan.,Department of Paediatrics, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, Shiga, 520-2192, Japan.,Department of Vascular Physiology, National Cerebral and Cardiovascular Centre Research Institute, 6-1 Kishibe-Shimmachi, Suita, Osaka, 564-8565, Japan
| | - Masahito Ohji
- Department of Ophthalmology, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, Shiga, 520-2192, Japan
| | - Yoshihiro Maruo
- Department of Paediatrics, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, Shiga, 520-2192, Japan
| | - Masaki Nishimura
- Molecular Neuroscience Research Centre (MNRC), Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, Shiga, 520-2192, Japan
| | - Masaki Mori
- Molecular Neuroscience Research Centre (MNRC), Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, Shiga, 520-2192, Japan. .,Department of Vascular Physiology, National Cerebral and Cardiovascular Centre Research Institute, 6-1 Kishibe-Shimmachi, Suita, Osaka, 564-8565, Japan.
| |
Collapse
|
9
|
Portal C, Wang Z, Scott DK, Wolosin JM, Iomini C. The c-Myc Oncogene Maintains Corneal Epithelial Architecture at Homeostasis, Modulates p63 Expression, and Enhances Proliferation During Tissue Repair. Invest Ophthalmol Vis Sci 2022; 63:3. [PMID: 35103750 PMCID: PMC8822362 DOI: 10.1167/iovs.63.2.3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose The transcription factor c-Myc (Myc) plays central regulatory roles in both self-renewal and differentiation of progenitors of multiple cell lineages. Here, we address its function in corneal epithelium (CE) maintenance and repair. Methods Myc ablation in the limbal–corneal epithelium was achieved by crossing a floxed Myc mouse allele (Mycfl/fl) with a mouse line expressing the Cre recombinase gene under the keratin (Krt) 14 promoter. CE stratification and protein localization were assessed by histology of paraffin and plastic sections and by immunohistochemistry of frozen sections, respectively. Protein levels and gene expression were determined by western blot and real-time quantitative PCR, respectively. CE wound closure was tracked by fluorescein staining. Results At birth, mutant mice appeared indistinguishable from control littermates; however, their rates of postnatal weight gain were 67% lower than those of controls. After weaning, mutants also exhibited spontaneous skin ulcerations, predominantly in the tail and lower lip, and died 45 to 60 days after birth. The mutant CE displayed an increase in stratal thickness, increased levels of Krt12 in superficial cells, and decreased exfoliation rates. Accordingly, the absence of Myc perturbed protein and mRNA levels of genes modulating differentiation and proliferation processes, including ΔNp63β, Ets1, and two Notch target genes, Hey1 and Maml1. Furthermore, Myc promoted CE wound closure and wound-induced hyperproliferation. Conclusions Myc regulates the balance among CE stratification, differentiation, and surface exfoliation and promotes the transition to the hyperproliferative state during wound healing. Its effect on this balance may be exerted through the control of multiple regulators of cell fate, including isoforms of tumor protein p63.
Collapse
Affiliation(s)
- Céline Portal
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Zheng Wang
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Donald K Scott
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - J Mario Wolosin
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, New York, United States
| | - Carlo Iomini
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States.,Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| |
Collapse
|
10
|
Bräutigam K, Reinhard S, Galván JA, Wartenberg M, Hewer E, Schürch CM. Systematic Investigation of SARS-CoV-2 Receptor Protein Distribution along Viral Entry Routes in Humans. Respiration 2022; 101:610-618. [PMID: 35038715 PMCID: PMC8805044 DOI: 10.1159/000521317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 11/24/2021] [Indexed: 12/02/2022] Open
Abstract
Background The novel beta-coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), enters the human body via mucosal surfaces of the upper and/or lower respiratory tract. Viral entry into epithelial cells is mediated via angiotensin-converting enzyme 2 (ACE2) and auxiliary molecules, but the precise anatomic site of infection still remains unclear. Methods Here, we systematically investigated the main SARS-CoV-2 receptor proteins ACE2 and transmembrane serine protease 2 (TMPRSS2), as well as 2 molecules potentially involved in viral entry, furin and CD147, in formalin-fixed, paraffin-embedded human tissues. Tissue microarrays incorporating a total of 879 tissue cores from conjunctival (n = 84), sinonasal (n = 95), and lung (bronchiolar/alveolar; n = 96) specimens were investigated for protein expression by immunohistochemistry. Results ACE2 and TMPRSS2 were expressed in ciliated epithelial cells of the conjunctivae and sinonasal tissues, with highest expression levels observed in the apical cilia. In contrast, in the lung, the expression of those molecules in bronchiolar and alveolar epithelial cells was much rarer and only very focal when present. Furin and CD147 were more uniformly expressed in all tissues analyzed, including the lung. Interestingly, alveolar macrophages consistently expressed high levels of all 4 molecules investigated. Conclusions Our study confirms and extends previous findings and contributes to a better understanding of potential SARS-CoV-2 infection sites along the human respiratory tract.
Collapse
Affiliation(s)
| | - Stefan Reinhard
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - José A Galván
- Institute of Pathology, University of Bern, Bern, Switzerland
| | | | - Ekkehard Hewer
- Institute of Pathology, University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Christian M Schürch
- Department of Pathology and Neuropathology, University Hospital and Comprehensive Cancer Center Tübingen, Tübingen, Germany
| |
Collapse
|
11
|
Abdelftah Z, Gaber AR, Abo-Eleneen RE, EL-Bakry AM. Microstructure characteristics of cornea of some birds: a comparative study. BENI-SUEF UNIVERSITY JOURNAL OF BASIC AND APPLIED SCIENCES 2021. [DOI: 10.1186/s43088-021-00155-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Light is the critical factor that affects the eye's morphology and auxiliary plans. The ecomorphological engineering of the cornea aids the physiological activities of the cornea during connections between photoreceptor neurons and light photons. Cornea was dissected free from the orbit from three avian species as ibis (Eudocium albus), duck (Anas platyrhynchus domesticus) and hawk (Buteo Buteo) and prepared for light and scanning electron microscopy and special stain for structural comparison related to function.
Results
The three investigated avian species are composed of three identical layers; epithelium, stroma, and endothelium, and two basement membranes; bowman's and Descemet’s membrane, separating two cellular layers, except for B. buteo which only has a Descemet’s membrane. The corneal layers in the investigated species display different affinity to stain with Periodic Acid Schiff stain. The external corneal surface secured by different normal epithelial cells ran from hexagonal to regular polygonal cells. Those epithelial cells are punctured by different diameter microholes and microplicae and microvilli of various length. Blebs are scarcely distributed over their surface. The present investigation utilized histological, histochemical and SEM examination.
Conclusions
The study presents a brief image/account of certain structures of cornea for three of Avian’s species. Data distinguish the anatomic structures of the owl's eye. The discussion explains the role of some functional anatomical structures all through the vision.
Collapse
|
12
|
Ye J, Sheahon KM, LeBoit PE, McCalmont TH, Lang UE. BAP1-inactivated melanocytic tumors show prominent centrosome amplification and associated loss of primary cilia. J Cutan Pathol 2021; 48:1353-1360. [PMID: 34085298 DOI: 10.1111/cup.14073] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 05/14/2021] [Accepted: 05/30/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND BRCA1-associated protein (BAP1) is a tumor suppressor whose loss is associated with various malignancies. The primary cilium is an organelle involved in signal transduction and cell cycle progression. Primary cilia have been shown to be absent in melanoma but retained to some extent in melanocytic nevi, and the severity of dysplasia influences the degree of cilia loss. Additionally, studies have revealed roles for BAP1 in centrosome and mitotic spindle formation. Because the primary cilium is nucleated on the mother centriole, we examined the connection between the presence of primary cilia and the formation of centrosomes in BAP1-inactivated melanocytic tumors (BIMTs). METHODS We evaluated the cilia and centrosomes in 11 BIMTs and five conventional melanocytic nevi using immunofluorescence staining of acetylated alpha-tubulin and gamma-tubulin. RESULTS We found that, compared to nevi, BIMTs show loss of primary cilia and amplification of centrosomes. Occasional nevi also showed increased centrioles; however, these foci of amplification were more likely to be ciliated than those in BIMTs. CONCLUSIONS Although centrosome amplification does not absolutely correlate with loss of primary cilia in melanocytic neoplasms, absence of BAP1 exacerbates the phenotype. Moreover, aberrant centrosome and cilia formation are likely critical in the pathogenesis of other BAP1-inactivated tumors.
Collapse
Affiliation(s)
- Julia Ye
- Department of Anatomic Pathology, University of California, San Francisco, California, USA
| | - Kathleen M Sheahon
- Department of Anatomic Pathology, University of California, San Francisco, California, USA
| | - Philip E LeBoit
- Department of Anatomic Pathology, University of California, San Francisco, California, USA.,Department of Dermatology, University of California, San Francisco, California, USA.,Helen Diller Comprehensive Cancer Center, University of California, San Francisco, California, USA
| | - Timothy H McCalmont
- Department of Anatomic Pathology, University of California, San Francisco, California, USA.,Department of Dermatology, University of California, San Francisco, California, USA.,Helen Diller Comprehensive Cancer Center, University of California, San Francisco, California, USA
| | - Ursula E Lang
- Department of Anatomic Pathology, University of California, San Francisco, California, USA.,Department of Dermatology, University of California, San Francisco, California, USA
| |
Collapse
|
13
|
Silencing of Histone Deacetylase 6 Decreases Cellular Malignancy and Contributes to Primary Cilium Restoration, Epithelial-to-Mesenchymal Transition Reversion, and Autophagy Inhibition in Glioblastoma Cell Lines. BIOLOGY 2021; 10:biology10060467. [PMID: 34073238 PMCID: PMC8228543 DOI: 10.3390/biology10060467] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/20/2021] [Accepted: 05/23/2021] [Indexed: 12/25/2022]
Abstract
Simple Summary Glioblastoma multiforme (GBM) is the most common as well as the most aggressive malignant brain tumor, with an overall survival of almost 15 months. Histone deacetylase 6 (HDAC6), an enzyme related to the deacetylation of α-tubulin, is overexpressed in GBM. The aim of our research was to study the effects of HDAC6 silencing in GBM cells. We first confirmed the overexpression of HDAC6 in GBM tissue (n = 40) against control brain (n = 10). Treatment with siHDAC6 diminished viability, clonogenic potential, and migration ability in GBM-derived cell lines. HDAC6 inhibition also reverted the mesenchymal phenotype, inhibited the Sonic Hedgehog pathway, restored primary cilium structure, and decreased autophagy. Thus, we confirm that HDAC6 is a good therapeutic target for GBM treatment. Abstract Glioblastoma multiforme, the most common type of malignant brain tumor as well as the most aggressive one, lacks an effective therapy. Glioblastoma presents overexpression of mesenchymal markers Snail, Slug, and N-Cadherin and of the autophagic marker p62. Glioblastoma cell lines also present increased autophagy, overexpression of mesenchymal markers, Shh pathway activation, and lack of primary cilia. In this study, we aimed to evaluate the role of HDAC6 in the pathogenesis of glioblastoma, as HDAC6 is the most overexpressed of all HDACs isoforms in this tumor. We treated glioblastoma cell lines with siHDAC6. HDAC6 silencing inhibited proliferation, migration, and clonogenicity of glioblastoma cell lines. They also reversed the mesenchymal phenotype, decreased autophagy, inhibited Shh pathway, and recovered the expression of primary cilia in glioblastoma cell lines. These results demonstrate that HDAC6 might be a good target for glioblastoma treatment.
Collapse
|
14
|
Abstract
As a transparent avascular tissue located at the front of the eyeball, the cornea is an important barrier to external damage. Both epithelial and endothelial cells of the cornea harbor primary cilia, which sense changes in the external environment and regulate intracellular signaling pathways. Accumulating evidence suggests that the primary cilium regulates corneal development in several ways, including participation in corneal epithelial stratification and maintenance of corneal endothelial cell morphology. In addition, the primary cilium has been implicated in the pathogenesis of several corneal diseases. In this review, we discuss recent findings that demonstrate the critical role of the primary cilium in corneal development. We also discuss the link between ciliary dysfunction and corneal diseases, which suggests that the primary cilium could be targeted to treat these diseases.
Collapse
Affiliation(s)
- Ting Song
- Institute of Biomedical Sciences, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, Shandong 250014, China
| | - Jun Zhou
- Institute of Biomedical Sciences, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, Shandong 250014, China.,State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, China. E-mail:
| |
Collapse
|
15
|
Hu HB, Song ZQ, Song GP, Li S, Tu HQ, Wu M, Zhang YC, Yuan JF, Li TT, Li PY, Xu YL, Shen XL, Han QY, Li AL, Zhou T, Chun J, Zhang XM, Li HY. LPA signaling acts as a cell-extrinsic mechanism to initiate cilia disassembly and promote neurogenesis. Nat Commun 2021; 12:662. [PMID: 33510165 PMCID: PMC7843646 DOI: 10.1038/s41467-021-20986-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 12/15/2020] [Indexed: 01/17/2023] Open
Abstract
Dynamic assembly and disassembly of primary cilia controls embryonic development and tissue homeostasis. Dysregulation of ciliogenesis causes human developmental diseases termed ciliopathies. Cell-intrinsic regulatory mechanisms of cilia disassembly have been well-studied. The extracellular cues controlling cilia disassembly remain elusive, however. Here, we show that lysophosphatidic acid (LPA), a multifunctional bioactive phospholipid, acts as a physiological extracellular factor to initiate cilia disassembly and promote neurogenesis. Through systematic analysis of serum components, we identify a small molecular-LPA as the major driver of cilia disassembly. Genetic inactivation and pharmacological inhibition of LPA receptor 1 (LPAR1) abrogate cilia disassembly triggered by serum. The LPA-LPAR-G-protein pathway promotes the transcription and phosphorylation of cilia disassembly factors-Aurora A, through activating the transcription coactivators YAP/TAZ and calcium/CaM pathway, respectively. Deletion of Lpar1 in mice causes abnormally elongated cilia and decreased proliferation in neural progenitor cells, thereby resulting in defective neurogenesis. Collectively, our findings establish LPA as a physiological initiator of cilia disassembly and suggest targeting the metabolism of LPA and the LPA pathway as potential therapies for diseases with dysfunctional ciliogenesis.
Collapse
Affiliation(s)
- Huai-Bin Hu
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Zeng-Qing Song
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Guang-Ping Song
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Sen Li
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Hai-Qing Tu
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Min Wu
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Yu-Cheng Zhang
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Jin-Feng Yuan
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Ting-Ting Li
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Pei-Yao Li
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Yu-Ling Xu
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Xiao-Lin Shen
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Qiu-Ying Han
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Ai-Ling Li
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Tao Zhou
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China
| | - Jerold Chun
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, USA
| | - Xue-Min Zhang
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China.
| | - Hui-Yan Li
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, China.
- School of Basic Medical Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
16
|
Paulson D, Harms R, Ward C, Latterell M, Pazour GJ, Fink DM. Loss of Primary Cilia Protein IFT20 Dysregulates Lymphatic Vessel Patterning in Development and Inflammation. Front Cell Dev Biol 2021; 9:672625. [PMID: 34055805 PMCID: PMC8160126 DOI: 10.3389/fcell.2021.672625] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/15/2021] [Indexed: 12/12/2022] Open
Abstract
Microenvironmental signals produced during development or inflammation stimulate lymphatic endothelial cells to undergo lymphangiogenesis, in which they sprout, proliferate, and migrate to expand the vascular network. Many cell types detect changes in extracellular conditions via primary cilia, microtubule-based cellular protrusions that house specialized membrane receptors and signaling complexes. Primary cilia are critical for receipt of extracellular cues from both ligand-receptor pathways and physical forces such as fluid shear stress. Here, we report the presence of primary cilia on immortalized mouse and primary adult human dermal lymphatic endothelial cells in vitro and on both luminal and abluminal domains of mouse corneal, skin, and mesenteric lymphatic vessels in vivo. The purpose of this study was to determine the effects of disrupting primary cilia on lymphatic vessel patterning during development and inflammation. Intraflagellar transport protein 20 (IFT20) is part of the transport machinery required for ciliary assembly and function. To disrupt primary ciliary signaling, we generated global and lymphatic endothelium-specific IFT20 knockout mouse models and used immunofluorescence microscopy to quantify changes in lymphatic vessel patterning at E16.5 and in adult suture-mediated corneal lymphangiogenesis. Loss of IFT20 during development resulted in edema, increased and more variable lymphatic vessel caliber and branching, as well as red blood cell-filled lymphatics. We used a corneal suture model to determine ciliation status of lymphatic vessels during acute, recurrent, and tumor-associated inflammatory reactions and wound healing. Primary cilia were present on corneal lymphatics during all of the mechanistically distinct lymphatic patterning events of the model and assembled on lymphatic endothelial cells residing at the limbus, stalk, and vessel tip. Lymphatic-specific deletion of IFT20 cell-autonomously exacerbated acute corneal lymphangiogenesis resulting in increased lymphatic vessel density and branching. These data are the first functional studies of primary cilia on lymphatic endothelial cells and reveal a new dimension in regulation of lymphatic vascular biology.
Collapse
Affiliation(s)
- Delayna Paulson
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD, United States
- BioSNTR, South Dakota State University, Brookings, SD, United States
| | - Rebecca Harms
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD, United States
- BioSNTR, South Dakota State University, Brookings, SD, United States
| | - Cody Ward
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD, United States
- BioSNTR, South Dakota State University, Brookings, SD, United States
| | - Mackenzie Latterell
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD, United States
- BioSNTR, South Dakota State University, Brookings, SD, United States
| | - Gregory J. Pazour
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, United States
| | - Darci M. Fink
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD, United States
- BioSNTR, South Dakota State University, Brookings, SD, United States
- *Correspondence: Darci M. Fink,
| |
Collapse
|
17
|
Wei H, Wang X, Niu X, Jiao R, Li X, Wang S. miR‑34c‑5p targets Notch1 and suppresses the metastasis and invasion of cervical cancer. Mol Med Rep 2020; 23:120. [PMID: 33300051 PMCID: PMC7751466 DOI: 10.3892/mmr.2020.11759] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 10/16/2020] [Indexed: 12/11/2022] Open
Abstract
Micro (mi)RNAs are crucial participants in the progression of cervical cancer (CC). Growing evidence indicates that miRNA (miR)-34c-5p is a pivotal tumor suppressor in numerous types of cancer and its functions in CC require further investigating. The present study demonstrated that there was a decreased level of miR-34c-5p in CC-associated cell lines compared with healthy control samples. It also demonstrated that miR-34c-5p targeted Notch1 and suppressed CC progression. Dual-Luciferase reporter assays verified the targeted relationship of miR-34c-5p and Notch1. The expression of Notch1 in HeLa cells was markedly reduced following miR-34c-5p overexpression and the proliferation, migration and invasion of HeLa cells were reduced although apoptosis was accelerated. However, overexpression of miR-34c-5p was reversed following the addition of Notch1, which supported the finding of the targeted relationship between miR-34c-5p and Notch1. Flow cytometry demonstrated that miR-34c-5p inhibited the proliferation of HeLa cells while accelerating apoptosis. The present study concluded that miR-34c-5p was a tumor suppressor in CC and may be a novel measure for the future treatment of CC.
Collapse
Affiliation(s)
- Huali Wei
- Department of Gynecology and Obstetrics, Emergency General Hospital, Beijing 100028, P.R. China
| | - Xiaolan Wang
- Department of Gynecology and Obstetrics, Emergency General Hospital, Beijing 100028, P.R. China
| | - Xiumin Niu
- Department of Gynecology and Obstetrics, Emergency General Hospital, Beijing 100028, P.R. China
| | - Ruili Jiao
- Department of Gynecology and Obstetrics, Chaoyang District Maternal and Child Health Hospital, Beijing 100020, P.R. China
| | - Xiaojuan Li
- Department of Medical Records and Statistics, Emergency General Hospital, Beijing 100028, P.R. China
| | - Sumei Wang
- Department of Gynecology and Obstetrics, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, P.R. China
| |
Collapse
|
18
|
Dugauquier A, Schrooyen M, Cordonnier M. Salzmann's nodular degeneration in a patient with Kartagener syndrome. J Fr Ophtalmol 2020; 44:e83-e86. [PMID: 33280904 DOI: 10.1016/j.jfo.2020.05.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 05/22/2020] [Accepted: 05/25/2020] [Indexed: 10/22/2022]
Affiliation(s)
- A Dugauquier
- Service d'ophtalmologie, hôpital Erasme, 808, route de Lennik, 1070 Bruxelles, Belgium.
| | - M Schrooyen
- Service d'ophtalmologie, hôpital Erasme, 808, route de Lennik, 1070 Bruxelles, Belgium; Service d'ophtalmologie, CHIREC, Bruxelles, Belgium
| | - M Cordonnier
- Service d'ophtalmologie, hôpital Erasme, 808, route de Lennik, 1070 Bruxelles, Belgium
| |
Collapse
|
19
|
Wang B, Liang Z, Liu P. Functional aspects of primary cilium in signaling, assembly and microenvironment in cancer. J Cell Physiol 2020; 236:3207-3219. [PMID: 33107052 PMCID: PMC7984063 DOI: 10.1002/jcp.30117] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 09/16/2020] [Accepted: 10/11/2020] [Indexed: 12/12/2022]
Abstract
The primary cilium is an antennae‐like structure extent outside the cell surface. It has an important role in regulating cell‐signaling transduction to affect proliferation, differentiation and migration. Evidence is accumulating that ciliary defects lead to ciliopathies and ciliary deregulation also play crucial roles in cancer formation and progression. Interestingly, restoring the cilia can suppress proliferation in some cancer cell. However, t he role of primary cilia in cancer still be debated. In this article, we review the role of the primary cilium in cancer through architecture, signaling pathways, cilia assembly and disassembly regulators, and summarized the new findings of the primary cilium in tumor microenvironments and different cancers, highlighting novel possibilities for therapeutic target in cancer.
Collapse
Affiliation(s)
- Bo Wang
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Zheyong Liang
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Peijun Liu
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
20
|
Poe AJ, Kulkarni M, Leszczynska A, Tang J, Shah R, Jami-Alahmadi Y, Wang J, Kramerov AA, Wohlschlegel J, Punj V, Ljubimov AV, Saghizadeh M. Integrated Transcriptome and Proteome Analyses Reveal the Regulatory Role of miR-146a in Human Limbal Epithelium via Notch Signaling. Cells 2020; 9:cells9102175. [PMID: 32993109 PMCID: PMC7650592 DOI: 10.3390/cells9102175] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/21/2020] [Accepted: 09/23/2020] [Indexed: 02/06/2023] Open
Abstract
MiR-146a is upregulated in the stem cell-enriched limbal region vs. central human cornea and can mediate corneal epithelial wound healing. The aim of this study was to identify miR-146a targets in human primary limbal epithelial cells (LECs) using genomic and proteomic analyses. RNA-seq combined with quantitative proteomics based on multiplexed isobaric tandem mass tag labeling was performed in LECs transfected with miR-146a mimic vs. mimic control. Western blot and immunostaining were used to confirm the expression of some targeted genes/proteins. A total of 251 differentially expressed mRNAs and 163 proteins were identified. We found that miR-146a regulates the expression of multiple genes in different pathways, such as the Notch system. In LECs and organ-cultured corneas, miR-146a increased Notch-1 expression possibly by downregulating its inhibitor Numb, but decreased Notch-2. Integrated transcriptome and proteome analyses revealed the regulatory role of miR-146a in several other processes, including anchoring junctions, TNF-α, Hedgehog signaling, adherens junctions, TGF-β, mTORC2, and epidermal growth factor receptor (EGFR) signaling, which mediate wound healing, inflammation, and stem cell maintenance and differentiation. Our results provide insights into the regulatory network of miR-146a and its role in fine-tuning of Notch-1 and Notch-2 expressions in limbal epithelium, which could be a balancing factor in stem cell maintenance and differentiation.
Collapse
Affiliation(s)
- Adam J. Poe
- Board of Governors Regenerative Medicine Institute, Eye Program, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (A.J.P.); (M.K.); (A.L.); (R.S.); (J.W.); (A.A.K.); (A.V.L.)
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Mangesh Kulkarni
- Board of Governors Regenerative Medicine Institute, Eye Program, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (A.J.P.); (M.K.); (A.L.); (R.S.); (J.W.); (A.A.K.); (A.V.L.)
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Aleksandra Leszczynska
- Board of Governors Regenerative Medicine Institute, Eye Program, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (A.J.P.); (M.K.); (A.L.); (R.S.); (J.W.); (A.A.K.); (A.V.L.)
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jie Tang
- Genomics Core, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA;
| | - Ruchi Shah
- Board of Governors Regenerative Medicine Institute, Eye Program, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (A.J.P.); (M.K.); (A.L.); (R.S.); (J.W.); (A.A.K.); (A.V.L.)
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Yasaman Jami-Alahmadi
- Department of Biological Chemistry, University of California, Los Angeles, CA 90095, USA; (Y.J.-A.); (J.W.)
| | - Jason Wang
- Board of Governors Regenerative Medicine Institute, Eye Program, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (A.J.P.); (M.K.); (A.L.); (R.S.); (J.W.); (A.A.K.); (A.V.L.)
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Andrei A. Kramerov
- Board of Governors Regenerative Medicine Institute, Eye Program, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (A.J.P.); (M.K.); (A.L.); (R.S.); (J.W.); (A.A.K.); (A.V.L.)
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - James Wohlschlegel
- Department of Biological Chemistry, University of California, Los Angeles, CA 90095, USA; (Y.J.-A.); (J.W.)
| | - Vasu Punj
- Department of Medicine, University of Southern California, Los Angeles, CA 90089, USA;
| | - Alexander V. Ljubimov
- Board of Governors Regenerative Medicine Institute, Eye Program, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (A.J.P.); (M.K.); (A.L.); (R.S.); (J.W.); (A.A.K.); (A.V.L.)
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Mehrnoosh Saghizadeh
- Board of Governors Regenerative Medicine Institute, Eye Program, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (A.J.P.); (M.K.); (A.L.); (R.S.); (J.W.); (A.A.K.); (A.V.L.)
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
- Correspondence: ; Tel.: +1-310-248-8696
| |
Collapse
|
21
|
Ma N, Zhou J. Functions of Endothelial Cilia in the Regulation of Vascular Barriers. Front Cell Dev Biol 2020; 8:626. [PMID: 32733899 PMCID: PMC7363763 DOI: 10.3389/fcell.2020.00626] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 06/23/2020] [Indexed: 12/12/2022] Open
Abstract
The vascular barrier between blood and tissues is a highly selective structure that is essential to maintain tissue homeostasis. Defects in the vascular barrier lead to a variety of cardiovascular diseases. The maintenance of vascular barriers is largely dependent on endothelial cells, but the precise mechanisms remain elusive. Recent studies reveal that primary cilia, microtubule-based structures that protrude from the surface of endothelial cells, play a critical role in the regulation of vascular barriers. Herein, we discuss recent advances on ciliary functions in the vascular barrier and suggest that ciliary signaling pathways might be targeted to modulate the vascular barrier.
Collapse
Affiliation(s)
- Nan Ma
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Jun Zhou
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China.,Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Institute of Biomedical Sciences, Shandong Normal University, Jinan, China
| |
Collapse
|
22
|
Mukherjee M, Ratnayake I, Janga M, Fogarty E, Scheidt S, Grassmeyer J, deRiso J, Chandrasekar I, Ahrenkiel P, Kopan R, Surendran K. Notch signaling regulates Akap12 expression and primary cilia length during renal tubule morphogenesis. FASEB J 2020; 34:9512-9530. [PMID: 32474964 PMCID: PMC7501208 DOI: 10.1096/fj.201902358rr] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 05/05/2020] [Accepted: 05/08/2020] [Indexed: 12/23/2022]
Abstract
Alagille syndrome patients present with loss of function mutations in either JAG1 or NOTCH2. About 40%-50% of patients have kidney abnormalities, and frequently display multicystic, dysplastic kidneys. Additionally, gain-of-function mutations in NOTCH2 are associated with cystic kidneys in Hajdu-Cheney syndrome patients. How perturbations in Notch signaling cause renal tubular cysts remains unclear. Here, we have determined that reduced Notch signaling mediated transcription by ectopic expression of dominant-negative mastermind-like (dnMaml) peptide in the nephrogenic epithelia from after the s-shaped body formation and in the developing collecting ducts results in proximal tubular and collecting duct cysts, respectively. An acute inhibition of Notch signaling for two days during kidney development is sufficient to disrupt tubule formation, and significantly increases Akap12 expression. Ectopic expression of Akap12 in renal epithelia results in abnormally long primary cilia similar to that observed in Notch-signaling-deficient epithelia. Both loss of Notch signaling and elevated Akap12 expression disrupt the ability of renal epithelial cells to form spherical structures with a single lumen when grown embedded in matrix. Interestingly, Akap12 can inhibit Notch signaling mediated transcription, which likely explains how both loss of Notch signaling and ectopic expression of Akap12 result in similar renal epithelial abnormalities. We conclude that Notch signaling regulates Akap12 expression while also ensuring normal primary cilia length and renal epithelial morphogenesis, and suggest that one aspect of diseases associated with defective Notch signaling, such as Alagille syndrome, maybe mechanistically related to ciliopathies.
Collapse
Affiliation(s)
- Malini Mukherjee
- Pediatrics and Rare Diseases Group, Sanford Research, 2301 East 60 Street North, Sioux Falls, SD 57104
- Malini Mukherjee, Ishara Ratnayake and Madhusudhana Janga made equal contributions
| | - Ishara Ratnayake
- Department of Nanoscience and Nanoengineering, South Dakota School of Mines and Technology, Rapid City, SD 57701
- Malini Mukherjee, Ishara Ratnayake and Madhusudhana Janga made equal contributions
| | - Madhusudhana Janga
- Pediatrics and Rare Diseases Group, Sanford Research, 2301 East 60 Street North, Sioux Falls, SD 57104
- Malini Mukherjee, Ishara Ratnayake and Madhusudhana Janga made equal contributions
| | - Eric Fogarty
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069
| | - Shania Scheidt
- Pediatrics and Rare Diseases Group, Sanford Research, 2301 East 60 Street North, Sioux Falls, SD 57104
| | | | - Jennifer deRiso
- Pediatrics and Rare Diseases Group, Sanford Research, 2301 East 60 Street North, Sioux Falls, SD 57104
| | - Indra Chandrasekar
- Pediatrics and Rare Diseases Group, Sanford Research, 2301 East 60 Street North, Sioux Falls, SD 57104
- Enabling Technologies Group, Sanford Research, 2301 East 60 Street North, Sioux Falls, SD 57104
| | - Phil Ahrenkiel
- Department of Nanoscience and Nanoengineering, South Dakota School of Mines and Technology, Rapid City, SD 57701
| | - Raphael Kopan
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229
| | - Kameswaran Surendran
- Pediatrics and Rare Diseases Group, Sanford Research, 2301 East 60 Street North, Sioux Falls, SD 57104
- Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, SD 57104, USA
| |
Collapse
|
23
|
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: 78] [Impact Index Per Article: 15.6] [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
|
24
|
Zhou P, Zhou J. The Primary Cilium as a Therapeutic Target in Ocular Diseases. Front Pharmacol 2020; 11:977. [PMID: 32676032 PMCID: PMC7333185 DOI: 10.3389/fphar.2020.00977] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 06/16/2020] [Indexed: 12/24/2022] Open
Abstract
Primary cilia are microtubule-based cellular structures located on the surfaces of most mammalian cells and play important roles in detecting external stimuli, signal transduction, and cell cycle regulation. Primary cilia are also present in several structures of the eye, and their abnormal development or dysfunction can cause various ocular diseases. The rapid development of proteomics and metabolomics technologies have helped in the identification of many ocular disease-related proteins, some of which are dysregulated in primary cilia. This review focuses on ciliary dysregulation in a number of ocular diseases and discusses the potential of targeting primary cilia in gene and stem cell therapy for these diseases.
Collapse
Affiliation(s)
- Peng Zhou
- Institute of Biomedical Sciences, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Jun Zhou
- Institute of Biomedical Sciences, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan, China.,State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| |
Collapse
|
25
|
Choudhury A, Neumann NM, Raleigh DR, Lang UE. Clinical Implications of Primary Cilia in Skin Cancer. Dermatol Ther (Heidelb) 2020; 10:233-248. [PMID: 31997226 PMCID: PMC7090118 DOI: 10.1007/s13555-020-00355-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Indexed: 12/25/2022] Open
Abstract
The primary cilium is a cell surface organelle that is an important component of cellular biology. While it was once believed to be a vestigial structure without biologic function, it is now known to have essential roles in critical cellular signaling pathways such as Hedgehog (HH) and Wnt. The HH and Wnt pathways are involved in pathogenesis of basal cell carcinoma and melanoma, respectively, and this knowledge is now beginning to inform therapeutic and diagnostic options for patients. The purpose of this review is to familiarize clinicians with primary cilia biology and how this complex cellular organelle has started to translate into clinical care.
Collapse
Affiliation(s)
- Abrar Choudhury
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
- Department of Radiation Oncology, University of California, San Francisco, CA, USA
| | - Neil M Neumann
- Department of Pathology, Dermatopathology Service, University of California, San Francisco, CA, USA
| | - David R Raleigh
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
- Department of Radiation Oncology, University of California, San Francisco, CA, USA
| | - Ursula E Lang
- Department of Pathology, Dermatopathology Service, University of California, San Francisco, CA, USA.
- Department of Dermatology, University of California, San Francisco, CA, USA.
| |
Collapse
|
26
|
Moscatelli G, Bovo S, Schiavo G, Mazzoni G, Bertolini F, Dall'Olio S, Fontanesi L. Genome-wide association studies for iris pigmentation and heterochromia patterns in Large White pigs. Anim Genet 2020; 51:409-419. [PMID: 32232994 DOI: 10.1111/age.12930] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/06/2020] [Indexed: 01/13/2023]
Abstract
Eye colour genetics have been extensively studied in humans since the rediscovery of Mendel's laws. This trait was first interpreted using simplistic genetic models but soon it was realised that it is more complex. In this study, we analysed eye colour variability in a Large White pig population (n = 897) and report the results of GWASs based on several comparisons including pigs having four main eye colour categories (three with both pigmented eyes of different brown grades: pale, 17.9%; medium, 14.8%; and dark, 54.3%; another one with both eyes completely depigmented, 3.8%) and heterochromia patterns (heterochromia iridis - depigmented iris sectors in pigmented irises, 3.2%; heterochromia iridum - one whole eye iris of depigmented phenotype and the other eye with the iris completely pigmented, 5.9%). Pigs were genotyped with the Illumina PorcineSNP60 BeadChip and GEMMA was used for the association analyses. The results indicated that SLC45A2 (on chromosome 16, SSC16), EDNRB (SSC11) and KITLG (SSC5) affect the different grades of brown pigmentation of the eyes, the bilateral eye depigmentation defect and the heterochromia iridis defect recorded in this white pig population respectively. These genes are involved in several mechanisms affecting pigmentation. Significant associations for the eye depigmented patterns were also identified for SNPs on two SSC4 regions (including two candidate genes: NOTCH2 and PREX2) and on SSC6, SSC8 and SSC14 (including COL17A1 as candidate gene). This study provided useful information to understand eye pigmentation mechanisms, further valuing the pig as animal model to study complex phenotypes in humans.
Collapse
Affiliation(s)
- G Moscatelli
- Division of Animal Sciences, Department of Agricultural and Food Sciences, University of Bologna, Viale G. Fanin 46, 40127, Bologna, Italy
| | - S Bovo
- Division of Animal Sciences, Department of Agricultural and Food Sciences, University of Bologna, Viale G. Fanin 46, 40127, Bologna, Italy
| | - G Schiavo
- Division of Animal Sciences, Department of Agricultural and Food Sciences, University of Bologna, Viale G. Fanin 46, 40127, Bologna, Italy
| | - G Mazzoni
- Department of Health Technology, Technical University of Denmark, Lyngby, 2800, Denmark
| | - F Bertolini
- National Institute of Aquatic Resources, Technical University of Denmark, Lyngby, 2800, Denmark
| | - S Dall'Olio
- Division of Animal Sciences, Department of Agricultural and Food Sciences, University of Bologna, Viale G. Fanin 46, 40127, Bologna, Italy
| | - L Fontanesi
- Division of Animal Sciences, Department of Agricultural and Food Sciences, University of Bologna, Viale G. Fanin 46, 40127, Bologna, Italy
| |
Collapse
|
27
|
Peixoto E, Richard S, Pant K, Biswas A, Gradilone SA. The primary cilium: Its role as a tumor suppressor organelle. Biochem Pharmacol 2020; 175:113906. [PMID: 32169416 DOI: 10.1016/j.bcp.2020.113906] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 03/06/2020] [Indexed: 02/08/2023]
Abstract
The primary cilium is an organelle that nearly all cells within the body contain. Its function is to sense the extracellular environment through its abundance of receptors and linked signaling pathways, working as an antenna. Ciliary defects lead to different pathologies. In particular, many tumors lose primary cilia, and this is linked with negative implications for the cell such as an increase in malignancy. In this work we will go through the knowledge of the role of primary cilia in normal conditions, how it is involved in diverse signaling pathways, and in disease, particularly in cancer, highlighting its tumor suppressor properties.
Collapse
Affiliation(s)
- Estanislao Peixoto
- The Hormel Institute, University of Minnesota, Austin, MN, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Seth Richard
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Kishor Pant
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Aalekhya Biswas
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Sergio A Gradilone
- The Hormel Institute, University of Minnesota, Austin, MN, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
28
|
Portal C, Rompolas P, Lwigale P, Iomini C. Primary cilia deficiency in neural crest cells models anterior segment dysgenesis in mouse. eLife 2019; 8:52423. [PMID: 31845891 PMCID: PMC6946567 DOI: 10.7554/elife.52423] [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: 10/03/2019] [Accepted: 12/17/2019] [Indexed: 02/06/2023] Open
Abstract
Defects affecting tissues of the anterior segment (AS) of the eye lead to a group of highly debilitating disorders called Anterior Segment Dysgenesis (ASD). Despite the identification of some causative genes, the pathogenesis of ASD remains unclear. Interestingly, several ciliopathies display conditions of the AS. Using conditional targeting of Ift88 with Wnt1-Cre, we show that primary cilia of neural crest cells (NCC), precursors of most AS structures, are indispensable for normal AS development and their ablation leads to ASD conditions including abnormal corneal dimensions, defective iridocorneal angle, reduced anterior chamber volume and corneal neovascularization. Mechanistically, NCC cilia ablation abolishes hedgehog (Hh) signaling in the periocular mesenchyme (POM) canonically activated by choroid-secreted Indian Hh, reduces proliferation of POM cells surrounding the retinal pigment epithelium and decreases the expression of Foxc1 and Pitx2, two transcription factors identified as major ASD causative genes. Thus, we uncovered a signaling axis linking cilia and ASD.
Collapse
Affiliation(s)
- Céline Portal
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Panteleimos Rompolas
- Department of Dermatology, Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| | - Peter Lwigale
- BioSciences Department, Rice University, Houston, United States
| | - Carlo Iomini
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, United States.,Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, United States
| |
Collapse
|
29
|
Hu X, Zhu S, Liu R, Miller JD, Merkley K, Tilton RG, Liu H. Sirt6 deficiency impairs corneal epithelial wound healing. Aging (Albany NY) 2019; 10:1932-1946. [PMID: 30070973 PMCID: PMC6128418 DOI: 10.18632/aging.101513] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 07/29/2018] [Indexed: 12/19/2022]
Abstract
Corneal transparency, dependent on the integrity of epithelial cells, is essential for vision. Corneal epithelial damage is one of the most commonly observed ocular conditions and proper wound healing is necessary for corneal transparency. Sirt6, a histone deacetylase, has been shown to regulate many cellular events including aging and inflammation. However, its specific role in corneal epithelial wound healing remains unknown. Here we demonstrated that Sirt6 was expressed in corneal epithelial cells and its expression decreased with age. In an in vivo corneal epithelial wound healing model, Sirt6 deficiency resulted in delayed and incomplete wound healing and was associated excessive inflammation in the corneal stroma and dysfunction of Notch signaling, leading to keratinization of the corneal epithelium and corneal opacity. Aging Sirt6-deficient mice spontaneously developed corneal keratitis with extensive infiltration of inflammatory cells into the cornea. In vitro experiments demonstrated that primary corneal epithelial cells with Sirt6 downregulation expressed increased basal levels of inflammatory genes and exhibited hyper-inflammatory reactivity to IL-1β and TNFα treatment. These results provide compelling evidence that Sirt6 is a critical regulator of inflammation in the cornea, and is responsible for corneal epithelial wound healing, thus contributing to the maintenance of epithelial integrity and corneal transparency.
Collapse
Affiliation(s)
- Xiaobing Hu
- Wuhan Hanyang Eyegood Ophthalmic Hospital, Wuhan, China.,Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Shuang Zhu
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Rong Liu
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, TX 77555, USA.,Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jordan D Miller
- Department of Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Kevin Merkley
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Ronald G Tilton
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, TX 77555, USA.,Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Hua Liu
- Department of Ophthalmology and Visual Sciences, University of Texas Medical Branch, Galveston, TX 77555, USA.,Center for Biomedical Engineering, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
30
|
Fulmer D, Toomer K, Guo L, Moore K, Glover J, Moore R, Stairley R, Lobo G, Zuo X, Dang Y, Su Y, Fogelgren B, Gerard P, Chung D, Heydarpour M, Mukherjee R, Body SC, Norris RA, Lipschutz JH. Defects in the Exocyst-Cilia Machinery Cause Bicuspid Aortic Valve Disease and Aortic Stenosis. Circulation 2019; 140:1331-1341. [PMID: 31387361 DOI: 10.1161/circulationaha.119.038376] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND Bicuspid aortic valve (BAV) disease is a congenital defect that affects 0.5% to 1.2% of the population and is associated with comorbidities including ascending aortic dilation and calcific aortic valve stenosis. To date, although a few causal genes have been identified, the genetic basis for the vast majority of BAV cases remains unknown, likely pointing to complex genetic heterogeneity underlying this phenotype. Identifying genetic pathways versus individual gene variants may provide an avenue for uncovering additional BAV causes and consequent comorbidities. METHODS We performed genome-wide association Discovery and Replication Studies using cohorts of 2131 patients with BAV and 2728 control patients, respectively, which identified primary cilia genes as associated with the BAV phenotype. Genome-wide association study hits were prioritized based on P value and validated through in vivo loss of function and rescue experiments, 3-dimensional immunohistochemistry, histology, and morphometric analyses during aortic valve morphogenesis and in aged animals in multiple species. Consequences of these genetic perturbations on cilia-dependent pathways were analyzed by Western and immunohistochemistry analyses, and assessment of aortic valve and cardiac function were determined by echocardiography. RESULTS Genome-wide association study hits revealed an association between BAV and genetic variation in human primary cilia. The most associated single-nucleotide polymorphisms were identified in or near genes that are important in regulating ciliogenesis through the exocyst, a shuttling complex that chaperones cilia cargo to the membrane. Genetic dismantling of the exocyst resulted in impaired ciliogenesis, disrupted ciliogenic signaling and a spectrum of cardiac defects in zebrafish, and aortic valve defects including BAV, valvular stenosis, and valvular calcification in murine models. CONCLUSIONS These data support the exocyst as required for normal ciliogenesis during aortic valve morphogenesis and implicate disruption of ciliogenesis and its downstream pathways as contributory to BAV and associated comorbidities in humans.
Collapse
Affiliation(s)
- Diana Fulmer
- Departments of Medicine (D.F., G.L., X.Z., Y.D., Y.S., R.A.N., J.H.L.), Medical University of South Carolina, Charleston.,Regenerative Medicine and Cell Biology (D.F., K.T., L.G., K.M., J.G., R. Moore, R.S., R.A.N.), Medical University of South Carolina, Charleston
| | - Katelynn Toomer
- Regenerative Medicine and Cell Biology (D.F., K.T., L.G., K.M., J.G., R. Moore, R.S., R.A.N.), Medical University of South Carolina, Charleston
| | - Lilong Guo
- Regenerative Medicine and Cell Biology (D.F., K.T., L.G., K.M., J.G., R. Moore, R.S., R.A.N.), Medical University of South Carolina, Charleston
| | - Kelsey Moore
- Regenerative Medicine and Cell Biology (D.F., K.T., L.G., K.M., J.G., R. Moore, R.S., R.A.N.), Medical University of South Carolina, Charleston
| | - Janiece Glover
- Regenerative Medicine and Cell Biology (D.F., K.T., L.G., K.M., J.G., R. Moore, R.S., R.A.N.), Medical University of South Carolina, Charleston
| | - Reece Moore
- Regenerative Medicine and Cell Biology (D.F., K.T., L.G., K.M., J.G., R. Moore, R.S., R.A.N.), Medical University of South Carolina, Charleston
| | - Rebecca Stairley
- Regenerative Medicine and Cell Biology (D.F., K.T., L.G., K.M., J.G., R. Moore, R.S., R.A.N.), Medical University of South Carolina, Charleston
| | - Glenn Lobo
- Departments of Medicine (D.F., G.L., X.Z., Y.D., Y.S., R.A.N., J.H.L.), Medical University of South Carolina, Charleston.,Ophthalmology (G.L.), Medical University of South Carolina, Charleston
| | - Xiaofeng Zuo
- Departments of Medicine (D.F., G.L., X.Z., Y.D., Y.S., R.A.N., J.H.L.), Medical University of South Carolina, Charleston
| | - Yujing Dang
- Departments of Medicine (D.F., G.L., X.Z., Y.D., Y.S., R.A.N., J.H.L.), Medical University of South Carolina, Charleston
| | - Yanhui Su
- Departments of Medicine (D.F., G.L., X.Z., Y.D., Y.S., R.A.N., J.H.L.), Medical University of South Carolina, Charleston
| | - Ben Fogelgren
- Department of Anatomy, Biochemistry, and Physiology, University of Hawaii at Manoa, Honolulu (B.F.)
| | - Patrick Gerard
- Department of Mathematical Sciences, Clemson University, SC (P.G.)
| | - Dongjun Chung
- Public Health Sciences (D.C.), Medical University of South Carolina, Charleston
| | - Mahyar Heydarpour
- Department of Anesthesiology, Brigham and Women's Hospital (M.H.), Harvard Medical School, Boston, MA
| | - Rupak Mukherjee
- Surgery (R. Mukherjee), Medical University of South Carolina, Charleston.,Departments of Research (R. Mukherjee), Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC
| | - Simon C Body
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center (S.C.B.), Harvard Medical School, Boston, MA
| | - Russell A Norris
- Departments of Medicine (D.F., G.L., X.Z., Y.D., Y.S., R.A.N., J.H.L.), Medical University of South Carolina, Charleston.,Regenerative Medicine and Cell Biology (D.F., K.T., L.G., K.M., J.G., R. Moore, R.S., R.A.N.), Medical University of South Carolina, Charleston
| | - Joshua H Lipschutz
- Departments of Medicine (D.F., G.L., X.Z., Y.D., Y.S., R.A.N., J.H.L.), Medical University of South Carolina, Charleston.,Medicine (J.H.L.), Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC
| |
Collapse
|
31
|
Yuan X, Liu M, Cao X, Yang S. Ciliary IFT80 regulates dental pulp stem cells differentiation by FGF/FGFR1 and Hh/BMP2 signaling. Int J Biol Sci 2019; 15:2087-2099. [PMID: 31592124 PMCID: PMC6775288 DOI: 10.7150/ijbs.27231] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Accepted: 02/19/2019] [Indexed: 01/09/2023] Open
Abstract
Primary cilia and intraflagellar transport (IFT) proteins control a wide variety of processes during development and tissue homeostasis. However, their potential roles in the regulation of stem cell differentiation and tooth development remain elusive. Here, we uncovered the critical roles of ciliary IFT80 in cilia formation and differentiation of dental pulp stem cells (DPSCs). IFT80-deficient DPSCs showed reduced fibroblast growth factor receptor 1 (FGFR1) expression, leading to the disruption of FGF2-FGFR1 signaling. We found, during DPSC differentiation, FGF2-FGFR1 signaling induces stress fiber rearrangement to promote cilia elongation, meanwhile stimulates PI3K-AKT signaling to aid Hh/bone morphogenetic protein 2 (BMP2) signaling activation. These signaling pathways and their coupling were disrupted in IFT80-deficient DPSCs, causing impaired differentiation. Our findings revealed a novel mechanism that ciliary protein regulates the odontogenic differentiation of DPSCs through FGF/FGFR1 and Hh/BMP2 signaling.
Collapse
Affiliation(s)
- Xue Yuan
- Department of Oral Biology, School of Dental Medicine, University of Buffalo, State University of New York, Buffalo, NY, United States
| | - Min Liu
- Department of Anatomy & Cell Biology, School of Dental Medicine, University of Pennsylvania, PA, United States
| | - Xu Cao
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Shuying Yang
- Department of Oral Biology, School of Dental Medicine, University of Buffalo, State University of New York, Buffalo, NY, United States
- Department of Anatomy & Cell Biology, School of Dental Medicine, University of Pennsylvania, PA, United States
| |
Collapse
|
32
|
Primary Cilium-Mediated Retinal Pigment Epithelium Maturation Is Disrupted in Ciliopathy Patient Cells. Cell Rep 2019; 22:189-205. [PMID: 29298421 PMCID: PMC6166245 DOI: 10.1016/j.celrep.2017.12.038] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 11/08/2017] [Accepted: 12/11/2017] [Indexed: 12/15/2022] Open
Abstract
Primary cilia are sensory organelles that protrude from the cell membrane. Defects in the primary cilium cause ciliopathy disorders, with retinal degeneration as a prominent phenotype. Here, we demonstrate that the retinal pigment epithelium (RPE), essential for photoreceptor development and function, requires a functional primary cilium for complete maturation and that RPE maturation defects in ciliopathies precede photoreceptor degeneration. Pharmacologically enhanced ciliogenesis in wild-type induced pluripotent stem cells (iPSC)-RPE leads to fully mature and functional cells. In contrast, ciliopathy patient-derived iPSC-RPE and iPSC-RPE with a knockdown of ciliary-trafficking protein remain immature, with defective apical processes, reduced functionality, and reduced adult-specific gene expression. Proteins of the primary cilium regulate RPE maturation by simultaneously suppressing canonical WNT and activating PKCδ pathways. A similar cilium-dependent maturation pathway exists in lung epithelium. Our results provide insights into ciliopathy-induced retinal degeneration, demonstrate a developmental role for primary cilia in epithelial maturation, and provide a method to mature iPSC epithelial cells for clinical applications. May-Simera et al. show that primary cilia regulate the maturation and polarization of human iPSC-RPE, mouse RPE, and human iPSC-lung epithelium through canonical WNT suppression and PKCδ activation. RPE cells derived from ciliopathy patients exhibit defective structure and function. These results provide insights into ciliopathy-induced retinal degeneration.
Collapse
|
33
|
Haws RM, McIntee TJ, Green CB. Cutaneous findings in Bardet‐Biedl syndrome. Int J Dermatol 2019; 58:1160-1164. [DOI: 10.1111/ijd.14412] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 11/26/2018] [Accepted: 01/29/2019] [Indexed: 11/28/2022]
|
34
|
Yuan X, Cao X, Yang S. IFT80 is required for stem cell proliferation, differentiation, and odontoblast polarization during tooth development. Cell Death Dis 2019; 10:63. [PMID: 30683845 PMCID: PMC6347632 DOI: 10.1038/s41419-018-0951-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 08/01/2018] [Indexed: 12/01/2022]
Abstract
Primary cilia and intraflagellar transport (IFT) proteins control a wide variety of processes during tissue development and homeostasis. However, their role in regulation of stem cell properties during tooth development remains elusive. Here, we revealed that dental pulp stem cells (DPSCs) express IFT80, which is required for maintaining DPSC properties. Mice with deletion of IFT80 in odontoblast lineage show impaired molar root development and delayed incisor eruption through reduced DPSC proliferation and differentiation, and disrupted odontoblast polarization. Impaired odontoblast differentiation resulted from disrupted hedgehog (Hh) signaling pathways. Decreased DPSC proliferation is associated with impaired fibroblast growth factor 2 (FGF2) signaling caused by loss of IFT80, leading to the disruption of FGF2-FGFR1-PI3K-AKT signaling in IFT80-deficient DPSCs. The results provide the first evidence that IFT80 controls tooth development through influencing cell proliferation, differentiation, and polarization, and Hh and FGF/AKT signaling pathways, demonstrating that IFT proteins are likely to be the new therapeutic targets for tooth and other tissue repair and regeneration.
Collapse
Affiliation(s)
- Xue Yuan
- Department of Oral Biology, School of Dental Medicine University of Buffalo, State University of New York, Buffalo, NY, USA
| | - Xu Cao
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Shuying Yang
- Department of Oral Biology, School of Dental Medicine University of Buffalo, State University of New York, Buffalo, NY, USA.
- Department of Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
35
|
Higgins M, Obaidi I, McMorrow T. Primary cilia and their role in cancer. Oncol Lett 2019; 17:3041-3047. [PMID: 30867732 PMCID: PMC6396132 DOI: 10.3892/ol.2019.9942] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 01/24/2018] [Indexed: 01/25/2023] Open
Abstract
Primary cilia are microtubule-based organelles that are expressed on almost all mammalian cells. It has become apparent that these structures are important signaling hubs that serve crucial roles in Wnt, hedgehog, extracellular signal-regulated kinase (ERK)/mitogen-activated protein kinase (MAPK) and Notch signaling pathways. A number of diseases have been found to involve dysfunctional primary cilia; collectively these diseases are called ciliopathies. In recent years, there has been more focus on the association between primary cilia and cancer, including renal, pancreatic and breast cancer. Numerous studies have demonstrated that various types of cancer cells fail to express cilia. Notably, it has also been indicated that a number of renal carcinogens induce a significant loss of cilia in renal epithelial cells. The present review focuses on the existing literature regarding primary cilia and their involvement with cancer signaling pathways, providing a brief overview of the structural features and functions of primary cilia, then discussing the evidence associating primary cilia with cancer, and presenting the available information on the ERK/MAPK, hedgehog and Wnt signaling pathways, and their involvement in primary cilia in association with cancer.
Collapse
Affiliation(s)
- Michael Higgins
- School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Ismael Obaidi
- School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| | - Tara McMorrow
- School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|
36
|
Vicente A, Byström B, Pedrosa Domellöf F. Altered Signaling Pathways in Aniridia-Related Keratopathy. ACTA ACUST UNITED AC 2018; 59:5531-5541. [DOI: 10.1167/iovs.18-25175] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- André Vicente
- Department of Clinical Science, Ophthalmology, Umeå University, Umeå, Sweden
| | - Berit Byström
- Department of Clinical Science, Ophthalmology, Umeå University, Umeå, Sweden
| | - Fátima Pedrosa Domellöf
- Department of Clinical Science, Ophthalmology, Umeå University, Umeå, Sweden
- Department of Integrative Medical Biology, Section for Anatomy, Umeå University, Umeå, Sweden
| |
Collapse
|
37
|
Eguether T, Hahne M. Mixed signals from the cell's antennae: primary cilia in cancer. EMBO Rep 2018; 19:embr.201846589. [PMID: 30348893 DOI: 10.15252/embr.201846589] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 08/08/2018] [Accepted: 09/24/2018] [Indexed: 02/03/2023] Open
Abstract
Primary cilia (PC) are antenna-like organelles that protrude from most mammalian cells. They are essential for the regulation of several signaling pathways such as Hedgehog and WNT It is therefore not surprising that a dysfunction of PC is frequently associated with pathologies. Originally, PC were found to be involved in a variety of diseases commonly referred to as ciliopathies including cystic kidney diseases. Evidence is accumulating that PC play also an important role in cancer formation and regulation, which is the focus of this review.
Collapse
Affiliation(s)
- Thibaut Eguether
- École Normale Supérieure, CNRS, INSERM, APHP, Laboratoire des Biomolécules (LBM), Sorbonne Université, PSL Research University, Paris, France
| | - Michael Hahne
- IGMM, CNRS, University of Montpellier, Montpellier, France
| |
Collapse
|
38
|
Wang Y, Mahesh P, Wang Y, Novo SG, Shihan MH, Hayward-Piatkovskyi B, Duncan MK. Spatiotemporal dynamics of canonical Wnt signaling during embryonic eye development and posterior capsular opacification (PCO). Exp Eye Res 2018; 175:148-158. [PMID: 29932883 DOI: 10.1016/j.exer.2018.06.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Revised: 06/14/2018] [Accepted: 06/18/2018] [Indexed: 02/07/2023]
Abstract
The appropriate spatial and temporal regulation of canonical Wnt signaling is vital for eye development. However, the literature often conflicts on the distribution of canonical Wnt signaling in the eye. Here, using a sensitive mouse transgenic reporter line, we report a detailed re-evaluation of the spatiotemporal dynamics of canonical Wnt signaling in the developing eye. Canonical Wnt activity was dynamic in the optic vesicle and later in the retina, while it was absent from the ectodermal precursors of the lens and corneal epithelium. However, later in corneal development, canonical Wnt reporter activity was detected in corneal stroma and endothelium precursors as they form from the neural crest, although this was lost around birth. Interestingly, while no canonical Wnt signaling was detected in the corneal limbus or basal cells at any developmental stage, it was robust in adult corneal wing and squamous epithelial cells. While canonical Wnt reporter activity was also absent from the postnatal lens, upon lens injury intended to model cataract surgery, it upregulated within 12 h in remnant lens epithelial cells, and co-localized with alpha smooth muscle actin in fibrotic lens epithelial cells from 48 h post-surgery onward. This pattern correlated with downregulation of the inhibitor of canonical Wnt signaling, Dkk3. These data demonstrate that canonical Wnt signaling is dynamic within the developing eye and upregulates in lens epithelial cells in response to lens injury. As canonical Wnt signaling can collaborate with TGFβ to drive fibrosis in other systems, these data offer the first evidence in a lens-injury model that canonical Wnt may synergize with TGFβ signaling to drive fibrotic posterior capsular opacification (PCO).
Collapse
Affiliation(s)
- Yichen Wang
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, United States
| | - Priyha Mahesh
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, United States
| | - Yan Wang
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, United States
| | - Samuel G Novo
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, United States
| | - Mahbubul H Shihan
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, United States
| | | | - Melinda K Duncan
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, United States.
| |
Collapse
|
39
|
Revenkova E, Liu Q, Gusella GL, Iomini C. The Joubert syndrome protein ARL13B binds tubulin to maintain uniform distribution of proteins along the ciliary membrane. J Cell Sci 2018; 131:jcs212324. [PMID: 29592971 PMCID: PMC5992585 DOI: 10.1242/jcs.212324] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 03/23/2018] [Indexed: 01/09/2023] Open
Abstract
Cilia-mediated signal transduction involves precise targeting and localization of selected molecules along the ciliary membrane. However, the molecular mechanism underlying these events is unclear. The Joubert syndrome protein ARL13B is a membrane-associated G-protein that localizes along the cilium and functions in protein transport and signaling. We identify tubulin as a direct interactor of ARL13B and demonstrate that the association occurs via the G-domain and independently from the GTPase activity of ARL13B. The G-domain is necessary for the interaction of ARL13B with the axoneme both in vitro and in vivo We further show that exogenously expressed mutants lacking the tubulin-binding G-domain (ARL13B-ΔGD) or whose GTPase domain is inactivated (ARL13B-T35N) retain ciliary localization, but fail to rescue ciliogenesis defects of null Arl13bhnn mouse embryonic fibroblasts (MEFs). However, while ARL13B-ΔGD and the membrane proteins Smoothened (SMO) and Somatostatin receptor-3 (SSTR3) distribute unevenly along the cilium of Arl13bhnn MEFs, ARL13B-T35N distributes evenly along the cilium and enables the uniform distribution of SMO and SSTR3. Thus, we propose a so far unknown function of ARL13B in anchoring ciliary membrane proteins to the axoneme through the direct interaction of its G-domain with tubulin.
Collapse
Affiliation(s)
- Ekaterina Revenkova
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Qing Liu
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - G Luca Gusella
- Department of Medicine, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Carlo Iomini
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| |
Collapse
|
40
|
Mansini AP, Peixoto E, Thelen KM, Gaspari C, Jin S, Gradilone SA. The cholangiocyte primary cilium in health and disease. Biochim Biophys Acta Mol Basis Dis 2018; 1864:1245-1253. [PMID: 28625917 PMCID: PMC5732091 DOI: 10.1016/j.bbadis.2017.06.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 06/08/2017] [Indexed: 12/14/2022]
Abstract
Cholangiocytes, like most cells, express primary cilia extending from their membranes. These organelles function as antennae which detect stimuli from bile and transmit the information into cells regulating several signaling pathways involved in secretion, proliferation and apoptosis. The ability of primary cilia to detect different signals is provided by ciliary associated proteins which are expressed in its membrane. Defects in the structure and/or function of these organelles lead to cholangiociliopathies that result in cholangiocyte hyperproliferation, altered fluid secretion and absorption. Since primary cilia dysfunction has been observed in several epithelial tumors, including cholangiocarcinoma (CCA), primary cilia have been proposed as tumor suppressor organelles. In addition, the loss of cilia is associated with dysregulation of several molecular pathways resulting in CCA development and progression. Thus, restoration of the primary cilia may be a potential therapeutic approach for several ciliopathies and CCA.
Collapse
Affiliation(s)
| | | | | | - Cesar Gaspari
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Sujeong Jin
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Sergio A Gradilone
- The Hormel Institute, University of Minnesota, Austin, MN, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
41
|
Abstract
Small cell lung cancer (SCLC) is a devastating and aggressive neuroendocrine carcinoma of the lung. It accounts for ~15% of lung cancer mortality and has had no improvement in standard treatment options for nearly 30 years. However, there is now hope for change with new therapies and modalities of therapy. Immunotherapies and checkpoint inhibitors are entering clinical practice, selected targeted therapies show promise, and "smart bomb"-based drug/radioconjugates have led to good response in early clinical trials. Additionally, new research insights into the genetics and tumor heterogeneity of SCLC alongside the availability of new tools such as patient-derived or circulating tumor cell xenografts offer the potential to shine light on this beshadowed cancer.
Collapse
|
42
|
Wheway G, Nazlamova L, Hancock JT. Signaling through the Primary Cilium. Front Cell Dev Biol 2018; 6:8. [PMID: 29473038 PMCID: PMC5809511 DOI: 10.3389/fcell.2018.00008] [Citation(s) in RCA: 323] [Impact Index Per Article: 46.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/23/2018] [Indexed: 12/13/2022] Open
Abstract
The presence of single, non-motile “primary” cilia on the surface of epithelial cells has been well described since the 1960s. However, for decades these organelles were believed to be vestigial, with no remaining function, having lost their motility. It wasn't until 2003, with the discovery that proteins responsible for transport along the primary cilium are essential for hedgehog signaling in mice, that the fundamental importance of primary cilia in signal transduction was realized. Little more than a decade later, it is now clear that the vast majority of signaling pathways in vertebrates function through the primary cilium. This has led to the adoption of the term “the cells's antenna” as a description for the primary cilium. Primary cilia are particularly important during development, playing fundamental roles in embryonic patterning and organogenesis, with a suite of inherited developmental disorders known as the “ciliopathies” resulting from mutations in genes encoding cilia proteins. This review summarizes our current understanding of the role of these fascinating organelles in a wide range of signaling pathways.
Collapse
Affiliation(s)
- Gabrielle Wheway
- Department of Applied Science, Faculty of Health and Applied Sciences, Centre for Research in Biosciences, University of the West of England, Bristol, United Kingdom
| | - Liliya Nazlamova
- Department of Applied Science, Faculty of Health and Applied Sciences, Centre for Research in Biosciences, University of the West of England, Bristol, United Kingdom
| | - John T Hancock
- Department of Applied Science, Faculty of Health and Applied Sciences, Centre for Research in Biosciences, University of the West of England, Bristol, United Kingdom
| |
Collapse
|
43
|
Toomer KA, Fulmer D, Guo L, Drohan A, Peterson N, Swanson P, Brooks B, Mukherjee R, Body S, Lipschutz JH, Wessels A, Norris RA. A role for primary cilia in aortic valve development and disease. Dev Dyn 2017; 246:625-634. [PMID: 28556366 DOI: 10.1002/dvdy.24524] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 05/17/2017] [Accepted: 05/18/2017] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Bicuspid aortic valve (BAV) disease is the most common congenital heart defect, affecting 0.5-1.2% of the population and causing significant morbidity and mortality. Only a few genes have been identified in pedigrees, and no single gene model explains BAV inheritance, thus supporting a complex genetic network of interacting genes. However, patients with rare syndromic diseases that stem from alterations in the structure and function of primary cilia ("ciliopathies") exhibit BAV as a frequent cardiovascular finding, suggesting primary cilia may factor broadly in disease etiology. RESULTS Our data are the first to demonstrate that primary cilia are expressed on aortic valve mesenchymal cells during embryonic development and are lost as these cells differentiate into collagen-secreting fibroblastic-like cells. The function of primary cilia was tested by genetically ablating the critical ciliogenic gene Ift88. Loss of Ift88 resulted in abrogation of primary cilia and increased fibrogenic extracellular matrix (ECM) production. Consequentially, stratification of ECM boundaries normally present in the aortic valve were lost and a highly penetrant BAV phenotype was evident at birth. CONCLUSIONS Our data support cilia as a novel cellular mechanism for restraining ECM production during aortic valve development and broadly implicate these structures in the etiology of BAV disease in humans. Developmental Dynamics 246:625-634, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Katelynn A Toomer
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina
| | - Diana Fulmer
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina
| | - Lilong Guo
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina
| | - Alex Drohan
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina
| | - Neal Peterson
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina
| | - Paige Swanson
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina
| | - Brittany Brooks
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina
| | - Rupak Mukherjee
- Division of Cardiothoracic Surgery, Department of Surgery, Medical University of South Carolina, Charleston, South Carolina.,Department of Medicine, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| | - Simon Body
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Joshua H Lipschutz
- Department of Medicine, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina.,Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Andy Wessels
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina
| | - Russell A Norris
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina.,Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
44
|
Wilcockson SG, Sutcliffe C, Ashe HL. Control of signaling molecule range during developmental patterning. Cell Mol Life Sci 2017; 74:1937-1956. [PMID: 27999899 PMCID: PMC5418326 DOI: 10.1007/s00018-016-2433-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 11/24/2016] [Accepted: 12/05/2016] [Indexed: 12/22/2022]
Abstract
Tissue patterning, through the concerted activity of a small number of signaling pathways, is critical to embryonic development. While patterning can involve signaling between neighbouring cells, in other contexts signals act over greater distances by traversing complex cellular landscapes to instruct the fate of distant cells. In this review, we explore different strategies adopted by cells to modulate signaling molecule range to allow correct patterning. We describe mechanisms for restricting signaling range and highlight how such short-range signaling can be exploited to not only control the fate of adjacent cells, but also to generate graded signaling within a field of cells. Other strategies include modulation of signaling molecule action by tissue architectural properties and the use of cellular membranous structures, such as signaling filopodia and exosomes, to actively deliver signaling ligands to target cells. Signaling filopodia can also be deployed to reach out and collect particular signals, thereby precisely controlling their site of action.
Collapse
Affiliation(s)
- Scott G Wilcockson
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK
| | - Catherine Sutcliffe
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK
| | - Hilary L Ashe
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK.
| |
Collapse
|
45
|
May-Simera H, Nagel-Wolfrum K, Wolfrum U. Cilia - The sensory antennae in the eye. Prog Retin Eye Res 2017; 60:144-180. [PMID: 28504201 DOI: 10.1016/j.preteyeres.2017.05.001] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 05/04/2017] [Accepted: 05/08/2017] [Indexed: 12/21/2022]
Abstract
Cilia are hair-like projections found on almost all cells in the human body. Originally believed to function merely in motility, the function of solitary non-motile (primary) cilia was long overlooked. Recent research has demonstrated that primary cilia function as signalling hubs that sense environmental cues and are pivotal for organ development and function, tissue hoemoestasis, and maintenance of human health. Cilia share a common anatomy and their diverse functional features are achieved by evolutionarily conserved functional modules, organized into sub-compartments. Defects in these functional modules are responsible for a rapidly growing list of human diseases collectively termed ciliopathies. Ocular pathogenesis is common in virtually all classes of syndromic ciliopathies, and disruptions in cilia genes have been found to be causative in a growing number of non-syndromic retinal dystrophies. This review will address what is currently known about cilia contribution to visual function. We will focus on the molecular and cellular functions of ciliary proteins and their role in the photoreceptor sensory cilia and their visual phenotypes. We also highlight other ciliated cell types in tissues of the eye (e.g. lens, RPE and Müller glia cells) discussing their possible contribution to disease progression. Progress in basic research on the cilia function in the eye is paving the way for therapeutic options for retinal ciliopathies. In the final section we describe the latest advancements in gene therapy, read-through of non-sense mutations and stem cell therapy, all being adopted to treat cilia dysfunction in the retina.
Collapse
Affiliation(s)
- Helen May-Simera
- Institute of Molecular Physiology, Cilia Biology, Johannes Gutenberg-Universität Mainz, 55099 Mainz, Germany
| | - Kerstin Nagel-Wolfrum
- Institute of Molecular Physiology, Molecular Cell Biology, Johannes Gutenberg-Universität Mainz, 55099 Mainz, Germany
| | - Uwe Wolfrum
- Institute of Molecular Physiology, Molecular Cell Biology, Johannes Gutenberg-Universität Mainz, 55099 Mainz, Germany.
| |
Collapse
|
46
|
Grisanti L, Revenkova E, Gordon RE, Iomini C. Primary cilia maintain corneal epithelial homeostasis by regulation of the Notch signaling pathway. J Cell Sci 2016. [DOI: 10.1242/jcs.193789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|